1
|
Seo ES, Lee JW, Cho HW, Ju HY, Cho YS, Lee S, Moon SH, Yoo KH, Lim DH, Sung KW. Response-adapted consolidation therapy strategy for patients with metastatic high-risk neuroblastoma: Results of the SMC NB-2014 study. Pediatr Blood Cancer 2024; 71:e31173. [PMID: 38965702 DOI: 10.1002/pbc.31173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/28/2024] [Accepted: 06/16/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Tandem high-dose chemotherapy and autologous stem cell transplantation (HDCT/auto-SCT) and incorporation of 131I-metaiodobenzylguanidine (131I-MIBG) treatment have shown positive outcomes in high-risk neuroblastoma. However, more optimized treatment strategies are still needed. PROCEDURE The NB-2014 study was a nonrandomized, prospective trial that examined survival outcomes in metastatic high-risk neuroblastoma patients using response-adapted consolidation therapy. We used post-induction residual 123I-MIBG status at metastatic sites as a treatment response marker. Patients achieving complete resolution of MIBG uptake at metastatic sites underwent a reduced first HDCT/auto-SCT with a 20% dose reduction in HDCT. After the first HDCT/auto-SCT, patients with remaining MIBG uptake received dose-escalated (18 mCi/kg) 131I-MIBG treatment. In contrast, those with complete resolution of MIBG at metastatic sites received a standard dose (12 mCi/kg) of 131I-MIBG. We compared survival and toxicity outcomes with a historical control group from the NB-2009. RESULTS Of 65 patients treated, 63% achieved complete resolution of MIBG uptake at metastatic sites following induction chemotherapy, while 29% of patients still had MIBG uptake at metastatic sites after the first HDCT/auto-SCT. The 3-year event-free survival (EFS) and overall survival (OS) rates were 68.2% ± 6.0% and 86.5% ± 4.5%, respectively. Compared to NB-2009, EFS was similar (p = .855); however, NB-2014 had a higher OS (p = .031), a lower cumulative incidence of treatment-related mortality (p = .036), and fewer acute and late toxicities. CONCLUSIONS Our results suggest that response-adaptive consolidation therapy based on chemotherapy response at metastatic sites facilitates better treatment tailoring, and appears promising for patients with metastatic high-risk neuroblastoma.
Collapse
Affiliation(s)
- Eun Seop Seo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hee Young Ju
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Seok Cho
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sanghoon Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Wang H, Chen X, He L. A narrative review of radiomics and deep learning advances in neuroblastoma: updates and challenges. Pediatr Radiol 2023; 53:2742-2755. [PMID: 37945937 DOI: 10.1007/s00247-023-05792-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023]
Abstract
Neuroblastoma is an extremely heterogeneous tumor that commonly occurs in children. The diagnosis and treatment of this tumor pose considerable challenges due to its varied clinical presentations and intricate genetic aberrations. Presently, various imaging modalities, including computed tomography, magnetic resonance imaging, and positron emission tomography, are utilized to assess neuroblastoma. Nevertheless, these conventional imaging modalities have limitations in providing quantitative information for accurate diagnosis and prognosis. Radiomics, an emerging technique, can extract intricate medical imaging information that is imperceptible to the human eye and transform it into quantitative data. In conjunction with deep learning algorithms, radiomics holds great promise in complementing existing imaging modalities. The aim of this review is to showcase the potential of radiomics and deep learning advancements to enhance the diagnostic capabilities of current imaging modalities for neuroblastoma.
Collapse
Affiliation(s)
- Haoru Wang
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan Road 2, Yuzhong District, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xin Chen
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan Road 2, Yuzhong District, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Ling He
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan Road 2, Yuzhong District, Chongqing, 400014, China.
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.
- Chongqing Key Laboratory of Pediatrics, Chongqing, China.
| |
Collapse
|
3
|
Wang H, Qin J, Chen X, Zhang T, Zhang L, Ding H, Pan Z, He L. Contrast-enhanced computed tomography radiomics in predicting primary site response to neoadjuvant chemotherapy in high-risk neuroblastoma. Abdom Radiol (NY) 2023; 48:976-986. [PMID: 36571609 DOI: 10.1007/s00261-022-03774-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/27/2022]
Abstract
PURPOSE To explore the clinical value of contrast-enhanced computed tomography (CECT) radiomics in predicting primary site response to neoadjuvant chemotherapy in high-risk neuroblastoma. MATERIALS AND METHODS Seventy patients were retrospectively included and separated into very good partial response (VGPR) group and non-VGPR group according to the changes in primary tumor volume. The clinical features with statistical difference between the two groups were used to construct the clinical models using a logistic regression (LR) algorithm. The radiomics models based on different radiomics features selected by Kruskal-Wallis (KW) test and recursive feature elimination (RFE) were established using support vector machine (SVM) and LR algorithms. The radiomics score (Radscore) and clinical features were integrated into the combined models. Leave-one-out cross-validation (LOOCV) was used to validate the predictive performance of models in the entire dataset. RESULTS The optimal clinical model achieved an area under the curve (AUC) of 0.767 [95% confidence interval (CI): 0.638, 0.896] and an accuracy of 0.771 after LOOCV. The AUCs of the best KW + SVM, KW + LR, RFE + SVM, and RFE + LR radiomics models were 0.816, 0.826, 0.853, and 0.850, respectively, and the corresponding AUCs after LOOCV were 0.780, 0.785, 0.755, and 0.772, respectively. The AUC and accuracy after LOOCV of the optimal combined model was 0.804 (95% CI: 0.694, 0.915) and 0.814, respectively. The Delong test showed a statistical difference in predictive performance between the optimal clinical and combined models after LOOCV (Z = 2.003, P = 0.045). The decision curve analysis showed that the combined model performs better than the clinical model. CONCLUSION The CECT radiomics models have a favorable predictive performance in predicting VGPR of high-risk neuroblastoma to neoadjuvant chemotherapy. When integrating radiomics features and clinical features, the predictive performance of the combined models can be further improved.
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 Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing, 400014, China
| | - Jinjie Qin
- 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 Pediatrics, 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 Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing, 400014, China
| | - Ting Zhang
- 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 Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing, 400014, China
| | - Li Zhang
- 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 Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing, 400014, China
| | - Hao 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 Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing, 400014, China
| | - Zhengxia Pan
- Department of Cardiothoracic Surgery, 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 Pediatrics, 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 Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing, 400014, China.
| |
Collapse
|
4
|
Mastrangelo S, Attinà G, Zagaria L, Romano A, Ruggiero A. Induction Regimen in High-Risk Neuroblastoma: A Pilot Study of Highly Effective Continuous Exposure of Tumor Cells to Radio-Chemotherapy Sequence for 1 Month. The Critical Role of Iodine-131-Metaiodobenzylguanidine. Cancers (Basel) 2022; 14:5170. [PMID: 36291955 PMCID: PMC9599979 DOI: 10.3390/cancers14205170] [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: 09/07/2022] [Revised: 10/16/2022] [Accepted: 10/20/2022] [Indexed: 11/29/2022] Open
Abstract
The prognosis of high-risk neuroblastoma (NB) continues to be poor. The early development of resistance often leads to disease recurrence. In the present study, an innovative induction regimen, including an intensive initial radio-chemotherapy sequence based on the use of iodine-131-metaiodobenzylguanidine (131-I-MIBG), was investigated. The duration of the regimen lasted only one month. Fifteen newly diagnosed patients aged >18 months with high-risk NB were treated with cisplatin, etoposide, cyclophosphamide, and vincristine, followed on day 10 by 131-I-MIBG (dose: 12−18.3 mCi/kg). Cisplatin and vincristine were administered on day 20 and 21 followed by the re-administration of vincristine, cyclophosphamide, and doxorubicin on day 29 and 30. Non-hematologic toxicity was not observed. Moderate hematologic toxicity was present probably attributable to chemotherapy. The evaluation of response was performed approximately 50 days after the initiation of treatment, yielding four complete responses, eight very good partial responses, one partial response, and two non-responses. Importantly, a complete metastatic response was achieved in 87% of patients. The present pilot study, which includes 131-I-MIBG, allows for a highly effective continuous exposure of tumor cells to both chemotherapy and radiotherapy. Furthermore, early high-dose chemotherapy followed by stem cell rescue may achieve high levels of tumor cell clearance and improve the prognosis of high-risk NB.
Collapse
Affiliation(s)
- Stefano Mastrangelo
- UOSD di Oncologia Pediatrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Largo F.sco Vito 1, 00168 Rome, Italy
| | - Giorgio Attinà
- UOSD di Oncologia Pediatrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Luca Zagaria
- UOC di Medicina Nucleare, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Alberto Romano
- UOSD di Oncologia Pediatrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Antonio Ruggiero
- UOSD di Oncologia Pediatrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Largo F.sco Vito 1, 00168 Rome, Italy
| |
Collapse
|
5
|
Gowin E, Jończyk-Potoczna K, Sosnowska-Sienkiewicz P, Belen Larque A, Kurzawa P, Januszkiewicz-Lewandowska D. Semi-Automatic Volumetric and Standard Three-Dimensional Measurements for Primary Tumor Evaluation and Response to Treatment Assessment in Pediatric Rhabdomyosarcoma. J Pers Med 2021; 11:jpm11080717. [PMID: 34442361 PMCID: PMC8399942 DOI: 10.3390/jpm11080717] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/11/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
Current prognostic classification of rhabdomyosarcoma in children requires precise measurements of the tumor. The purpose of the study was to compare the standard three-dimensional (3D) measurements with semi-automatic tumor volume measurement method concerning assessment of the primary tumor size and the degree of response to treatment for rhabdomyosarcoma in children. Magnetic Resonance Imaging data on 31 children with treated rhabdomyosarcoma based on the Cooperative Weichteilsarkom Studiengruppe (CWS) guidance was evaluated. Tumor sizes were measured by two methods: 3D standard measurements and semi-automatic tumor volume measurement (VOI) at diagnosis, and after 9 and 17/18 weeks of the induction chemotherapy. Response to treatment and prediction values were assessed. The tumor volume medians calculated using VOI were significantly higher in comparison with those calculated using the 3D method both during the diagnosis as well as after 9 weeks of the chemotherapy and during the 17-18th week of the treatment. The volume measurements based on the generalized estimating equations on the VOI method were significantly better than the 3D method (p = 0.037). The volumetric measurements alone can hardly be considered an unequivocal marker used to make decisions on modification of the therapy in patients with rhabdomyosarcoma.
Collapse
Affiliation(s)
- Ewelina Gowin
- Department of Health Promotion, Poznan University of Medical Sciences, Święcickiego 6 Street, 60-781 Poznan, Poland;
| | - Katarzyna Jończyk-Potoczna
- Department of Pediatric Radiology, Poznan University of Medical Sciences, Szpitalna Street 27/33, 60-572 Poznan, Poland;
| | - Patrycja Sosnowska-Sienkiewicz
- Department of Pediatric Surgery, Traumatology and Urology, Poznan University of Medical Sciences, Szpitalna Street 27/33, 60-572 Poznan, Poland
- Correspondence: or ; Tel.: +48-61-849-15-78; Fax: +48-61-849-52-28
| | - Anna Belen Larque
- Department of Pathology, Hospital Clínic, Villarroel, 170, 08036 Barcelona, Spain;
| | - Paweł Kurzawa
- Department of Pathology, Hospital of Lord’s Transfiguration, University of Medical Sciences, Długa Street 1/2, 61-848 Poznan, Poland;
| | - Danuta Januszkiewicz-Lewandowska
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Szpitalna 27/33, 60-572 Poznan, Poland;
- Department of Medical Diagnostics, Dobra 38a, 60-595 Poznan, Poland
| |
Collapse
|
6
|
Pediatric Rhabdomyosarcomas: Three-Dimensional Radiological Assessments after Induction Chemotherapy Predict Survival Better than One-Dimensional and Two-Dimensional Measurements. Cancers (Basel) 2020; 12:cancers12123808. [PMID: 33348683 PMCID: PMC7766999 DOI: 10.3390/cancers12123808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/12/2020] [Accepted: 12/14/2020] [Indexed: 11/16/2022] Open
Abstract
Radiological response to neoadjuvant chemotherapy is currently used to assess the efficacy of treatment in pediatric patients with rhabdomyosarcoma (RMS), but the association between early tumor response on imaging and survival is still controversial. The aim of this study was to investigate the prognostic value of assessing radiological response after induction therapy in pediatric RMS, comparing four different methods. This retrospective, two-center study was conducted on 66 non-metastatic RMS patients. Two radiologists measured tumor size on pre- and post-treatment magnetic resonance (MR) or computed tomography (CT) images using four methods: considering maximal diameter with the 1D-RECIST (Response Evaluation Criteria in Solid Tumors); multiplying the two maximal diameters with the 2D-WHO (World Health Organization); multiplying the three maximal diameters with the 3D-EpSSG (European pediatric Soft tissue sarcoma Study Group); obtaining a software-assisted volume assessment with the 3D-Osirix. Each patient was classified as a responder or non-responder based on the proposed thresholds for each method. Tumor response was compared with survival using Kaplan-Meier plots, the log-rank test, and Cox's regression. Agreement between methods and observers (weighted-κ) was also calculated. The 5-year event-free survival (5yr-EFS) calculated with the Kaplan-Meier plots was significantly longer for responders than for non-responders with all the methods, but the 3D assessments differentiated between the two groups better than the 1D-RECIST or 2D-WHO (p1D-RECIST = 0.018, p2D-WHO = 0.007, p3D-EpSSG and p3D-Osirix < 0.0001). Comparing the 5yr-EFS of responders and non-responders also produced adjusted hazard ratios of 3.57 (p = 0.0158) for the 1D-RECIST, 5.05 for the 2D-WHO (p = 0.0042), 14.40 for the 3D-EpSSG (p < 0.0001) and 11.60 for the 3D-Osirix (p < 0.0001), indicating that the volumetric measurements were significantly more strongly associated with EFS. Inter-method agreement was excellent between the 3D-EpSSG and the 3D-Osirix (κ = 0.98), and moderate for the other comparisons (0.5 < κ < 0.8). The 1D-RECIST and the 2D-WHO tended to underestimate response to treatment. Inter-observer agreement was excellent with all methods (κ > 0.8) except for the 2D-WHO (κ = 0.7). In conclusion, early tumor response was confirmed as a significant prognostic factor in RMS, and the 3D-EpSSG and 3D-Osirix methods predicted response to treatment better than the 1D-RECIST or 2D-WHO measurements.
Collapse
|
7
|
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: 0.5] [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
|
8
|
Chui C. Effects of preoperative chemotherapy on neuroblastoma with MYCN amplification: a surgeon’s perspective. WORLD JOURNAL OF PEDIATRIC SURGERY 2020; 3:e000129. [DOI: 10.1136/wjps-2020-000129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 11/04/2022] Open
Abstract
BackgroundPreoperative chemotherapy plays an important role in the surgical management of unresectable neuroblastoma. Its response to chemotherapy has been variable due to the tumor’s heterogeneity. We aimed to evaluate the effects of preoperative chemotherapy on MYCN-amplified (MYCNA) neuroblastoma that would impact on surgical resection.MethodsPatients with MYCNA neuroblastoma who received preoperative chemotherapy followed by surgical resection performed at our center were included. The tools of response evaluated included tumor volume reduction (TVR), reduction in image-defined risk factors (IDRFs), percentage tumor necrosis (Nec), and surgical complications.ResultsAmong 62 patients evaluated, mean age was 3.0 (range, 0.9–11.8) years, and primary tumors were distributed in the abdomen (n=59), pelvis (n=2), and thorax (n=1). The patients were in stages L2 (n=14) and M (n=48). Surgery was performed after median of 4 (range, 2–10) cycles of chemotherapy. On completion of preoperative chemotherapy, 41 (66.1%) patients had TVR >65%, 24 (42.9%) responded with reduced IDRFs, 47 (75.8%) tumors had Nec >50%, and 27 patients suffered 31 surgical complications. Majority (83.9%) continued to have IDRFs at surgery. IDRFs commonly encountered were encasement of renal pedicles (n=50), superior mesenteric artery (n=46), celiac axis (n=45), and aorta/vena cava (n=44), and most remained refractory to resolution. Patients with TVR >65% were associated with Nec >50% (87.5% vs 54.5%, p=0.004) and reduced IDRFs (46.3% vs 19%, p=0.035), but not with the incidence of surgical complications.ConclusionsMajority of MYCNA neuroblastomas were highly chemosensitive as they experienced high TVR, reduced IDRFs, and high Nec, and hence created favorable conditions for surgical resection. Poor responders and persistent IDRFs that were commonly refractory to preoperative chemotherapy remained a surgical challenge.
Collapse
|
9
|
Lim H, Son MH, Hyun JK, Cho HW, Ju HY, Lee JW, Yoo KH, Sung KW, Koo HH. Clinical Significance of Segmental Chromosomal Aberrations in Patients with Neuroblastoma: First Report in Korean Population. J Korean Med Sci 2020; 35:e82. [PMID: 32281311 PMCID: PMC7152533 DOI: 10.3346/jkms.2020.35.e82] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/05/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND This study aimed to investigate the incidence and clinical significance of segmental chromosomal aberrations (SCAs) in Korean patients with neuroblastoma. METHODS Patients diagnosed with neuroblastoma from 2012 to 2018 were included for retrospective review. Fluorescence in situ hybridization (FISH) was used to analyze four SCAs (MYCN amplification, 1p deletion, 11q deletion, and 17q gain). Clinical characteristics at diagnosis, early tumor response (reduction in primary tumor volume and neuron-specific enolase level after the first three cycles of chemotherapy), and survival rates were compared according to SCAs. RESULTS Among 173 patients with FISH results, 92 (53.2%) had at least one of the four SCAs, while 25 (14.5%) had two co-aberrations, and eight (4.6%) had three co-aberrations. SCAs detected in our study were MYCN amplification (n = 17, 9.8%), 1p deletion (n = 26, 15.2%), 11q deletion (n = 44, 25.6%), and 17q gain (n = 46, 27.1%). Patients with MYCN amplification showed a better early response but a worse survival than those without (5-year overall survival: 46.2% ± 13.1% vs. 88.6% ± 3.4%). Furthermore, 1p deletion was associated with a better early response but a worse survival; however, it was not an independent factor for survival. We could not find any prognostic significance associated with 11q deletion or 17q gain. CONCLUSION This is the first study investigating SCAs in Korean neuroblastoma patients. Prognostic significance of SCAs other than MYCN amplification was different from those reported in western countries. Further study with a larger cohort and longer follow-up is needed to confirm our findings.
Collapse
Affiliation(s)
- Hana Lim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Meong Hi Son
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Ju Kyung Hyun
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Young Ju
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
10
|
Abstract
Although intensive multimodal treatment has improved outcomes for patients with high-risk neuroblastoma, the specific role of primary tumor resection remains controversial. Many studies have been designed to determine whether the extent of surgical resection impacts survival; however, these reports have demonstrated conflicting results. There is also ongoing debate regarding the timing of primary tumor resection, with subtle differences in the approach between the large pediatric oncology cooperative consortia. Most of the published literature to date has been approached from a surgical viewpoint. Although most evidence supports surgery as part of the local control approach for high-risk neuroblastoma, recommendations for timing and extent of surgical resection are not consistent. This review summarizes our current understanding from the perspectives of both the pediatric oncologist and pediatric surgeons and discusses how the objectives of neuroblastoma primary surgical resection are different from that of other malignancies. Furthermore, this commentary will address how retrospective surgical outcome data may be interpreted in the setting of modern era high-risk neuroblastoma treatment.
Collapse
|
11
|
Seo E, Kim JS, Ma YE, Cho HW, Ju HY, Lee SH, Lee JW, Yoo KH, Sung KW, Koo HH. Differential Clinical Significance of Neurotrophin-3 Expression according to MYCN Amplification and TrkC Expression in Neuroblastoma. J Korean Med Sci 2019; 34:e254. [PMID: 31602824 PMCID: PMC6786962 DOI: 10.3346/jkms.2019.34.e254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/23/2019] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Neurotrophin-3 (NT-3), a member of the NT family, has only been considered an ancillary compound that provides anti-apoptotic benefits by inactivating tropomyosin receptor kinase C (TrkC)-induced apoptotic signals. However, little is known about the clinical relevance of NT-3 expression itself in neuroblastoma. The purpose of this study was to assess NT-3 expression in patients with neuroblastoma and its relevance to clinicopathologic findings and treatment outcomes. METHODS In this study, expression of NT-3 and TrkC was analyzed using immunohistochemistry in 240 patients with newly diagnosed neuroblastoma. RESULTS The results of the study revealed that NT-3 expression was associated with older age at diagnosis, localized tumors, and more differentiated tumors but was not associated with early treatment response (degree of residual tumor volume after three cycles of chemotherapy) and progression-free survival (PFS). However, when analysis was confined to patients with MYCN amplified tumors, NT-3 expression was associated with better early treatment response with borderline significance (P = 0.092) and higher PFS (86.9% vs. 58.2%; P = 0.044). In multivariate analysis in patients with MYCN amplified tumors, NT-3 was independent prognostic factor (hazard ratio, 0.246; 95% confidence interval, 0.061-0.997; P = 0.050). In another subgroup analysis, the early treatment response was better if NT-3 was expressed in patients without TrkC expression (P = 0.053) while it was poorer in patients with TrkC expression (P = 0.023). CONCLUSION This study suggests that NT-3 expression in neuroblastoma has its own clinical significance independent of TrkC expression, and its prognostic significance differs depending on the status of MYCN amplification and/or TrkC expression.
Collapse
Affiliation(s)
- Eunseop Seo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung Sun Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Sungkyunkwan University, Samsung Advanced Institute for Health Sciences & Technology, Seoul, Korea
| | - Young Eun Ma
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Young Ju
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo Hyun Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
12
|
Furman WL, Federico SM, McCarville MB, Shulkin BL, Davidoff AM, Krasin MJ, Sahr N, Sykes A, Wu J, Brennan RC, Bishop MW, Helmig S, Stewart E, Navid F, Triplett B, Santana VM, Bahrami A, Anthony G, Yu AL, Hank J, Gillies SD, Sondel PM, Leung WH, Pappo AS. A Phase II Trial of Hu14.18K322A in Combination with Induction Chemotherapy in Children with Newly Diagnosed High-Risk Neuroblastoma. Clin Cancer Res 2019; 25:6320-6328. [PMID: 31601569 DOI: 10.1158/1078-0432.ccr-19-1452] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/12/2019] [Accepted: 08/13/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE We sought to evaluate whether combining a humanized antidisialoganglioside mAb (hu14.18K322A) with induction chemotherapy improves early responses and outcomes in children with newly diagnosed high-risk neuroblastoma. PATIENTS AND METHODS We conducted a prospective nonrandomized, single-arm, two-stage, phase II clinical trial. Six courses of induction chemotherapy were coadministered with hu14.18K322A and followed with granulocyte-macrophage colony-stimulating factor (GM-CSF) and low-dose IL2. Consolidation was performed with a busulfan/melphalan preparative regimen. An additional course of hu14.18K322A was administered with parent-derived natural killer cells, when available, during consolidation. Hu14.18K322A, GM-CSF, IL2, and isotretinoin were then administered. Secondary outcomes included reduced tumor volume and semiquantitative 123I-metaiodobenzylguanidine scoring [i.e., Curie scores (CS)] at the end of induction. RESULTS Forty-two patients received hu14.18K322A and induction chemotherapy. This regimen was well tolerated, with continuous-infusion narcotics adjusted to patient tolerance. Partial responses (PR) or better after the first two chemoimmunotherapy courses occurred in 32 patients [76.2%; 95% confidence interval (CI), 60.6-88.0]. This was accompanied by primary tumor volume reductions (median, -76%; range, -100% to 5%). Of 35 patients with stage IV disease who completed induction, 31 had end-of-induction CSs of 2 or less. No patients experienced progression during induction. Two-year event-free survival (EFS) was 85.7% (95% CI, 70.9-93.3). CONCLUSIONS Adding hu14.18K322A to induction chemotherapy produced early PR or better in most patients, reduced tumor volumes, improved CSs at the end of induction, and yielded an encouraging 2-year EFS. These results, if validated in a larger study, may change the standard of care for children with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Wayne L Furman
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee.
| | - Sara M Federico
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Barry L Shulkin
- Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Matthew J Krasin
- Radiation Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Natasha Sahr
- Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - April Sykes
- Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jianrong Wu
- Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Rachel C Brennan
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Sara Helmig
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Elizabeth Stewart
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Fariba Navid
- Division of Hematology, Oncology and Bone Marrow Transplant, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Brandon Triplett
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Victor M Santana
- Clinical Trials Administration, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Armita Bahrami
- Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Gwendolyn Anthony
- Cancer Center Administration, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Alice L Yu
- University of California, San Diego and Moores Cancer Center and Genomics Research Center, Academia Sinica, Taiwan
| | - Jacquelyn Hank
- Departments of Pediatrics and Human Oncology, University of Wisconsin, Madison, Wisconsin
| | | | - Paul M Sondel
- Departments of Pediatrics and Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Wing H Leung
- Department of Hematology Oncology, KK Women's and Children's Hospital, Duke-NUS, Singapore
| | - Alberto S Pappo
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
13
|
Tang X, He Q, Qu H, Sun G, Liu J, Gao L, Shi J, Ye J, Liang Y. Post-therapy pathologic tumor volume predicts survival in gastric cancer patients who underwent neoadjuvant chemotherapy and gastrectomy. BMC Cancer 2019; 19:797. [PMID: 31409315 PMCID: PMC6693132 DOI: 10.1186/s12885-019-6012-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/02/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To demonstrate that post-therapy pathological tumor volume (ypTV) should be considered as an independent prognostic factor in advanced gastric cancer (GC) patients who underwent neoadjuvant chemotherapy (NAC) and gastrectomy. METHODS A total of 253 GC patients who received gastrectomy between January 2010 and December 2016 in our hospital were enrolled in this study. Clinicopathologic factors were evaluated using univariable and multivariable analysis. ypTV was calculated using π* (tumor diameter/2)2 *tumor invasion depth (cm3). RESULTS Cut-point survival analysis demonstrated that the appropriate cut-offs for ypTV were 3, 6, 10, and 19 (cm3). Patients with tumor volumes of 0-3.0, 3.1-6.0, 6.1-10.0, 10.1-19.0, ≥19.1 cm3 were defined as ypTV1, 2, 3, 4a and 4b. Using multivariable analysis, the tumor volume (ypTV stage, P < 0.05), ypN stage (P < 0.05), response to NAC (P < 0.05), vascular invasion (P < 0.05) and ypTvNM staging (P < 0.05) were independent prognostic factors. Kaplan-Meier analysis demonstrated that the 8th AJCC/UICC ypTNM staging was not a significant predictor for survival (P > 0.05); however, our newly defined ypTvNM staging was a significant predictor for survival (P < 0.05). CONCLUSIONS ypTV should be considered as an independent prognostic factor for GC patients after NAC. ypTvNM staging should be recommended to improve the accuracy of prognostic prediction for GC patients who received NAC plus gastrectomy.
Collapse
Affiliation(s)
- Xiaolong Tang
- Department of General Surgery, Qilu Hospital of Shandong University, No.107, West of Wenhua Street, Lixia District, Jinan, 250012, China
| | - Qingsi He
- Department of General Surgery, Qilu Hospital of Shandong University, No.107, West of Wenhua Street, Lixia District, Jinan, 250012, China
| | - Hui Qu
- Department of General Surgery, Qilu Hospital of Shandong University, No.107, West of Wenhua Street, Lixia District, Jinan, 250012, China.
| | - Guorui Sun
- Department of General Surgery, Qilu Hospital of Shandong University, No.107, West of Wenhua Street, Lixia District, Jinan, 250012, China
| | - Jia Liu
- Department of Health Management Center, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Lei Gao
- Qilu Medical College of Shandong University, Jinan, 250011, Shandong, China
| | - Jingbo Shi
- Qilu Medical College of Shandong University, Jinan, 250011, Shandong, China
| | - Jianhong Ye
- Qilu Medical College of Shandong University, Jinan, 250011, Shandong, China
| | - Yahang Liang
- Qilu Medical College of Shandong University, Jinan, 250011, Shandong, China
| |
Collapse
|
14
|
Fawzy M, Hamoda A, Elhemaly A, Elkinaai N, Soliman S, Reda H, Elmenawi S, Moussa E. Does Salvage Chemotherapy Regimen Intensity Embark on Clearance of Bone Marrow Neuroblastoma? Asian Pac J Cancer Prev 2019; 20:1519-1524. [PMID: 31128057 PMCID: PMC6857875 DOI: 10.31557/apjcp.2019.20.5.1519] [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] [Indexed: 11/25/2022] Open
Abstract
Introduction:: Neuroblastoma (NBL) is the most common extracranial solid tumor in children. It accounts for 15% of the deaths from cancer in the pediatric age group. Approximately half of the newly diagnosed children are at “high risk” (HR) of treatment failure. This study aim was to evaluate the impact of salvage chemotherapy ICE (ifosfamide, carboplatin, and etoposide) versus TC (topotecan/cyclophosphamide) when administered to NBL HR patients having residual bone marrow disease after primary tumor control on the first line treatment regimen. Materials and Methods: The present retrospective study included two groups of eligible stage 4 NBL patients with persistent bone marrow disease. Group (1), 29 patients, received ICE whereas less intensive TC was administered to Group (2), 32 patients. Data analysis included epidemiological variables, pathology subtype, MYCN gene status, primary tumor response and their correlation with bone marrow disease clearance on each regimen. Results: A higher tendency of complete bone marrow clearance was reported in patients who received ICE compared to TC; 41.4% versus 25.0%, respectively. However, the difference was not statistically significant (p= 0.174). Conclusion: TC regimen appears to be a good alternative to ICE as salvage treatment in an attempt to clear NBL bone marrow residual, with the privilege of being less toxic and can be given on outpatient basis. Further randomized trials of larger study sample size with survival impact analysis are warranted.
Collapse
Affiliation(s)
- Mohamed Fawzy
- Department of Pediatric Oncology, Children Cancer Hospital Egypt-57357 and National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Asmaa Hamoda
- Department of Pediatric Oncology, Children Cancer Hospital Egypt-57357 and National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Ahmed Elhemaly
- Department of Pediatric Oncology, Children Cancer Hospital Egypt-57357 and National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Naglaa Elkinaai
- Department of Pathology, Children Cancer Hospital Egypt-57357 and National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Sonya Soliman
- Department of Clinical Pathology, Children Cancer Hospital Egypt-57357 and National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Hala Reda
- Department of Clinical Pathology, Children Cancer Hospital Egypt-57357 and National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Salma Elmenawi
- Clinical Research Unit, Research Department, Children Cancer Hospita Egypt-57357 and National Cancer Institute (NCI), Cairo University, Cairo, Egypt.
| | - Emad Moussa
- Department of Pediatric Oncology, Children Cancer Hospital Egypt-57357 and National Cancer Institute (NCI), Cairo University, Cairo, Egypt.,Menoufeya Faculty of Medicine, Cairo, Egypt
| |
Collapse
|
15
|
Peschmann AL, Beer M, Ammann B, Dreyhaupt J, Kneer K, Beer AJ, Beltinger C, Steinbach D, Cario H, Neubauer H. Quantitative DWI predicts event-free survival in children with neuroblastic tumours: preliminary findings from a retrospective cohort study. Eur Radiol Exp 2019; 3:6. [PMID: 30701332 PMCID: PMC6353978 DOI: 10.1186/s41747-019-0087-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/04/2019] [Indexed: 01/18/2023] Open
Abstract
Background Quantitative diffusion-weighted imaging (DWI) probes into tissue microstructure in solid tumours. In this retrospective ethically approved study, we investigated DWI as a potential non-invasive predictor of tumour dignity and prognosis in paediatric patients with neuroblastic tumours. Methods Nineteen consecutive patients with neuroblastoma (NB, n = 15), ganglioneuroblastoma (GNB, n = 1) and ganglioneuroma (GN, n = 3) underwent 3-T magnetic resonance imaging at first diagnosis and after 3-month follow-up, following a protocol including DWI (b = 50 and 800 s/mm2) in addition to standard sequences. All DWI scans were analysed for tumour volume assessment and apparent diffusion coefficient (ADC) calculation. Correlation with tumour pathology and risk factors (bone-marrow metastases, MYCN-amplification and 1p-deletion), therapeutic regime (observation versus chemotherapy) and clinical follow-up was evaluated. Results At baseline, mean ADC in NB was lower than in GNB/GN (0.76 vs. 1.47 × 10−3 mm2/s, p = 0.003). An ADC cutoff ≤ 1.05 identified malignant disease with 100.0% sensitivity (95% confidence interval [CI] 29.2–100.0%) and 93.8% specificity (95% CI 69.8–99.8%). Initial ADC was < 0.80 in all NB patients with eventual tumour relapse. During follow-up, tumour ADC values increased in the observation group (NB/GN) without relapse (p = 0.043). In eventually relapsing tumours, ADC values at follow-up tended to decrease further despite reduction in tumour volume. Conclusions ADC values at first presentation differed significantly between malignant and benign neuroblastic tumours. Low baseline ADC was predictive of tumour progression and relapse in NB patients. With therapy, increasing ADC values appeared to predict relapse-free survival, while a decreasing ADC during therapy was an indicator of poor prognosis.
Collapse
Affiliation(s)
- Anna-Lydia Peschmann
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Albert-Schweitzer-Allee 23, 89081, Ulm, Germany
| | - Meinrad Beer
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Albert-Schweitzer-Allee 23, 89081, Ulm, Germany
| | - Bettina Ammann
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Albert-Schweitzer-Allee 23, 89081, Ulm, Germany
| | - Jens Dreyhaupt
- Department of Biometrics, University Hospital Ulm, 89081, Ulm, Germany
| | - Katharina Kneer
- Department of Nuclear Medicine, University Hospital Ulm, 89081, Ulm, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, University Hospital Ulm, 89081, Ulm, Germany
| | - Christian Beltinger
- Department of Paediatrics and Adolescent Medicine, University Hospital Ulm, 89081, Ulm, Germany
| | - Daniel Steinbach
- Department of Paediatrics and Adolescent Medicine, University Hospital Ulm, 89081, Ulm, Germany
| | - Holger Cario
- Department of Paediatrics and Adolescent Medicine, University Hospital Ulm, 89081, Ulm, Germany
| | - Henning Neubauer
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Albert-Schweitzer-Allee 23, 89081, Ulm, Germany.
| |
Collapse
|
16
|
Lee JW, Son MH, Cho HW, Ma YE, Yoo KH, Sung KW, Koo HH. Clinical significance of MYCN amplification in patients with high-risk neuroblastoma. Pediatr Blood Cancer 2018; 65:e27257. [PMID: 29797634 DOI: 10.1002/pbc.27257] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 11/11/2022]
Abstract
BACKGROUND This study investigated the clinical significance of MYCN amplification within high-risk neuroblastoma (NB). METHODS Medical records of 135 patients who were diagnosed with high-risk NB from 2004 to 2016 were reviewed. RESULTS Fifty-one (38%) patients had MYCN amplified tumors, and the remaining 84 (62%) had nonamplified tumors. MYCN amplification was associated with abdominal primary site, less differentiated pathology, higher levels of lactate dehydrogenase and neuron-specific enolase (NSE), lower vanillylmandelic acid level, and larger primary tumor volume at diagnosis. MYCN amplification was associated with a better early response (faster reduction of primary tumor volume and NSE level). The proportion of patients in complete response or very good partial response after induction treatment was relatively higher in MYCN amplified tumors than in nonamplified tumors; however, all progressions during induction treatment occurred only in MYCN amplified tumors (P = 0.007). The time to progression was shorter (median 1.5 years vs. 1.9 years, P = 0.037) and survival after relapse/progression was worse in MYCN amplified tumors (3 year overall survival: 7.7 ± 7.4% vs. 20.5 ± 8.8%, P = 0.046). There was no difference in event-free survival and overall survival between MYCN amplified and nonamplified tumors. CONCLUSION MYCN amplification was associated with more aggressive features at diagnosis and a better early response, but a higher progression rate during induction treatment and lower chance of survival after relapse/progression. There was no difference in survival rates according to MYCN amplification in patients with high-risk NB.
Collapse
Affiliation(s)
- Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Meong Hi Son
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Young Eun Ma
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
17
|
Vaarwerk B, van der Lee JH, Breunis WB, Orbach D, Chisholm JC, Cozic N, Jenney M, van Rijn RR, McHugh K, Gallego S, Glosli H, Devalck C, Gaze MN, Kelsey A, Bergeron C, Stevens MCG, Oberlin O, Minard-Colin V, Merks JHM. Prognostic relevance of early radiologic response to induction chemotherapy in pediatric rhabdomyosarcoma: A report from the International Society of Pediatric Oncology Malignant Mesenchymal Tumor 95 study. Cancer 2017; 124:1016-1024. [DOI: 10.1002/cncr.31157] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/18/2017] [Accepted: 10/24/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Bas Vaarwerk
- Department of Pediatric Oncology; Emma Children's Hospital/Academic Medical Center; Amsterdam the Netherlands
| | - Johanna H. van der Lee
- Pediatric Clinical Research Office; Emma Children's Hospital/Academic Medical Center; Amsterdam the Netherlands
| | - Willemijn B. Breunis
- Department of Pediatric Oncology; Emma Children's Hospital/Academic Medical Center; Amsterdam the Netherlands
| | - Daniel Orbach
- Department of Pediatric, Adolescent, and Young Adult Oncology; Curie Institute; Paris France
| | - Julia C. Chisholm
- Children and Young People's Department; Royal Marsden Hospital; Sutton United Kingdom
| | - Nathalie Cozic
- Department of Biostatistics and Epidemiology; Gustave-Roussy; Villejuif France
| | - Meriel Jenney
- Department of Pediatric Oncology; Children's Hospital for Wales; Cardiff United Kingdom
| | - Rick R. van Rijn
- Pediatric Radiology; Emma Children's Hospital/Academic Medical Center; Amsterdam the Netherlands
| | - Kieran McHugh
- Department of Radiology; Great Ormond Street Hospital for Children; London United Kingdom
| | - Soledad Gallego
- Pediatric Oncology; Vall d'Hebron University Hospital; Barcelona Spain
| | - Heidi Glosli
- Department of Pediatric and Adolescent Medicine; Oslo University Hospital; Oslo Norway
| | - Christine Devalck
- Pediatric Hematology Oncology Department; Children's University Hospital; Brussels Belgium
| | - Mark N. Gaze
- Department of Oncology; University College London Hospitals NHS Foundation Trust; London United Kingdom
| | - Anna Kelsey
- Pathology Department; Royal Manchester Children's Hospital; Manchester United Kingdom
| | | | - Michael C. G. Stevens
- Department of Pediatric Oncology; Bristol Royal Hospital for Children; Bristol United Kingdom
| | - Odile Oberlin
- Department of Pediatric and Adolescent Oncology; Gustave-Roussy; Villejuif France
| | | | - Johannes H. M. Merks
- Department of Pediatric Oncology; Emma Children's Hospital/Academic Medical Center; Amsterdam the Netherlands
| |
Collapse
|
18
|
Radiogenomics of neuroblastomas: Relationships between imaging phenotypes, tumor genomic profile and survival. PLoS One 2017; 12:e0185190. [PMID: 28945781 PMCID: PMC5612658 DOI: 10.1371/journal.pone.0185190] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 09/05/2017] [Indexed: 01/01/2023] Open
Abstract
Purpose This study investigated relationships between neuroblastomas (NBs) imaging phenotypes, tumor genomic profile and patient outcome. Patients and methods This IRB-approved retrospective observational study included 133 NB patients (73 M, 60 F; median age 15 months, range 0–151) treated in a single institution between 1998 and 2012. A consensus review of imaging (CT-scan, MRI) categorized tumors according to both the primarily involved compartment (i.e., neck, chest, abdomen or pelvis) and the sympathetic anatomical structure the tumors rose from (i.e., cervical, paravertebral or periarterial chains, or adrenal gland). Tumor shape, volume and image-defined surgical risk factors (IDRFs) at diagnosis were recorded. Genomic profiles were assessed using array-based comparative genomic hybridization and divided into three groups: “numerical-only chromosome alterations” (NCA), “segmental chromosome alterations” (SCA) and “MYCN amplification” (MNA). Statistical analyses included Kruskal–Wallis, Chi2 and Fisher’s exact tests and the Kaplan-Meier method with log-rank tests and Cox model for univariate and multivariate survival analyses. Results A significant association between the sympathetic structure origin of tumors and genomic profiles was demonstrated. NBs arising from cervical sympathetic chains were all NCA. Paravertebral NBs were NCA or SCA in 75% and 25%, respectively and none were MNA. Periarterial NBs were NCA, SCA or MNA in 33%, 56% and 11%, respectively. Adrenal NBs were NCA, SCA or MNA in 16%, 36% and 48%, respectively. Among MNA NBs, 92% originated from the adrenal gland. The sympathetic anatomical classification was significantly better correlated to overall survival than the compartmental classification (P < .0003). The tumor volume of MNA NBs was significantly higher than NCA or SCA NBs (P < .0001). Patients with initial volume less than 160 mL had significantly better overall survival (P < .009). A “single mass” pattern was significantly more frequent in NCA NBs (P = .0003). The number of IDRFs was significantly higher in MNA NBs (P < .0001). Conclusion Imaging phenotypes of neuroblastomas, including tumor origin along the sympathetic system, correlate with tumor genomic profile and patient outcome.
Collapse
|
19
|
Zhen Z, Yang K, Ye L, You Z, Chen R, Liu Y. Decorin gene upregulation mediated by an adeno-associated virus vector increases intratumoral uptake of nab-paclitaxel in neuroblastoma via inhibition of stabilin-1. Invest New Drugs 2017. [PMID: 28631095 DOI: 10.1007/s10637-017-0477-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The availability of effective medication for the treatment of refractory or recurrent neuroblastoma remains limited. This study sought to investigate the effects of increased decorin (DCN) expression on the intratumoral uptake of nab-paclitaxel as a potential novel approach to NB. Correlation between the clinical characteristics of neuroblastoma and the expression of DCN, secreted protein acidic and rich in cysteine (SPARC) and stabilin-1 was evaluated. The anticancer effect of recombinant adeno-associated virus-DCN (rAAV-DCN) was assessed in vivo and in vitro. And the effect of rAAV-DCN on the intratumoral uptake of paclitaxel was also studied in neuroblastoma-grafted nude mice. Overall, 12.5%, 17.7%, and 71.9% of the tumors stained positive for DCN, SPARC and stabilin-1 respectively and correlated to age, stage and N-MYC status in 96 children and adolescents with neuroblastoma. Transfected neuroblastoma cells stably expressed DCN, with in vivo and in vitro studies demonstrating rAAV-DCN sensitized the anticancer effect of nab-paclitaxel. Systemic rAAV-DCN in neuroblastoma-grafted nude mice inhibited stabilin-1, up-regulated SPARC, and increased the intratumoral uptake of paclitaxel. Macrophage depletion or anti-stabilin-1 monoclonal antibody increased the intratumoral uptake of nab-paclitaxel and its anticancer effects to a degree comparable to that achieved by systemic rAAV-DCN. The systemic administration of rAAV-DCN up-regulates DCN in neuroblastoma and accelerates the intratumoral uptake of nab-paclitaxel by inhibiting stabilin-1 mediated SPARC degradation.
Collapse
Affiliation(s)
- Zijun Zhen
- State Key Laboratory of Oncology in South China, Guangzhou, China. .,Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China. .,Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.
| | - Kaibin Yang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.,Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, China
| | - Litong Ye
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.,Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, China
| | - Zhiyao You
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.,Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, China
| | - Rirong Chen
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.,Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, China
| | - Ying Liu
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.,Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, China
| |
Collapse
|
20
|
Zhen Z, Yang K, Ye L, You Z, Chen R, Liu Y, He Y. Suberoylanilide hydroxamic acid sensitizes neuroblastoma to paclitaxel by inhibiting thioredoxin-related protein 14-mediated autophagy. Cancer Sci 2017; 108:1485-1492. [PMID: 28498513 PMCID: PMC5497723 DOI: 10.1111/cas.13279] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/01/2017] [Accepted: 05/08/2017] [Indexed: 12/21/2022] Open
Abstract
Paclitaxel is not as effective for neuroblastoma as most of the front‐line chemotherapeutics due to drug resistance. This study explored the regulatory mechanism of paclitaxel‐associated autophagy and potential solutions to paclitaxel resistance in neuroblastoma. The formation of autophagic vesicles was detected by scanning transmission electron microscopy and flow cytometry. The autophagy‐associated proteins were assessed by western blot. Autophagy was induced and the autophagy‐associated proteins LC3‐I, LC3‐II, Beclin 1, and thioredoxin‐related protein 14 (TRP14), were found to be upregulated in neuroblastoma cells that were exposed to paclitaxel. The inhibition of Beclin 1 or TRP14 by siRNA increased the sensitivity of the tumor cells to paclitaxel. In addition, Beclin 1‐mediated autophagy was regulated by TRP14. Furthermore, the TRP14 inhibitor suberoylanilide hydroxamic acid (SAHA) downregulated paclitaxel‐induced autophagy and enhanced the anticancer effects of paclitaxel in normal control cancer cells but not in cells with upregulated Beclin 1 and TRP14 expression. Our findings showed that paclitaxel‐induced autophagy in neuroblastoma cells was regulated by TRP14 and that SAHA could sensitize neuroblastoma cells to paclitaxel by specifically inhibiting TRP14.
Collapse
Affiliation(s)
- Zijun Zhen
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Kaibin Yang
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, China
| | - Litong Ye
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, China
| | - Zhiyao You
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, China
| | - Rirong Chen
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, China
| | - Ying Liu
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, China
| | - Youjian He
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.,Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
21
|
Yao YMM, Donoho GP, Iversen PW, Zhang Y, Van Horn RD, Forest A, Novosiadly RD, Webster YW, Ebert P, Bray S, Ting JC, Aggarwal A, Henry JR, Tiu RV, Plowman GD, Peng SB. Mouse PDX Trial Suggests Synergy of Concurrent Inhibition of RAF and EGFR in Colorectal Cancer with BRAF or KRAS Mutations. Clin Cancer Res 2017; 23:5547-5560. [DOI: 10.1158/1078-0432.ccr-16-3250] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 05/17/2017] [Accepted: 06/07/2017] [Indexed: 11/16/2022]
|
22
|
Lee JW, Lee S, Cho HW, Ma Y, Yoo KH, Sung KW, Koo HH, Cho EJ, Lee SK, Lim DH. Incorporation of high-dose 131I-metaiodobenzylguanidine treatment into tandem high-dose chemotherapy and autologous stem cell transplantation for high-risk neuroblastoma: results of the SMC NB-2009 study. J Hematol Oncol 2017; 10:108. [PMID: 28511709 PMCID: PMC5432997 DOI: 10.1186/s13045-017-0477-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 05/05/2017] [Indexed: 11/10/2022] Open
Abstract
Background In our previous SMC NB-2004 study of patients with high-risk neuroblastomas, which incorporated total-body irradiation (TBI) with second high-dose chemotherapy and autologous stem cell transplantation (HDCT/auto-SCT), the survival rate was encouraging; however, short- and long-term toxicities were significant. In the present SMC NB-2009 study, only TBI was replaced with 131I-meta-iodobenzylguanidine (MIBG) treatment in order to reduce toxicities. Methods From January 2009 to December 2013, 54 consecutive patients were assigned to receive tandem HDCT/auto-SCT after nine cycles of induction chemotherapy. The CEC (carboplatin + etoposide + cyclophosphamide) regimen and the TM (thiotepa + melphalan) regimen with (for metastatic MIBG avid tumors) or without (for localized or MIBG non-avid tumors) 131I-MIBG treatment (18 or 12 mCi/kg) were used for tandem HDCT/auto-SCT. Local radiotherapy, differentiation therapy with 13-cis-retinoic acid, and immunotherapy with interleukin-2 were administered after tandem HDCT/auto-SCT. Results Fifty-two patients underwent the first HDCT/auto-SCT and 47 patients completed tandem HDCT/auto-SCT. There was no significant immediate toxicity during the 131I-MIBG infusion. Acute toxicities during the tandem HDCT/auto-SCT were less severe in the NB-2009 study than in the NB-2004 study. Late effects such as growth hormone deficiency, cataracts, and glomerulopathy evaluated at 3 years after the second HDCT/auto-SCT were also less significant in the NB-2009 study than in NB-2004 study. There was no difference in the 5-year event-free survival (EFS) between the two studies (67.5 ± 6.7% versus 58.3 ± 6.9%, P = 0.340). Conclusions Incorporation of high-dose 131I-MIBG treatment into tandem HDCT/auto-SCT could reduce short- and long-term toxicities associated with TBI, without jeopardizing the survival rate. Trial registration ClinicalTrials.gov NCT03061656
Collapse
Affiliation(s)
- Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea
| | - Sanghoon Lee
- Department of Pediatric Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea
| | - Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea
| | - Youngeun Ma
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea.
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea
| | - Eun Joo Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea
| | - Suk-Koo Lee
- Department of Pediatric Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 135-710, Republic of Korea
| |
Collapse
|
23
|
Trout AT, Towbin AJ, Klingbeil L, Weiss BD, von Allmen D. Single and multidimensional measurements underestimate neuroblastoma response to therapy. Pediatr Blood Cancer 2017; 64:18-24. [PMID: 27440309 DOI: 10.1002/pbc.26159] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/26/2016] [Accepted: 06/28/2016] [Indexed: 11/08/2022]
Abstract
BACKGROUND Changes in three-dimensional (3D) measurements of neuroblastoma are used to assess response. Linear measurements may not accurately characterize tumor size due to the infiltrative character of these tumors. The purpose of this study was to assess the accuracy of one-dimensional (1D), two-dimensional (2D), and 3D measurements in characterizing neuroblastoma response compared to a reference standard of tumor volume. PROCEDURE We retrospectively reviewed imaging for 34 patients with stage 3 or 4 neuroblastoma. Blinded readers contoured or made linear measurements of tumors. Correlation coefficients were used to compare linear measurements to volumetric and 3D measurements. Bland-Altman analyses were used to assess bias between measurements. Sensitivity and specificity for patient events and survival were calculated for each measurement technique. RESULTS Mean patient age was 2.9 ± 3.0 years (range 0-15 years). There was strong correlation between volumetric and 1D (r = 0.78, P < 0.0001), 2D (r = 0.86, P < 0.0001), and 3D (r = 0.88, P < 0.0001) measurements. Mean bias between volumetric measurements and 1D, 2D, and 3D measurements was 37.1% (95% limits: 6.2-67.9%), 16.1% (95% limits: -11.7-43.8%), and 7.7% (95% limits: -19.7-35.1%), respectively. 1D and 2D measurements undercategorized response versus volumetric change in 88.2% (30/34) and 29.4% (10/34) of cases. 3D measurements incorrectly characterized response in 16.7% (4/24) of cases versus volumetric change. 3D measurements were highly sensitive for patient events and survival, but all measurement techniques had poor specificity. CONCLUSIONS 3D measurements most accurately quantify neuroblastoma size response versus volumetric change in patients with stage 3 and 4 neuroblastoma. 1D and 2D measurements underrepresent tumor response.
Collapse
Affiliation(s)
- Andrew T Trout
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Alexander J Towbin
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Lindsey Klingbeil
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Brian D Weiss
- Cancer and Blood Disease Institute, Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Daniel von Allmen
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
24
|
Angelini P, Irwin MS. Measuring primary tumor response in neuroblastoma: More than using a ruler. Pediatr Blood Cancer 2017; 64:11-12. [PMID: 27762062 DOI: 10.1002/pbc.26274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 08/24/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Paola Angelini
- Division of Haematology-Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Ontario, Canada
| | - Meredith S Irwin
- Division of Haematology-Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Ontario, Canada
| |
Collapse
|
25
|
Ma Y, Lee JW, Park SJ, Yi ES, Choi YB, Yoo KH, Sung KW, Koo HH. Detection of MYCN Amplification in Serum DNA Using Conventional Polymerase Chain Reaction. J Korean Med Sci 2016; 31:1392-6. [PMID: 27510381 PMCID: PMC4974179 DOI: 10.3346/jkms.2016.31.9.1392] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/03/2016] [Indexed: 01/11/2023] Open
Abstract
Neuroblastoma (NB) is the most common extra-cranial solid tumor of childhood and is characterized by a wide range of clinical behaviors. Amplification of MYCN is a well-known poor prognostic factor in NB patients. As the MYCN amplification status is usually tested using tumor specimens, lengthy and invasive procedures are unavoidable. To evaluate the possibility of detecting MYCN amplification without invasive procedure, we performed conventional polymerase chain reaction (PCR) analysis to identify MYCN amplification using the preserved serum DNA. PCR of serum DNA was done in 105 NB patients whose MYCN status had been confirmed by fluorescence in situ hybridization. MYCN amplification was evaluated as the ratio of signal intensities between MYCN and NAGK (M/N ratio). When regarding the tissue FISH results as a reference, 10 patients had MYCN-amplified (MNA) NB, and 95 had non-MNA NB. The M/N ratio of the MNA group (median 2.56, range 1.01-3.58) was significantly higher than that of the non-MNA group (median 0.97, range 0.67-5.18) (P < 0.001). In the receiver operating characteristic curve analysis, the area under the curve was 0.957 (95% confidence interval 0.898-1.000; P < 0.001), and it showed 90.9% sensitivity and 97.9% specificity with the selected cut-off value set as 1.6. The detection of MYCN amplification using conventional PCR analysis of serum samples seems to be a simple and promising method to evaluate the MYCN status of NB patients. Further study with a larger set of patients is needed to confirm the accuracy of this result.
Collapse
Affiliation(s)
- Youngeun Ma
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo Jin Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun Sang Yi
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Bae Choi
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
26
|
Bagatell R, McHugh K, Naranjo A, Van Ryn C, Kirby C, Brock P, Lyons KA, States LJ, Rojas Y, Miller A, Volchenboum SL, Simon T, Krug B, Sarnacki S, Valteau-Couanet D, von Schweinitz D, Kammer B, Granata C, Pio L, Park JR, Nuchtern J. Assessment of Primary Site Response in Children With High-Risk Neuroblastoma: An International Multicenter Study. J Clin Oncol 2016; 34:740-6. [PMID: 26755515 DOI: 10.1200/jco.2015.63.2042] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The International Neuroblastoma Response Criteria (INRC) require serial measurements of primary tumors in three dimensions, whereas the Response Evaluation Criteria in Solid Tumors (RECIST) require measurement in one dimension. This study was conducted to identify the preferred method of primary tumor response assessment for use in revised INRC. PATIENTS AND METHODS Patients younger than 20 years with high-risk neuroblastoma were eligible if they were diagnosed between 2000 and 2012 and if three primary tumor measurements (antero-posterior, width, cranio-caudal) were recorded at least twice before resection. Responses were defined as ≥ 30% reduction in longest dimension as per RECIST, ≥ 50% reduction in volume as per INRC, or ≥ 65% reduction in volume. RESULTS Three-year event-free survival for all patients (N = 229) was 44% and overall survival was 58%. The sensitivity of both volume response measures (ability to detect responses in patients who survived) exceeded the sensitivity of the single dimension measure, but the specificity of all response measures (ability to identify lack of response in patients who later died) was low. In multivariable analyses, none of the response measures studied was predictive of outcome, and none was predictive of the extent of resection. CONCLUSION None of the methods of primary tumor response assessment was predictive of outcome. Measurement of three dimensions followed by calculation of resultant volume is more complex than measurement of a single dimension. Primary tumor response in children with high-risk neuroblastoma should therefore be evaluated in accordance with RECIST criteria, using the single longest dimension.
Collapse
Affiliation(s)
- Rochelle Bagatell
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy.
| | - Kieran McHugh
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Arlene Naranjo
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Collin Van Ryn
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Chaim Kirby
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Penelope Brock
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Karen A Lyons
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Lisa J States
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Yesenia Rojas
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Alexandra Miller
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Sam L Volchenboum
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Thorsten Simon
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Barbara Krug
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Sabine Sarnacki
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Dominique Valteau-Couanet
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Dietrich von Schweinitz
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Birgit Kammer
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Claudio Granata
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Luca Pio
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Julie R Park
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| | - Jed Nuchtern
- Rochelle Bagatell, Lisa J. States, and Alexandra Miller, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA; Arlene Naranjo and Collin Van Ryn, University of Florida, Gainesville, FL; Chaim Kirby and Sam L. Volchenboum, University of Chicago, Chicago, IL; Karen A. Lyons, Yesenia Rojas, and Jed Nuchtern, Texas Children's Hospital and Baylor College of Medicine, Houston, TX; Julie R. Park, Seattle Children's Hospital and University of Washington, Seattle, WA; Kieran McHugh and Penelope Brock, Great Ormond Street Hospital for Children, London, United Kingdom; Thorsten Simon and Barbara Krug, University of Cologne Medical School, Cologne; Dietrich von Schweinitz and Birgit Kammer, Dr von Hauner Children's Hospital and University of Munich, Munich, Germany; Sabine Sarnacki, Hopital Necker-Enfants Malades and Paris Descartes University; Dominique Valteau-Couanet, Institut Gustave Roussy, Universite Paris Sud, Paris, France; and Claudio Granata and Luca Pio, Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
27
|
Choi YB, Bae GE, Lee NH, Kim JS, Lee SH, Yoo KH, Sung KW, Koo HH. Clinical Significance of Persistent Tumor in Bone Marrow during Treatment of High-risk Neuroblastoma. J Korean Med Sci 2015; 30:1062-7. [PMID: 26240483 PMCID: PMC4520936 DOI: 10.3346/jkms.2015.30.8.1062] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/30/2015] [Indexed: 11/30/2022] Open
Abstract
The records of 63 high-risk neuroblastoma patients with bone marrow (BM) tumors at diagnosis were retrospectively reviewed. All patients received nine cycles of induction chemotherapy followed by tandem high-dose chemotherapy and autologous stem cell transplantation (HDCT/auto-SCT). Follow-up BM examination was performed every three cycles during induction chemotherapy and every three months for one year after the second HDCT/auto-SCT. BM tumor cells persisted in 48.4%, 37.7%, 23.3%, and 20.4% of patients after three, six, and nine cycles of induction chemotherapy and three months after the second HDCT/auto-SCT, respectively. There was no difference in progression-free survival (PFS) rate between patients with persistent BM tumor and those without during the induction treatment. However, after tandem HDCT/auto-SCT, the PFS rate was worse in patients with persistent BM tumor than in those without (probability of 5-yr PFS 14.7% ± 13.4% vs. 64.2% ± 8.3%, P = 0.009). Persistent BM tumor during induction treatment is not associated with a worse prognosis when intensive tandem HDCT/auto-SCT is given as consolidation treatment. However, persistent BM tumor after tandem HDCT/auto-SCT is associated with a worse prognosis. Therefore, further treatment might be needed in patients with persistent BM tumor after tandem HDCT/auto-SCT.
Collapse
Affiliation(s)
- Young Bae Choi
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Go Eun Bae
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Na Hee Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung-Sun Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo Hyun Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
28
|
Gajer JM, Furdas SD, Gründer A, Gothwal M, Heinicke U, Keller K, Colland F, Fulda S, Pahl HL, Fichtner I, Sippl W, Jung M. Histone acetyltransferase inhibitors block neuroblastoma cell growth in vivo. Oncogenesis 2015; 4:e137. [PMID: 25664930 PMCID: PMC4338425 DOI: 10.1038/oncsis.2014.51] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 12/09/2014] [Indexed: 12/25/2022] Open
Abstract
We have previously described novel histone acetyltransferase (HAT) inhibitors that block neuroblastoma cell growth in vitro. Here we show that two selected pyridoisothiazolone HAT inhibitors, PU139 and PU141, induce cellular histone hypoacetylation and inhibit growth of several neoplastic cell lines originating from different tissues. Broader in vitro selectivity profiling shows that PU139 blocks the HATs Gcn5, p300/CBP-associated factor (PCAF), CREB (cAMP response element-binding) protein (CBP) and p300, whereas PU141 is selective toward CBP and p300. The pan-inhibitor PU139 triggers caspase-independent cell death in cell culture. Both inhibitors block growth of SK-N-SH neuroblastoma xenografts in mice and the PU139 was shown to synergize with doxorubicin in vivo. The latter also reduces histone lysine acetylation in vivo at concentrations that block neoplastic xenograft growth. This is one of the very few reports on hypoacetylating agents with in vivo anticancer activity.
Collapse
Affiliation(s)
- J M Gajer
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - S D Furdas
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Freiburg Institute of Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany
| | - A Gründer
- Section of Molecular Hematology, Department of Hematology/Oncology, University Hospital Freiburg, Freiburg, Germany
| | - M Gothwal
- Section of Molecular Hematology, Department of Hematology/Oncology, University Hospital Freiburg, Freiburg, Germany
| | - U Heinicke
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| | - K Keller
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - F Colland
- Hybrigenics, 3-5 impasse Reille, Paris, France
| | - S Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
- German Cancer Consortium (DKTK), Frankfurt, Germany
- German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - H L Pahl
- Section of Molecular Hematology, Department of Hematology/Oncology, University Hospital Freiburg, Freiburg, Germany
| | - I Fichtner
- Experimental Pharmacology and Oncology Berlin-Buch GmbH, Berlin-Buch, Germany
| | - W Sippl
- Freiburg Institute of Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany
- Department of Pharmaceutical Chemistry, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - M Jung
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Frankfurt, Germany
- German Cancer Consortium (DKTK), Freiburg, Germany
| |
Collapse
|
29
|
Dantonello TM, Stark M, Timmermann B, Fuchs J, Selle B, Linderkamp C, Handgretinger R, Hagen R, Feuchtgruber S, Kube S, Kosztyla D, Kazanowska B, Ladenstein R, Niggli F, Ljungman G, Bielack SS, Klingebiel T, Koscielniak E. Tumour volume reduction after neoadjuvant chemotherapy impacts outcome in localised embryonal rhabdomyosarcoma. Pediatr Blood Cancer 2015; 62:16-23. [PMID: 25263634 DOI: 10.1002/pbc.25207] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 07/07/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Response (tumour volume reduction) to induction chemotherapy has been used to stratify secondary local and systemic treatment of Intergroup Rhabdomyosarcoma Study Group III (IRSG-III) embryonal rhabdomyosarcoma (RME) in consecutive CWS-trials. To evaluate its actual impact we studied response-related treatment and outcomes. PROCEDURE Patients with IRSG-III RME <21 years and non-response (NR, <33% volume reduction) in five consecutive CWS-trials were analysed and compared with partial responders (PAR, ≥ 33% reduction). The NR was reviewed and sub-classified as Objective Response (OR, <0%-33% reduction) or Stable/Progressive Disease (SPD). RESULTS Fifty-nine of 529 patients had NR (n = 34 OR, n = 25 SPD). Primary risk-factors including age, tumour size, and TN-classification did not differ between NR and PAR groups but NR had more patients with unfavourable sites comparatively (P = 0.04). There were no differences in primary risk-factors between OR and SPD. Significant factors associated with poor outcome in multivariate analysis were NR, TN-classification, age >10 years, tumour size >5 cm and therapy in older trials. After response assessment n = 24 NR continued to receive induction chemotherapy, n = 32 received other combinations and n = 3 no further chemotherapy. Forty-two non-responders were irradiated, and the tumours were completely resected in n = 20. After a median follow-up of 8 years, 34 NR are alive. Seventeen of 21 failures leading to disease-related deaths were locoregional. The five-year overall survival rate (OS) was 76 ± 4% for PAR, 79 ± 14% for OR, but only 40 ± 19% for SPD (P < 0.001). CONCLUSION Response to induction chemotherapy appears to be an important surrogate marker of poor outcome in patients with SPD largely due to ineffective local control.
Collapse
Affiliation(s)
- Tobias M Dantonello
- Paediatrics 5 (oncology, hematology, immunology), Olgahospital, Klinikum Stuttgart, Stuttgart, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|