1
|
Yamamoto N, Ishizawa K, Umemoto M, Nishimura A, Fujikawa T, Inoue S, Nakatani N, Tamura A, Nino N, Uemura S, Hasegawa D, Kosaka Y, Nishimura N. Evaluation of Minimal Residual Disease in Patients with Neuroblastoma. Mol Diagn Ther 2025:10.1007/s40291-025-00788-4. [PMID: 40450177 DOI: 10.1007/s40291-025-00788-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2025] [Indexed: 06/03/2025]
Abstract
Neuroblastoma (NB) is a pediatric extracranial solid tumor that accounts for approximately 15% of all pediatric cancer deaths. More than 50% of patients with newly diagnosed NB are classified into a high-risk group with an approximately 50% long-term survival rate. Although most high-risk patients with NB achieve remission, more than half may have minimal residual disease (MRD) that eventually causes relapse. Looking towards an optimal outcome, the accurate evaluation of MRD in patients with NB (NB-MRD) is essential to monitor the treatment response and disease burden. Over the past decades, the quantification of NB-associated messenger RNA (NB-mRNA) by reverse transcriptase-polymerase chain reaction has become widely used to detect NB-MRD, owing to the lack of recurrent genomic aberrations in NB cells. To achieve a more accurate and sensitive detection, the current NB-MRD assays quantify a set of NB-mRNAs to detect NB cells in bone marrow, peripheral blood, and peripheral blood stem cell samples. Among a growing number of NB-MRD assays, several assays quantitating different but overlapping sets of NB-mRNAs are reported to have a significant prognostic value. However, the clinical significance of NB-MRD remains to be established. In this review, we summarize the recent progress in NB-MRD and evaluate its clinical value.
Collapse
Affiliation(s)
- Nobuyuki Yamamoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kikyo Ishizawa
- Department of Public Health, Kobe University Graduate School of Health Science, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Mayuno Umemoto
- Department of Public Health, Kobe University Graduate School of Health Science, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Akihiro Nishimura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoko Fujikawa
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shotaro Inoue
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoko Nakatani
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akihiro Tamura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Nanako Nino
- Department of Hematology and Oncology, Kobe Children Hospital, Kobe, Japan
| | - Suguru Uemura
- Department of Hematology and Oncology, Kobe Children Hospital, Kobe, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Kobe Children Hospital, Kobe, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Kobe Children Hospital, Kobe, Japan
| | - Noriyuki Nishimura
- Department of Public Health, Kobe University Graduate School of Health Science, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan.
| |
Collapse
|
2
|
Simon T, Thole T, Castelli S, Timmermann B, Jazmati D, Schwarz R, Fuchs J, Warmann S, Hubertus J, Schmidt M, Rogasch J, Körber F, Vokuhl C, Schäfer J, Schulte JH, Deubzer H, Rosswog C, Fischer M, Lang P, Langer T, Astrahantseff K, Lode H, Hero B, Eggert A. GPOH Guidelines for Diagnosis and First-line Treatment of Patients with Neuroblastic Tumors, update 2025. KLINISCHE PADIATRIE 2025; 237:117-140. [PMID: 40345224 DOI: 10.1055/a-2556-4302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
The clinical course of neuroblastoma is more heterogeneous than any other malignant disease. Many low-risk patients experience regression after limited or even no chemotherapy. However, more than half of high-risk patients die from disease despite intensive multimodal treatment. Precise disease characterization for each patient at diagnosis is key for risk-adapted treatment. The guidelines presented here incorporate results from national and international clinical trials to produce recommendations for diagnosing and treating neuroblastoma patients in German hospitals outside of clinical trials.
Collapse
Affiliation(s)
- Thorsten Simon
- Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - Theresa Thole
- Pediatric Oncology and Hematology, Charité University Hospital Berlin, Berlin, Germany
| | - Sveva Castelli
- Pediatric Oncology and Hematology, Charité University Hospital Berlin, Berlin, Germany
| | - Beate Timmermann
- Westgerman Protontherapycenter Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Danny Jazmati
- Department of Radiation Oncology, Heinrich Heine University Düsseldorf, Dusseldorf, Germany
| | | | - Jörg Fuchs
- Pediatric Surgery and Urology, University of Tübingen, Tübingen, Germany
| | - Steven Warmann
- Department of Pediatric Surgery, Charité University Hospital Berlin, Berlin, Germany
| | - Jochen Hubertus
- Department of Pediatric Surgery, Marien-Hospital Witten, Witten, Germany
| | | | - Julian Rogasch
- Nuclear Medicine, Charité University Hospital Berlin, Berlin, Germany
| | - Friederike Körber
- Institut und Poliklinik für Radiologische Diagnostik, Kinderradiologie, University of Cologne, Cologne, Germany
| | - Christian Vokuhl
- Pediatric Pathology, Institute for Pathology, University of Bonn, Bonn, Germany
| | - Jürgen Schäfer
- Diagnostic and Interventional Radiology, University of Tübingen, Tübingen, Germany
| | | | - Hedwig Deubzer
- Pediatric Oncology and Hematology, Charité University Hospital Berlin, Berlin, Germany
| | - Carolina Rosswog
- Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
- Experimental Pediatric Oncology, University of Cologne, Cologne, Germany
| | - Matthias Fischer
- Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
- Experimental Pediatric Oncology, University of Cologne, Cologne, Germany
| | - Peter Lang
- Pediatric Oncology and Hematology, University of Tübingen, Tübingen, Germany
| | - Thorsten Langer
- Childrens' Hospital, University Hospital Schleswig-Holstein Lübeck Campus, Lübeck, Germany
| | - Kathy Astrahantseff
- Pediatric Oncology and Hematology, Charité University Hospital Berlin, Berlin, Germany
| | - Holger Lode
- Pediatric Oncology and Hematology, University of Greifswald, Greifswald, Germany
| | - Barbara Hero
- Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - Angelika Eggert
- Pediatric Oncology and Hematology, Charité University Hospital Berlin, Berlin, Germany
| |
Collapse
|
3
|
van Zogchel LMJ, Decarolis B, van Wezel EM, Zappeij-Kannegieter L, Gelineau NU, Schumacher-Kuckelkorn R, Simon T, Berthold F, van Noesel MM, Fiocco M, van der Schoot CE, Hero B, Stutterheim J, Tytgat GAM. Sensitive liquid biopsy monitoring correlates with outcome in the prospective international GPOH-DCOG high-risk neuroblastoma RT-qPCR validation study. J Exp Clin Cancer Res 2024; 43:331. [PMID: 39722049 DOI: 10.1186/s13046-024-03261-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Liquid biopsies offer less burdensome sensitive disease monitoring. Bone marrow (BM) metastases, common in various cancers including neuroblastoma, is associated with poor outcomes. In pediatric high-risk neuroblastoma most patients initially respond to treatment, but in the majority the disease recurs with only 40% long-term survivors, stressing the need for more sensitive detection of disseminated disease during therapy. METHODS To validate sensitive neuroblastoma mRNA RT-qPCR BM testing, we prospectively assessed serial BM samples from 345 international high-risk neuroblastoma patients, treated in trials NB2004 (GPOH) or NBL2009 (DCOG), using PHOX2B, TH, DDC, CHRNA3, and GAP43 RT-qPCR mRNA markers and BM GD2-immunocytology. Association between BM-infiltration levels and event-free survival (EFS) and overall survival (OS) was estimated by using Cox regression models and Kaplan-Meier's methodology. RESULTS BM infiltration >10% by RT-qPCR at diagnosis was prognostic for survival (adjusted hazard ratio (HR) 1.82 [95%CI 1.25-2.63] and 2.04 [1.33-3.14] for EFS and OS, respectively). Any post-induction RT-qPCR positivity correlated with poor EFS and OS, with a HR of 2.10 [1.27-3.49] and 1.76 [1.01-3.08] and 5-years EFS of 26.6% [standard error 5.2%] versus 60.4% [6.7] and OS of 43.8% [5.9] versus 65.7% [6.6] for RT-qPCR-positive patients versus RT-qPCR-negative patients. In contrast, post-induction immunocytology positivity was not associated with EFS or OS (HR 1.22 [0.68-2.19] and 1.26 [0.54-2.42]). CONCLUSION This study validates the association of not clearing of BM metastases by sensitive RT-qPCR detection with very poor outcome. We therefore propose implementation of RT-qPCR for minimal residual disease testing in neuroblastoma to guide therapy.
Collapse
Affiliation(s)
- Lieke M J van Zogchel
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Boris Decarolis
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | - Esther M van Wezel
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Lily Zappeij-Kannegieter
- Department of Immunocytology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Nina U Gelineau
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Roswitha Schumacher-Kuckelkorn
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | - Thorsten Simon
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | - Frank Berthold
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Division Imaging & Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marta Fiocco
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- Department of Biomedical data Science, Section Medical Statistics, Leiden University Medical Center, Leiden, The Netherlands
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Barbara Hero
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | | | - Godelieve A M Tytgat
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands.
- Department of Genetics, Utrecht University Medical Center, Utrecht, the Netherlands.
| |
Collapse
|
4
|
Janssen FW, Lak NSM, Janda CY, Kester LA, Meister MT, Merks JHM, van den Heuvel-Eibrink MM, van Noesel MM, Zsiros J, Tytgat GAM, Looijenga LHJ. A comprehensive overview of liquid biopsy applications in pediatric solid tumors. NPJ Precis Oncol 2024; 8:172. [PMID: 39097671 PMCID: PMC11297996 DOI: 10.1038/s41698-024-00657-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/15/2024] [Indexed: 08/05/2024] Open
Abstract
Liquid biopsies are emerging as an alternative source for pediatric cancer biomarkers with potential applications during all stages of patient care, from diagnosis to long-term follow-up. While developments within this field are reported, these mainly focus on dedicated items such as a specific liquid biopsy matrix, analyte, and/or single tumor type. To the best of our knowledge, a comprehensive overview is lacking. Here, we review the current state of liquid biopsy research for the most common non-central nervous system pediatric solid tumors. These include neuroblastoma, renal tumors, germ cell tumors, osteosarcoma, Ewing sarcoma, rhabdomyosarcoma and other soft tissue sarcomas, and liver tumors. Within this selection, we discuss the most important or recent studies involving liquid biopsy-based biomarkers, anticipated clinical applications, and the current challenges for success. Furthermore, we provide an overview of liquid biopsy-based biomarker publication output for each tumor type based on a comprehensive literature search between 1989 and 2023. Per study identified, we list the relevant liquid biopsy-based biomarkers, matrices (e.g., peripheral blood, bone marrow, or cerebrospinal fluid), analytes (e.g., circulating cell-free and tumor DNA, microRNAs, and circulating tumor cells), methods (e.g., digital droplet PCR and next-generation sequencing), the involved pediatric patient cohort, and proposed applications. As such, we identified 344 unique publications. Taken together, while the liquid biopsy field in pediatric oncology is still behind adult oncology, potentially relevant publications have increased over the last decade. Importantly, steps towards clinical implementation are rapidly gaining ground, notably through validation of liquid biopsy-based biomarkers in pediatric clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Michael T Meister
- Princess Máxima Center, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Johannes H M Merks
- Princess Máxima Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center, Utrecht, the Netherlands
- Wilhelmina Children's Hospital-Division of CHILDHEALTH, University Medical Center Utrech, University of Utrecht, Utrecht, the Netherlands
| | - Max M van Noesel
- Princess Máxima Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | | | - Godelieve A M Tytgat
- Princess Máxima Center, Utrecht, the Netherlands
- Department of Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Leendert H J Looijenga
- Princess Máxima Center, Utrecht, the Netherlands.
- Department of Pathology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
5
|
Nakatani N, Win KHN, Mon CY, Fujikawa T, Uemura S, Saito A, Ishida T, Mori T, Hasegawa D, Kosaka Y, Inoue S, Nishimura A, Nino N, Tamura A, Yamamoto N, Nozu K, Nishimura N. Distinct Expression Profiles of Neuroblastoma-Associated mRNAs in Peripheral Blood and Bone Marrow of Non-High-Risk and High-Risk Neuroblastoma Patients. BIOLOGY 2024; 13:345. [PMID: 38785826 PMCID: PMC11117621 DOI: 10.3390/biology13050345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/03/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024]
Abstract
Non-high-risk (non-HR) neuroblastoma (NB) patients have excellent outcomes, with more than a 90% survival rate, whereas HR NB patients expect less than a 50% survival rate. Metastatic disease is the principal cause of death among both non-HR and HR NB patients. Previous studies have reported the significant but limited prognostic value of quantitative PCR (qPCR)-based assays, measuring overlapping but different sets of neuroblastoma-associated mRNAs (NB-mRNAs), to detect metastatic disease in both non-HR and HR patient samples. A droplet digital PCR (ddPCR)-based assay measuring seven NB-mRNAs (CRMP1, DBH, DDC, GAP43, ISL1, PHOX2B, and TH mRNAs) was recently developed and exhibited a better prognostic value for HR patient samples than qPCR-based assays. However, it remained to be tested on non-HR patient samples. In the present study, we employed the ddPCR-based assay to study peripheral blood (PB) and bone marrow (BM) samples collected at diagnosis from eight non-HR and eleven HR cases and characterized the expression profiles of NB-mRNAs. The most highly expressed NB-mRNAs in PB and BM differed between non-HR and HR cases, with the CRMP1 mRNA being predominant in non-HR cases and the GAP43 mRNA in HR cases. The levels of NB-mRNAs in PB and BM were 5 to 1000 times lower in non-HR cases than in HR cases. The PB to BM ratio of NB-mRNAs was 10 to 100 times higher in non-HR cases compared to HR cases. The present case series suggests that non-HR and HR NB patients have the distinct expression profiles of NB-mRNAs in their PB and BM.
Collapse
Affiliation(s)
- Naoko Nakatani
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (N.N.); (T.F.); (S.I.); (A.N.); (A.T.); (N.Y.); (K.N.)
| | - Kaung Htet Nay Win
- Department of Public Health, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan; (K.H.N.W.); (C.Y.M.)
| | - Cho Yee Mon
- Department of Public Health, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan; (K.H.N.W.); (C.Y.M.)
| | - Tomoko Fujikawa
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (N.N.); (T.F.); (S.I.); (A.N.); (A.T.); (N.Y.); (K.N.)
| | - Suguru Uemura
- Department of Hematology/Oncology, Kobe Children’s Hospital, Kobe 650-0047, Japan; (S.U.); (A.S.); (T.I.); (T.M.); (D.H.); (Y.K.); (N.N.)
| | - Atsuro Saito
- Department of Hematology/Oncology, Kobe Children’s Hospital, Kobe 650-0047, Japan; (S.U.); (A.S.); (T.I.); (T.M.); (D.H.); (Y.K.); (N.N.)
| | - Toshiaki Ishida
- Department of Hematology/Oncology, Kobe Children’s Hospital, Kobe 650-0047, Japan; (S.U.); (A.S.); (T.I.); (T.M.); (D.H.); (Y.K.); (N.N.)
| | - Takeshi Mori
- Department of Hematology/Oncology, Kobe Children’s Hospital, Kobe 650-0047, Japan; (S.U.); (A.S.); (T.I.); (T.M.); (D.H.); (Y.K.); (N.N.)
| | - Daiichiro Hasegawa
- Department of Hematology/Oncology, Kobe Children’s Hospital, Kobe 650-0047, Japan; (S.U.); (A.S.); (T.I.); (T.M.); (D.H.); (Y.K.); (N.N.)
| | - Yoshiyuki Kosaka
- Department of Hematology/Oncology, Kobe Children’s Hospital, Kobe 650-0047, Japan; (S.U.); (A.S.); (T.I.); (T.M.); (D.H.); (Y.K.); (N.N.)
| | - Shotaro Inoue
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (N.N.); (T.F.); (S.I.); (A.N.); (A.T.); (N.Y.); (K.N.)
| | - Akihiro Nishimura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (N.N.); (T.F.); (S.I.); (A.N.); (A.T.); (N.Y.); (K.N.)
| | - Nanako Nino
- Department of Hematology/Oncology, Kobe Children’s Hospital, Kobe 650-0047, Japan; (S.U.); (A.S.); (T.I.); (T.M.); (D.H.); (Y.K.); (N.N.)
| | - Akihiro Tamura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (N.N.); (T.F.); (S.I.); (A.N.); (A.T.); (N.Y.); (K.N.)
| | - Nobuyuki Yamamoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (N.N.); (T.F.); (S.I.); (A.N.); (A.T.); (N.Y.); (K.N.)
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (N.N.); (T.F.); (S.I.); (A.N.); (A.T.); (N.Y.); (K.N.)
| | - Noriyuki Nishimura
- Department of Public Health, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan; (K.H.N.W.); (C.Y.M.)
| |
Collapse
|
6
|
Nishimura N, Ishida T, Yokota I, Matsumoto K, Shichino H, Fujisaki H, Sarashina T, Kamijo T, Takimoto T, Iehara T, Tajiri T, on behalf of the JCCG Neuroblastoma Committee. Minimal Residual Disease Detected by the 7NB-mRNAs ddPCR Assay Is Associated with Disease Progression in High-Risk Neuroblastoma Patients: A Prospective Multicenter Observational Study in Japan. BIOLOGY 2023; 12:1350. [PMID: 37887060 PMCID: PMC10604505 DOI: 10.3390/biology12101350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
High-risk neuroblastoma (HR-NB) patients remain far from obtaining optimal outcomes, with more than 50% relapse/regrowth rate despite current intensive multimodal therapy. This originated from the activation/proliferation of chemoresistant minimal residual disease (MRD). MRD with a significant prognostic was reported by several quantitative PCR (qPCR) or droplet digital PCR (ddPCR) assays quantitating different sets of NB-associated mRNAs (NB-mRNAs). The 7NB-mRNAs ddPCR assay quantitating CRMP1, DBH, DDC, GAP43, ISL1, PHOX2B, and TH mRNAs was reported to outperform other qPCR assays by a retrospective in-house observational study. In the present study, the Japan Children's Cancer Group (JCCG) Neuroblastoma Committee conducted a prospective multicenter observational study aimed at evaluating a prognostic value of MRD in bone marrow (BM-MRD) and peripheral blood (PB-MRD) detected by 7NB-mRNAs ddPCR assay. Between August 2018 and August 2022, 7 HR-NB patients who registered for JCCG clinical trials (JN-H-11 and JN-H-15) were enrolled. A total of 19 BM and 19 PB samples were collected, and 4/15 BM and 4/15 PB samples were classified as progressive disease (PD)/non-PD samples. BM-MRD and PB-MRD estimated area under curve (AUC) of 0.767 and 0.800 with a significant accuracy (AUC > 0.7). The present study validated a prognostic value of BM-MRD obtained by a previous study (AUC 0.723) and revealed the significant accuracy of PB-MRD as well as BM-MRD.
Collapse
Affiliation(s)
- Noriyuki Nishimura
- Department of Public Health, Graduate School of Health Science, Kobe University, Kobe 654-0142, Japan
| | - Toshiaki Ishida
- Department of Hematology/Oncology, Kobe Children’s Hospital, Kobe 650-0047, Japan;
| | - Isao Yokota
- Department of Biostatistics, Faculty of Medicine, Hokkaido University, Sapporo 060-0808, Japan;
| | - Kimikazu Matsumoto
- Children’s Cancer Center, National Center for Child Health and Development, Tokyo 157-8535, Japan;
| | - Hiroyuki Shichino
- Department of Pediatrics, National Center for Global Health and Medicine, Tokyo 162-8655, Japan;
| | - Hiroyuki Fujisaki
- Department of Pediatric Hematology/Oncology, Osaka City General Hospital, Osaka 534-0021, Japan;
| | - Takeo Sarashina
- Department of Pediatrics, Asahikawa Medical University, Asahikawa, 078-8510, Japan;
| | - Takehiko Kamijo
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama 362-0806, Japan;
| | - Tetsuya Takimoto
- Department of Childhood Cancer Data Management, National Center for Child Health and Development, Tokyo 157-8535, Japan;
| | - Tomoko Iehara
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan;
| | - Tatsuro Tajiri
- Department of Pediatric Surgery, Faculty of Medical Sciences, Kyushu University of Medicine, Fukuoka 812-8582, Japan;
| | | |
Collapse
|
7
|
Gelineau NU, van Barneveld A, Samim A, Van Zogchel L, Lak N, Tas ML, Matser Y, Mavinkurve-Groothuis AMC, van Grotel M, Zsiros J, van Eijkelenburg NKA, Knops RRG, van Ewijk R, Langenberg KPS, Krijger RD, Hiemcke-Jiwa LS, Van Paemel R, Cornelli L, De Preter K, De Wilde B, Van Der Schoot E, Tytgat G. Case series on clinical applications of liquid biopsy in pediatric solid tumors: towards improved diagnostics and disease monitoring. Front Oncol 2023; 13:1209150. [PMID: 37664065 PMCID: PMC10473251 DOI: 10.3389/fonc.2023.1209150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Background and aims Solid tumors account for about 30% of all pediatric cancers. The diagnosis is typically based on histological and molecular analysis of a primary tumor biopsy. Liquid biopsies carry several advantages over conventional tissue biopsy. However, their use for genomic analysis and response monitoring of pediatric solid tumors is still in experimental stages and mostly performed retrospectively without direct impact on patient management. In this case series we discuss six clinical cases of children with a solid tumor for whom a liquid biopsy assay was performed and demonstrate the potential of liquid biopsy for future clinical decision making. Methods We performed quantitative real-time PCR (RT-qPCR), droplet digital PCR (ddPCR) or reduced representation bisulphite sequencing of cell-free DNA (cfRRBS) on liquid biopsies collected from six pediatric patients with a solid tumor treated between 2017 and 2023 at the Princess Máxima Center for Pediatric Oncology in the Netherlands. Results were used to aid in clinical decision making by contribution to establish a diagnosis, by prognostication and response to therapy monitoring. Results In three patients cfRRBS helped to establish the diagnosis of a rhabdomyosarcoma, an Ewing sarcoma and a neuroblastoma (case 1-3). In two patients, liquid biopsies were used for prognostication, by MYCN ddPCR in a patient with neuroblastoma and by RT-qPCR testing rhabdomyosarcoma-specific mRNA in bone marrow of a patient with a rhabdomyosarcoma (case 4 and 5). In case 6, mRNA testing demonstrated disease progression and assisted clinical decision making. Conclusion This case series illustrates the value of liquid biopsy. We further demonstrate and recommend the use of liquid biopsies to be used in conjunction with conventional methods for the determination of metastatic status, prognostication and monitoring of treatment response in patients with pediatric solid tumors.
Collapse
Affiliation(s)
- Nina U. Gelineau
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
| | | | - Atia Samim
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
| | - Lieke Van Zogchel
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
| | - Nathalie Lak
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
| | - Michelle L. Tas
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
| | - Yvette Matser
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
| | | | - Martine van Grotel
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
| | - Jószef Zsiros
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
| | | | - Rutger R. G. Knops
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
| | - Roelof van Ewijk
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
| | | | - Ronald De Krijger
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Laura S. Hiemcke-Jiwa
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ruben Van Paemel
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Ghent University, Ghent, Belgium
- Research Institute, Ghent University, Ghent, East Flanders, Belgium
| | - Lotte Cornelli
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- VIB-UGent Center for Medical Biotechnology, Gent, Belgium
| | - Katleen De Preter
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Research Institute, Ghent University, Ghent, East Flanders, Belgium
- VIB-UGent Center for Medical Biotechnology, Gent, Belgium
| | - Bram De Wilde
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Ghent University, Ghent, Belgium
- Research Institute, Ghent University, Ghent, East Flanders, Belgium
| | - Ellen Van Der Schoot
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
| | - Godelieve Tytgat
- Princess Máxima Center for Pediatric Oncology Research, Utrecht, Netherlands
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
| |
Collapse
|
8
|
Nino N, Ishida T, Nakatani N, Lin KS, Win KHN, Mon CY, Nishimura A, Inoue S, Tamura A, Yamamoto N, Uemura S, Saito A, Mori T, Hasegawa D, Kosaka Y, Nozu K, Nishimura N. Minimal residual disease detected by droplet digital PCR in peripheral blood stem cell grafts has a prognostic impact on high-risk neuroblastoma patients. Heliyon 2022; 8:e10978. [PMID: 36276741 PMCID: PMC9578974 DOI: 10.1016/j.heliyon.2022.e10978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
More than half of high-risk neuroblastoma (NB) patients have experienced relapse due to the activation of chemoresistant minimal residual disease (MRD) even though they are treated by high-dose chemotherapy with autologous peripheral blood stem cell (PBSC) transplantation. Although MRD in high-risk NB patients can be evaluated by quantitative PCR with several sets of neuroblastoma-associated mRNAs (NB-mRNAs), the prognostic significance of MRD in PBSC grafts (PBSC-MRD) is unclear. In the present study, we collected 20 PBSC grafts from 20 high-risk NB patients and evaluated PBSC-MRD detected by droplet digital PCR (ddPCR) with 7NB-mRNAs (CRMP1, DBH, DDC, GAP43, ISL1, PHOX2B, and TH mRNA). PBSC-MRD in 11 relapsed patients was significantly higher than that in 9 non-relapsed patients. Patients with a higher PBSC-MRD had a lower 3-year event-free survival (P = 0.0148). The present study suggests that PBSC-MRD detected by ddPCR with 7NB-mRNAs has a prognostic impact on high-risk NB patients.
Collapse
Affiliation(s)
- Nanako Nino
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Toshiaki Ishida
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Naoko Nakatani
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kyaw San Lin
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kaung Htet Nay Win
- Department of Public Health, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Cho Yee Mon
- Department of Public Health, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Akihiro Nishimura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shotaro Inoue
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akihiro Tamura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Nobuyuki Yamamoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Suguru Uemura
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Atsuro Saito
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Takeshi Mori
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriyuki Nishimura
- Department of Public Health, Kobe University Graduate School of Health Science, Kobe, Japan,Corresponding author.
| |
Collapse
|
9
|
Fan H, Xing T, Hong H, Duan C, Zhao W, Zhao Q, Wang X, Huang C, Zhu S, Jin M, Su Y, Gao C, Ma X. The expression of PHOX2B in bone marrow and peripheral blood predicts adverse clinical outcome in non-high-risk neuroblastoma. Pediatr Hematol Oncol 2022; 39:343-356. [PMID: 34752187 DOI: 10.1080/08880018.2021.1995090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Paired-like homeobox 2B (PHOX2B) is a highly sensitive and specific biomarker for diagnosing neuroblastoma, as well as detecting minimal residual disease in neuroblastoma. The clinical significance of PHOX2B expression in bone marrow (BM) and peripheral blood (PB) samples of newly diagnosed patients with very low-, low- and intermediate-risk neuroblastoma remains unknown, to the best of our knowledge. The expression level of PHOX2B in paired BM and PB samples of patients with newly diagnosed neuroblastoma was validated using reverse transcription-quantitative polymerase chain reaction (RTqPCR). Among the 132 patients, 26 exhibited a positive PHOX2B expression BM (19.7%) and 11 in PB (8.3%) samples. PHOX2B was highly expressed in BM and PB samples from patients aged <18 months, with International Neuroblastoma Risk Group Staging System stages M and MS, 1p loss of heterozygosity, and high levels of lactate dehydrogenase, serum ferritin and neuron-specific enolase (p < 0.05). In all eligible patients, the 2-year event-free survival (EFS) and overall survival (OS) rates were 94.7 ± 2.0% and 97.7 ± 1.3%, respectively. However, the 2-year EFS rates were significantly decreased to 76.9 ± 8.3% and 63.6 ± 14.5% in patients with a positive PHOX2B expression in BM and PB samples, respectively (p < 0.05). Similarly, the 2-year OS rates were also decreased to 88.5 ± 6.3% and 81.8 ± 11.6% in patients with a positive PHOX2B expression in BM and PB samples, respectively (p < 0.05). In conclusion, a positive PHOX2B expression in BM and PB samples at diagnosis had a strong adverse prognostic effect on patients with non-high-risk neuroblastoma.
Collapse
Affiliation(s)
- Hongjun Fan
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Ocology, Key Laboratory of Major Diseases in Children, Ministry of Education, 56 Nan Lishi Road, Xicheng District, Beijing, China
| | - Tianyu Xing
- Hematologic Disease Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Huimin Hong
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Ocology, Key Laboratory of Major Diseases in Children, Ministry of Education, 56 Nan Lishi Road, Xicheng District, Beijing, China
| | - Chao Duan
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Ocology, Key Laboratory of Major Diseases in Children, Ministry of Education, 56 Nan Lishi Road, Xicheng District, Beijing, China
| | - Wen Zhao
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Ocology, Key Laboratory of Major Diseases in Children, Ministry of Education, 56 Nan Lishi Road, Xicheng District, Beijing, China
| | - Qian Zhao
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Ocology, Key Laboratory of Major Diseases in Children, Ministry of Education, 56 Nan Lishi Road, Xicheng District, Beijing, China
| | - Xisi Wang
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Ocology, Key Laboratory of Major Diseases in Children, Ministry of Education, 56 Nan Lishi Road, Xicheng District, Beijing, China
| | - Cheng Huang
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Ocology, Key Laboratory of Major Diseases in Children, Ministry of Education, 56 Nan Lishi Road, Xicheng District, Beijing, China
| | - Shuai Zhu
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Ocology, Key Laboratory of Major Diseases in Children, Ministry of Education, 56 Nan Lishi Road, Xicheng District, Beijing, China
| | - Mei Jin
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Ocology, Key Laboratory of Major Diseases in Children, Ministry of Education, 56 Nan Lishi Road, Xicheng District, Beijing, China
| | - Yan Su
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Ocology, Key Laboratory of Major Diseases in Children, Ministry of Education, 56 Nan Lishi Road, Xicheng District, Beijing, China
| | - Chao Gao
- Hematologic Disease Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Xiaoli Ma
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Ocology, Key Laboratory of Major Diseases in Children, Ministry of Education, 56 Nan Lishi Road, Xicheng District, Beijing, China
| |
Collapse
|
10
|
van Zogchel LMJ, Lak NSM, Verhagen OJHM, Tissoudali A, Gussmalla Nuru M, Gelineau NU, Zappeij-Kannengieter L, Javadi A, Zijtregtop EAM, Merks JHM, van den Heuvel-Eibrink M, Schouten-van Meeteren AYN, Stutterheim J, van der Schoot CE, Tytgat GAM. Novel Circulating Hypermethylated RASSF1A ddPCR for Liquid Biopsies in Patients With Pediatric Solid Tumors. JCO Precis Oncol 2021; 5:PO.21.00130. [PMID: 34820594 PMCID: PMC8608265 DOI: 10.1200/po.21.00130] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/06/2021] [Accepted: 10/08/2021] [Indexed: 12/19/2022] Open
Abstract
Liquid biopsies can be used to investigate tumor-derived DNA, circulating in the cell-free DNA (cfDNA) pool in blood. We aimed to develop a droplet digital polymerase chain reaction (ddPCR) assay detecting hypermethylation of tumor suppressor gene RASSF1A as a simple standard test to detect various pediatric tumor types in small volume blood samples and to evaluate this test for monitoring treatment response of patients with high-risk neuroblastoma. The circulating tumor marker hypermethylated RASSF1A can be detected in the plasma of pediatric patients with solid tumors![]()
Collapse
Affiliation(s)
- Lieke M J van Zogchel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Nathalie S M Lak
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Onno J H M Verhagen
- Department of Immunocytology, Sanquin Diagnostic Services, Amsterdam, the Netherlands
| | - Ahmed Tissoudali
- Department of Immunohematology Diagnostics, Sanquin Diagnostic Services, Amsterdam, the Netherlands
| | - Mohammed Gussmalla Nuru
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Nina U Gelineau
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Lily Zappeij-Kannengieter
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Department of Immunocytology, Sanquin Diagnostic Services, Amsterdam, the Netherlands
| | - Ahmad Javadi
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Eline A M Zijtregtop
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Pediatric Oncology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | | | | | | | | | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | | |
Collapse
|
11
|
Uemura S, Lin KS, Mon Thwin KK, Nakatani N, Ishida T, Yamamoto N, Tamura A, Saito A, Mori T, Hasegawa D, Kosaka Y, Nino N, Nagano C, Takafuji S, Iijima K, Nishimura N. Limited correlation between tumor markers and minimal residual disease detected by seven neuroblastoma-associated mRNAs in high-risk neuroblastoma patients. Mol Clin Oncol 2021; 15:137. [PMID: 34055352 DOI: 10.3892/mco.2021.2299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/18/2020] [Indexed: 01/23/2023] Open
Abstract
Vanillylmandelic acid (VMA), homovanillic acid (HVA), neuron-specific enolase (NSE) and lactate dehydrogenase (LDH) are classical tumor markers and are used as standard clinical evaluations for patients with neuroblastoma (NB). Minimal residual disease (MRD) can be monitored by quantifying several sets of NB-associated mRNAs in the bone marrow (BM) and peripheral blood (PB) of patients with NB. Although MRD in BM and PB has been revealed to be a strong prognostic factor that is independent of standard clinical evaluations, its interrelation with tumor markers remains uncharacterized. The present study determined the levels of tumor markers (VMA, HVA, NSE and LDH) and MRD (BM-MRD and PB-MRD) in 133 pairs of concurrently collected BM, PB and urine samples from 19 patients with high-risk NB. The patients were evaluated during the entire course of treatment, which included 10 diagnoses, 32 treatments, 36 post-treatment, 9 relapses and 46 post-relapse sample pairs. The level of BM-MRD and PB-MRD was determined by quantifying 7 NB-mRNAs (collapsin response mediator protein 1, dopamine beta-hydroxylase, dopa decarboxylase, growth-associated protein 43, ISL LIM homeobox 1, pairedlike homeobox 2b and tyrosine hydroxylase) using droplet digital PCR. In overall sample pairs, tumor markers (VMA, HVA, NSE and LDH) demonstrated weak but significant correlations (P<0.011) with BM-MRD and PB-MRD. In subgroups according to each patient evaluation, the degree of correlation between tumor markers and MRD became stronger in patients with adrenal gland tumors, BM metastasis at diagnosis and relapse/regrowth compared with overall sample pairs. In contrast, tumor markers demonstrated variable correlations with MRD in subgroups according to each sample evaluation (BM infiltration at sampling, collection time point and disease status). The results suggested that tumor markers may demonstrate limited correlation with MRD in patients with high-risk NB.
Collapse
Affiliation(s)
- Suguru Uemura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Kyaw San Lin
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Khin Kyae Mon Thwin
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Naoko Nakatani
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Toshiaki Ishida
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Hyogo 650-0047, Japan
| | - Nobuyuki Yamamoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Akihiro Tamura
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Hyogo 650-0047, Japan
| | - Atsuro Saito
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Hyogo 650-0047, Japan
| | - Takeshi Mori
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Hyogo 650-0047, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Hyogo 650-0047, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Hyogo 650-0047, Japan
| | - Nanako Nino
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - China Nagano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Satoru Takafuji
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Noriyuki Nishimura
- Department of Public Health, Kobe University Graduate School of Health Science, Kobe, Hyogo 654-0142, Japan
| |
Collapse
|
12
|
Lin KS, Uemura S, Thwin KKM, Nakatani N, Ishida T, Yamamoto N, Tamura A, Saito A, Mori T, Hasegawa D, Kosaka Y, Nino N, Nagano C, Takafuji S, Iijima K, Nishimura N. Minimal residual disease in high-risk neuroblastoma shows a dynamic and disease burden-dependent correlation between bone marrow and peripheral blood. Transl Oncol 2021; 14:101019. [PMID: 33993097 PMCID: PMC8138775 DOI: 10.1016/j.tranon.2021.101019] [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: 08/31/2020] [Revised: 10/20/2020] [Accepted: 01/12/2021] [Indexed: 11/04/2022] Open
Abstract
Bone marrow is the most frequent site of metastasis and relapse for neuroblastoma. Minimal residual disease has been identified in bone marrow and peripheral blood (BM-MRD and PB-MRD) by quantifying several sets of neuroblastoma-associated mRNAs. BM-MRD has significant prognostic information for high-risk neuroblastoma. BM-MRD and PB-MRD show a dynamic and disease burden-dependent correlation in high-risk neuroblastoma.
Neuroblastoma (NB) is the most common extracranial solid tumor in children and originates from sympathoadrenal or Schwann cell precursors derived from neural crest. These neural crest derivatives also constitute the hematopoietic and mesenchymal stem cells in bone marrow (BM) that is the most frequent site of NB metastasis and relapse. In NB patients, NB cells have been pathologically detected in BM and peripheral blood (PB), and minimal residual disease (MRD) in BM and PB (BM-MRD and PB-MRD) can be monitored by quantitating several sets of NB-associated mRNAs (NB-mRNAs). Although previous studies have shown varying degrees of correlation between BM-MRD and PB-MRD, the underlying factors and/or mechanisms remains unknown. In the present study, we determined the levels of BM-MRD and PB-MRD by quantitating seven NB-mRNAs in 133 pairs of concurrently collected BM and PB samples from 19 high-risk NB patients with clinical disease evaluation, and examined their correlation in overall and subgroups of sample pairs. The levels of BM-MRD and PB-MRD were moderately (r = 0.418, p < 0.001) correlated with each other in overall sample pairs. The correlation became strong (r = 0.725, p < 0.001), weak (r = 0.284, p = 0.008), and insignificant (p = 0.194) in progression, stable, and remission subgroups of sample pairs, respectively. It also became stronger in subgroups of sample pairs with poor treatment responses and poor prognostic factors. Present study suggests that MRD in high-risk NB shows a dynamic and disease burden-dependent correlation between BM and PB.
Collapse
Affiliation(s)
- Kyaw San Lin
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Suguru Uemura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Khin Kyae Mon Thwin
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoko Nakatani
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Toshiaki Ishida
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Nobuyuki Yamamoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akihiro Tamura
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Atsuro Saito
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Takeshi Mori
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Nanako Nino
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - China Nagano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoru Takafuji
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriyuki Nishimura
- Department of Public Health, Kobe University Graduate School of Health Science, Kobe, Japan.
| |
Collapse
|
13
|
Hochheuser C, Windt LJ, Kunze NY, de Vos DL, Tytgat GA, Voermans C, Timmerman I. Mesenchymal Stromal Cells in Neuroblastoma: Exploring Crosstalk and Therapeutic Implications. Stem Cells Dev 2021; 30:59-78. [PMID: 33287630 PMCID: PMC7826431 DOI: 10.1089/scd.2020.0142] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma (NB) is the second most common solid cancer in childhood, accounting for 15% of cancer-related deaths in children. In high-risk NB patients, the majority suffers from metastasis. Despite intensive multimodal treatment, long-term survival remains <40%. The bone marrow (BM) is among the most common sites of distant metastasis in patients with high-risk NB. In this environment, small populations of tumor cells can persist after treatment (minimal residual disease) and induce relapse. Therapy resistance of these residual tumor cells in BM remains a major obstacle for the cure of NB. A detailed understanding of the microenvironment and its role in tumor progression is of utmost importance for improving the treatment efficiency of NB. In BM, mesenchymal stromal cells (MSCs) constitute an important part of the microenvironment, where they support hematopoiesis and modulate immune responses. Their role in tumor progression is not completely understood, especially for NB. Although MSCs have been found to promote epithelial-mesenchymal transition, tumor growth, and metastasis and to induce chemoresistance, some reports point toward a tumor-suppressive effect of MSCs. In this review, we aim to compile current knowledge about the role of MSCs in NB development and progression. We evaluate arguments that depict tumor-supportive versus -suppressive properties of MSCs in the context of NB and give an overview of factors involved in MSC-NB crosstalk. A focus lies on the BM as a metastatic niche, since that is the predominant site for NB metastasis and relapse. Finally, we will present opportunities and challenges for therapeutic targeting of MSCs in the BM microenvironment.
Collapse
Affiliation(s)
- Caroline Hochheuser
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Laurens J. Windt
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Nina Y. Kunze
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Dieuwke L. de Vos
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Carlijn Voermans
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Ilse Timmerman
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
14
|
Chen X, Wu H, Feng J, Li Y, Lv J, Shi W, Fan W, Xiao L, Sun D, Jiang M, Shi M. Transcriptome profiling unveils GAP43 regulates ABC transporters and EIF2 signaling in colorectal cancer cells. BMC Cancer 2021; 21:24. [PMID: 33402155 PMCID: PMC7786480 DOI: 10.1186/s12885-020-07728-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/11/2020] [Indexed: 02/08/2023] Open
Abstract
Background The growth- and plasticity-associated protein-43 (GAP43) is biasedly expressed in indigestive system and nervous system. Recent study has shown that GAP43 is responsible for the development of neuronal growth and axonal regeneration in normal nervous tissue, while serves as a specific biomarker of relapsed or refractory neuroblastoma. However, its expression pattern and function in digestive system cancer remains to be clarified. Methods In this study, we examined the GAP43 status with qRT-PCR and bisulfite genomic sequencing in colorectal cancer (CRC). We investigated the effect of overexpressed GAP43 in CRC cells with RNA-seq. The RNA-seq data was analyzed with DAVID and IPA. Results GAP43 was downregulated in CRC compared to the adjacent tissues. DNA methylase inhibitor 5-Aza-CdR treatment could significantly induce GAP43, indicated that the silencing of GAP43 gene in CRC is closely related to DNA methylation. Bisulfite genomic sequencing confirmed the promoter methylation of GAP43 in CRC. To explore the transcriptional alterations by overexpressed GAP43 in CRC, we performed RNA-seq and found that upregulated genes were significantly enriched in the signaling pathways of ABC transporters and ECM-receptor interaction, while downregulated genes were significantly enriched in Ribosome signaling pathway. Further Ingenuity Pathway Analysis (IPA) showed that EIF2 signaling pathway was significantly repressed by overexpression of GAP43. Conclusion Our findings provide a novel mechanistic insight of GAP43 in CRC. Transcriptome profiling of overexpressed GAP43 in CRC uncovered the functional roles of GAP43 in the development of human CRC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-020-07728-x.
Collapse
Affiliation(s)
- Xi Chen
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Hongjin Wu
- Department of Clinical Laboratory, Hangzhou Cancer Hospital, Hangzhou,, 320000, Zhejiang, China.,The NHC Key Laboratory of Drug Addition Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, 650000, China
| | - Jia Feng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Ying Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Jiao Lv
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Weikai Shi
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Weiwei Fan
- Department of Infectious and Medicine, Heilongjiang Provincial Hospital, Harbin, 150036, China
| | - Li Xiao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Danmeng Sun
- Department of Pediatrics, Data People's hospital, Shenmu, 719301, China
| | - Mingfeng Jiang
- Department of Clinical Laboratory, Hangzhou Cancer Hospital, Hangzhou,, 320000, Zhejiang, China.
| | - Ming Shi
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
15
|
van Zogchel LMJ, Zappeij-Kannegieter L, Javadi A, Lugtigheid M, Gelineau NU, Lak NSM, Zwijnenburg DA, Koster J, Stutterheim J, van der Schoot CE, Tytgat GAM. Specific and Sensitive Detection of Neuroblastoma mRNA Markers by Multiplex RT-qPCR. Cancers (Basel) 2021; 13:E150. [PMID: 33466359 PMCID: PMC7796198 DOI: 10.3390/cancers13010150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/27/2020] [Accepted: 12/31/2020] [Indexed: 12/16/2022] Open
Abstract
mRNA RT-qPCR is shown to be a very sensitive technique to detect minimal residual disease (MRD) in patients with neuroblastoma. Multiple mRNA markers are known to detect heterogeneous neuroblastoma cells in bone marrow (BM) or blood from patients. However, the limited volumes of BM and blood available can hamper the detection of multiple markers. To make optimal use of these samples, we developed a multiplex RT-qPCR for the detection of MRD in neuroblastoma. GUSB and PHOX2B were tested as single markers. The adrenergic markers TH, GAP43, CHRNA3 and DBH and mesenchymal markers POSTN, PRRX1 and FMO3 were tested in multiplex. Using control blood and BM, we established new thresholds for positivity. Comparison of multiplex and singleplex RT-qPCR results from 21 blood and 24 BM samples from neuroblastoma patients demonstrated a comparable sensitivity. With this multiplex RT-qPCR, we are able to test seven different neuroblastoma mRNA markers, which overcomes tumor heterogeneity and improves sensitivity of MRD detection, even in those samples of low RNA quantity. With resources and time being saved, reduction in sample volume and consumables can assist in the introduction of MRD by RT-qPCR into clinical practice.
Collapse
Affiliation(s)
- Lieke M. J. van Zogchel
- Princess Maxima Center for Pediatric Oncology, Department of Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (L.M.J.v.Z.); (N.U.G.); (N.S.M.L.); (J.S.)
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands; (L.Z.-K.); (A.J.); (M.L.); (C.E.v.d.S.)
| | - Lily Zappeij-Kannegieter
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands; (L.Z.-K.); (A.J.); (M.L.); (C.E.v.d.S.)
| | - Ahmad Javadi
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands; (L.Z.-K.); (A.J.); (M.L.); (C.E.v.d.S.)
| | - Marjolein Lugtigheid
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands; (L.Z.-K.); (A.J.); (M.L.); (C.E.v.d.S.)
| | - Nina U. Gelineau
- Princess Maxima Center for Pediatric Oncology, Department of Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (L.M.J.v.Z.); (N.U.G.); (N.S.M.L.); (J.S.)
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands; (L.Z.-K.); (A.J.); (M.L.); (C.E.v.d.S.)
| | - Nathalie S. M. Lak
- Princess Maxima Center for Pediatric Oncology, Department of Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (L.M.J.v.Z.); (N.U.G.); (N.S.M.L.); (J.S.)
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands; (L.Z.-K.); (A.J.); (M.L.); (C.E.v.d.S.)
| | - Danny A. Zwijnenburg
- Academic Medical Center, Department of Oncogenomics, 1105 AZ Amsterdam, The Netherlands; (D.A.Z.); (J.K.)
| | - Jan Koster
- Academic Medical Center, Department of Oncogenomics, 1105 AZ Amsterdam, The Netherlands; (D.A.Z.); (J.K.)
| | - Janine Stutterheim
- Princess Maxima Center for Pediatric Oncology, Department of Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (L.M.J.v.Z.); (N.U.G.); (N.S.M.L.); (J.S.)
| | - C. Ellen van der Schoot
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands; (L.Z.-K.); (A.J.); (M.L.); (C.E.v.d.S.)
| | - Godelieve A. M. Tytgat
- Princess Maxima Center for Pediatric Oncology, Department of Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (L.M.J.v.Z.); (N.U.G.); (N.S.M.L.); (J.S.)
| |
Collapse
|
16
|
van Zogchel LMJ, van Wezel EM, van Wijk J, Stutterheim J, Bruins WSC, Zappeij-Kannegieter L, Slager TJE, Schumacher-Kuckelkorn R, Verly IRN, van der Schoot CE, Tytgat GAM. Hypermethylated RASSF1A as Circulating Tumor DNA Marker for Disease Monitoring in Neuroblastoma. JCO Precis Oncol 2020; 4:1900261. [PMID: 32923888 PMCID: PMC7446415 DOI: 10.1200/po.19.00261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2020] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Circulating tumor DNA (ctDNA) has been used for disease monitoring in several types of cancer. The aim of our study was to investigate whether ctDNA can be used for response monitoring in neuroblastoma. METHODS One hundred forty-nine plasma samples from 56 patients were analyzed by quantitative polymerase chain reaction (qPCR) for total cell free DNA (cfDNA; albumin and β-actin) and ctDNA (hypermethylated RASSF1A). ctDNA results were compared with mRNA-based minimal residual disease (qPCR) in bone marrow (BM) and blood and clinical patient characteristics. RESULTS ctDNA was detected at diagnosis in all patients with high-risk and stage M neuroblastoma and in 3 of 7 patients with localized disease. The levels of ctDNA were highest at diagnosis, decreased during induction therapy, and not detected before or after autologous stem-cell transplantation. At relapse, the amount of ctDNA was comparable to levels at diagnosis. There was an association between ctDNA and blood or BM mRNA, with concordant results when tumor burden was high or no tumor was detected. The discrepancies indicated either low-level BM infiltration (ctDNA negative/mRNA positive) or primary tumor/soft tissue lesions with no BM involvement (ctDNA positive/mRNA negative). CONCLUSION ctDNA can be used for monitoring disease in patients with neuroblastoma. In high-risk patients and all patients with stage M at diagnosis, ctDNA is present. Our data indicate that at low tumor load, testing of both ctDNA and mRNA increases the sensitivity of molecular disease monitoring. It is likely that ctDNA can originate from both primary tumor and metastases and may be of special interest for disease monitoring in patients who experience relapse in other organs than BM.
Collapse
Affiliation(s)
- Lieke M J van Zogchel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Sanquin Research, Department of Experimental Immunohematology, and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Esther M van Wezel
- Sanquin Research, Department of Experimental Immunohematology, and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Jalenka van Wijk
- Sanquin Research, Department of Experimental Immunohematology, and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | | | - Wassilis S C Bruins
- Sanquin Research, Department of Experimental Immunohematology, and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Lily Zappeij-Kannegieter
- Sanquin Research, Department of Experimental Immunohematology, and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Tirza J E Slager
- Department of Pediatric Oncology, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | | | - Iedan R N Verly
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - C Ellen van der Schoot
- Sanquin Research, Department of Experimental Immunohematology, and Landsteiner Laboratory, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | | |
Collapse
|
17
|
Thwin KK, Ishida T, Uemura S, Yamamoto N, Lin KS, Tamura A, Kozaki A, Saito A, Kishimoto K, Mori T, Hasegawa D, Kosaka Y, Nino N, Takafuji S, Iijima K, Nishimura N. Level of Seven Neuroblastoma-Associated mRNAs Detected by Droplet Digital PCR Is Associated with Tumor Relapse/Regrowth of High-Risk Neuroblastoma Patients. J Mol Diagn 2020; 22:236-246. [DOI: 10.1016/j.jmoldx.2019.10.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 07/25/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023] Open
|
18
|
The pitfalls and promise of liquid biopsies for diagnosing and treating solid tumors in children: a review. Eur J Pediatr 2020; 179:191-202. [PMID: 31897843 PMCID: PMC6971142 DOI: 10.1007/s00431-019-03545-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/29/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022]
Abstract
Cell-free DNA profiling using patient blood is emerging as a non-invasive complementary technique for cancer genomic characterization. Since these liquid biopsies will soon be integrated into clinical trial protocols for pediatric cancer treatment, clinicians should be informed about potential applications and advantages but also weaknesses and potential pitfalls. Small retrospective studies comparing genetic alterations detected in liquid biopsies with tumor biopsies for pediatric solid tumor types are encouraging. Molecular detection of tumor markers in cell-free DNA could be used for earlier therapy response monitoring and residual disease detection as well as enabling detection of pathognomonic and therapeutically relevant genomic alterations.Conclusion: Existing analyses of liquid biopsies from children with solid tumors increasingly suggest a potential relevance for molecular diagnostics, prognostic assessment, and therapeutic decision-making. Gaps remain in the types of tumors studied and value of detection methods applied. Here we review the current stand of liquid biopsy studies for pediatric solid tumors with a dedicated focus on cell-free DNA analysis. There is legitimate hope that integrating fully validated liquid biopsy-based innovations into the standard of care will advance patient monitoring and personalized treatment of children battling solid cancers.What is Known:• Liquid biopsies are finding their way into routine oncological screening, diagnosis, and disease monitoring in adult cancer types fast.• The most widely adopted source for liquid biopsies is blood although other easily accessible body fluids, such as saliva, pleural effusions, urine, or cerebrospinal fluid (CSF) can also serve as sources for liquid biopsiesWhat is New:• Retrospective proof-of-concept studies in small cohorts illustrate that liquid biopsies in pediatric solid tumors yield tremendous potential to be used in diagnostics, for therapy response monitoring and in residual disease detection.• Liquid biopsy diagnostics could tackle some long-standing issues in the pediatric oncology field; they can enable accurate genetic diagnostics in previously unbiopsied tumor types like renal tumors or brain stem tumors leading to better treatment strategies.
Collapse
|
19
|
van Wezel EM, van Zogchel LMJ, van Wijk J, Timmerman I, Vo NK, Zappeij-Kannegieter L, deCarolis B, Simon T, van Noesel MM, Molenaar JJ, van Groningen T, Versteeg R, Caron HN, van der Schoot CE, Koster J, van Nes J, Tytgat GAM. Mesenchymal Neuroblastoma Cells Are Undetected by Current mRNA Marker Panels: The Development of a Specific Neuroblastoma Mesenchymal Minimal Residual Disease Panel. JCO Precis Oncol 2019; 3:1800413. [PMID: 34036221 PMCID: PMC8133311 DOI: 10.1200/po.18.00413] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2019] [Indexed: 12/29/2022] Open
Abstract
Patients with neuroblastoma in molecular remission remain at considerable risk for disease recurrence. Studies have found that neuroblastoma tissue contains adrenergic (ADRN) and mesenchymal (MES) cells; the latter express low levels of commonly used markers for minimal residual disease (MRD). We identified MES-specific MRD markers and studied the dynamics of these markers during treatment. PATIENTS AND METHODS Microarray data were used to identify genes differentially expressed between ADRN and MES cell lines. Candidate genes were then studied using real-time quantitative polymerase chain reaction in cell lines and control bone marrow and peripheral blood samples. After selecting a panel of markers, serial bone marrow, peripheral blood, and peripheral blood stem cell samples were obtained from patients with high-risk neuroblastoma and tested for marker expression; survival analyses were also performed. RESULTS PRRX1, POSTN, and FMO3 mRNAs were used as a panel for specifically detecting MES mRNA in patient samples. MES mRNA was detected only rarely in peripheral blood; moreover, the presence of MES mRNA in peripheral blood stem cell samples was associated with low event-free survival and overall survival. Of note, during treatment, serial bone marrow samples obtained from 29 patients revealed a difference in dynamics between MES mRNA markers and ADRN mRNA markers. Furthermore, MES mRNA was detected in a higher percentage of patients with recurrent disease than in those who remained disease free (53% v 32%, respectively; P = .03). CONCLUSION We propose that the markers POSTN and PRRX1, in combination with FMO3, be used for real-time quantitative polymerase chain reaction-based detection of MES neuroblastoma mRNA in patient samples because these markers have a unique pattern during treatment and are more prevalent in patients with poor outcome. Together with existing markers of MRD, these new markers should be investigated further in large prospective studies.
Collapse
Affiliation(s)
- Esther M van Wezel
- Sanquin Research Amsterdam, the Netherlands.,Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Lieke M J van Zogchel
- Sanquin Research Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jalenka van Wijk
- Sanquin Research Amsterdam, the Netherlands.,Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Ilse Timmerman
- Sanquin Research Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | | | | | - Thorsten Simon
- Children's Hospital University of Cologne, Cologne, Germany
| | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Rogier Versteeg
- Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Huib N Caron
- Amsterdam University Medical Center, Amsterdam, the Netherlands
| | | | - Jan Koster
- Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Johan van Nes
- Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Godelieve A M Tytgat
- Amsterdam University Medical Center, Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
20
|
Uemura S, Ishida T, Thwin KKM, Yamamoto N, Tamura A, Kishimoto K, Hasegawa D, Kosaka Y, Nino N, Lin KS, Takafuji S, Mori T, Iijima K, Nishimura N. Dynamics of Minimal Residual Disease in Neuroblastoma Patients. Front Oncol 2019; 9:455. [PMID: 31214500 PMCID: PMC6558004 DOI: 10.3389/fonc.2019.00455] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 05/14/2019] [Indexed: 12/11/2022] Open
Abstract
Neuroblastoma is a common extracranial solid tumor of neural crest (NC) origin that accounts for up to 15% of all pediatric cancer deaths. The disease arises from a transient population of NC cells that undergo an epithelial-mesenchymal transition (EMT) and generate diverse cell-types and tissues. Patients with neuroblastoma are characterized by their extreme heterogeneity ranging from spontaneous regression to malignant progression. More than half of newly diagnosed patients present highly metastatic tumors and are stratified into a high-risk group with dismal outcome. As many as 20% of high-risk patients have residual disease that is refractory or progressive during induction chemotherapy. Although a majority of high-risk patients achieve remission, larger part of those patients has minimal residual disease (MRD) that causes relapse even after additional consolidation therapy. MRD is composed of drug-resistant tumor cells and dynamically presented as cancer stem cells (CSCs) in residual tumors, circulating tumor cells (CTCs) in peripheral blood (PB), and disseminated tumor cells (DTCs) in bone marrow (BM) and other metastatic sites. EMT appears to be a key mechanism for cancer cells to acquire MRD phenotypes and malignant aggressiveness. Due to the restricted availability of residual tumors, PB and BM have been used to isolate and analyze CTCs and DTCs to evaluate MRD in cancer patients. In addition, recent technical advances make it possible to use circulating tumor DNA (ctDNA) shed from tumor cells into PB for MRD evaluation. Because MRD can be detected by tumor-specific antigens, genetic or epigenetic changes, and mRNAs, numerous assays using different methods and samples have been reported to detect MRD in cancer patients. In contrast to the tumor-specific gene-rearrangement-positive acute lymphoblastic leukemia (ALL) and the oncogenic fusion-gene-positive chronic myelogenous leukemia (CML) and several solid tumors, the clinical significance of MRD remains to be established in neuroblastoma. Given the extreme heterogeneity of neuroblastoma, dynamics of MRD in neuroblastoma patients will hold a key to the clinical validation. In this review, we summarize the biology and detection methods of cancer MRD in general and evaluate the available assays and clinical significance of neuroblastoma MRD to clarify its dynamics in neuroblastoma patients.
Collapse
Affiliation(s)
- Suguru Uemura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Toshiaki Ishida
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Khin Kyae Mon Thwin
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Nobuyuki Yamamoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akihiro Tamura
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Kenji Kishimoto
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Nanako Nino
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kyaw San Lin
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoru Takafuji
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takeshi Mori
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriyuki Nishimura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
21
|
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
|
22
|
Peinemann F, van Dalen EC, Enk H, Tytgat GAM, Cochrane Childhood Cancer Group. Anti-GD2 antibody-containing immunotherapy postconsolidation therapy for people with high-risk neuroblastoma treated with autologous haematopoietic stem cell transplantation. Cochrane Database Syst Rev 2019; 4:CD012442. [PMID: 31016728 PMCID: PMC6479178 DOI: 10.1002/14651858.cd012442.pub2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Neuroblastoma is a rare malignant disease that primarily affects children. The tumours mainly develop in the adrenal medullary tissue, and an abdominal mass is the most common presentation. High-risk disease is characterised by metastasis and other primary tumour characteristics resulting in increased risk for an adverse outcome. The GD2 carbohydrate antigen is expressed on the cell surface of neuroblastoma tumour cells and is thus a promising target for anti-GD2 antibody-containing immunotherapy. OBJECTIVES To assess the efficacy of anti-GD2 antibody-containing postconsolidation immunotherapy after high-dose chemotherapy (HDCT) and autologous haematopoietic stem cell transplantation (HSCT) compared to standard therapy after HDCT and autologous HSCT in people with high-risk neuroblastoma. Our primary outcomes were overall survival and treatment-related mortality. Our secondary outcomes were progression-free survival, event-free survival, early toxicity, late non-haematological toxicity, and health-related quality of life. SEARCH METHODS We searched the electronic databases CENTRAL (2018, Issue 9), MEDLINE (PubMed), and Embase (Ovid) on 20 September 2018. We searched trial registries and conference proceedings on 28 October 2018. Further searches included reference lists of recent reviews and relevant articles as well as contacting experts in the field. There were no limits on publication year or language. SELECTION CRITERIA Randomised controlled trials evaluating anti-GD2 antibody-containing immunotherapy after HDCT and autologous HSCT in people with high-risk neuroblastoma. DATA COLLECTION AND ANALYSIS Two review authors independently performed study selection, abstracted data on study and participant characteristics, and assessed risk of bias and GRADE. Any differences were resolved by discussion, with third-party arbitration unnecessary. We performed analyses according to the guidelines of the Cochrane Handbook for Systematic Reviews of Interventions. We used the five GRADE considerations, that is study limitations, consistency of effect, imprecision, indirectness, and publication bias, to judge the quality of the evidence. MAIN RESULTS We identified one randomised controlled trial that included 226 people with high-risk neuroblastoma who were pre-treated with autologous HSCT. The study randomised 113 participants to receive immunotherapy including isotretinoin, granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-2, and ch14.18, a type of anti-GD2 antibody also known as dinutuximab. The study randomised another 113 participants to receive standard therapy including isotretinoin.The results on overall survival favoured the dinutuximab-containing immunotherapy group (hazard ratio (HR) 0.50, 95% confidence interval (CI) 0.31 to 0.80; P = 0.004). The results on event-free survival also favoured the dinutuximab-containing immunotherapy group (HR 0.61, 95% CI 0.41 to 0.92; P = 0.020). Randomised data on adverse events were not reported separately. The study did not report progression-free survival, late non-haematological toxicity, and health-related quality of life as separate endpoints. We graded the quality of the evidence as moderate. AUTHORS' CONCLUSIONS The evidence base favours dinutuximab-containing immunotherapy compared to standard therapy concerning overall survival and event-free survival in people with high-risk neuroblastoma pre-treated with autologous HSCT. Randomised data on adverse events are lacking, therefore more research is needed before definitive conclusions can be made regarding this outcome.
Collapse
Affiliation(s)
- Frank Peinemann
- Children's Hospital, University of ColognePediatric Oncology and HematologyKerpener Str. 62CologneGermany50937
| | - Elvira C van Dalen
- Princess Máxima Center for Pediatric OncologyHeidelberglaan 25UtrechtNetherlands3584 CS
| | - Heike Enk
- c/o Cochrane Childhood CancerAmsterdamNetherlands
| | - Godelieve AM Tytgat
- Princess Máxima Center for Pediatric OncologyHeidelberglaan 25UtrechtNetherlands3584 CS
| | | |
Collapse
|
23
|
Zhang F, Ying L, Jin J, Feng J, Chen K, Huang M, Wu Y, Yu H, Su D. GAP43, a novel metastasis promoter in non-small cell lung cancer. J Transl Med 2018; 16:310. [PMID: 30419922 PMCID: PMC6233536 DOI: 10.1186/s12967-018-1682-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/06/2018] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Brain metastasis is an extremely serious sequela with a dismal prognosis in non-small cell lung cancer (NSCLC). The present study aimed to identify novel biomarkers and potential therapeutic targets for brain metastases of NSCLC. METHODS We performed high-throughput Luminex assays to profile the transcriptional levels of 36 genes in 70 operable NSCLC patients, among whom 37 developed brain metastases as the first relapse within 3 years after surgery. The Cox proportional hazards regression model was used to evaluate the association between genes and brain metastases. Wound healing assay and transwell assay was carried out to estimate the function of target gene in vitro. And left ventricular injection on nude mice was used to evaluate the effect of target gene in vivo. RESULTS Growth-associated protein 43 (GAP43) was found to be related to brain metastasis. Multivariate Cox regression analysis showed that NSCLC patients with elevated GAP43 had a 3.29-fold increase in the risk for brain metastasis compared with those with low levels (95% confidence interval: 1.55-7.00; P = 0.002). Kaplan-Meier survival curves revealed that GAP43 was also associated with overall survival. Analysis of a cohort of 1926 NSCLC patients showed similar results: patients with high levels of GAP43 had worse progression-free and overall survival rates. Furthermore, in vitro experiments showed that GAP43 facilitated cell migration. Animal studies demonstrated that GAP43-silenced NSCLC cells were less likely to metastasize to the brain and bone than control cells. Immunofluorescence and F-actin/G-actin in vivo assays indicated that GAP43 knockdown triggered depolymerization of the F-actin cytoskeleton. Rho GTPase activation assays showed that Rac1 was deactivated after GAP43 was silenced. CONCLUSIONS Our findings suggest that GAP43 is an independent predictor of NSCLC brain metastasis and that it may facilitate metastasis by regulating the Rac1/F-actin pathway.
Collapse
Affiliation(s)
- Fanrong Zhang
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, China
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, No. 1 East Banshan Road, Hangzhou, 310022 China
| | - Lisha Ying
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, No. 1 East Banshan Road, Hangzhou, 310022 China
| | - Jiaoyue Jin
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, No. 1 East Banshan Road, Hangzhou, 310022 China
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jianguo Feng
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, No. 1 East Banshan Road, Hangzhou, 310022 China
| | - Kaiyan Chen
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, No. 1 East Banshan Road, Hangzhou, 310022 China
| | - Minran Huang
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, No. 1 East Banshan Road, Hangzhou, 310022 China
| | - Yingxue Wu
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, No. 1 East Banshan Road, Hangzhou, 310022 China
| | - Herbert Yu
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI USA
| | - Dan Su
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, No. 1 East Banshan Road, Hangzhou, 310022 China
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
24
|
Druy AE, Shorikov EV, Tsaur GA, Popov AM, Zaychikov AN, Tuponogov SN, Saveliev LI, Tytgat GAM, Fechina LG. Prospective investigation of applicability and the prognostic significance of bone marrow involvement in patients with neuroblastoma detected by quantitative reverse transcription PCR. Pediatr Blood Cancer 2018; 65:e27354. [PMID: 30007008 DOI: 10.1002/pbc.27354] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/14/2018] [Accepted: 06/17/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Detection of bone marrow (BM) involvement in patients with neuroblastoma is crucial for staging and defining prognosis. Furthermore, the persistence of residual tumor cells in the BM is associated with an unfavorable outcome. METHODS Expression of PHOX2B, TH, ELAVL4, and B4GALNT1 (GD2-synthase) was analyzed by quantitative polymerase chain reaction in neuroblastoma cell lines, control BM samples, and in BM samples from patients. The threshold level of expression for each gene was established through receiver operator characteristic analysis and used to determine the diagnostic test performance. The prognostic significance of BM involvement was assessed by survival rates calculations. The median of follow-up time was 36.1 months. RESULTS Neither PHOX2B nor TH expression was detected in control BM, while expression of ELAVL4 was found in 20 (76.9%) and GD2-synthase in 15 (57.7%) of 26 samples. The overall correct predictive value for TH, ELAVL4, and GD2-synthase, based on thresholds levels, was 0.952, 0.828, and 0.767, respectively, whereas the overall correct predictive value for PHOX2B was 0.994. The PHOX2B/TH expression in diagnostic BM of patients with neuroblastoma corresponded with a decreased survival rate (P < 0.001) in the total cohort and in different risk groups. Predominance of normalized expression of PHOX2B over TH > 1.68 in the diagnostic BM samples demonstrated an adverse prognostic effect (P = 0.006). Persistence of PHOX2B/TH expression in the BM during and after induction chemotherapy resulted in dismal outcome (P = 0.022 and P = 0.012). CONCLUSION PHOX2B and TH are the most optimal markers for detection of BM involvement, allowing identification of high-risk patients. Predominance of PHOX2B expression over TH has a strong adverse prognostic impact.
Collapse
Affiliation(s)
- Alexander E Druy
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation.,Research Institute of Medical Cell Technologies, Yekaterinburg, Russian Federation
| | - Egor V Shorikov
- PET-Technology Center of Nuclear Medicine, Yekaterinburg, Russian Federation
| | - Grigory A Tsaur
- Research Institute of Medical Cell Technologies, Yekaterinburg, Russian Federation.,Regional Children's Hospital N1, Yekaterinburg, Russian Federation.,Department of Immunochemistry, Ural Federal University named after the First President of Russia B.N. Yeltsin, Yekaterinburg, Russian Federation
| | - Alexander M Popov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | | | | | - Leonid I Saveliev
- Research Institute of Medical Cell Technologies, Yekaterinburg, Russian Federation.,Regional Children's Hospital N1, Yekaterinburg, Russian Federation
| | - Godelieve A M Tytgat
- Department of Pediatric Oncology, Emma Children's Hospital (EKZ/AMC), Amsterdam, The Netherlands.,Princess Máxima Centre for Pediatric Oncology (PMC), Utrecht, The Netherlands
| | - Larisa G Fechina
- Regional Children's Hospital N1, Yekaterinburg, Russian Federation
| |
Collapse
|
25
|
Corrias MV, Parodi S, Tchirkov A, Lammens T, Vicha A, Pasqualini C, Träger C, Yáñez Y, Dallorso S, Varesio L, Luksch R, Laureys G, Valteau-Couanet D, Canete A, Pöetschger U, Ladenstein R, Burchill SA. Event-free survival of infants and toddlers enrolled in the HR-NBL-1/SIOPEN trial is associated with the level of neuroblastoma mRNAs at diagnosis. Pediatr Blood Cancer 2018; 65:e27052. [PMID: 29603574 DOI: 10.1002/pbc.27052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/25/2018] [Accepted: 02/22/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND The purpose of this study was to evaluate whether levels of neuroblastoma mRNAs in bone marrow and peripheral blood from stage M infants (≤12 months of age at diagnosis, MYCN amplified) and toddlers (between 12 and 18 months, any MYCN status) predict event-free survival (EFS). METHODS Bone marrow aspirates and peripheral blood samples from 97 infants/toddlers enrolled in the European High-Risk Neuroblastoma trial were collected at diagnosis in PAXgene™ blood RNA tubes. Samples were analyzed by reverse transcription quantitative polymerase chain reaction according to standardized procedures. RESULTS Bone marrow tyrosine hydroxylase (TH) or paired-like homeobox 2b (PHOX2B) levels in the highest tertile were associated with worse EFS; hazard ratios, adjusted for age and MYCN status, were 1.5 and 1.8 respectively. Expression of both TH and PHOX2B in the highest tertile predicted worse outcome (p = 0.015), and identified 20 (23%) infants/toddlers with 5-year EFS of 20% (95%CI: 4%-44%). Prognostic significance was maintained after adjusting for over-fitting bias (p = 0.038), age and MYCN status. In peripheral blood, PHOX2B levels in the highest tertile predicted a two-fold increased risk of an event (p = 0.032), and identified 23 (34%) infants/toddlers with 5-year EFS of 29% (95%CI: 12%-48%). Time-dependent receiver operating characteristic analysis confirmed the prognostic value of combined TH and PHOX2B in bone marrow and of PHOX2B in peripheral blood during the first year of follow-up. CONCLUSIONS High levels of bone marrow TH and PHOX2B and of peripheral blood PHOX2B at diagnosis allow early identification of a group of high-risk infant and toddlers with neuroblastoma who may be candidates for alternative treatments. Integration with additional biomarkers, as well as validation in additional international trials is warranted.
Collapse
Affiliation(s)
- Maria V Corrias
- Unit of Experimental Therapy in Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | - Stefano Parodi
- Unit of Experimental Therapy in Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | - Andrei Tchirkov
- CHU Clermont-Ferrand, Service de Cytogénétique Médicale and Université Clermont Auvergne, Clermont-Ferrand, France
| | - Tim Lammens
- Department of Pediatric Hematology/Oncology, Ghent University Hospital, Ghent, Belgium
| | - Ales Vicha
- Department of Pediatric Hematology and Oncology, 2nd Medical Faculty Charles University and Faculty Hospital Motol, Prague, Czech Republic
| | - Claudia Pasqualini
- Department of Child and Adolescent Cancer, Institut Gustave Roussy, Villejuif, France
| | - Catarina Träger
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Yania Yáñez
- Oncología Pediátrica, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Sandro Dallorso
- Unit of Experimental Therapy in Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | - Luigi Varesio
- Unit of Experimental Therapy in Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | - Roberto Luksch
- Department of Pediatric Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Genevieve Laureys
- Department of Pediatric Hematology/Oncology, Ghent University Hospital, Ghent, Belgium
| | | | - Adela Canete
- Oncología Pediátrica, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Ulrike Pöetschger
- Department of Pediatric Oncology, CCRI/St. Anna Children's Hospital, Vienna, Austria
| | - Ruth Ladenstein
- Department of Pediatric Oncology, CCRI/St. Anna Children's Hospital, Vienna, Austria
| | - Susan A Burchill
- Children's Cancer Research Group, Leeds Institute of Cancer and Pathology, Leeds, United Kingdom
| |
Collapse
|
26
|
Li C, Zhang J, Chen S, Huang S, Wu S, Zhang L, Zhang F, Wang H. Prognostic value of metabolic indices and bone marrow uptake pattern on preoperative 18F-FDG PET/CT in pediatric patients with neuroblastoma. Eur J Nucl Med Mol Imaging 2017; 45:306-315. [PMID: 29110068 DOI: 10.1007/s00259-017-3851-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/05/2017] [Indexed: 01/21/2023]
Abstract
PURPOSE To evaluate the prognostic value of metabolic parameters and bone marrow uptake (BMU) patterns on pretherapeutic 18-F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) in pediatric patients with neuroblastoma (NB). PATIENTS AND METHODS Forty-seven pediatric patients with newly diagnosed neuroblastoma who underwent 18F-FDG PET/CT were retrospectively reviewed. Clinicopathological factors and metabolic parameters including maximum standardized uptake value (SUVmax), metabolic tumor volume (MTV), total lesion glycolysis (TLG) and bone marrow uptake patterns on PET/CT were compared to predict recurrence-free survival (RFS) and overall survival (OS) by univariate and multivariate analysis. RESULTS During the follow-up period, 27 (57.4%) patients experienced recurrence. MTV (P = 0.001), TLG (P = 0.004) and BMU patterns (P = 0.025) remained significant predictive factors for tumor recurrence, along with tumor size, histology, stage, lactate dehydrogenase (LDH) and other distant metastasis (except bone metastasis). Univariate analysis showed that histology, stage, tumor size (>37.25 cm), other distant metastasis, MTV (>88.10cm3) and TLG (>1045.2 g) and BMU patterns correlated with both RFS and OS (P < 0.05). On multivariate analysis, TLG remained the only independent prognostic factor for RFS (P = 0.016) and OS (P = 0.012), and BMU patterns and MTV were statistically significant for OS (P = 0.024 and P = 0.038, respectively). CONCLUSION Pretherapeutic 18F-FDG PET/CT can provide reliable prognostic information for neuroblastoma pediatric patients, and patients with high MTV, TLG and focal bone marrow (unifocal and multifocal) uptake on PET/CT may have inferior outcomes during subsequent treatment.
Collapse
Affiliation(s)
- Chao Li
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Jian Zhang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.,Shanghai Universal Medical Imaging Diagnostic Center, 406 Guilin Road, Shanghai, 201103, China
| | - Suyun Chen
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Shuo Huang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Shuqi Wu
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Linlin Zhang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Fengxian Zhang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Hui Wang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
27
|
Rifatbegovic F, Frech C, Abbasi MR, Taschner-Mandl S, Weiss T, Schmidt WM, Schmidt I, Ladenstein R, Ambros IM, Ambros PF. Neuroblastoma cells undergo transcriptomic alterations upon dissemination into the bone marrow and subsequent tumor progression. Int J Cancer 2017; 142:297-307. [PMID: 28921546 PMCID: PMC5725737 DOI: 10.1002/ijc.31053] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/28/2017] [Accepted: 08/02/2017] [Indexed: 12/12/2022]
Abstract
Neuroblastoma is the most common extracranial solid tumor in childhood. The vast majority of metastatic (M) stage patients present with disseminated tumor cells (DTCs) in the bone marrow (BM) at diagnosis and relapse. Although these cells represent a major obstacle in the treatment of neuroblastoma patients, insights into their expression profile remained elusive. The present RNA‐Seq study of stage 4/M primary tumors, enriched BM‐derived diagnostic and relapse DTCs, as well as the corresponding BM‐derived mononuclear cells (MNCs) from 53 patients revealed 322 differentially expressed genes in DTCs as compared to the tumors (q < 0.001, |log2FC|>2). Particularly, the levels of transcripts encoded by mitochondrial DNA were elevated in DTCs, whereas, for example, genes involved in angiogenesis were downregulated. Furthermore, 224 genes were highly expressed in DTCs and only slightly, if at all, in MNCs (q < 8 × 10−75 log2FC > 6). Interestingly, we found the transcriptome of relapse DTCs largely resembling those of diagnostic DTCs with only 113 differentially expressed genes under relaxed cut‐offs (q < 0.01, |log2FC|>0.5). Notably, relapse DTCs showed a positional enrichment of 31 downregulated genes on chromosome 19, including five tumor suppressor genes: SIRT6, BBC3/PUMA, STK11, CADM4 and GLTSCR2. This first RNA‐Seq analysis of neuroblastoma DTCs revealed their unique expression profile in comparison to the tumors and MNCs, and less pronounced differences between diagnostic and relapse DTCs. The latter preferentially affected downregulation of genes encoded by chromosome 19. As these alterations might be associated with treatment failure and disease relapse, further functional studies on DTCs should be considered. What's new? More than 90% of patients diagnosed with stage 4 metastatic (4/M) neuroblastoma present with disseminated tumor cells (DTCs) in the bone marrow (BM). Despite treatment, a substantial fraction of these patients experience disease relapse. Here, sequencing analysis of tumor tissue, BM‐derived mononuclear cells (MNCs), and DTCs from stage 4/M neuroblastoma patients indicates that numerous genes are differentially expressed in DTCs but are not or are only slightly altered in tumors and MNCs. Moreover, DTCs exhibited significant downregulation of tumor suppressor genes specifically on chromosome 19. Further studies are needed to determine whether DTC transcriptomic alterations are associated with neuroblastoma relapse.
Collapse
Affiliation(s)
- Fikret Rifatbegovic
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Christian Frech
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - M Reza Abbasi
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Sabine Taschner-Mandl
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Tamara Weiss
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Wolfgang M Schmidt
- Neuromuscular Research Department, Medical University of Vienna, Vienna, Austria
| | - Iris Schmidt
- Neuromuscular Research Department, Medical University of Vienna, Vienna, Austria
| | - Ruth Ladenstein
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Inge M Ambros
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Peter F Ambros
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
Marachelian A, Villablanca JG, Liu CW, Liu B, Goodarzian F, Lai HA, Shimada H, Tran HC, Parra JA, Gallego R, Bedrossian N, Young S, Czarnecki S, Kennedy R, Weiss BD, Goldsmith K, Granger M, Matthay KK, Groshen S, Asgharzadeh S, Sposto R, Seeger RC. Expression of Five Neuroblastoma Genes in Bone Marrow or Blood of Patients with Relapsed/Refractory Neuroblastoma Provides a New Biomarker for Disease and Prognosis. Clin Cancer Res 2017; 23:5374-5383. [PMID: 28559462 DOI: 10.1158/1078-0432.ccr-16-2647] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 02/13/2017] [Accepted: 05/23/2017] [Indexed: 11/16/2022]
Abstract
Purpose: We determined whether quantifying neuroblastoma-associated mRNAs (NB-mRNAs) in bone marrow and blood improves assessment of disease and prediction of disease progression in patients with relapsed/refractory neuroblastoma.Experimental Design: mRNA for CHGA, DCX, DDC, PHOX2B, and TH was quantified in bone marrow and blood from 101 patients concurrently with clinical disease evaluations. Correlation between NB-mRNA (delta cycle threshold, ΔCt, for the geometric mean of genes from the TaqMan Low Density Array NB5 assay) and morphologically defined tumor cell percentage in bone marrow, 123I-meta-iodobenzylguanidine (MIBG) Curie score, and CT/MRI-defined tumor longest diameter was determined. Time-dependent covariate Cox regression was used to analyze the relationship between ΔCt and progression-free survival (PFS).Results: NB-mRNA was detectable in 83% of bone marrow (185/223) and 63% (89/142) of blood specimens, and their ΔCt values were correlated (Spearman r = 0.67, P < 0.0001), although bone marrow Ct was 7.9 ± 0.5 Ct stronger than blood Ct When bone marrow morphology, MIBG, or CT/MRI were positive, NB-mRNA was detected in 99% (99/100), 88% (100/113), and 81% (82/101) of bone marrow samples. When all three were negative, NB-mRNA was detected in 55% (11/20) of bone marrow samples. Bone marrow NB-mRNA correlated with bone marrow morphology or MIBG positivity (P < 0.0001 and P = 0.007). Bone marrow and blood ΔCt values correlated with PFS (P < 0.001; P = 0.001) even when bone marrow was morphologically negative (P = 0.001; P = 0.014). Multivariate analysis showed that bone marrow and blood ΔCt values were associated with PFS independently of clinical disease and MYCN gene status (P < 0.001; P = 0.055).Conclusions: This five-gene NB5 assay for NB-mRNA improves definition of disease status and correlates independently with PFS in relapsed/refractory neuroblastoma. Clin Cancer Res; 23(18); 5374-83. ©2017 AACR.
Collapse
Affiliation(s)
- Araz Marachelian
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Children's Center for Cancer and Blood Diseases, Los Angeles, California.
- Children's Hospital Los Angeles, Los Angeles, California
| | - Judith G Villablanca
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Children's Center for Cancer and Blood Diseases, Los Angeles, California
- Children's Hospital Los Angeles, Los Angeles, California
| | - Cathy W Liu
- Children's Hospital Los Angeles, Los Angeles, California
| | - Betty Liu
- Children's Hospital Los Angeles, Los Angeles, California
| | - Fariba Goodarzian
- Children's Hospital Los Angeles, Los Angeles, California
- Department of Radiology, Keck School of Medicine, University of Southern California, Children's Center for Cancer and Blood Diseases, Los Angeles, California
| | - Hollie A Lai
- Children's Hospital Los Angeles, Los Angeles, California
- Department of Radiology, Keck School of Medicine, University of Southern California, Children's Center for Cancer and Blood Diseases, Los Angeles, California
| | - Hiroyuki Shimada
- Children's Hospital Los Angeles, Los Angeles, California
- Department of Pathology, Keck School of Medicine, University of Southern California, Children's Center for Cancer and Blood Diseases, Los Angeles, California
| | - Hung C Tran
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Children's Center for Cancer and Blood Diseases, Los Angeles, California
- Children's Hospital Los Angeles, Los Angeles, California
| | - Jaime A Parra
- Children's Hospital Los Angeles, Los Angeles, California
| | | | | | - Sabrina Young
- Children's Hospital Los Angeles, Los Angeles, California
| | | | | | - Brian D Weiss
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kelly Goldsmith
- Aflac Cancer Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia
| | | | - Katherine K Matthay
- University of California, San Francisco Children's Hospital, San Francisco, California
| | - Susan Groshen
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Children's Center for Cancer and Blood Diseases, Los Angeles, California
| | - Shahab Asgharzadeh
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Children's Center for Cancer and Blood Diseases, Los Angeles, California
- Children's Hospital Los Angeles, Los Angeles, California
- Department of Pathology, Keck School of Medicine, University of Southern California, Children's Center for Cancer and Blood Diseases, Los Angeles, California
| | - Richard Sposto
- Children's Hospital Los Angeles, Los Angeles, California
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Children's Center for Cancer and Blood Diseases, Los Angeles, California
| | - Robert C Seeger
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Children's Center for Cancer and Blood Diseases, Los Angeles, California
- Children's Hospital Los Angeles, Los Angeles, California
| |
Collapse
|
29
|
Burchill SA, Beiske K, Shimada H, Ambros PF, Seeger R, Tytgat GAM, Brock PR, Haber M, Park JR, Berthold F. Recommendations for the standardization of bone marrow disease assessment and reporting in children with neuroblastoma on behalf of the International Neuroblastoma Response Criteria Bone Marrow Working Group. Cancer 2017; 123:1095-1105. [PMID: 27984660 DOI: 10.1002/cncr.30380] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/24/2016] [Accepted: 08/15/2016] [Indexed: 11/10/2022]
Abstract
BACKGROUND The current study was conducted to expedite international standardized reporting of bone marrow disease in children with neuroblastoma and to improve equivalence of care. METHODS A multidisciplinary International Neuroblastoma Response Criteria Bone Marrow Working Group was convened by the US National Cancer Institute in January 2012 with representation from Europe, North America, and Australia. Practical transferable recommendations to standardize the reporting of bone marrow disease were developed. RESULTS To the authors' knowledge, the current study is the first to comprehensively present consensus criteria for the collection, analysis, and reporting of the percentage area of bone marrow parenchyma occupied by tumor cells in trephine-biopsies. The quantitative analysis of neuroblastoma content in bone marrow aspirates by immunocytology and reverse transcriptase-quantitative polymerase chain reaction are revised. The inclusion of paired-like homeobox 2b (PHOX2B) for immunohistochemistry and reverse transcriptase-quantitative polymerase chain reaction is recommended. Recommendations for recording bone marrow response are provided. The authors endorse the quantitative assessment of neuroblastoma cell content in bilateral core needle biopsies-trephines and aspirates in all children with neuroblastoma, with the exception of infants, in whom the evaluation of aspirates alone is advised. It is interesting to note that 5% disease is accepted as an internationally achievable level for disease assessment. CONCLUSIONS The quantitative assessment of neuroblastoma cells is recommended to provide data from which evidence-based numerical criteria for the reporting of bone marrow response can be realized. This is particularly important in the minimal disease setting and when neuroblastoma detection in bone marrow is intermittent, where clinical impact has yet to be validated. The wide adoption of these harmonized criteria will enhance the ability to compare outcomes from different trials and facilitate collaborative trial design. Cancer 2017;123:1095-1105. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Susan A Burchill
- Children's Cancer Research Group, Leeds Institute of Cancer and Pathology, St James's University Hospital, Leeds, United Kingdom
| | - Klaus Beiske
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Hiroyuki Shimada
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - Peter F Ambros
- CCRI, Children's Cancer Research Institute, Vienna, Austria
| | - Robert Seeger
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | | | - Penelope R Brock
- Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom
| | - Michelle Haber
- Children's Cancer Institute for Medical Research, Cancer Research Centre, Sydney, New South Wales, Australia
| | - Julie R Park
- Department of Pediatrics, School of Medicine, University of Washington, Seattle Children's Hospital, Seattle, Washington
| | - Frank Berthold
- Department of Pediatric Oncology and Hematology, Center for Integrated Oncology, University of Cologne, Cologne, Germany
| |
Collapse
|
30
|
Brownhill SC, Burchill SA. PCR-based amplification of circulating RNAs as prognostic and predictive biomarkers - Focus on neuroblastoma. Pract Lab Med 2017; 7:41-44. [PMID: 28856217 PMCID: PMC5575362 DOI: 10.1016/j.plabm.2016.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 04/16/2016] [Indexed: 01/18/2023] Open
Abstract
Metastatic disease is a major challenge for cancer cure, haematogenous spread and subsequent growth of tumour cells at distant sites being the cause of most cancer deaths. Molecular characterization and detection of the tumour cells responsible for haematogenous spread may increase understanding of the biology of metastasis, help improve patient management and allow evaluation of novel treatments to prevent and eradicate this disease. The bone marrow is a common site to which tumour cells metastasize, from which they may re-circulate to other organs with a favourable microenvironment for growth. The detection of tumour cells in blood suggests one route for metastasis, and provides an accessible, minimally invasive liquid sample through which it may be possible to monitor and detect minimal disease and early signs of metastasis. Significant improvements in the sensitivity and specificity of tumour cell detection have been made, such that it is now possible to unambiguously detect a single tumour cell in over 10 million normal cells. However, the clinical impact of such low level disease and how to interpret the natural variation that can arise from sequential sampling of bone marrow aspirates and blood is currently largely unknown. This commentary will focus on the technical advancements and application of reverse transcriptase polymerase chain reaction to detect cancer mRNAs in bone marrow and blood, and discuss the potential clinical impact of this test in neuroblastoma.
Collapse
Affiliation(s)
- Sam C Brownhill
- Children's Cancer Research Group, Leeds Institute of Cancer and Pathology, School of Medicine and Health, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Sue A Burchill
- Children's Cancer Research Group, Leeds Institute of Cancer and Pathology, School of Medicine and Health, University of Leeds, Leeds LS9 7TF, United Kingdom
| |
Collapse
|
31
|
Hirase S, Saitoh A, Hartomo TB, Kozaki A, Yanai T, Hasegawa D, Kawasaki K, Kosaka Y, Matsuo M, Yamamoto N, Mori T, Hayakawa A, Iijima K, Nishio H, Nishimura N. Early detection of tumor relapse/regrowth by consecutive minimal residual disease monitoring in high-risk neuroblastoma patients. Oncol Lett 2016; 12:1119-1123. [PMID: 27446404 DOI: 10.3892/ol.2016.4682] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 01/12/2016] [Indexed: 12/21/2022] Open
Abstract
Neuroblastoma is an aggressive pediatric tumor accounting for ~15% of cancer-associated mortalities in children. Despite the current intensive therapy, >50% of high-risk patients experience tumor relapse or regrowth caused by the activation of minimal residual disease (MRD). Although several MRD detection protocols using various reverse transcription-quantitative polymerase chain reaction (RT-qPCR) markers have been reported to evaluate the therapeutic response and disease status of neuroblastoma patients, their clinical significance remains elusive. The present study reports two high-risk neuroblastoma patients, whose MRD was consecutively monitored using 11 RT-qPCR markers (CHRNA3, CRMP1, DBH, DCX, DDC, GABRB3, GAP43, ISL1, KIF1A, PHOX2B and TH) during their course of treatment. The two patients initially responded to the induction therapy and reached MRD-negative status. The patients' MRD subsequently became positive with no elevation of their urinary homovanillic acid, urinary vanillylmandelic acid and serum neuron-specific enolase levels at 13 or 19 weeks prior to the clinical diagnosis of tumor relapse or regrowth. The present cases highlight the possibility of consecutive MRD monitoring using 11 markers to enable an early detection of tumor relapse or regrowth in high-risk neuroblastoma patients.
Collapse
Affiliation(s)
- Satoshi Hirase
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Atsuro Saitoh
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe 654-0081, Japan
| | - Tri Budi Hartomo
- Department of Epidemiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Aiko Kozaki
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe 654-0081, Japan
| | - Tomoko Yanai
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe 654-0081, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe 654-0081, Japan
| | - Keiichiro Kawasaki
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe 654-0081, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe 654-0081, Japan
| | - Masafumi Matsuo
- Department of Medical Rehabilitation, Kobe Gakuin University, Kobe 651-2180, Japan
| | - Nobuyuki Yamamoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Takeshi Mori
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Akira Hayakawa
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Hisahide Nishio
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Department of Epidemiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Noriyuki Nishimura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Department of Epidemiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| |
Collapse
|
32
|
Yáñez Y, Hervás D, Grau E, Oltra S, Pérez G, Palanca S, Bermúdez M, Márquez C, Cañete A, Castel V. TH and DCX mRNAs in peripheral blood and bone marrow predict outcome in metastatic neuroblastoma patients. J Cancer Res Clin Oncol 2016; 142:573-80. [PMID: 26498952 DOI: 10.1007/s00432-015-2054-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/09/2015] [Indexed: 12/26/2022]
Abstract
PURPOSE In metastatic neuroblastoma (NB) patients, accurate risk stratification and disease monitoring would reduce relapse probabilities. This study aims to evaluate the independent prognostic significance of detecting tyrosine hydroxylase (TH) and doublecortin (DCX) mRNAs by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) in peripheral blood (PB) and bone marrow (BM) samples from metastatic NB patients. PROCEDURES RT-qPCR was performed on PB and BM samples from metastatic NB patients at diagnosis, post-induction therapy and at the end of treatment for TH and DCX mRNAs detection. RESULTS High levels of TH and DCX mRNAs when detected in PB and BM at diagnosis independently predicted worse outcome in a cohort of 162 metastatic NB. In the subgroup of high-risk metastatic NB, TH mRNA detected in PB remained as independent predictor of EFS and OS at diagnosis. After the induction therapy, high levels of TH mRNA in PB and DCX mRNA in BM independently predicted poor EFS and OS. Furthermore TH mRNA when detected in BM predicted worse EFS. TH mRNA in PB samples at the end of treatment is an independent predictor of worse outcome. CONCLUSION TH and DCX mRNAs levels in PB and BM assessed by RT-qPCR should be considered in new pre-treatment risk stratification strategies to reliable estimate outcome differences in metastatic NB patients. In those high-risk metastatic NB, TH and DCX mRNA quantification could be used for the assessment of response to treatment and for early detection of progressive disease or relapses.
Collapse
Affiliation(s)
- Yania Yáñez
- Unidad de Oncología Pediátrica, Hospital La Fe, Avda. Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - David Hervás
- Unidad de Bioestadística, Hospital La Fe, Valencia, Spain
| | - Elena Grau
- Unidad de Oncología Pediátrica, Hospital La Fe, Avda. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Silvestre Oltra
- Unidad de Genética y Diagnóstico Prenatal, Hospital La Fe, Valencia, Spain
| | - Gema Pérez
- Laboratorio de Biología Molecular, Hospital La Fe, Valencia, Spain
| | - Sarai Palanca
- Laboratorio de Biología Molecular, Hospital La Fe, Valencia, Spain
| | - Mar Bermúdez
- Servicio de Oncología Pediátrica, Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Catalina Márquez
- Servicio de Oncología Pediátrica, Hospital Virgen del Rocio, Seville, Spain
| | - Adela Cañete
- Unidad de Oncología Pediátrica, Hospital La Fe, Avda. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Victoria Castel
- Unidad de Oncología Pediátrica, Hospital La Fe, Avda. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| |
Collapse
|
33
|
van Wezel EM, Decarolis B, Stutterheim J, Zappeij-Kannegieter L, Berthold F, Schumacher-Kuckelkorn R, Simon T, Fiocco M, van Noesel MM, Caron HN, van der Schoot CE, Hero B, Tytgat GAM. Neuroblastoma messenger RNA is frequently detected in bone marrow at diagnosis of localised neuroblastoma patients. Eur J Cancer 2016; 54:149-158. [PMID: 26796600 DOI: 10.1016/j.ejca.2015.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 09/25/2015] [Accepted: 11/09/2015] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The clinical importance of the detection of neuroblastoma messenger RNA (mRNA) in bone marrow (BM) of localised neuroblastoma patients at diagnosis remains unclear. In this prospective multicentre study, BM samples of a large cohort, were studied using real-time quantitative polymerase chain reaction (qPCR). METHODS BM samples at diagnosis from 160 patients with localised neuroblastoma were prospectively collected at Dutch and German centres between 2009 and 2013. qPCR was performed using five neuroblastoma specific markers. The association with other biological factors and the prognostic impact of BM positivity and clinical response was assessed. RESULTS In 58 out of 160 patients neuroblastoma mRNA was detected in BM. In 47 of the 58 positive samples only one marker was found positive. BM positivity was significantly associated with MYCN amplification (p = 0.02) and deletion of chromosome 1p (p = 0.04). In total 31 patients had an event, of which only five patients had progression to stage IV. BM positivity was not associated with an unfavourable outcome. However, the detection of more than one marker was associated with an unfavourable outcome (systemic or local relapse) (event free survival 48% versus 85%; p = 0.03) in the whole cohort and in the observation group. CONCLUSIONS BM positivity was associated with unfavourable biological factors and might represent more aggressive tumours. Patients with qPCR positive BM should not be upstaged, because of very few systemic events in the cohort. However, for patients with more than one marker positive a more careful follow-up is advisable. These results need to be verified in a very large cohort of localised patients.
Collapse
Affiliation(s)
- Esther M van Wezel
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the AMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Oncology, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Boris Decarolis
- Children's Hospital, University of Cologne, Pediatric Hematology and Oncology, Germany
| | - Janine Stutterheim
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the AMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Oncology, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Lily Zappeij-Kannegieter
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Frank Berthold
- Children's Hospital, University of Cologne, Pediatric Hematology and Oncology, Germany
| | | | - Thorsten Simon
- Children's Hospital, University of Cologne, Pediatric Hematology and Oncology, Germany
| | - Marta Fiocco
- Department of Biostatistics, Leiden University Medical Center and Dutch Childhood Oncology Group, The Hague, The Netherlands
| | - Max M van Noesel
- Department of Pediatric Oncology, Sophia Children's Hospital, Erasmus Medical Center, Rotterdam, The Netherlands; Prinses Máxima Centrum, Utrecht, The Netherlands
| | - Huib N Caron
- Department of Pediatric Oncology, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, The Netherlands
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Barbara Hero
- Children's Hospital, University of Cologne, Pediatric Hematology and Oncology, Germany
| | - Godelieve A M Tytgat
- Department of Pediatric Oncology, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, The Netherlands; Prinses Máxima Centrum, Utrecht, The Netherlands.
| |
Collapse
|
34
|
Yamamoto N, Kozaki A, Hartomo TB, Yanai T, Hasegawa D, Kawasaki K, Kosaka Y, Matsuo M, Hirase S, Mori T, Hayakawa A, Iijima K, Nishio H, Nishimura N. Differential expression of minimal residual disease markers in peripheral blood and bone marrow samples from high-risk neuroblastoma patients. Oncol Lett 2015; 10:3228-3232. [PMID: 26722317 DOI: 10.3892/ol.2015.3710] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 07/16/2015] [Indexed: 12/19/2022] Open
Abstract
Neuroblastoma is an aggressive solid tumor that leads to tumor relapse in more than half of high-risk patients. Minimal residual disease (MRD) is primarily responsible for tumor relapses and may be detected in peripheral blood (PB) and bone marrow (BM) samples. To evaluate the disease status and treatment response, a number of MRD detection protocols based on either common or distinct markers for PB and BM samples have been reported. However, the correlation between the expression of MRD markers in PB and BM samples remains elusive in the clinical samples. In the present study, the expression of 11 previously validated MRD markers (CHRNA3, CRMP1, DBH, DCX, DDC, GABRB3, GAP43, ISL1, KIF1A, PHOX2B and TH) was determined in 23 pairs of PB and BM samples collected from seven high-risk neuroblastoma patients at the same time point, and the sample was scored as MRD-positive if one of the MRD markers exceeded the normal range. Although the number of MRD-positive samples was not significantly different between PB and BM samples, the two most sensitive markers for PB samples (CRMP1 and KIF1A) were different from those for BM samples (PHOX2B and DBH). There was no statistically significant correlation between the expression of MRD markers in the PB and BM samples. These results suggest that MRD markers were differentially expressed in PB and BM samples from high-risk neuroblastoma patients.
Collapse
Affiliation(s)
- Nobuyuki Yamamoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Aiko Kozaki
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe 654-0081, Japan
| | - Tri Budi Hartomo
- Department of Epidemiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Tomoko Yanai
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe 654-0081, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe 654-0081, Japan
| | - Keiichiro Kawasaki
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe 654-0081, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe 654-0081, Japan
| | - Masafumi Matsuo
- Department of Medical Rehabilitation, Kobe Gakuin University, Kobe 651-2180, Japan
| | - Satoshi Hirase
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Takeshi Mori
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Akira Hayakawa
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Hisahide Nishio
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan ; Department of Epidemiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Noriyuki Nishimura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan ; Department of Epidemiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| |
Collapse
|
35
|
van Wezel EM, Stutterheim J, Vree F, Zappeij-Kannegieter L, Decarolis B, Hero B, Berthold F, Schumacher-Kuckelkorn R, Simon T, Fiocco M, Voermans C, van Noesel MM, Caron HN, van der Schoot CE, Tytgat GAM. Minimal residual disease detection in autologous stem cell grafts from patients with high risk neuroblastoma. Pediatr Blood Cancer 2015; 62:1368-73. [PMID: 25939774 DOI: 10.1002/pbc.25507] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 02/18/2015] [Indexed: 11/10/2022]
Abstract
BACKGROUND The clinical significance of minimal residual disease (MRD) detected by real-time quantitative PCR (qPCR) in autologous stem cell grafts in high risk neuroblastoma is still controversial. In this retrospective multicenter study, autologous stem cell grafts of a large cohort were studied using a panel of RNA markers. PROCEDURE From 104 patients with high risk neuroblastoma, who received autologous stem cell transplantation as first line treatment, 66 peripheral blood stem cells (PBSC) and 38 CD34+ selected grafts were retrospectively collected at 2 Dutch and 12 German centers between 1997 and 2010. To investigate graft contamination qPCR was performed by using 5 neuroblastoma specific markers (PHOX2B, TH, DDC, CHRNA3, and DBH). RESULTS In PBSC 6/66 (9%) and in CD34+ selected grafts 3/38 (8%) samples were contaminated. Graft contamination was not associated with an unfavorable outcome (5-years OS, 66% vs. 50.5%; P=0.6 and 5-years EFS, 22% vs. 35%, P=0.7). In multivariate Cox analysis BM MRD at time of harvest was significantly associated with survival (P=0.008 OS and P=0.002 EFS), but graft contamination was still not associated with an unfavorable outcome (P=0.9 OS and P=1 EFS). CONCLUSIONS Graft contamination is very infrequent in this retrospective cohort of patients with no or minimal BM disease prior to stem cell collection and does not influence outcome in univariate and multivariate analysis. The presence of MRD at time of harvest is a strong outcome predictor. However, these results will have to be verified in a large prospective study.
Collapse
Affiliation(s)
- Esther M van Wezel
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands and Landsteiner Laboratory of the AMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital, Academical Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Janine Stutterheim
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands and Landsteiner Laboratory of the AMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital, Academical Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Florentien Vree
- Department of Pediatric Oncology, Emma Children's Hospital, Academical Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Lily Zappeij-Kannegieter
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands and Landsteiner Laboratory of the AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Boris Decarolis
- Children's Hospital, University of Cologne, Pediatric Hematology and Oncology, Cologne, Germany
| | - Barbara Hero
- Children's Hospital, University of Cologne, Pediatric Hematology and Oncology, Cologne, Germany
| | - Frank Berthold
- Children's Hospital, University of Cologne, Pediatric Hematology and Oncology, Cologne, Germany
| | | | - Thorsten Simon
- Children's Hospital, University of Cologne, Pediatric Hematology and Oncology, Cologne, Germany
| | - Marta Fiocco
- Department of Biostatistics, Leiden University Medical Center and Dutch Childhood Oncology Group, The Hague, Netherlands
| | - Carlijn Voermans
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands and Landsteiner Laboratory of the AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Max M van Noesel
- Department of Pediatric Oncology, Sophia Children's Hospital, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Huib N Caron
- Department of Pediatric Oncology, Emma Children's Hospital, Academical Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, The Netherlands and Landsteiner Laboratory of the AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Godelieve A M Tytgat
- Department of Pediatric Oncology, Emma Children's Hospital, Academical Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
36
|
Mora J, Cruz O, Lavarino C, Rios J, Vancells M, Parareda A, Salvador H, Suñol M, Carrasco R, Guillen A, Mañé S, de Torres C. Results of induction chemotherapy in children older than 18 months with stage-4 neuroblastoma treated with an adaptive-to-response modified N7 protocol (mN7). Clin Transl Oncol 2015; 17:521-9. [PMID: 25596034 DOI: 10.1007/s12094-014-1273-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 12/24/2014] [Indexed: 12/17/2022]
Abstract
PURPOSE We report the response rate in children older than 18 months with stage 4 Neuroblastoma, using a modified dose-intensive, response-adaptive, induction mN7 protocol. METHODS From 2005 to 2012, 24 patients were treated with the mN7 protocol. Phase 1 included five MSKCC N7 cycles and surgery and two high-dose cyclophosphamide-topotecan (HD-CT) cycles for those who did not achieve complete remission (CR) and negative bone marrow (BM) minimal residual disease (MRD) status (CR+MRD-). Phase 2 consisted of myeloablative doses of topotecan, thiotepa and carboplatin plus hyperfractionated RT. Phase 3 included isotretinoin and 3F8 immunotherapy plus GM-CSF. BM MRD was monitored using GD2 synthase, PHOX2B and cyclin D1 mRNAs. RESULTS After 3 cycles, all patients showed BM complete histological clearance and 6 (25 %) were MRD-. Twenty of 21 s-look surgeries achieved macroscopic complete resection. After 5 cycles and surgery, (123)I-MIBG scan was negative in 15 (62.5 %) cases, BM disease by histology was negative in 23 (96 %) and 10 (42 %) patients were MRD-. Twelve (50 %) pts were in CR, 2 in very good partial response (VGPR), 9 partial response (PR) and one had progressive disease. With 2 HD-CT extra cycles, 17 (71 %) pts achieved CR+MRD- status moving to phase 2. Overall and event-free survival at 3 years for the 17 patients who achieved CR+MRD- is 65 and 53 %, respectively, median follow-up 47 months. Seven (29 %) patients never achieved CR+MRD-. Univariate Cox regression analysis shows CR+MRD- status after mN7 induction as the only statistically significant prognostic factor to predict overall survival. CONCLUSIONS mN7 induction regimen produced a CR+MRD- rate of 71 %. CR+MRD- status following induction was the only predictive marker of long-term survival.
Collapse
Affiliation(s)
- J Mora
- Department of Oncology, Hospital Sant Joan de Déu, Barcelona, Passeig Sant Joan de Déu, 2, Esplugues de Llobregat, 08950, Barcelona, Spain,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
van Wezel EM, Zwijnenburg D, Zappeij-Kannegieter L, Bus E, van Noesel MM, Molenaar JJ, Versteeg R, Fiocco M, Caron HN, van der Schoot CE, Koster J, Tytgat GA. Whole-Genome Sequencing Identifies Patient-Specific DNA Minimal Residual Disease Markers in Neuroblastoma. J Mol Diagn 2015; 17:43-52. [DOI: 10.1016/j.jmoldx.2014.09.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 08/27/2014] [Accepted: 09/05/2014] [Indexed: 12/21/2022] Open
|
38
|
Viprey VF, Gregory WM, Corrias MV, Tchirkov A, Swerts K, Vicha A, Dallorso S, Brock P, Luksch R, Valteau-Couanet D, Papadakis V, Laureys G, Pearson AD, Ladenstein R, Burchill SA. Neuroblastoma mRNAs predict outcome in children with stage 4 neuroblastoma: a European HR-NBL1/SIOPEN study. J Clin Oncol 2014; 32:1074-83. [PMID: 24590653 DOI: 10.1200/jco.2013.53.3604] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024] Open
Abstract
PURPOSE To evaluate the hypothesis that detection of neuroblastoma mRNAs by reverse transcriptase quantitative polymerase chain reaction (RTqPCR) in peripheral blood (PB) and bone marrow aspirates (BM) from children with stage 4 neuroblastoma are clinically useful biomarkers of risk. METHODS RTqPCR for paired-like homeobox 2b (PHOX2B), tyrosine hydroxylase (TH), and doublecortin (DCX) mRNA in PB and BM of children enrolled onto the High-Risk Neuroblastoma Trial-1 of the European Society of Pediatric Oncology Neuroblastoma Group (HR-NBL1/SIOPEN) was performed at diagnosis and after induction therapy. RESULTS High levels of TH, PHOX2B, or DCX mRNA in PB or BM at diagnosis strongly predicted for worse event-free survival (EFS) and overall survival (OS) in a cohort of 290 children. After induction therapy, high levels of these mRNAs predicted worse EFS and OS in BM but not in PB. Combinations of mRNAs in BM did not add to the predictive power of any single mRNA. However, in the original (n = 182) and validation (n = 137) PB cohorts, high TH (log10TH > 0.8) or high PHOX2B (log10PHOX2B > 0.28) identify 19% of children as ultrahigh risk, with 5-year EFS and OS rates of 0%; OS rate was 25% (95% CI, 16% to 36%) and EFS rate was 38% (95% CI, 28% to 49%) in the remaining children. The magnitude of reduction in mRNA level between diagnosis and postinduction therapy in BM or PB was not of additional predictive value. CONCLUSION High levels of TH and PHOX2B mRNA in PB at diagnosis objectively identify children with ultrahigh-risk disease who may benefit from novel treatment approaches. The level of TH, PHOX2B, and DCX mRNA in BM and/or PB at diagnosis might contribute to an algorithm to improve stratification of children for treatment.
Collapse
Affiliation(s)
- Virginie F Viprey
- Virginie F. Viprey and Susan A. Burchill, Leeds Institute of Cancer and Pathology; Walter M. Gregory, Clinical Trials Research Unit, University of Leeds, Leeds; Penelope Brock, Great Ormond Street Hospital, London; Andrew D. Pearson, Institute of Cancer Research/Royal Marsden National Health Service Foundation Trust, Sutton, United Kingdom; Maria V. Corrias and Sandro Dallorso, Gaslini Institute, Genoa; Roberto Luksch, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milano, Italy; Andrei Tchirkov, Centre Hospitalier Universitaire Clermont-Ferrand and Clermont Université, Université d'Auvergne, Clermont-Ferrand; Dominique Valteau-Couanet, Institut Gustave Roussy, Villejuif, France; Katrien Swerts and Genevieve Laureys, University Hospital Ghent, Ghent, Belgium; Ales Vicha, Charles University and University Hospital Motol, Prague, Czech Republic; Vassilios Papadakis, Agia Sofia Children's Hospital, Athens, Greece; and Ruth Ladenstein, Children's Cancer Research Institute/St Anna Children's Hospital, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Kreissman SG, Seeger RC, Matthay KK, London WB, Sposto R, Grupp SA, Haas-Kogan DA, Laquaglia MP, Yu AL, Diller L, Buxton A, Park JR, Cohn SL, Maris JM, Reynolds CP, Villablanca JG. Purged versus non-purged peripheral blood stem-cell transplantation for high-risk neuroblastoma (COG A3973): a randomised phase 3 trial. Lancet Oncol 2013; 14:999-1008. [PMID: 23890779 DOI: 10.1016/s1470-2045(13)70309-7] [Citation(s) in RCA: 221] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Myeloablative chemoradiotherapy and immunomagnetically purged autologous bone marrow transplantation has been shown to improve outcome for patients with high-risk neuroblastoma. Currently, peripheral blood stem cells (PBSC) are infused after myeloablative therapy, but the effect of purging is unknown. We did a randomised study of tumour-selective PBSC purging in stem-cell transplantation for patients with high-risk neuroblastoma. METHODS Between March 16, 2001, and Feb 24, 2006, children and young adults (<30 years) with high-risk neuroblastoma were randomly assigned at diagnosis by a web-based system (in a 1:1 ratio) to receive either non-purged or immunomagnetically purged PBSC. Randomisation was done in blocks stratified by International Neuroblastoma Staging System stage, age, MYCN status, and International Neuroblastoma Pathology classification. Patients and treating physicians were not masked to treatment assignment. All patients were treated with six cycles of induction chemotherapy, myeloablative consolidation, and radiation therapy to the primary tumour site plus meta-iodobenzylguanidine avid metastases present before myeloablative therapy, followed by oral isotretinoin. PBSC collection was done after two induction cycles. For purging, PBSC were mixed with carbonyl iron and phagocytic cells removed with samarium cobalt magnets. Remaining cells were mixed with immunomagnetic beads prepared with five monoclonal antibodies targeting neuroblastoma cell surface antigens and attached cells were removed using samarium cobalt magnets. Patients underwent autologous stem-cell transplantation with PBSC as randomly assigned after six cycles of induction therapy. The primary endpoint was event-free survival and was analysed by intention-to-treat. The trial is registered with ClinicalTrials.gov, number NCT00004188. FINDINGS 495 patients were enrolled, of whom 486 were randomly assigned to treatment: 243 patients to receive non-purged PBSC and 243 to received purged PBSC. PBSC were collected from 229 patients from the purged group and 236 patients from the non-purged group, and 180 patients from the purged group and 192 from the non-purged group received transplant. 5-year event-free survival was 40% (95% CI 33-46) in the purged group versus 36% (30-42) in the non-purged group (p=0·77); 5-year overall survival was 50% (95% CI 43-56) in the purged group compared with 51% (44-57) in the non-purged group (p=0·81). Toxic deaths occurred in 15 patients during induction (eight in the purged group and seven in the non-purged group) and 12 during consolidation (eight in the purged group and four in the non-purged group). The most common adverse event reported was grade 3 or worse stomatitis during both induction (87 of 242 patients in the purged group and 93 of 243 patients in the non-purged group) and consolidation (131 of 177 in the purged group vs 145 of 191 in the non-purged group). Serious adverse events during induction were grade 3 or higher decreased cardiac function (four of 242 in the purged group and five of 243 in the non-purged group) and elevated creatinine (five of 242 in the purged group and six of 243 non-purged group) and during consolidation were sinusoidal obstructive syndrome (12 of 177 in the purged group and 17 of 191 in the non-purged group), acute vascular leak (11 of 177 in the purged group and nine of 191 in the non-purged group), and decreased cardiac function (one of 177 in the purged group and four of 191 in the non-purged group). INTERPRETATION Immunomagnetic purging of PBSC for autologous stem-cell transplantation did not improve outcome, perhaps because of incomplete purging or residual tumour in patients. Non-purged PBSC are acceptable for support of myeloablative therapy of high-risk neuroblastoma.
Collapse
|
40
|
HARTOMO TRIBUDI, KOZAKI AIKO, HASEGAWA DAIICHIRO, VAN HUYEN PHAM THI, YAMAMOTO NOBUYUKI, SAITOH ATSURO, ISHIDA TOSHIAKI, KAWASAKI KEIICHIRO, KOSAKA YOSHIYUKI, OHASHI HIROKI, YAMAMOTO TOMOTO, MORIKAWA SATORU, HIRASE SATOSHI, KUBOKAWA IKUKO, MORI TAKESHI, YANAI TOMOKO, HAYAKAWA AKIRA, TAKESHIMA YASUHIRO, IIJIMA KAZUMOTO, MATSUO MASAFUMI, NISHIO HISAHIDE, NISHIMURA NORIYUKI. Minimal residual disease monitoring in neuroblastoma patients based on the expression of a set of real-time RT-PCR markers in tumor-initiating cells. Oncol Rep 2013; 29:1629-36. [DOI: 10.3892/or.2013.2286] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 12/28/2012] [Indexed: 11/06/2022] Open
|
41
|
Owens C, Irwin M. Neuroblastoma: the impact of biology and cooperation leading to personalized treatments. Crit Rev Clin Lab Sci 2012; 49:85-115. [PMID: 22646747 DOI: 10.3109/10408363.2012.683483] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Neuroblastoma is the most common extra-cranial solid tumor in children. It is a heterogeneous disease, consisting of neural crest-derived tumors with remarkably different clinical behaviors. It can present in a wide variety of ways, including lesions which have the potential to spontaneously regress, or as an extremely aggressive form of metastatic cancer which is resistant to all forms of modern therapy. They can arise anywhere along the sympathetic nervous system. The median age of presentation is approximately 18 months of age. Urinary catecholamines (HVA and VMA) are extremely sensitive and specific tumor markers and are used in diagnosis, treatment response assessment and post-treatment surveillance. The largest national treatment groups from North America, Europe and Japan have formed the International Neuroblastoma Risk Group Task Force (INRG) to identify prognostic factors, to understand the mechanisms of tumorigenesis in this rare disease and to develop multi-modality therapies to improve outcomes and decrease treatment-related toxicities. This international cooperation has resulted in a significant leap in our understanding of the molecular pathogenesis of neuroblastoma. Lower staged disease can be cured if the lesion is resectable. Treatment of unresectable disease (loco-regional and metastatic) is stratified depending on clinical features (age at presentation, staging investigations) and specific tumor biological markers that include histopathological analyses, chromosomal abnormalities and the quantification of expression of an oncogene (MYCN). Modern treatment of high-risk neuroblastoma is the paradigm for the evolution of therapy in pediatric oncology. Outcomes have improved substantially with multi-modality therapy, including chemotherapy, surgery, radiation therapy, myeloablative therapy with stem cell transplant, immunotherapy and differentiation therapy; these comprise the standard of care worldwide. In addition, newer targeted therapies are being tested in phase I/II trials. If successful these agents will be incorporated into mainstream treatment programs.
Collapse
Affiliation(s)
- Cormac Owens
- Division of Haematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | |
Collapse
|
42
|
Affiliation(s)
- Stephan A Grupp
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.
| | | | | |
Collapse
|