1
|
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
|
2
|
Chiangjong W, Panachan J, Keadsanti S, Newburg DS, Morrow AL, Hongeng S, Chutipongtanate S. Development of red blood cell-derived extracellular particles as a biocompatible nanocarrier of microRNA-204 (REP-204) to harness anti-neuroblastoma effect. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 60:102760. [PMID: 38852882 DOI: 10.1016/j.nano.2024.102760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/14/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in the pediatric population with a high degree of heterogeneity in clinical outcomes. Upregulation of the tumor suppressor miR-204 in neuroblastoma is associated with good prognosis. Although miR-204 has been recognized as a potential therapeutic candidate, its delivery is unavailable. We hypothesized that REP-204, the red blood cell-derived extracellular particles (REP) with miR-204 loading, can suppress neuroblastoma cells in vitro. After miR-204 loading by electroporation, REP-204, but not REP carriers, inhibited the viability, migration, and 3D spheroid growth of neuroblastoma cells regardless of MYCN amplification status. SWATH-proteomics revealed that REP-204 treatment may trigger a negative regulation of mRNA splicing by the spliceosome, suppression of amino acid metabolism and protein production, and prevent SLIT/ROBO signaling-mediated cell migration, to halt neuroblastoma tumor growth and metastasis. The therapeutic efficacy of REP-204 should be further investigated in preclinical models and clinical studies.
Collapse
Affiliation(s)
- Wararat Chiangjong
- Pediatric Translational Research Unit, Division of Evidence-based Pediatrics and Research, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand.
| | - Jirawan Panachan
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Sujitra Keadsanti
- Center of Excellence for Antibody Research, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - David S Newburg
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, United States
| | - Ardythe L Morrow
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, United States; Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Somchai Chutipongtanate
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, United States; Extracellular Vesicle Working Group, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA.
| |
Collapse
|
3
|
Borenäs M, Umapathy G, Lind DE, Lai WY, Guan J, Johansson J, Jennische E, Schmidt A, Kurhe Y, Gabre JL, Aniszewska A, Strömberg A, Bemark M, Hall MN, den Eynden JV, Hallberg B, Palmer RH. ALK signaling primes the DNA damage response sensitizing ALK-driven neuroblastoma to therapeutic ATR inhibition. Proc Natl Acad Sci U S A 2024; 121:e2315242121. [PMID: 38154064 PMCID: PMC10769851 DOI: 10.1073/pnas.2315242121] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
High-risk neuroblastoma (NB) is a significant clinical challenge. MYCN and Anaplastic Lymphoma Kinase (ALK), which are often involved in high-risk NB, lead to increased replication stress in cancer cells, suggesting therapeutic strategies. We previously identified an ATR (ataxia telangiectasia and Rad3-related)/ALK inhibitor (ATRi/ALKi) combination as such a strategy in two independent genetically modified mouse NB models. Here, we identify an underlying molecular mechanism, in which ALK signaling leads to phosphorylation of ATR and CHK1, supporting an effective DNA damage response. The importance of ALK inhibition is supported by mouse data, in which ATRi monotreatment resulted in a robust initial response, but subsequent relapse, in contrast to a 14-d ALKi/ATRi combination treatment that resulted in a robust and sustained response. Finally, we show that the remarkable response to the 14-d combined ATR/ALK inhibition protocol reflects a robust differentiation response, reprogramming tumor cells to a neuronal/Schwann cell lineage identity. Our results identify an ability of ATR inhibition to promote NB differentiation and underscore the importance of further exploring combined ALK/ATR inhibition in NB, particularly in high-risk patient groups with oncogene-induced replication stress.
Collapse
Affiliation(s)
- Marcus Borenäs
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
| | - Ganesh Umapathy
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
| | - Dan E. Lind
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
| | - Wei-Yun Lai
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
| | - Jikui Guan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
| | - Joel Johansson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
| | - Eva Jennische
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, Basel University, Basel4056, Switzerland
| | - Yeshwant Kurhe
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
| | - Jonatan L. Gabre
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
- Department of Human Structure and Repair, Anatomy and Embryology Unit, Ghent University, Ghent9000, Belgium
| | - Agata Aniszewska
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
| | - Anneli Strömberg
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
| | - Mats Bemark
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg SE-405 30, Sweden
| | | | - Jimmy Van den Eynden
- Department of Human Structure and Repair, Anatomy and Embryology Unit, Ghent University, Ghent9000, Belgium
| | - Bengt Hallberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
| | - Ruth H. Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, GothenburgSE-405 30, Sweden
| |
Collapse
|
4
|
Grupel D, Sagi O, Nissan I, Grossman R, Yair M, Moran-Gilad J, Danino D. Challenging diagnosis of Mycolicibacterium cosmeticum/canariasense infection: A case report and literature review. J Clin Tuberc Other Mycobact Dis 2023; 33:100393. [PMID: 37637323 PMCID: PMC10450513 DOI: 10.1016/j.jctube.2023.100393] [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: 08/29/2023] Open
Abstract
We present the case of an immunocompromised child with Mycolicibacterium cosmeticum/ canariasense infection. Our case highlights the difficulty in adequate speciation. Most isolates described in the literature were identified using 16 s-rRNA PCR, which if performed on our sample would at best be inconclusive. Misidentifications could have a real impact on the body of evidence collected on these isolates thus far.
Collapse
Affiliation(s)
- Daniel Grupel
- Soroka University Medical Center, Infectious Disease Unit, Beer-Sheva, Israel
- Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel
| | - Orly Sagi
- Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel
- Soroka University Medical Center, Microbiology Laboratory, Beer-Sheva, Israel
| | - Israel Nissan
- Public Health Services, National Public Health Laboratory, Ministry of Health, Tel Aviv, Israel
| | - Rona Grossman
- Public Health Services, National Public Health Laboratory, Ministry of Health, Tel Aviv, Israel
| | - Motro Yair
- Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel
| | - Jacob Moran-Gilad
- Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel
- Soroka University Medical Center, Microbiology Laboratory, Beer-Sheva, Israel
| | - Dana Danino
- Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel
- Soroka University Medical Center, Pediatric Infectious Disease Unit, Beer-Sheva, Israel
| |
Collapse
|
5
|
Chicco D, Haupt R, Garaventa A, Uva P, Luksch R, Cangelosi D. Computational intelligence analysis of high-risk neuroblastoma patient health records reveals time to maximum response as one of the most relevant factors for outcome prediction. Eur J Cancer 2023; 193:113291. [PMID: 37708628 DOI: 10.1016/j.ejca.2023.113291] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 07/24/2023] [Accepted: 08/09/2023] [Indexed: 09/16/2023]
Abstract
OBJECTIVE Seek new candidate prognostic markers for neuroblastoma outcome, relapse or progression. MATERIALS AND METHODS In this multicentre and retrospective study, Random Forests coupled with recursive feature elimination techniques were applied to electronic records (55 clinical features) of 3034 neuroblastoma patients. To assess model performance and feature importance, dataset was split into a training set (80%) and a test set (20%). RESULTS In the test set, the mean Matthews correlation coefficient for the Random Forests models was greater than 0.46. Feature importance analysis revealed that, together with maximum response to first-line treatment (D_MAX_RESP), time to maximum response to first-line treatment (TIME_MAX_RESP.days) is a relevant predictor of both patients' outcome and relapse\progression. We showed the prognostic value of the max response to first-line treatment in clinically relevant subsets of high-, intermediate-, and low-risk patients for both overall and relapse-free survival (Log-rank p-value<0.0001). In high-risk patients older than 18 months and stage 4 tumour achieving a complete response or very good partial response, patients who exhibited a D_MAX_RESP greater than 9 months showed a better prognosis with respect to patients achieving D_MAX_RESP earlier than 9 months (overall survival): hazard ratio 3.3 95% confidence interval 1.8-5.9, Log-rank p-value p < 0.0001; relapse-free survival: 3.2 95%CI 1.8-5.6, Log-rank p-value p < 0.0001). CONCLUSION Our findings evidence the emerging role of the TIME_MAX_RESP.days in addition to the D_MAX_RESP as relevant predictors of outcome and relapse\progression in neuroblastoma with potential clinical impact on the management and treatment of patients.
Collapse
Affiliation(s)
- Davide Chicco
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Dipartimento di Informatica Sistemistica e Comunicazione, Università di Milano-Bicocca, Milan, Italy
| | - Riccardo Haupt
- DOPO Clinic, Department of Hematology/Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Paolo Uva
- Unità di Bioinformatica Clinica, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Roberto Luksch
- S.C. Pediatria oncologica, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Davide Cangelosi
- Unità di Bioinformatica Clinica, IRCCS Istituto Giannina Gaslini, Genoa, Italy.
| |
Collapse
|
6
|
Muñoz JP, Larrosa C, Chamorro S, Perez-Jaume S, Simao M, Sanchez-Sierra N, Varo A, Gorostegui M, Castañeda A, Garraus M, Lopez-Miralles S, Mora J. Early Salvage Chemo-Immunotherapy with Irinotecan, Temozolomide and Naxitamab Plus GM-CSF (HITS) for Patients with Primary Refractory High-Risk Neuroblastoma Provide the Best Chance for Long-Term Outcomes. Cancers (Basel) 2023; 15:4837. [PMID: 37835531 PMCID: PMC10571514 DOI: 10.3390/cancers15194837] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/26/2023] [Accepted: 10/01/2023] [Indexed: 10/15/2023] Open
Abstract
Patients with high-risk neuroblastoma (HR-NB) who are unable to achieve a complete response (CR) to induction therapy have worse outcomes. We investigated the combination of humanized anti-GD2 mAb naxitamab (Hu3F8), irinotecan (I), temozolomide (T), and sargramostim (GM-CSF)-HITS-against primary resistant HR-NB. Eligibility criteria included having a measurable chemo-resistant disease at the end of induction (EOI) treatment. Patients were excluded if they had progressive disease (PD) during induction. Prior anti-GD2 mAb and/or I/T therapy was permitted. Each cycle, administered four weeks apart, comprised Irinotecan 50 mg/m2/day intravenously (IV) plus Temozolomide 150 mg/m2/day orally (days 1-5); naxitamab 2.25 mg/kg/day IV on days 2, 4, 8 and 10, (total 9 mg/kg or 270 mg/m2 per cycle), and GM-CSF 250 mg/m2/day subcutaneously was used (days 6-10). Toxicity was measured using CTCAE v4.0 and responses through the modified International Neuroblastoma Response Criteria (INRC). Thirty-four patients (median age at treatment initiation, 4.9 years) received 164 (median 4; 1-12) HITS cycles. Toxicities included myelosuppression and diarrhea, which was expected with I/T, and pain and hypertension, expected with naxitamab. Grade ≥3-related toxicities occurred in 29 (85%) of the 34 patients; treatment was outpatient. The best responses were CR = 29% (n = 10); PR = 3% (n = 1); SD = 53% (n = 18); PD = 5% (n = 5). For cohort 1 (early treatment), the best responses were CR = 47% (n = 8) and SD = 53% (n = 9). In cohort 2 (late treatment), the best responses were CR = 12% (n = 2); PR = 6% (n = 1); SD = 53% (n = 9); and PD = 29% (n = 5). Cohort 1 had a 3-year OS of 84.8% and EFS 54.4%, which are statistically significant improvements (EFS p = 0.0041 and OS p = 0.0037) compared to cohort 2. In conclusion, naxitamab-based chemo-immunotherapy is effective against primary chemo-resistant HR-NB, increasing long-term outcomes when administered early during the course of treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jaume Mora
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (J.P.M.); (C.L.); (S.C.); (S.P.-J.); (M.S.); (N.S.-S.); (A.V.); (M.G.); (A.C.); (M.G.); (S.L.-M.)
| |
Collapse
|
7
|
Zhu K, Xia Y, Tian X, He Y, Zhou J, Han R, Guo H, Song T, Chen L, Tian X. Characterization and therapeutic perspectives of differentiation-inducing therapy in malignant tumors. Front Genet 2023; 14:1271381. [PMID: 37745860 PMCID: PMC10514561 DOI: 10.3389/fgene.2023.1271381] [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: 08/02/2023] [Accepted: 08/30/2023] [Indexed: 09/26/2023] Open
Abstract
Cancer is a major public health issue globally and is one of the leading causes of death. Although available treatments improve the survival rate of some cases, many advanced tumors are insensitive to these treatments. Cancer cell differentiation reverts the malignant phenotype to its original state and may even induce differentiation into cell types found in other tissues. Leveraging differentiation-inducing therapy in high-grade tumor masses offers a less aggressive strategy to curb tumor progression and heightens chemotherapy sensitivity. Differentiation-inducing therapy has been demonstrated to be effective in a variety of tumor cells. For example, differentiation therapy has become the first choice for acute promyelocytic leukemia, with the cure rate of more than 90%. Although an appealing concept, the mechanism and clinical drugs used in differentiation therapy are still in their nascent stage, warranting further investigation. In this review, we examine the current differentiation-inducing therapeutic approach and discuss the clinical applications as well as the underlying biological basis of differentiation-inducing agents.
Collapse
Affiliation(s)
- Kangwei Zhu
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuren Xia
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xindi Tian
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuchao He
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jun Zhou
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Chiyoda, Japan
| | - Ruyu Han
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hua Guo
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Tianqiang Song
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lu Chen
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiangdong Tian
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
8
|
Wieczorek A, Żebrowska U, Ussowicz M, Sokół A, Stypińska M, Dembowska-Bagińska B, Pawińska-Wąsikowska K, Balwierz W. Dinutuximab Beta Maintenance Therapy in Patients with High-Risk Neuroblastoma in First-Line and Refractory/Relapsed Settings-Real-World Data. J Clin Med 2023; 12:5252. [PMID: 37629294 PMCID: PMC10455178 DOI: 10.3390/jcm12165252] [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: 07/13/2023] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Dinutuximab beta is approved for the maintenance treatment of patients with high-risk neuroblastoma (HR-NB), including patients with relapsed/refractory (R/R) disease. However, the data on its use in real-world clinical practice is limited. We retrospectively reviewed the clinical records of 54 patients with HR-NB who received maintenance therapy with dinutuximab beta in first-line (37 patients) or R/R settings (17 patients) at three centers in Poland. Of the 37 patients who received first-line treatment, twenty-eight had a complete response, two had a partial response, three had progressive disease, and four relapsed at the end of treatment. The median overall survival (OS) was 24.37 months, and the three-year progression-free survival (PFS) and OS were 0.63 and 0.80, respectively. Of the 17 patients in the R/R group, 11 had a complete response, two had a partial response, one had stable disease, and three had progressive disease or relapsed at the end of treatment. The median OS was 33.1 months and the three-year PFS and OS were 0.75 and 0.86, respectively. Treatment was generally well tolerated, including in patients with co-morbidities and those who had experienced toxicities with previous therapies. These findings demonstrate that the use of dinutuximab beta is feasible and beneficial as a first-line or R/R treatment in routine clinical practice in Poland.
Collapse
Affiliation(s)
- Aleksandra Wieczorek
- Department of Pediatric Oncology and Hematology, Jagiellonian University Medical College, 30-663 Krakow, Poland
- Department of Pediatric Oncology and Hematology, University Children’s Hospital of Krakow, 30-663 Krakow, Poland
| | - Urszula Żebrowska
- Department of Pediatric Oncology and Hematology, University Children’s Hospital of Krakow, 30-663 Krakow, Poland
| | - Marek Ussowicz
- Department of Pediatric Bone Marrow Transplantation, Oncology and Hematology, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Agnieszka Sokół
- Department of Pediatric Bone Marrow Transplantation, Oncology and Hematology, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Marzena Stypińska
- Department of Oncology, Children Memorial Health Institute, 04-730 Warsaw, Poland
| | | | - Katarzyna Pawińska-Wąsikowska
- Department of Pediatric Oncology and Hematology, Jagiellonian University Medical College, 30-663 Krakow, Poland
- Department of Pediatric Oncology and Hematology, University Children’s Hospital of Krakow, 30-663 Krakow, Poland
| | - Walentyna Balwierz
- Department of Pediatric Oncology and Hematology, Jagiellonian University Medical College, 30-663 Krakow, Poland
- Department of Pediatric Oncology and Hematology, University Children’s Hospital of Krakow, 30-663 Krakow, Poland
| |
Collapse
|
9
|
Rahavi SM, Aletaha M, Farrokhi A, Lorentzian A, Lange PF, Maxwell CA, Lim CJ, Reid GSD. Adaptation of the Th-MYCN Mouse Model of Neuroblastoma for Evaluation of Disseminated Disease. Int J Mol Sci 2023; 24:12071. [PMID: 37569447 PMCID: PMC10419036 DOI: 10.3390/ijms241512071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
High-risk neuroblastoma remains a profound clinical challenge that requires eradication of neuroblastoma cells from a variety of organ sites, including bone marrow, liver, and CNS, to achieve a cure. While preclinical modeling is a powerful tool for the development of novel cancer therapies, the lack of widely available models of metastatic neuroblastoma represents a significant barrier to the development of effective treatment strategies. To address this need, we report a novel luciferase-expressing derivative of the widely used Th-MYCN mouse. While our model recapitulates the non-metastatic neuroblastoma development seen in the parental transgenic strain, transplantation of primary tumor cells from disease-bearing mice enables longitudinal monitoring of neuroblastoma growth at distinct sites in immune-deficient or immune-competent recipients. The transplanted tumors retain GD2 expression through many rounds of serial transplantation and are sensitive to GD2-targeted immune therapy. With more diverse tissue localization than is seen with human cell line-derived xenografts, this novel model for high-risk neuroblastoma could contribute to the optimization of immune-based treatments for this deadly disease.
Collapse
Affiliation(s)
- Seyed M. Rahavi
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Maryam Aletaha
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Ali Farrokhi
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, 950 W28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Amanda Lorentzian
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Philipp F. Lange
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pathology, University of British Columbia, 950 W28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Christopher A. Maxwell
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, 950 W28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Chinten James Lim
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, 950 W28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Gregor S. D. Reid
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, 950 W28th Avenue, Vancouver, BC V5Z 4H4, Canada
| |
Collapse
|
10
|
Wieczorek A, Zaniewska-Tekieli A, Ehlert K, Pawinska-Wasikowska K, Balwierz W, Lode H. Dinutuximab beta combined with chemotherapy in patients with relapsed or refractory neuroblastoma. Front Oncol 2023; 13:1082771. [PMID: 36816982 PMCID: PMC9936065 DOI: 10.3389/fonc.2023.1082771] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Prognosis in children with refractory and relapsed high-risk neuroblastoma is poor. Only a minority of patients obtain remission when treated with second-line chemotherapy regimens. Chemotherapy combined with anti-GD2 antibodies has previously been shown to increase response and survival rates. We retrospectively analyzed a cohort of 25 patients with relapsed or refractory high-risk neuroblastoma who were treated with irinotecan/temozolomide chemotherapy in combination with the anti-GD2 antibody dinutuximab beta. The therapy resulted in an objective response rate of 64%, with 32% of patients achieving a complete response. Response to treatment was observed in patients with refractory disease (n=5) and those with first (n=12) or consecutive (n=8) relapses, including patients with progressing disease. In four patients, best response was achieved after more than 5 cycles, suggesting that some patients may benefit from prolonged chemotherapy and dinutuximab beta treatment. Fourteen of our 25 patients had previously received dinutuximab beta, four of whom achieved complete response and six partial response (objective response rate 71%). The therapy was well tolerated, even in heavily pre-treated patients and those who had previously received dinutuximab beta treatment. Toxicities were comparable to those previously reported for the individual therapies, and no discontinuations due to toxicities occurred. Combination of chemotherapy with dinutuximab beta is a promising treatment option for patients with relapsed or refractory high-risk neuroblastoma and should be further explored in clinical studies.
Collapse
Affiliation(s)
- Aleksandra Wieczorek
- Pediatric Oncology and Hematology, Jagiellonian University Medical College, Krakow, Poland
- Pediatric Oncology and Hematology, University Children's Hospital of Krakow, Krakow, Poland
| | - Anna Zaniewska-Tekieli
- Pediatric Oncology and Hematology, University Children's Hospital of Krakow, Krakow, Poland
| | - Karoline Ehlert
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Katarzyna Pawinska-Wasikowska
- Pediatric Oncology and Hematology, Jagiellonian University Medical College, Krakow, Poland
- Pediatric Oncology and Hematology, University Children's Hospital of Krakow, Krakow, Poland
| | - Walentyna Balwierz
- Pediatric Oncology and Hematology, Jagiellonian University Medical College, Krakow, Poland
- Pediatric Oncology and Hematology, University Children's Hospital of Krakow, Krakow, Poland
| | - Holger Lode
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
11
|
Clark-Corrigall J, Myssina S, Michaelis M, Cinatl J, Ahmed S, Carr-Wilkinson J, Carr-Wilkinson J. Elevated Expression of LGR5 and WNT Signaling Factors in Neuroblastoma Cells With Acquired Drug Resistance. Cancer Invest 2023; 41:173-182. [PMID: 36318235 DOI: 10.1080/07357907.2022.2136682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Neuroblastoma (NB) is a pediatric solid cancer with high fatality, relapses, and acquired resistance to chemotherapy, that requires new therapeutic approaches to improve survival. LGR5 is a receptor that potentiates WNT/signaling pathway and has been reported to promote development and survival in several adult cancers. In this study we investigated LGR5 expression in a panel of NB cell lines with acquired resistance to vincristine or doxorubicin. We show LGR5-LRP6 cooperation with enhanced expression in drug resistant NB cell lines compared to parental cells, suggesting a role for LGR5 in the emergence of drug resistance, warranting further investigation.
Collapse
Affiliation(s)
- John Clark-Corrigall
- School of Nursing and Health Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, UK
| | - Svetlana Myssina
- School of Nursing and Health Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, UK
| | - Martin Michaelis
- School of Biosciences and Industrial Biotechnology Centre, University of Kent, Canterbury, UK
| | - Jindrich Cinatl
- Institut für Medizinische Virologie, Klinikum der Goethe-Universität, Frankfurt am Main, Germany
| | - Shafiq Ahmed
- School of Medicine, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, UK
| | - Jane Carr-Wilkinson
- School of Medicine, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, UK
| | - Jane Carr-Wilkinson
- School of Medicine, Faculty of Health Sciences and Wellbeing, University of Sunderland, United Kingdom
| |
Collapse
|
12
|
Olgun N, Cecen E, Ince D, Kizmazoglu D, Baysal B, Onal A, Ozdogan O, Guleryuz H, Cetingoz R, Demiral A, Olguner M, Celik A, Kamer S, Ozer E, Altun Z, Aktas S. Dinutuximab beta plus conventional chemotherapy for relapsed/refractory high-risk neuroblastoma: A single-center experience. Front Oncol 2022; 12:1041443. [PMID: 36620564 PMCID: PMC9816792 DOI: 10.3389/fonc.2022.1041443] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Background Relapsed/refractory high-risk neuroblastoma has a dismal prognosis. Anti-GD2-mediated chemo-immunotherapy has a notable anti-tumor activity in patients with relapsed/refractory high-risk neuroblastoma. The purpose of this study was to analyze the efficacy and safety of the combination of immunotherapy with dinutuximab beta (DB) and chemotherapy in patients with relapsed/refractory high-risk neuroblastoma. Methods All patients received the Turkish Pediatric Oncology Group NB 2009 national protocol for HR-NB treatment at the time of diagnosis. Salvage treatments were administered after progression or relapse. The patients who could not achieve remission in primary or metastatic sites were included in the study. The most common chemotherapy scheme was irinotecan and temozolomide. DB was administered intravenously for 10 days through continuous infusion with 10 mg/m2 per day. The patients received 2 to 14 successive cycles with duration of 28 days each. Disease assessment was performed after cycles 2, 4, and 6 and every 2 to 3 cycles thereafter. Results Between January 2020 and March 2022, nineteen patients received a total of 125 cycles of DB and chemotherapy. Objective responses were achieved in 12/19 (63%) patients, including complete remission in 6/19 and partial response in 6/19. Stable disease was observed in two patients. The remaining five patients developed bone/bone marrow and soft tissue progression after 2-4 cycles of treatment. The most common Grade ≥3 toxicities were leukopenia, thrombocytopenia, hypertransaminasemia, fever, rash/itching and capillary leak syndrome, respectively. Conclusion Our study results suggest that DB-based chemo-immunotherapy seems to be suitable with encouraging response rates in patients with relapsed/refractory high-risk neuroblastoma.
Collapse
Affiliation(s)
- Nur Olgun
- Department of Pediatric Oncology, Dokuz Eylul University Institute of Oncology, Izmir, Türkiye,*Correspondence: Nur Olgun,
| | - Emre Cecen
- Department of Pediatric Oncology, Dokuz Eylul University Institute of Oncology, Izmir, Türkiye
| | - Dilek Ince
- Department of Pediatric Oncology, Dokuz Eylul University Institute of Oncology, Izmir, Türkiye
| | - Deniz Kizmazoglu
- Department of Pediatric Oncology, Dokuz Eylul University Institute of Oncology, Izmir, Türkiye
| | - Birsen Baysal
- Department of Pediatric Oncology, Dokuz Eylul University Institute of Oncology, Izmir, Türkiye
| | - Ayse Onal
- Department of Pediatric Oncology, Dokuz Eylul University Institute of Oncology, Izmir, Türkiye
| | - Ozhan Ozdogan
- Department of Nuclear Medicine, Dokuz Eylul University School of Medicine, Izmir, Türkiye
| | - Handan Guleryuz
- Department of Radiology, Dokuz Eylul University School of Medicine, Izmir, Türkiye
| | - Riza Cetingoz
- Department of Radiation Oncology, Dokuz Eylul University School of Medicine, Izmir, Türkiye
| | - Ayse Demiral
- Department of Radiation Oncology, Dokuz Eylul University School of Medicine, Izmir, Türkiye
| | - Mustafa Olguner
- Department of Pediatric Surgery, Dokuz Eylul University School of Medicine, Izmir, Türkiye
| | - Ahmet Celik
- Department of Pediatric Surgery, Ege University School of Medicine, Izmir, Türkiye
| | - Serra Kamer
- Department of Radiation Oncology, Ege University School of Medicine, Izmir, Türkiye
| | - Erdener Ozer
- Department of Pathology, Dokuz Eylul University School of Medicine, Izmir, Türkiye
| | - Zekiye Altun
- Department of Basic Oncology, Dokuz Eylul University Institute of Oncology, Izmir, Türkiye
| | - Safiye Aktas
- Department of Basic Oncology, Dokuz Eylul University Institute of Oncology, Izmir, Türkiye
| |
Collapse
|
13
|
Yan Z, Liu Q, Cao Z, Wang J, Zhang H, Liu J, Zou L. Multi-omics integration reveals a six-malignant cell maker gene signature for predicting prognosis in high-risk neuroblastoma. Front Neuroinform 2022; 16:1034793. [DOI: 10.3389/fninf.2022.1034793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
BackgroundNeuroblastoma is the most common extracranial solid tumor of childhood, arising from the sympathetic nervous system. High-risk neuroblastoma (HRNB) remains a major therapeutic challenge with low survival rates despite the intensification of therapy. This study aimed to develop a malignant-cell marker gene signature (MMGS) that might serve as a prognostic indicator in HRNB patients.MethodsMulti-omics datasets, including mRNA expression (single-cell and bulk), DNA methylation, and clinical information of HRNB patients, were used to identify prognostic malignant cell marker genes. MMGS was established by univariate Cox analysis, LASSO, and stepwise multivariable Cox regression analysis. Kaplan–Meier (KM) curve and time-dependent receiver operating characteristic curve (tROC) were used to evaluate the prognostic value and performance of MMGS, respectively. MMGS further verified its reliability and accuracy in the independent validation set. Finally, the characteristics of functional enrichment, tumor immune features, and inflammatory activity between different MMGS risk groups were also investigated.ResultsWe constructed a prognostic model consisting of six malignant cell maker genes (MAPT, C1QTNF4, MEG3, NPW, RAMP1, and CDT1), which stratified patients into ultra-high-risk (UHR) and common-high-risk (CHR) group. Patients in the UHR group had significantly worse overall survival (OS) than those in the CHR group. MMGS was verified as an independent predictor for the OS of HRNB patients. The area under the curve (AUC) values of MMGS at 1-, 3-, and 5-year were 0.78, 0.693, and 0.618, respectively. Notably, functional enrichment, tumor immune features, and inflammatory activity analyses preliminarily indicated that the poor prognosis in the UHR group might result from the dysregulation of the metabolic process and immunosuppressive microenvironment.ConclusionThis study established a novel six-malignant cell maker gene prognostic model that can be used to predict the prognosis of HRNB patients, which may provide new insight for the treatment and personalized monitoring of HRNB patients.
Collapse
|
14
|
Abbas AA, Samkari AMN. High-Risk Neuroblastoma: Poor Outcomes Despite Aggressive Multimodal
Therapy. CURRENT CANCER THERAPY REVIEWS 2022. [DOI: 10.2174/1573394717666210805114226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
Neuroblastoma (NBL) is a highly malignant embryonal tumor that originates from the
primordial neural crest cells. NBL is the most common tumor in infants and the most common extracranial
solid tumor in children. The tumor is more commonly diagnosed in children of 1-4 years
of age. NBL is characterized by enigmatic clinical behavior that ranges from spontaneous regression
to an aggressive clinical course leading to frequent relapses and death. Based on the likelihood
of progression and relapse, the International Neuroblastoma Risk Group classification system categorized
NBL into very low risk, low risk, intermediate risk, and high risk (HR) groups. HR NBL is
defined based on the patient's age (> 18 months), disease metastasis, tumor histology, and MYCN
gene amplification. HR NBL is diagnosed in nearly 40% of patients, mainly those > 18 months of
age, and is associated with aggressive clinical behavior. Treatment strategies involve the use of intensive
chemotherapy (CTR), surgical resection, high dose CTR with hematopoietic stem cell support,
radiotherapy, biotherapy, and immunotherapy with Anti-ganglioside 2 monoclonal antibodies.
Although HR NBL is now better characterized and aggressive multimodal therapy is applied, the
outcomes of treatment are still poor, with overall survival and event-free survival of approximately
40% and 30% at 3-years, respectively. The short and long-term side effects of therapy are tremendous.
HR NBL carries a high mortality rate accounting for nearly 15% of pediatric cancer deaths.
However, most mortalities are attributed to the high frequency of disease relapse (50%) and disease
reactiveness to therapy (20%). Newer treatment strategies are therefore urgently needed. Recent
discoveries in the field of biology and molecular genetics of NBL have led to the identification
of several targets that can improve the treatment results. In this review, we discuss the different
aspects of the epidemiology, biology, clinical presentations, diagnosis, and treatment of HR
NBL, in addition to the recent developments in the management of the disease.
Collapse
Affiliation(s)
- Adil Abdelhamed Abbas
- College of Medicine King Saud bin Abdulaziz, University for Health Sciences Consultant Pediatric Hematology / Oncology
& BMT The Pediatric Hematology/Oncology Section Princess Nourah Oncology Centre King Abdulaziz Medical
City, Jeddah, Saudi Arabia
| | - Alaa Mohammed Noor Samkari
- College of Medicine King Saud bin Abdulaziz, University for Health Sciences Consultant
Anatomical Pathologist Department of Laboratory Medicine King Abdulaziz Medical City, Jeddah, Saudi Arabia
| |
Collapse
|
15
|
Treis D, Umapathy G, Fransson S, Guan J, Mendoza-García P, Siaw JT, Wessman S, Gordon Murkes L, Stenman JJE, Djos A, Elfman LHM, Johnsen JI, Hallberg B, Palmer RH, Martinsson T, Kogner P. Sustained Response to Entrectinib in an Infant With a Germline ALKAL2 Variant and Refractory Metastatic Neuroblastoma With Chromosomal 2p Gain and Anaplastic Lymphoma Kinase and Tropomyosin Receptor Kinase Activation. JCO Precis Oncol 2022; 6:e2100271. [PMID: 35085006 PMCID: PMC8830523 DOI: 10.1200/po.21.00271] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Personalized molecular workup enabled successful ALK inhibitor treatment in a child with resistant neuroblastoma.![]()
Collapse
Affiliation(s)
- Diana Treis
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, and Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Ganesh Umapathy
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Susanne Fransson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jikui Guan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Patricia Mendoza-García
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joachim T. Siaw
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sandra Wessman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pathology, Karolinska University Hospital, Stockholm, Sweden
| | - Lena Gordon Murkes
- Department of Pediatric Radiology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Jakob J. E. Stenman
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, and Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
- Department of Pediatric Surgery, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Djos
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lotta H. M. Elfman
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, and Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, and Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Bengt Hallberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ruth H. Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tommy Martinsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, and Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
- Per Kogner, MD, PhD; Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet , Tomtebodavägen 18A, 171 77 Stockholm, Sweden;
| |
Collapse
|
16
|
Szydzik J, Lind DE, Arefin B, Kurhe Y, Umapathy G, Siaw JT, Claeys A, Gabre JL, Van den Eynden J, Hallberg B, Palmer RH. ATR inhibition enables complete tumour regression in ALK-driven NB mouse models. Nat Commun 2021; 12:6813. [PMID: 34819497 PMCID: PMC8613282 DOI: 10.1038/s41467-021-27057-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 11/03/2021] [Indexed: 01/23/2023] Open
Abstract
High-risk neuroblastoma (NB) often involves MYCN amplification as well as mutations in ALK. Currently, high-risk NB presents significant clinical challenges, and additional therapeutic options are needed. Oncogenes like MYCN and ALK result in increased replication stress in cancer cells, offering therapeutically exploitable options. We have pursued phosphoproteomic analyses highlighting ATR activity in ALK-driven NB cells, identifying the BAY1895344 ATR inhibitor as a potent inhibitor of NB cell growth and proliferation. Using RNA-Seq, proteomics and phosphoproteomics we characterize NB cell and tumour responses to ATR inhibition, identifying key components of the DNA damage response as ATR targets in NB cells. ATR inhibition also produces robust responses in mouse models. Remarkably, a 2-week combined ATR/ALK inhibition protocol leads to complete tumor regression in two independent genetically modified mouse NB models. These results suggest that NB patients, particularly in high-risk groups with oncogene-induced replication stress, may benefit from ATR inhibition as therapeutic intervention. Effective therapeutic options are still needed in neuroblastoma treatment. Here, the authors, through a comprehensive proteomics analysis, identify ATR as a potential therapeutic target of neuroblastoma and demonstrate the efficacy of the ATR inhibitor BAY1895344 in combination with the ALK tyrosine kinase inhibitor lorlatinib.
Collapse
Affiliation(s)
- Joanna Szydzik
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden
| | - Dan E Lind
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden
| | - Badrul Arefin
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden
| | - Yeshwant Kurhe
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden
| | - Ganesh Umapathy
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden
| | - Joachim Tetteh Siaw
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden
| | - Arne Claeys
- Department of Human Structure and Repair, Anatomy and Embryology Unit, Ghent University, 9000, Ghent, Belgium
| | - Jonatan L Gabre
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden.,Department of Human Structure and Repair, Anatomy and Embryology Unit, Ghent University, 9000, Ghent, Belgium
| | - Jimmy Van den Eynden
- Department of Human Structure and Repair, Anatomy and Embryology Unit, Ghent University, 9000, Ghent, Belgium.
| | - Bengt Hallberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden.
| | - Ruth H Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden.
| |
Collapse
|
17
|
Merlos Rodrigo MA, Casar B, Michalkova H, Jimenez Jimenez AM, Heger Z, Adam V. Extending the Applicability of In Ovo and Ex Ovo Chicken Chorioallantoic Membrane Assays to Study Cytostatic Activity in Neuroblastoma Cells. Front Oncol 2021; 11:707366. [PMID: 34540673 PMCID: PMC8440826 DOI: 10.3389/fonc.2021.707366] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose The chick chorioallantoic membrane (CAM) assay can provide an alternative versatile, cost-effective, and ethically less controversial in vivo model for reliable screening of drugs. In the presented work, we demonstrate that CAM assay (in ovo and ex ovo) can be simply employed to delineate the effects of cisplatin (CDDP) and ellipticine (Elli) on neuroblastoma (Nbl) cells in terms of their growth and metastatic potential. Methods The Nbl UKF-NB-4 cell line was established from recurrent bone marrow metastases of high-risk Nbl (stage IV, MYCN amplification, 7q21 gain). Ex ovo and in ovo CAM assays were optimized to evaluate the antimetastatic activity of CDDP and Elli. Immunohistochemistry, qRT-PCR, and DNA isolation were performed. Results Ex ovo CAM assay was employed to study whether CDDP and Elli exhibit any inhibitory effects on growth of Nbl xenograft in ex ovo CAM assay. Under the optimal conditions, Elli and CDDP exhibited significant inhibition of the size of the primary tumor. To study the efficiency of CDDP and Elli to inhibit primary Nbl tumor growth, intravasation, and extravasation in the organs, we adapted the in ovo CAM assay protocol. In in ovo CAM assay, both studied compounds (CDDP and Elli) exhibited significant (p < 0.001) inhibitory activity against extravasation to all investigated organs including distal CAM. Conclusions Taken together, CAM assay could be a helpful and highly efficient in vivo approach for high-throughput screening of libraries of compounds with expected anticancer activities.
Collapse
Affiliation(s)
- Miguel Angel Merlos Rodrigo
- Research Group for Molecular Biology and Nanomedicine, Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia.,Central European Institute of Technology, Brno University of Technology, Brno, Czechia
| | - Berta Casar
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas (CSIC) - Universidad de Cantabria, Santander, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Hana Michalkova
- Research Group for Molecular Biology and Nanomedicine, Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia.,Central European Institute of Technology, Brno University of Technology, Brno, Czechia
| | - Ana Maria Jimenez Jimenez
- Research Group for Molecular Biology and Nanomedicine, Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia.,Central European Institute of Technology, Brno University of Technology, Brno, Czechia
| | - Zbynek Heger
- Research Group for Molecular Biology and Nanomedicine, Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia.,Central European Institute of Technology, Brno University of Technology, Brno, Czechia
| | - Vojtech Adam
- Research Group for Molecular Biology and Nanomedicine, Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia.,Central European Institute of Technology, Brno University of Technology, Brno, Czechia
| |
Collapse
|
18
|
Bauters T, Heenen D, Norga K, Van Damme A, Uyttebroeck A, Laureys G. Off-label drug use in paediatric haemato-oncology patients: financial implications and proposed solutions for Belgian patients. Eur J Pediatr 2021; 180:3067-3071. [PMID: 34137919 DOI: 10.1007/s00431-021-04150-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 11/28/2022]
Abstract
Treatment of children with cancer requires access to and reimbursement of effective drugs. Children with haemato-oncological diseases are often treated according to established treatment recommendations or in the framework of late-phase clinical trials. These often involve the use of drugs authorised for adults but which, however, have been used for many years in paediatrics with no perspective of authorisation in children. In Belgium, medicines are predominantly reimbursed based on their authorised indication. As a consequence, many drugs used in paediatric haemato-oncology are used off-label, despite their status of 'standard of care'. As reimbursement is often not available, alternative ways for funding need to be explored, which causes a significant administrative burden for healthcare providers and emotional distress for the parents. Solutions to organise a systematic reimbursement of standard of care off-label used drugs are described.Conclusion: A number of structural solutions are proposed, and we hope that they might guide health authorities to provide a solution to the problem caused by the lack of reimbursement of some standard of care medicines for children with cancer. What is Known: • Off-label drug use is frequently observed in paediatric haemato-oncology and compromises-in some countries-reimbursement. What is New: • An estimation of the impact of non-reimbursed drugs in Belgium is provided. • Some solutions are presented to overcome this problem in Belgium.
Collapse
Affiliation(s)
- T Bauters
- Tiene Bauters, Pediatric Haematology, Oncology and Stem Cell Transplantation, Ghent University Hospital, C. Heymanslaan 10, 9000, Ghent, Belgium.
| | - D Heenen
- KickCancer, Public Interest Foundation, Brussels, Belgium
| | - K Norga
- Pediatric Haematology/Oncology Unit, Queen Mathilde Mother and Child Center, Antwerp University, Antwerp, Belgium.,Hospital and Faculty of Medicine and Health Sciences, Antwerp University, Antwerp, Belgium
| | - A Van Damme
- Pediatric Haematology and Oncology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - A Uyttebroeck
- Pediatric Haematology and Oncology, University Hospitals Leuven, Leuven, Belgium
| | - G Laureys
- Tiene Bauters, Pediatric Haematology, Oncology and Stem Cell Transplantation, Ghent University Hospital, C. Heymanslaan 10, 9000, Ghent, Belgium
| |
Collapse
|
19
|
Saito Y, Urashima M, Takahashi Y, Ogawa A, Kiyotani C, Yuza Y, Koh K, Watanabe K, Kosaka Y, Goto H, Kikuta A, Okada K, Koga Y, Fujimura J, Inoue M, Sato A, Atsuta Y, Matsumoto K. Effect of high-dose chemotherapy plus stem cell rescue on the survival of patients with neuroblastoma modified by MYCN gene gain/amplification and remission status: a nationwide registration study in Japan. Bone Marrow Transplant 2021; 56:2173-2182. [PMID: 33911201 DOI: 10.1038/s41409-021-01303-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 03/24/2021] [Accepted: 04/09/2021] [Indexed: 02/05/2023]
Abstract
In high-risk neuroblastoma, the presence of an MYCN gain/amplification (MYCN-GA) is not always a risk factor of cancer-specific death. We herein examined the effect modification of high-dose chemotherapy with autologous hematopoietic stem cell rescue (HDC-autoSCR) in terms of the interaction between MYCN status and remission status (complete remission or very good partial remission [CR/VGPR] vs. partial remission or less [≤PR]). The present study recruited patient data from 1992 to 2017 in the Japan Society of Hematopoietic Cell Transplantation's national registry. The MYCN status was known in 586 of 950 patients with a single course of HDC-autoSCR. Cumulative hazard curves for neuroblastoma-specific death showed that a subgroup with MYCN-GA and ≤PR had a significantly poorer prognosis than three other subgroups, namely, the MYCN-NGA/ ≤ PR, MYCN-NGA/CR/VGPR, and MYCN-GA/CR/VGPR subgroups even after adjusting for non-infants and stage IV disease (hazard ratio: 2.79; 95% confidence interval: 1.91-4.09; P < 0.001). The interaction between MYCN-GA and ≤PR was significant (pinteraction = 0.006). Hence, the patients with MYCN-GA with non-remission status at HDC-autoSCR had a significantly poorer prognosis than the other subgroups, suggesting that HDC-autoSCR may be effective in patients with CR/VGPR regardless of MYCN gene status and in patients with MYCN-NGA regardless of remission status.
Collapse
Affiliation(s)
- Yuya Saito
- Department of Hematology/Oncology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan. .,Division of Molecular Epidemiology, Jikei University School of Medicine, Tokyo, Japan.
| | - Mitsuyoshi Urashima
- Division of Molecular Epidemiology, Jikei University School of Medicine, Tokyo, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Ogawa
- Department of Pediatrics, Niigata Cancer Center Hospital, Niigata, Japan
| | - Chikako Kiyotani
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Yuki Yuza
- Department of Hematology/Oncology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Kenichiro Watanabe
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Hiroaki Goto
- Division of Hematology/Oncology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Atsushi Kikuta
- Department of Pediatric Oncology, Fukushima Medical University Hospital, Fukushima, Japan
| | - Keiko Okada
- Department of Pediatric Hematology/Oncology, Osaka City General Hospital, Osaka, Japan
| | - Yuhki Koga
- Department of Pediatrics, Kyushu University Hospital, Fukuoka, Japan
| | - Junya Fujimura
- Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Masami Inoue
- Department of Hematology/Oncology, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Atsushi Sato
- Department of Hematology and Oncology, Miyagi Children's Hospital, Sendai, Japan
| | - Yoshiko Atsuta
- Department of Healthcare Administration, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Japanese Data Center for Hematopoietic Cell Transplantation, Nagoya, Japan
| | - Kimikazu Matsumoto
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
20
|
Škubník J, Pavlíčková VS, Ruml T, Rimpelová S. Vincristine in Combination Therapy of Cancer: Emerging Trends in Clinics. BIOLOGY 2021; 10:849. [PMID: 34571726 PMCID: PMC8468923 DOI: 10.3390/biology10090849] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023]
Abstract
Treatment of blood malignancies and other cancer diseases has been mostly unfeasible, so far. Therefore, novel treatment regimens should be developed and the currently used ones should be further elaborated. A stable component in various cancer treatment regimens consists of vincristine, an antimitotic compound of natural origin. Despite its strong anticancer activity, mostly, it cannot be administered as monotherapy due to its unspecific action and severe side effects. However, vincristine is suitable for combination therapy. Multidrug treatment regimens including vincristine are standardly applied in the therapy of non-Hodgkin lymphoma and other malignancies, in which it is combined with drugs of different mechanisms of action, mainly with DNA-interacting compounds (for example cyclophosphamide), or drugs interfering with DNA synthesis (for example methotrexate). Besides, co-administration of vincristine with monoclonal antibodies has also emerged, the typical example of which is the anti-CD20 antibody rituximab. Although in some combination anticancer therapies, vincristine has been replaced with other drugs exhibiting lesser side effects, though, in most cases, it is still irreplaceable. This is strongly evidenced by the number of active clinical trials evaluating vincristine in combination cancer therapy. Therefore, in this article, we have reviewed the most common cancer treatment regimens employing vincristine and bring an overview of current trends in the clinical development of this compound.
Collapse
Affiliation(s)
| | | | | | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 3, 166 28 Prague, Czech Republic; (J.Š.); (V.S.P.); (T.R.)
| |
Collapse
|
21
|
Garaventa A, Poetschger U, Valteau-Couanet D, Luksch R, Castel V, Elliott M, Ash S, Chan GCF, Laureys G, Beck-Popovic M, Vettenranta K, Balwierz W, Schroeder H, Owens C, Cesen M, Papadakis V, Trahair T, Schleiermacher G, Ambros P, Sorrentino S, Pearson ADJ, Ladenstein RL. Randomized Trial of Two Induction Therapy Regimens for High-Risk Neuroblastoma: HR-NBL1.5 International Society of Pediatric Oncology European Neuroblastoma Group Study. J Clin Oncol 2021; 39:2552-2563. [PMID: 34152804 DOI: 10.1200/jco.20.03144] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/07/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Induction therapy is a critical component of the therapy of high-risk neuroblastoma. We aimed to assess if the Memorial Sloan Kettering Cancer Center (MSKCC) N5 induction regimen (MSKCC-N5) would improve metastatic complete response (mCR) rate and 3-year event-free survival (EFS) compared with rapid COJEC (rCOJEC; cisplatin [C], vincristine [O], carboplatin [J], etoposide [E], and cyclophosphamide [C]). PATIENTS AND METHODS Patients (age 1-20 years) with stage 4 neuroblastoma or stage 4/4s aged < 1 year with MYCN amplification were eligible for random assignment to rCOJEC or MSKCC-N5. Random assignment was stratified according to national group and metastatic sites. Following induction, therapy comprised primary tumor resection, high-dose busulfan and melphalan, radiotherapy to the primary tumor site, and isotretinoin with ch14.18/CHO (dinutuximab beta) antibody with or without interleukin-2 immunotherapy. The primary end points were mCR rate and 3-year EFS. RESULTS A total of six hundred thirty patients were randomly assigned to receive rCOJEC (n = 313) or MSKCC-N5 (n = 317). Median age at diagnosis was 3.2 years (range, 1 month to 20 years), and 16 were younger than 1 year of age with MYCN amplification. mCR rate following rCOJEC induction (32%, 86/272 evaluable patients) was not significantly different from 35% (99/281) with MSKCC-N5 (P = .368), and 3-year EFS was 44% ± 3% for rCOJEC compared with 47% ± 3% for MSKCC-N5 (P = .527). Three-year overall survival was 60% ± 3% for rCOJEC compared with 65% ± 3% for MSKCC-N5 (P = .379). Toxic death rates with both regimens were 1%. However, nonhematologic CTC grade 3 and 4 toxicities were higher with MSKCC-N5: 68% (193/283) versus 48% (129/268) (P < .001); infection 35% versus 25% (P = .011); stomatitis 25% versus 3% (P < .001); nausea and vomiting 17% versus 7% (P < .001); and diarrhea 7% versus 3% (P = .011). CONCLUSION No difference in outcome was observed between rCOJEC and MSKCC-N5; however, acute toxicity was less with rCOJEC, and therefore rCOJEC is the preferred induction regimen for International Society of Pediatric Oncology European Neuroblastoma Group.
Collapse
Affiliation(s)
| | | | | | - Roberto Luksch
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Victoria Castel
- Pediatric Oncology Unit, Hospital Universitari I Politecnic La Fe, Valencia, Spain
| | - Martin Elliott
- Leeds Teaching Hospitals, NHS Trust, Leeds, United Kingdom
| | - Shifra Ash
- Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Godfrey C F Chan
- University of Hong Kong and Hong Kong Children's Hospital, Hong Kong SAR, China
| | | | | | - Kim Vettenranta
- Children's Hospital, University of Helsinki, Helsinki, Finland
| | | | - Henrik Schroeder
- Department of Paediatrics, University Hospital of Aarhus, Denmark
| | | | | | | | | | | | - Peter Ambros
- Children's Cancer Research Institute, Vienna, Austria
| | | | - Andrew D J Pearson
- Retired. Institute of Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom
| | - Ruth Lydia Ladenstein
- Department of Paediatrics, St Anna Children's Hospital and Children's Cancer Research Institute, Medical University, Vienna, Austria
| |
Collapse
|
22
|
Early Use of Dinutuximab Beta in Patients with High-Risk Neuroblastoma. Case Rep Pediatr 2021; 2021:6610955. [PMID: 34239748 PMCID: PMC8235958 DOI: 10.1155/2021/6610955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 05/06/2021] [Accepted: 06/09/2021] [Indexed: 11/17/2022] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor in children, accounting for 15% of all pediatric cancer deaths. High-risk neuroblastoma (HRNB) is a particularly difficult-to-treat form of the disease that requires aggressive multimodality therapy, including induction chemotherapy, consolidation therapy with high-dose chemotherapy and autologous stem cell transplant, and maintenance therapy with dinutuximab beta. Despite treatment advances, the prognosis of these patients remains poor. As a better response to induction therapy has been associated with prolonged survival in patients with HRNB, we hypothesized that early use of dinutuximab beta-post-induction chemotherapy-may improve patient outcomes. We describe here our experience of administering at least one cycle of dinutuximab beta post-induction and prior to surgery in three children with HRNB who did not demonstrate a complete response to induction chemotherapy. All three patients achieved complete remission. Early use of dinutuximab beta may therefore have the potential to improve outcomes in patients with HRNB.
Collapse
|
23
|
Uva P, Bosco MC, Eva A, Conte M, Garaventa A, Amoroso L, Cangelosi D. Connectivity Map Analysis Indicates PI3K/Akt/mTOR Inhibitors as Potential Anti-Hypoxia Drugs in Neuroblastoma. Cancers (Basel) 2021; 13:cancers13112809. [PMID: 34199959 PMCID: PMC8200206 DOI: 10.3390/cancers13112809] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/17/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022] Open
Abstract
Neuroblastoma (NB) is one of the deadliest pediatric cancers, accounting for 15% of deaths in childhood. Hypoxia is a condition of low oxygen tension occurring in solid tumors and has an unfavorable prognostic factor for NB. In the present study, we aimed to identify novel promising drugs for NB treatment. Connectivity Map (CMap), an online resource for drug repurposing, was used to identify connections between hypoxia-modulated genes in NB tumors and compounds. Two sets of 34 and 21 genes up- and down-regulated between hypoxic and normoxic primary NB tumors, respectively, were analyzed with CMap. The analysis reported a significant negative connectivity score across nine cell lines for 19 compounds mainly belonging to the class of PI3K/Akt/mTOR inhibitors. The gene expression profiles of NB cells cultured under hypoxic conditions and treated with the mTORC complex inhibitor PP242, referred to as the Mohlin dataset, was used to validate the CMap findings. A heat map representation of hypoxia-modulated genes in the Mohlin dataset and the gene set enrichment analysis (GSEA) showed an opposite regulation of these genes in the set of NB cells treated with the mTORC inhibitor PP242. In conclusion, our analysis identified inhibitors of the PI3K/Akt/mTOR signaling pathway as novel candidate compounds to treat NB patients with hypoxic tumors and a poor prognosis.
Collapse
Affiliation(s)
- Paolo Uva
- Clinical Bioinformatics Unit, Scientific Direction, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy;
- Italian Institute of Technology, Via Morego 30, 16163 Genova, Italy
| | - Maria Carla Bosco
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy; (M.C.B.); (A.E.)
| | - Alessandra Eva
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy; (M.C.B.); (A.E.)
| | - Massimo Conte
- UOC Oncologia, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy; (M.C.); (A.G.); (L.A.)
| | - Alberto Garaventa
- UOC Oncologia, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy; (M.C.); (A.G.); (L.A.)
| | - Loredana Amoroso
- UOC Oncologia, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy; (M.C.); (A.G.); (L.A.)
| | - Davide Cangelosi
- Clinical Bioinformatics Unit, Scientific Direction, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy;
- Correspondence:
| |
Collapse
|
24
|
Chung C, Boterberg T, Lucas J, Panoff J, Valteau-Couanet D, Hero B, Bagatell R, Hill-Kayser CE. Neuroblastoma. Pediatr Blood Cancer 2021; 68 Suppl 2:e28473. [PMID: 33818884 PMCID: PMC8785544 DOI: 10.1002/pbc.28473] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 01/01/2023]
Abstract
The survival of patients with high-risk neuroblastoma has improved significantly with the use of intensive multimodality treatment regimens, including chemotherapy, surgery, radiation therapy, myeloablative chemotherapy followed by stem cell rescue, and immunotherapy. This report summarizes the current treatment strategies used in the COG and SIOP for children with neuroblastoma. The improved global collaboration and the adoption of a uniform International Neuroblastoma Risk Group Staging System will help facilitate comparison of homogeneous pretreatment cohorts across clinical trials. Future research strategies regarding the indications for and dosages of radiation therapy to the primary and metastatic sites, and the integration of meta-iodobenzyl guanidine therapy into the multimodal treatment program, are discussed.
Collapse
Affiliation(s)
- Christine Chung
- Department of Radiation Oncology, Diablo Valley Oncology and Hematology, Pleasant Hill, California
| | - Tom Boterberg
- Department of Radiation Oncology Ghent University Hospital, Gent, Belgium
| | - John Lucas
- Department of Radiation Oncology St Jude Children's Research Hospital, Memphis, Tennessee
| | - Joseph Panoff
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health, South Florida, Florida
| | - Dominique Valteau-Couanet
- Department of Childhood and Adolescent Cancer Gustave Roussy Cancer Campus, Villejuif-Grand, Paris, France
| | - Barbara Hero
- Department of Pediatric Hematology and Oncology, University Children's Hospital, Cologne, Germany
| | - Rochelle Bagatell
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christine E Hill-Kayser
- Department of Radiation Oncology University of Pennsylvania and the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
25
|
Jain R, Trehan A, Menon P, Kapoor R, Kakkar N, Radhika S, Saxena AK, Mittal BR, Varma N, Samujh R, Bansal D. Survival in patients with high-risk neuroblastoma treated without autologous stem cell transplant or dinutuximab beta. Pediatr Hematol Oncol 2021; 38:291-304. [PMID: 33622164 DOI: 10.1080/08880018.2020.1850955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The majority of patients with high-risk neuroblastoma (HR-NB) in low- and middle-income countries (LMIC) do not have access to autologous stem cell transplant (ASCT) and dinutuximab. Consolidation with nonmyeloablative chemotherapy is not well-defined, and the outcomes are variable. We report a single-center outcome of patients with HR-NB, treated with nonmyeloablative consolidation. A tabulated compilation of similar reports is included. A retrospective chart review of patients with HR-NB was performed from January 2009 till June 2016. Patients were treated on the backbone of HR-NBL1/SIOPEN protocol. Treatment included induction with rapid-COJEC, surgery, followed by consolidation. Consolidation involved 4 cycles of topotecan, vincristine, and doxorubicin (TVD) instead of ASCT. Infusion of vincristine and doxorubicin were modified for ease and to enable administration in the clinic. Subsequent treatment included radiotherapy to the primary tumor and differentiation therapy with isotretinoin. Over 7½ years, 28 patients with HR-NB were treated. Two (7%) patients had therapy-related mortality. A relapse or disease progression occurred in 11 (39%) patients at a median duration of 17 months (IQR: 5, 18). Treatment abandonment was observed in 4 (14%) patients. The median follow-up of disease-free patients was 49 months (IQR: 45, 79). Patients with relapse were not treated further. A 4-year EFS of 29.3% was observed when 4-cycles of TVD were administered instead of ASCT in patients with HR-NB. The study and the review will aid decision-making for care of patients in LMIC while considering the options of treatment for HR-NB if access to ACST and dinutuximab is lacking.
Collapse
Affiliation(s)
- Richa Jain
- Pediatric Hematology-Oncology Unit, Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amita Trehan
- Pediatric Hematology-Oncology Unit, Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Prema Menon
- Department of Pediatric Surgery, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rakesh Kapoor
- Department of Radiotherapy, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Nandita Kakkar
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Srinivasan Radhika
- Department of Cytopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Akshay Kumar Saxena
- Department of Radiodiagnosis and Imaging, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bhagwant Rai Mittal
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Neelam Varma
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ram Samujh
- Department of Pediatric Surgery, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepak Bansal
- Pediatric Hematology-Oncology Unit, Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
26
|
Kumar A, Rocke JPJ, Kumar BN. Evolving treatments in high-risk neuroblastoma. Expert Opin Orphan Drugs 2020. [DOI: 10.1080/21678707.2020.1865918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Abhinav Kumar
- Division of Medicine, University College London Medical School, London, UK
| | - John P J Rocke
- ENT Department, Royal Albert Edward Infirmary, Wigan, UK
| | - B Nirmal Kumar
- ENT Department, Wrightington, Wigan & Leigh Teaching NHS, Wigan, UK
| |
Collapse
|
27
|
Berlanga P, Pasqualini C, Pötschger U, Sangüesa C, Castellani MR, Cañete A, Luksch R, Elliot M, Schreier G, Kropf M, Morgenstern D, Papadakis V, Ash S, Ruud E, Brock P, Wieczorek A, Kogner P, Trahair T, Ambros P, Boterberg T, Castel V, Valteau-Couanet D, Ladenstein R. Central nervous system relapse in high-risk stage 4 neuroblastoma: The HR-NBL1/SIOPEN trial experience. Eur J Cancer 2020; 144:1-8. [PMID: 33316634 DOI: 10.1016/j.ejca.2020.10.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/08/2020] [Accepted: 10/20/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND There is rising concern on the impact of new strategies, such as high-dose chemotherapy (HDC) and immunotherapy, on the pattern of relapse in high-risk neuroblastoma (HR-NBL). Our aim is to evaluate the incidence and identify risk factors for first recurrence in the central nervous system (CNS) in HR-NBL. PATIENTS AND METHODS Data from patients with stage 4V HR-NBL included from February 2002 to June 2015 in the prospective HR-NBL trial of the European International Society of Pediatric Oncology Neuroblastoma Group were analysed. Characteristics at diagnosis, treatment and the pattern of first relapse were studied. CNS imaging at relapse was centrally reviewed. RESULTS The 1977 included patients had a median age of 3 years (1 day-20 years); 1163 were boys. Among the 1161 first relapses, 53 were in the CNS, with an overall incidence of 2.7%, representing 6.2% of all metastatic relapses. One- and three-year post-relapse overall survival was 25 ± 6% and 8 ± 4%, respectively. Higher risk of CNS recurrence was associated with female sex (hazard ratio [HR] = 2.0 [95% confidence interval {CI}: 1.1-3.5]; P = 0.016), MYCN-amplification (HR = 2.4 [95% CI: 1.2-4.4]; P = 0.008), liver (HR = 2.5 [95% CI: 1.2-5.1]; P = 0.01) or >1 metastatic compartment involvement (HR = 7.1 [95% CI: 1.0-48.4]; P = 0.047) at diagnosis. Neither HDC nor immunotherapy was associated with higher risk of CNS recurrence. Stable incidence of CNS relapse was reported over time. CONCLUSIONS The risk of CNS recurrence is linked to both patient and disease characteristics, with neither impact of HDC nor immunotherapy. These findings support the current treatment strategy and do not justify a CNS prophylactic treatment.
Collapse
Affiliation(s)
- P Berlanga
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Paris-Saclay University, Paris, France.
| | - C Pasqualini
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Paris-Saclay University, Paris, France
| | - U Pötschger
- Department for Studies and Statistics and Integrated Research, Children's Cancer Research Institute, Vienna, Austria
| | - C Sangüesa
- Pediatric Radiology Unit, Hospital Universitario y Politecnico La Fe, Valencia, Spain
| | - M R Castellani
- Nuclear Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - A Cañete
- Pediatric Oncology Unit, Hospital Universitario y Politecnico La Fe, Valencia, Spain
| | - R Luksch
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M Elliot
- Pediatric Oncology, Leeds Teaching Hospital NHS Trust, Leeds, UK
| | - G Schreier
- Centre for Health and Bioresources, AIT Austrian Institute of Technology GmbH, Graz, Austria
| | - M Kropf
- Centre for Health and Bioresources, AIT Austrian Institute of Technology GmbH, Graz, Austria
| | - D Morgenstern
- Division of Pediatric Hematology/Oncology, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - V Papadakis
- Paediatric Hematology/Oncology, Agia Sofia Children's Hospital, Athens, Greece
| | - S Ash
- Department of Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Sackler Faculty of Medicine Tel Aviv University, Petach Tikvah, Israel
| | - E Ruud
- Department of Paediatric Medicine, Rikshospitalet, Oslo, Norway
| | - P Brock
- Department of Pediatric Oncology, Great Ormond Street Hospital, London, UK
| | - A Wieczorek
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - P Kogner
- Department of Women's and Children's Health, Karolinska Institutet, 17177, Stockholm, Sweden
| | - T Trahair
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - P Ambros
- Department of Tumor Biology, Children's Cancer Research Institute, Vienna, Austria
| | - T Boterberg
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - V Castel
- Pediatric Oncology Unit, Hospital Universitario y Politecnico La Fe, Valencia, Spain
| | - D Valteau-Couanet
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Paris-Saclay University, Paris, France
| | - R Ladenstein
- St Anna Children's Hospital, Vienna, Austria; Department for Studies and Statistics and Integrated Research, Vienna, Austria; Children's Cancer Research Institute, Vienna, Austria
| |
Collapse
|
28
|
A promoter-driven assay for INSM1-associated signaling pathway in neuroblastoma. Cell Signal 2020; 76:109785. [PMID: 32966884 DOI: 10.1016/j.cellsig.2020.109785] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/01/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
Aggressive form of neuroblastoma (NB) is a malignant childhood cancer derived from granule neuron precursors and sympatho-adrenal lineage with N-MYC amplification. An insulinoma associated-1 (INSM1) transcription factor has emerged as a NB biomarker that plays critical role in facilitating tumor cell growth and transformation. N-myc activates INSM1 in NB was discovered. In order to elucidate the signaling pathways associated with INSM1 expression and NB tumor cell growth, we developed an INSM1 promoter-driven luciferase assay for new drug discovery. Promoter-driven luciferase assay demonstrated high Z' factor (>0.7). Performance measures using signal-to-noise ratio, signal window, and Z' factor confirmed this assay is a robust screening method. A panel of FDA-approved oncology drugs set (147 compounds) was subjected to the INSM1 promoter-driven luciferase assay. The positive-hit compounds were validated for effects on cell viability and INSM1 expression. Screening a FDA-approved oncology drugs set revealed multiple inhibitors against various signaling pathways via suppression of INSM1 promoter activity. The positive-hit compounds consist of 9 signaling pathway inhibitors negatively regulates INSM1 expression and cell viability in NB. These inhibitors target DNA/RNA/protein synthesis, tubulin assembly, and histone deacetylase signaling pathways. The outcome of this assay indicates that the newly identified pathways could be critical for NB growth and transformation. This technology and the positive-hits of multiple pathways represent a promising option of identifying re-purposed FDA-approved drugs valuable for NB treatment in the context of a NB-specific marker, INSM1.
Collapse
|
29
|
Jin Z, Lu Y, Wu Y, Che J, Dong X. Development of differentiation modulators and targeted agents for treating neuroblastoma. Eur J Med Chem 2020; 207:112818. [PMID: 32937281 DOI: 10.1016/j.ejmech.2020.112818] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/10/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
Neuroblastoma (NB) is one of the most common pediatric malignancies. Easy metastasis, poor prognosis, and a high degree of heterogeneity of NB hinder its successful treatment. Several different therapeutic strategies have been developed to overcome these problems, including differentiation and targeted therapy. In this review, we summarize the recent development of differentiation modulators and targeted agents for treating NB. Several promising targets of NB were also discussed.
Collapse
Affiliation(s)
- Zegao Jin
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Yang Lu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Yizhe Wu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Jinxin Che
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China.
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310058, PR China; Cancer Center of Zhejiang University, Hangzhou, 310058, PR China.
| |
Collapse
|
30
|
Holmes K, Pötschger U, Pearson ADJ, Sarnacki S, Cecchetto G, Gomez-Chacon J, Squire R, Freud E, Bysiek A, Matthyssens LE, Metzelder M, Monclair T, Stenman J, Rygl M, Rasmussen L, Joseph JM, Irtan S, Avanzini S, Godzinski J, Björnland K, Elliott M, Luksch R, Castel V, Ash S, Balwierz W, Laureys G, Ruud E, Papadakis V, Malis J, Owens C, Schroeder H, Beck-Popovic M, Trahair T, Forjaz de Lacerda A, Ambros PF, Gaze MN, McHugh K, Valteau-Couanet D, Ladenstein RL. Influence of Surgical Excision on the Survival of Patients With Stage 4 High-Risk Neuroblastoma: A Report From the HR-NBL1/SIOPEN Study. J Clin Oncol 2020; 38:2902-2915. [PMID: 32639845 DOI: 10.1200/jco.19.03117] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To evaluate the impact of surgeon-assessed extent of primary tumor resection on local progression and survival in patients in the International Society of Pediatric Oncology Europe Neuroblastoma Group High-Risk Neuroblastoma 1 trial. PATIENTS AND METHODS Patients recruited between 2002 and 2015 with stage 4 disease > 1 year or stage 4/4S with MYCN amplification < 1 year who had completed induction without progression, achieved response criteria for high-dose therapy (HDT), and had no resection before induction were included. Data were collected on the extent of primary tumor excision, severe operative complications, and outcome. RESULTS A total of 1,531 patients were included (median observation time, 6.1 years). Surgeon-assessed extent of resection included complete macroscopic excision (CME) in 1,172 patients (77%) and incomplete macroscopic resection (IME) in 359 (23%). Surgical mortality was 7 (0.46%) of 1,531. Severe operative complications occurred in 142 patients (9.7%), and nephrectomy was performed in 124 (8.8%). Five-year event-free survival (EFS) ± SE (0.40 ± 0.01) and overall survival (OS; 0.45 ± 0.02) were significantly higher with CME compared with IME (5-year EFS, 0.33 ± 0.03; 5-year OS, 0.37 ± 0.03; P < .001 and P = .004). The cumulative incidence of local progression (CILP) was significantly lower after CME (0.17 ± 0.01) compared with IME (0.30 ± 0.02; P < .001). With immunotherapy, outcomes were still superior with CME versus IME (5-year EFS, 0.47 ± 0.02 v 0.39 ± 0.04; P = .038); CILP was 0.14 ± 0.01 after CME and 0.27 ± 0.03 after IME (P < .002). A hazard ratio of 1.3 for EFS associated with IME compared with CME was observed before and after the introduction of immunotherapy (P = .030 and P = .038). CONCLUSION In patients with stage 4 high-risk neuroblastoma who have responded to induction therapy, CME of the primary tumor is associated with improved survival and local control after HDT, local radiotherapy (21 Gy), and immunotherapy.
Collapse
Affiliation(s)
- Keith Holmes
- Paediatric Surgery, St George's Hospital London and Royal Marsden Hospital, Sutton, United Kingdom
| | - Ulrike Pötschger
- Children's Cancer Research Institute, Department of Paediatrics, Medical University of Vienna, Vienna, Austria
| | - Andrew D J Pearson
- Institute of Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom
| | - Sabine Sarnacki
- Department of Pediatric Surgery, Necker Enfants-Malades Hospital, Assistance Publique Hôpitaux de Paris, University de Paris, Paris, France
| | - Giovanni Cecchetto
- Pediatric Surgery, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Javier Gomez-Chacon
- Paediatric Oncology, Paediatric Surgical Oncology Unit, Hospital Universitario La FE, Valencia, Spain
| | - Roly Squire
- Paediatric Oncology, Leeds Teaching Hospital, Leeds, United Kingdom
| | - Enrique Freud
- Schneider Children's Medical Center of Israel, Petach, Tikvah, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adam Bysiek
- Department of Pediatric Surgery, University Children's Hospital, Kraków, Poland
| | - Lucas E Matthyssens
- Department of Gastrointestinal and Paediatric Surgery, Princess Elisabeth Children's Hospital, Ghent University Hospital, Ghent, Belgium
| | - Martin Metzelder
- Paediatric Surgery, Medical University of Vienna, Vienna, Austria
| | - Tom Monclair
- Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | - Michal Rygl
- University Hospital Motol, Prague, Czech Republic
| | - Lars Rasmussen
- Department of Surgical Gastroenterology A, Odense University Hospital, Odense, Denmark
| | | | - Sabine Irtan
- Sorbonne University, Department of Visceral and Neonatal Pediatric Surgery, Armand Trousseau Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Stefano Avanzini
- Pediatric Surgery Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Jan Godzinski
- Department of Paediatric Surgery, Marciniak Hospital, and Department of Paediatric Traumatology and Emergency Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Kristin Björnland
- Oslo University Hospital Rikshospitalet, Oslo, Norway.,University of Oslo, Oslo, Norway
| | - Martin Elliott
- Paediatric Oncology, Leeds Teaching Hospital, Leeds, United Kingdom
| | - Roberto Luksch
- Paediatric Oncology, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Victoria Castel
- Paediatric Oncology, Paediatric Surgical Oncology Unit, Hospital Universitario La FE, Valencia, Spain
| | - Shifra Ash
- Schneider Children's Medical Center of Israel, Petach, Tikvah, Israel
| | | | - Geneviève Laureys
- Department of Paediatric Haematology and Oncology, Princess Elisabeth Children's Hospital, Ghent University Hospital, Ghent, Belgium
| | - Ellen Ruud
- Oslo University Hospital Rikshospitalet, Oslo, Norway.,University of Oslo, Oslo, Norway
| | | | - Josef Malis
- University Hospital Motol, Prague, Czech Republic
| | - Cormac Owens
- Paediatric Haematology/Oncology, Our Lady's Children's Hospital, Crumlin, Dublin, Republic of Ireland
| | | | | | - Toby Trahair
- Sydney Children's Hospital, Randwick, New South Wales, Australia
| | | | - Peter F Ambros
- Children's Cancer Research Institute, Department of Paediatrics, Medical University of Vienna, Vienna, Austria
| | - Mark N Gaze
- University College Hospital, London, United Kingdom
| | - Kieran McHugh
- Paediatric Oncology, Great Ormond Street Hospital, London, United Kingdom
| | | | - Ruth Lydia Ladenstein
- St Anna Children's Hospital and Children's Cancer Research Institute, Department of Paediatrics, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
31
|
George SL, Parmar V, Lorenzi F, Marshall LV, Jamin Y, Poon E, Angelini P, Chesler L. Novel therapeutic strategies targeting telomere maintenance mechanisms in high-risk neuroblastoma. J Exp Clin Cancer Res 2020; 39:78. [PMID: 32375866 PMCID: PMC7201617 DOI: 10.1186/s13046-020-01582-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
The majority of high-risk neuroblastomas can be divided into three distinct molecular subgroups defined by the presence of MYCN amplification, upstream TERT rearrangements or alternative lengthening of telomeres (ALT). The common defining feature of all three subgroups is altered telomere maintenance; MYCN amplification and upstream TERT rearrangements drive high levels of telomerase expression whereas ALT is a telomerase independent telomere maintenance mechanism. As all three telomere maintenance mechanisms are independently associated with poor outcomes, the development of strategies to selectively target either telomerase expressing or ALT cells holds great promise as a therapeutic approach that is applicable to the majority of children with aggressive disease.Here we summarise the biology of telomere maintenance and the molecular drivers of aggressive neuroblastoma before describing the most promising therapeutic strategies to target both telomerase expressing and ALT cancers. For telomerase-expressing neuroblastoma the most promising targeted agent to date is 6-thio-2'-deoxyguanosine, however clinical development of this agent is required. In osteosarcoma cell lines with ALT, selective sensitivity to ATR inhibition has been reported. However, we present data showing that in fact ALT neuroblastoma cells are more resistant to the clinical ATR inhibitor AZD6738 compared to other neuroblastoma subtypes. More recently a number of additional candidate compounds have been shown to show selectivity for ALT cancers, such as Tetra-Pt (bpy), a compound targeting the telomeric G-quadruplex and pifithrin-α, a putative p53 inhibitor. Further pre-clinical evaluation of these compounds in neuroblastoma models is warranted.In summary, telomere maintenance targeting strategies offer a significant opportunity to develop effective new therapies, applicable to a large proportion of children with high-risk neuroblastoma. In parallel to clinical development, more pre-clinical research specifically for neuroblastoma is urgently needed, if we are to improve survival for this common poor outcome tumour of childhood.
Collapse
Affiliation(s)
- S L George
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, London, UK.
- Children and Young People's Unit, Royal Marsden NHS Foundation Trust, London, UK.
| | - V Parmar
- Children and Young People's Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - F Lorenzi
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - L V Marshall
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, London, UK
- Children and Young People's Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Y Jamin
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - E Poon
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - P Angelini
- Children and Young People's Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - L Chesler
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, London, UK
- Children and Young People's Unit, Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
32
|
Wang Y, Stear JH, Swain A, Xu X, Bryce NS, Carnell M, Alieva IB, Dugina VB, Cripe TP, Stehn J, Hardeman EC, Gunning PW. Drug Targeting the Actin Cytoskeleton Potentiates the Cytotoxicity of Low Dose Vincristine by Abrogating Actin-Mediated Repair of Spindle Defects. Mol Cancer Res 2020; 18:1074-1087. [PMID: 32269073 DOI: 10.1158/1541-7786.mcr-19-1122] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/09/2020] [Accepted: 04/03/2020] [Indexed: 11/16/2022]
Abstract
Antimicrotubule vinca alkaloids are widely used in the clinic but their toxicity is often dose limiting. Strategies that enhance their effectiveness at lower doses are needed. We show that combining vinca alkaloids with compounds that target a specific population of actin filaments containing the cancer-associated tropomyosin Tpm3.1 result in synergy against a broad range of tumor cell types. We discovered that low concentrations of vincristine alone induce supernumerary microtubule asters that form transient multi-polar spindles in early mitosis. Over time these asters can be reconstructed into functional bipolar spindles resulting in cell division and survival. These microtubule asters are organized by the nuclear mitotic apparatus protein (NuMA)-dynein-dynactin complex without involvement of centrosomes. However, anti-Tpm3.1 compounds at nontoxic concentrations inhibit this rescue mechanism resulting in delayed onset of anaphase, formation of multi-polar spindles, and apoptosis during mitosis. These findings indicate that drug targeting actin filaments containing Tpm3.1 potentiates the anticancer activity of low-dose vincristine treatment. IMPLICATIONS: Simultaneously inhibiting Tpm3.1-containing actin filaments and microtubules is a promising strategy to potentiate the anticancer activity of low-dose vincristine.
Collapse
Affiliation(s)
- Yao Wang
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jeffrey H Stear
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Ashleigh Swain
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Xing Xu
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Nicole S Bryce
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Michael Carnell
- Biomedical Imaging Facility, Mark Wainwright Analytical Center, University of New South Wales, Sydney, New South Wales, Australia
| | - Irina B Alieva
- Department of Electron Microscopy, A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vera B Dugina
- Department of Mathematical Methods in Biology, A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Justine Stehn
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Edna C Hardeman
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Peter W Gunning
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
33
|
Avitabile M, Lasorsa VA, Cantalupo S, Cardinale A, Cimmino F, Montella A, Capasso D, Haupt R, Amoroso L, Garaventa A, Quattrone A, Corrias MV, Iolascon A, Capasso M. Association of PARP1 polymorphisms with response to chemotherapy in patients with high-risk neuroblastoma. J Cell Mol Med 2020; 24:4072-4081. [PMID: 32103589 PMCID: PMC7171401 DOI: 10.1111/jcmm.15058] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/17/2020] [Accepted: 01/27/2020] [Indexed: 12/25/2022] Open
Abstract
The genetic aetiology and the molecular mechanisms that characterize high‐risk neuroblastoma are still little understood. The majority of high‐risk neuroblastoma patients do not take advantage of current induction therapy. So far, one of the main reasons liable for cancer therapeutic failure is the acquisition of resistance to cytotoxic anticancer drugs, because of the DNA repair system of tumour cells. PARP1 is one of the main DNA damage sensors involved in the DNA repair system and genomic stability. We observed that high PARP1 mRNA level is associated with unfavourable prognosis in 3 public gene expression NB patients’ datasets and in 20 neuroblastomas analysed by qRT‐PCR. Among 4983 SNPs in PARP1, we selected two potential functional SNPs. We investigated the association of rs907187, in PARP1 promoter, and rs2048426 in non‐coding region with response chemotherapy in 121 Italian patients with high‐risk NB. Results showed that minor G allele of rs907187 associated with induction response of patients (P = .02) and with decrease PARP1 mRNA levels in NB cell line (P = .003). Furthermore, rs907187 was predicted to alter the binding site of E2F1 transcription factor. Specifically, allele G had low binding affinity with E2F1 whose expression positively correlates with PARP1 expression and associated with poor prognosis of patients with NB. By contrast, we did not find genetic association for the SNP rs2048426. These data reveal rs907187 as a novel potential risk variant associated with the failure of induction therapy for high‐risk NB.
Collapse
Affiliation(s)
- Marianna Avitabile
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Vito Alessandro Lasorsa
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | | | | | | | | | - Dalila Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Riccardo Haupt
- UOS Epidemiology, Biostatistics and Committees, Genova, Italy
| | - Loredana Amoroso
- Department of Pediatric Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Alberto Garaventa
- Department of Pediatric Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Alessandro Quattrone
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Maria Valeria Corrias
- Laboratory of Experimental Therapy in Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Mario Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy.,IRCCS SDN, Naples, Italy
| |
Collapse
|
34
|
Investigation of the Role of Dinutuximab Beta-Based Immunotherapy in the SIOPEN High-Risk Neuroblastoma 1 Trial (HR-NBL1). Cancers (Basel) 2020; 12:cancers12020309. [PMID: 32013055 PMCID: PMC7072500 DOI: 10.3390/cancers12020309] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/17/2020] [Accepted: 01/25/2020] [Indexed: 11/18/2022] Open
Abstract
To explore the effects of immunotherapy in the International Society of Paediatric Oncology Europe Neuroblastoma Group SIOPEN high-risk neuroblastoma 1 trial (HR-NBL1 trial), two cohorts were studied: one prior to and one after the introduction of dinutuximab beta. All patients received standard induction and high-dose therapy (HDT) with autologous stem cell rescue (ASCR); the local control comprised surgery and radiotherapy to the primary tumour site, followed by isotretinoin. A landmark timepoint of 109 days, resulting from the median time between ASCR and initiation of immunotherapy, was used to define patients’ eligibility in the pre-immunotherapy analysis cohort. Median follow-up was 5.8 years (inter-quartile range (IQR): 4.2–8.2 years) for 844 eligible patients balanced for risk factors, such as age, sex, stage 4, MYCN amplification and response prior to HDT. The five-year event-free and overall survival (95% confidence interval (CI) of 466 patients not receiving immunotherapy was 42% (38–47%) and 50% (46–55%) but was 57% (51–62%) and 64% (59–69%) for 378 patients receiving immunotherapy (p < 0.001). A multivariate analysis identified absence of immunotherapy (p = 0.0002, hazard ratio (HR) 1.573); type of HDT (p = 0.0029, HR 1.431); less than complete response prior to maintenance therapy (p = 0.0043, HR 1.494) and >1 metastatic compartment at diagnosis (p < 0.001, HR 2.665) as risk factors for relapse or progression. Results suggest an important role for dinutuximab beta-based immunotherapy within the treatment concepts applied in HR-NBL1/SIOPEN.
Collapse
|
35
|
Berthold F, Faldum A, Ernst A, Boos J, Dilloo D, Eggert A, Fischer M, Frühwald M, Henze G, Klingebiel T, Kratz C, Kremens B, Krug B, Leuschner I, Schmidt M, Schmidt R, Schumacher-Kuckelkorn R, von Schweinitz D, Schilling FH, Theissen J, Volland R, Hero B, Simon T. Extended induction chemotherapy does not improve the outcome for high-risk neuroblastoma patients: results of the randomized open-label GPOH trial NB2004-HR. Ann Oncol 2020; 31:422-429. [PMID: 32067684 DOI: 10.1016/j.annonc.2019.11.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Long-term survival of high-risk neuroblastoma patients is still below 50% despite intensive multimodal treatment. This trial aimed to address whether the addition of two topotecan-containing chemotherapy courses compared to standard induction therapy improves event-free survival (EFS) of these patients. PATIENTS AND METHODS An open-label, multicenter, prospective randomized controlled trial was carried out at 58 hospitals in Germany and Switzerland. Patients aged 1-21 years with stage 4 neuroblastoma and patients aged 6 months to 21 years with MYCN-amplified tumors were eligible. The primary endpoint was EFS. Patients were randomly assigned to standard induction therapy with six chemotherapy courses or to experimental induction chemotherapy starting with two additional courses of topotecan, cyclophosphamide, and etoposide followed by standard induction chemotherapy (eight courses in total). After induction chemotherapy, all patients received high-dose chemotherapy with autologous hematopoietic stem cell rescue and isotretinoin for consolidation. Radiotherapy was applied to patients with active tumors at the end of induction chemotherapy. RESULTS Of 536 patients enrolled in the trial, 422 were randomly assigned to the control arm (n = 211) and the experimental arm (n = 211); the median follow-up time was 3.32 years (interquartile range 1.65-5.92). At data lock, the 3-year EFS of experimental and control patients was 34% and 32% [95% confidence Interval (CI) 28% to 40% and 26% to 38%; P = 0.258], respectively. Similarly, the 3-year overall survival of the patients did not differ [54% and 48% (95% CI 46% to 62% and 40% to 56%), respectively; P = 0.558]. The response to induction chemotherapy was not different between the arms. The median number of non-fatal toxicities per patient was higher in the experimental group while the median number of toxicities per chemotherapy course was not different. CONCLUSION While the burden for the patients was increased by prolonging the induction chemotherapy and the toxicity, the addition of two topotecan-containing chemotherapy courses did not improve the EFS of high-risk neuroblastoma patients and thus cannot be recommended. CLINICAL TRIALS. GOV NUMBER NCT number 03042429.
Collapse
Affiliation(s)
- F Berthold
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany.
| | - A Faldum
- Institute of Medical Statistics and Clinical Research, University of Muenster, Muenster, Germany
| | - A Ernst
- Institute of Medical Statistics and Computational Biology (IMSB), University of Cologne, Cologne, Germany
| | - J Boos
- Department of Pediatric Oncology and Hematology, University of Muenster, Muenster, Germany
| | - D Dilloo
- Department of Pediatric Oncology and Hematology, University of Bonn, Bonn, Germany
| | - A Eggert
- Department of Pediatric Oncology and Hematology, Charité Universitätsmedizin Berlin and Berlin Institute of Health, Berlin, Germany
| | - M Fischer
- Department of Experimental Pediatric Oncology and Center for Molecular Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - M Frühwald
- Swabian Children's Cancer Center, Children's Hospital, University Hospital Augsburg, Augsburg, Germany
| | - G Henze
- Department of Pediatric Oncology and Hematology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - T Klingebiel
- Department of Children and Adolescents, University Hospital, Goethe University Frankfurt (Main), Frankfurt am Main, Germany
| | - C Kratz
- Department of Pediatric Oncology and Hematology, Medicinal University, Hannover, Germany
| | - B Kremens
- Department of Pediatric Oncology and Hematology, University of Essen, Essen, Germany
| | - B Krug
- Institute of Diagnostic and Interventional Radiology, University of Cologne, Cologne, Germany
| | - I Leuschner
- Children's Tumor Registry, Institute of Pathology, University of Kiel, Kiel, Germany
| | - M Schmidt
- Department of Nuclear Medicine, University of Cologne, Cologne, Germany
| | - R Schmidt
- Institute of Medical Statistics and Clinical Research, University of Muenster, Muenster, Germany
| | | | - D von Schweinitz
- Department of Pediatric Surgery, University of Munich, Munich, Germany
| | - F H Schilling
- Department of Pediatric Oncology and Hematology, Olgahospital Stuttgart, Stuttgart, Germany
| | - J Theissen
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - R Volland
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - B Hero
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - T Simon
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| |
Collapse
|
36
|
Cardiac MRI: a Promising Diagnostic Tool to Detect Cancer Therapeutics–Related Cardiac Dysfunction. CURRENT CARDIOVASCULAR IMAGING REPORTS 2019. [DOI: 10.1007/s12410-019-9489-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
37
|
Novel Therapies for Relapsed and Refractory Neuroblastoma. CHILDREN-BASEL 2018; 5:children5110148. [PMID: 30384486 PMCID: PMC6262328 DOI: 10.3390/children5110148] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 12/17/2022]
Abstract
While recent increases in our understanding of the biology of neuroblastoma have allowed for more precise risk stratification and improved outcomes for many patients, children with high-risk neuroblastoma continue to suffer from frequent disease relapse, and despite recent advances in our understanding of neuroblastoma pathogenesis, the outcomes for children with relapsed neuroblastoma remain poor. These children with relapsed neuroblastoma, therefore, continue to need novel treatment strategies based on a better understanding of neuroblastoma biology to improve outcomes. The discovery of new tumor targets and the development of novel antibody- and cell-mediated immunotherapy agents have led to a large number of clinical trials for children with relapsed neuroblastoma, and additional clinical trials using molecular and genetic tumor profiling to target tumor-specific aberrations are ongoing. Combinations of these new therapeutic modalities with current treatment regimens will likely be needed to improve the outcomes of children with relapsed and refractory neuroblastoma.
Collapse
|
38
|
Valteau-Couanet D, Schleiermacher G, Sarnacki S, Pasqualini C. Prise en charge des neuroblastomes de haut risque : l’expérience du groupe européen SIOPEN. Bull Cancer 2018; 105:918-924. [DOI: 10.1016/j.bulcan.2018.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 09/14/2018] [Accepted: 09/17/2018] [Indexed: 10/28/2022]
|
39
|
Felgenhauer J, Tomino L, Selich-Anderson J, Bopp E, Shah N. Dual BRD4 and AURKA Inhibition Is Synergistic against MYCN-Amplified and Nonamplified Neuroblastoma. Neoplasia 2018; 20:965-974. [PMID: 30153557 PMCID: PMC6111011 DOI: 10.1016/j.neo.2018.08.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/02/2018] [Accepted: 08/02/2018] [Indexed: 11/29/2022] Open
Abstract
A majority of cases of high-risk neuroblastoma, an embryonal childhood cancer, are driven by MYC or MYCN-driven oncogenic signaling. While considered to be directly “undruggable” therapeutically, MYC and MYCN can be repressed transcriptionally by inhibition of Bromodomain-containing protein 4 (BRD4) or destabilized posttranslationally by inhibition of Aurora Kinase A (AURKA). Preclinical and early-phase clinical studies of BRD4 and AURKA inhibitors, however, show limited efficacy against neuroblastoma when used alone. We report our studies on the concomitant use of the BRD4 inhibitor I-BET151 and AURKA inhibitor alisertib. We show that, in vitro, the drugs act synergistically to inhibit viability in four models of high-risk neuroblastoma. We demonstrate that this synergy is driven, in part, by the ability of I-BET151 to mitigate reflexive upregulation of AURKA, MYC, and MYCN in response to AURKA inhibition. We then demonstrate that I-BET151 and alisertib are effective in prolonging survival in four xenograft neuroblastoma models in vivo, and this efficacy is augmented by the addition of the antitubule chemotherapeutic vincristine. These data suggest that epigenetic and posttranslational inhibition of MYC/MYCN-driven pathways may have significant clinical efficacy against neuroblastoma.
Collapse
Affiliation(s)
- Joshua Felgenhauer
- Nationwide Children's Hospital, Center for Childhood Cancer and Blood Disorders, 700 Children's Drive, Columbus, OH, 43205
| | - Laura Tomino
- Nationwide Children's Hospital, Center for Childhood Cancer and Blood Disorders, 700 Children's Drive, Columbus, OH, 43205
| | - Julia Selich-Anderson
- Nationwide Children's Hospital, Center for Childhood Cancer and Blood Disorders, 700 Children's Drive, Columbus, OH, 43205
| | - Emily Bopp
- The Ohio State University, College of Arts and Sciences, 186 University Hall, Columbus, OH, 43210
| | - Nilay Shah
- Nationwide Children's Hospital, Center for Childhood Cancer and Blood Disorders, 700 Children's Drive, Columbus, OH, 43205; The Ohio State University, College of Medicine, Department of Pediatrics, 370 W 9th Ave, Columbus, OH 43210.
| |
Collapse
|
40
|
Morandi F, Amoroso L, Dondero A, Castriconi R, Parodi S, Luksch R, Casale F, Castellano A, Garaventa A, Moretta A, Bottino C, Ponzoni M, Corrias MV. Updated clinical and biological information from the two-stage phase II study of imatinib mesylate in subjects with relapsed/refractory neuroblastoma. Oncoimmunology 2018; 7:e1468953. [PMID: 30357053 DOI: 10.1080/2162402x.2018.1468953] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/17/2018] [Accepted: 04/19/2018] [Indexed: 12/12/2022] Open
Abstract
Several studies support the notion that the kinase inhibitor Imatinib mesylate exerts off-target effects on cells of the immune system. After our first report of continuous daily oral administration in subjects with relapsed/refractory neuroblastoma (NB, EudraCT: 2005-005778-63), here we update the clinical information and report additional information on potential surrogate markers for prediction of efficacy. Peripheral blood (PB) samples collected at study entry and after the first and second cycle of Imatinib mesylate treatment were tested for IFN-γ, TNF-α, TGF-β, IL-10, CXCL12 and soluble (s) B7-H6 plasma levels. In addition, paired PB and bone marrow (BM) samples collected at study entry and after the second Imatinib cycle were evaluated for CXCL12, CXCR4 and NKp30 isoform mRNA levels. Correlation between each parameter level and response/outcome was then evaluated. Out of the six subjects still alive at the time of the first report, thee died after additional therapy, two for NB progression and one for a second malignancy. Three are presently alive and cured from NB at 10 years after the first Imatinib cycle. Of these, one achieved complete response (CR) during Imatinib treatment and never relapsed, one had a local relapse removed by surgery and the third received TVD as rescue therapy. Response and outcome were associated with low Imatinib exposure, whereas none of the tested immunological and molecular parameters was predictive of response/outcome. However, after Imatinib treatment NKp30 isoform mRNA levels significantly increase in BM samples, indicating that Imatinib mesylate exerted an off-target effect on NK cells in vivo. Imatinib mesylate efficacy in relapsed/refractory NB has been confirmed at a longer follow-up, supporting its inclusion in new Phase II trials for these subjects, that should envisage collection of samples to evaluate the predictive power of other potential surrogate markers of efficacy.
Collapse
Affiliation(s)
- Fabio Morandi
- Experimental Therapy in Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Loredana Amoroso
- Pediatric Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Alessandra Dondero
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genoa, Italy
| | - Roberta Castriconi
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genoa, Italy.,Centre of Excellence for Biomedical Research, Università degli Studi di Genova, Genoa, Italy
| | - Stefano Parodi
- Epidemiology and Biostatistics Section, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Roberto Luksch
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Aurora Castellano
- Department of Hematology-Oncology, IRCCS Ospedale Bambino Gesù, Rome, Italy
| | | | - Alessandro Moretta
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genoa, Italy.,Centre of Excellence for Biomedical Research, Università degli Studi di Genova, Genoa, Italy
| | - Cristina Bottino
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genoa, Italy.,Laboratory of Clinical and Experimental Immunology, Istituto Giannina Gaslini, Genoa, Italy
| | - Mirco Ponzoni
- Experimental Therapy in Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | |
Collapse
|
41
|
Johnsen JI, Dyberg C, Fransson S, Wickström M. Molecular mechanisms and therapeutic targets in neuroblastoma. Pharmacol Res 2018; 131:164-176. [PMID: 29466695 DOI: 10.1016/j.phrs.2018.02.023] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/20/2022]
Abstract
Neuroblastoma is the most common extracranical tumor of childhood and the most deadly tumor of infancy. It is characterized by early age onset and high frequencies of metastatic disease but also the capacity to spontaneously regress. Despite intensive therapy, the survival for patients with high-risk neuroblastoma and those with recurrent or relapsed disease is low. Hence, there is an urgent need to develop new therapies for these patient groups. The molecular pathogenesis based on high-throughput omics technologies of neuroblastoma is beginning to be resolved which have given the opportunity to develop personalized therapies for high-risk patients. Here we discuss the potential of developing targeted therapies against aberrantly expressed molecules detected in sub-populations of neuroblastoma patients and how these selected targets can be drugged in order to overcome treatment resistance, improve survival and quality of life for these patients and also the possibilities to transfer preclinical research into clinical testing.
Collapse
Affiliation(s)
- John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital Solna, 171 77 Stockholm, Sweden.
| | - Cecilia Dyberg
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital Solna, 171 77 Stockholm, Sweden
| | - Susanne Fransson
- Department of Pathology and Genetics, Sahlgrenska Academy at the University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Malin Wickström
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital Solna, 171 77 Stockholm, Sweden
| |
Collapse
|
42
|
Amoroso L, Haupt R, Garaventa A, Ponzoni M. Investigational drugs in phase II clinical trials for the treatment of neuroblastoma. Expert Opin Investig Drugs 2017; 26:1281-1293. [PMID: 28906153 DOI: 10.1080/13543784.2017.1380625] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Neuroblastoma (NB) is an embryonal tumor originating from undifferentiated neural crest cell, highly heterogeneous ranging from spontaneous regression to progression despite multimodal treatments. Approximately, 20% of patients are refractory to frontline therapy and 50% will relapse/progress after an initial response. The overall five year survival for high-risk neuroblastoma ranges from 35-45%. Despite enhanced understanding of NB biology and the addition of myeloablative chemotherapy, isotretinoin and immunotherapy, survival for high risk NB remains less than 50%. Areas covered: This review summarizes and gives a critical overview of phase II trials investigating therapies for relapsed-refractory and high risk neuroblastoma. Expert opinion: Several novel molecules have been developed and are currently under investigation for the treatment of NB. The trend of novel targeted agents is one towards individualized, tailored therapy, based on the molecular and biological differences that characterize tumors that seem similar based solely on histological analysis. The task of developing new molecules is particularly difficult for NB, given the recurrent development of new patterns of drug resistance. However, even if current research is focused towards identifying the best treatments for each children and young adult with a NB defined disease, a deeper knowledge of the molecular biology and genetics is needed.
Collapse
Affiliation(s)
- Loredana Amoroso
- a Department of Pediatric Oncology , Istituto G.Gaslini , Genova , Italy
| | - Riccardo Haupt
- b Epidemiology and Biostatistics Unit , Istituto G.Gaslini , Genova , Italy
| | - Alberto Garaventa
- a Department of Pediatric Oncology , Istituto G.Gaslini , Genova , Italy
| | - Mirco Ponzoni
- c Experimental Therapy Unit in Oncology , Istituto G. Gaslini , Genova , Italy
| |
Collapse
|