1
|
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: 2.0] [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
|
2
|
Tagalakis AD, Jayarajan V, Maeshima R, Ho KH, Syed F, Wu L, Aldossary AM, Munye MM, Mistry T, Ogunbiyi OK, Sala A, Standing JF, Moghimi SM, Stoker AW, Hart SL. Integrin-Targeted, Short Interfering RNA Nanocomplexes for Neuroblastoma Tumor-Specific Delivery Achieve MYCN Silencing with Improved Survival. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2104843. [PMID: 35712226 PMCID: PMC9178728 DOI: 10.1002/adfm.202104843] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Indexed: 06/15/2023]
Abstract
The authors aim to develop siRNA therapeutics for cancer that can be administered systemically to target tumors and retard their growth. The efficacy of systemic delivery of siRNA to tumors with nanoparticles based on lipids or polymers is often compromised by their rapid clearance from the circulation by the liver. Here, multifunctional cationic and anionic siRNA nanoparticle formulations are described, termed receptor-targeted nanocomplexes (RTNs), that comprise peptides for siRNA packaging into nanoparticles and receptor-mediated cell uptake, together with lipids that confer nanoparticles with stealth properties to enhance stability in the circulation, and fusogenic properties to enhance endosomal release within the cell. Intravenous administration of RTNs in mice leads to predominant accumulation in xenograft tumors, with very little detected in the liver, lung, or spleen. Although non-targeted RTNs also enter the tumor, cell uptake appears to be RGD peptide-dependent indicating integrin-mediated uptake. RTNs with siRNA against MYCN (a member of the Myc family of transcription factors) in mice with MYCN-amplified neuroblastoma tumors show significant retardation of xenograft tumor growth and enhanced survival. This study shows that RTN formulations can achieve specific tumor-targeting, with minimal clearance by the liver and so enable delivery of tumor-targeted siRNA therapeutics.
Collapse
Affiliation(s)
- Aristides D. Tagalakis
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
- Present address:
Department of BiologyEdge Hill UniversityOrmskirkL39 4QPUK
| | - Vignesh Jayarajan
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Ruhina Maeshima
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Kin H. Ho
- Department of InflammationInfection and ImmunityUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Farhatullah Syed
- Department of InflammationInfection and ImmunityUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Lin‐Ping Wu
- Centre for Pharmaceutical Nanotechnology and NanotoxicologyFaculty of Health and Medical SciencesUniversity of CopenhagenUniversitetsparken 2Copenhagen2100Denmark
- Present address:
Guangzhou institute of Biomedicine and HealthChinese Academy of SciencesGuangzhou510530People's Republic of China
| | - Ahmad M. Aldossary
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
- Present address:
National Center for BiotechnologyKing Abdulaziz City for Science and TechnologyRiyadh11442Saudi Arabia
| | - Mustafa M. Munye
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
- Present address:
Cell and Gene Therapy Catapult12th Floor Tower Wing, Guy's Hospital, Great Maze PondLondonSE1 9RTUK
| | - Talisa Mistry
- Department of HistopathologyGreat Ormond Street Hospital for ChildrenNHS Foundation TrustLondonWC1N 3JHUK
| | - Olumide Kayode Ogunbiyi
- Department of HistopathologyGreat Ormond Street Hospital for ChildrenNHS Foundation TrustLondonWC1N 3JHUK
| | - Arturo Sala
- Department of Life SciencesBrunel University LondonKingston LaneMiddlesexUB8 3PHUK
| | - Joseph F. Standing
- Department of InflammationInfection and ImmunityUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Seyed M. Moghimi
- Centre for Pharmaceutical Nanotechnology and NanotoxicologyFaculty of Health and Medical SciencesUniversity of CopenhagenUniversitetsparken 2Copenhagen2100Denmark
- Present address:
School of Pharmacy, and Translational and Clinical Research Institute, the Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneNE1 7RUUK
- Present address:
Colorado Center for Nanomedicine and Nanosafety, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
| | - Andrew W. Stoker
- Department of Developmental Biology and CancerUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| | - Stephen L. Hart
- Department of Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthUniversity College London30 Guilford StreetLondonWC1N 1EHUK
| |
Collapse
|
3
|
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: 6.5] [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
|
4
|
Broomfield LM, Alonso-Moreno C, Martin E, Shafir A, Posadas I, Ceña V, Castro-Osma JA. Aminophosphine ligands as a privileged platform for development of antitumoral ruthenium(ii) arene complexes. Dalton Trans 2017; 46:16113-16125. [DOI: 10.1039/c7dt03369a] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The potential utility of aminophosphine ligands in both high-throughput testing and rational design of new anticancer metallodrugs.
Collapse
Affiliation(s)
- L. M. Broomfield
- Institute of Chemical Research of Catalonia (ICIQ)
- Barcelona Institute of Science and Technology
- Tarragona
- Spain
| | - C. Alonso-Moreno
- Departamento de Química Inorgánica
- Orgánica y Bioquímica
- Facultad de Farmacia
- Universidad de Castilla-La Man-cha
- 02071-Albacete
| | - E. Martin
- Institute of Chemical Research of Catalonia (ICIQ)
- Barcelona Institute of Science and Technology
- Tarragona
- Spain
| | - A. Shafir
- Institute of Chemical Research of Catalonia (ICIQ)
- Barcelona Institute of Science and Technology
- Tarragona
- Spain
| | - I. Posadas
- CIBERNED
- Instituto de Salud Carlos III
- Madrid
- Spain
- Unidad Asociada Neurodeath CSIC-UCLM
| | - V. Ceña
- CIBERNED
- Instituto de Salud Carlos III
- Madrid
- Spain
- Unidad Asociada Neurodeath CSIC-UCLM
| | - J. A. Castro-Osma
- Departamento de Química Inorgánica
- Orgánica y Bioquímica
- Facultad de Farmacia
- Universidad de Castilla-La Man-cha
- 02071-Albacete
| |
Collapse
|
5
|
Peinemann F, Kahangire DA, van Dalen EC, Berthold F. Rapid COJEC versus standard induction therapies for high-risk neuroblastoma. Cochrane Database Syst Rev 2015; 2015:CD010774. [PMID: 25989478 PMCID: PMC10501324 DOI: 10.1002/14651858.cd010774.pub2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Neuroblastoma is a rare malignant disease and mainly affects infants and very young children. The tumors mainly develop in the adrenal medullary tissue and an abdominal mass is the most common presentation. The high-risk group is characterized by metastasis and other characteristics that increase the risk for an adverse outcome. In the rapid COJEC induction schedule, higher single doses of selected drugs than standard induction schedules are administered over a substantially shorter treatment period, with shorter intervals between cycles. Shorter intervals and higher doses increase the dose intensity of chemotherapy and might improve survival. OBJECTIVES The aim of this study was to evaluate the efficacy and adverse events of the rapid COJEC induction schedule as compared to standard induction schedules in patients with high-risk neuroblastoma (as defined by the International Neuroblastoma Risk Group (INRG) classification system). Outcomes of interest were complete response, early toxicity and treatment-related mortality as primary endpoints and overall survival, progression- and event-free survival, late non-hematological toxicity, and health-related quality of life as secondary endpoints. SEARCH METHODS We searched the electronic databases CENTRAL (2014, Issue 11), MEDLINE (PubMed), and EMBASE (Ovid) for articles from inception to 11 November 2014. Further searches included trial registries, conference proceedings, and reference lists of recent reviews and relevant articles. We did not apply limits on publication year or languages. SELECTION CRITERIA Randomized controlled trials evaluating the rapid COJEC induction schedule for high-risk neuroblastoma patients compared to standard induction schedules. DATA COLLECTION AND ANALYSIS Two review authors performed study selection, abstracted data on study and patient characteristics, and assessed risk of bias independently. We resolved differences by discussion or by appeal to a third review author. We performed analyses according to the guidelines of the Cochrane Handbook for Systematic Reviews of Interventions. We used the five GRADE considerations, study limitations, consistency of effect, imprecision, indirectness, and publication bias, to judge the quality of the evidence. We downgraded for risk of bias and imprecision MAIN RESULTS We identified one randomized controlled trial (CCLG-ENSG-5) that included 262 patients with high-risk neuroblastoma who were randomized to receive either rapid COJEC (N = 130) or standard OPEC/COJEC (N = 132) induction chemotherapy. We graded the evidence as low quality; we downgraded for risk of bias and imprecision.There was no clear evidence of a difference between the treatment groups in complete response (risk ratio (RR) 0.99, 95% confidence interval (CI) 0.71 to 1.38), treatment-related mortality (RR 1.21, 95% CI 0.33 to 4.39), overall survival (hazard ratio (HR) 0.83, 95% CI 0.63 to 1.10), and event-free survival (HR 0.86, 95% CI 0.65 to 1.13). We calculated the HRs using the complete follow-up period of the trial.Febrile neutropenia (two or more episodes), proven fungal infections, septicemia (one or more episodes), gastrointestinal toxicity (grade 3 or 4), renal toxicity (glomerular filtration rate < 80 ml/min per body surface area of 1.73 m(2)), neurological toxicity (grade 3 or 4), and ototoxicity (Brock grade 2 to 4) were addressed as early toxicities (during pre-operative chemotherapy). For febrile neutropenia, septicemia, and renal toxicity, a statistically significant difference in favor of the standard treatment arm was identified; for all other early toxicities no clear evidence of a difference between treatment groups was identified. With regard to late non-hematological toxicities (median follow-up 12.7 years; range 6.9 to 16.5 years), the study provided data on any complication, renal toxicity (glomerular filtration rate < 80 ml/min per body surface area of 1.73m(2)), ototoxicity (Brock grade 1 to 4), endocrine complications, neurocognitive complications (i.e. behavioral, speech, or learning difficulties), and second malignancies. For endocrine complications and neurocognitive complications, a statistically significant difference in favor of the rapid COJEC arm was found; for all other late non-hematological toxicities no clear evidence of a difference between treatment groups was identified.Data on progression-free survival and health-related quality of life were not reported. AUTHORS' CONCLUSIONS We identified one randomized controlled trial that evaluated rapid COJEC versus standard induction therapy in patients with high-risk neuroblastoma. No clear evidence of a difference in complete response, treatment-related mortality, overall survival, and event-free survival between the treatment alternatives was found. This could be the result of low power or too short a follow-up period. Results of both early and late toxicities were ambiguous. Information on progression-free survival and health-related quality of life were not available. This trial was performed in the 1990s. Since then, many changes in, for example, treatment and risk classification have occurred. Therefore, based on the currently available evidence, we are uncertain about the effects of rapid COJEC and standard induction therapy in patients with high-risk neuroblastoma. More research is needed for a definitive conclusion.
Collapse
Affiliation(s)
- Frank Peinemann
- University of CologneChildren's HospitalKerpener Str. 62CologneNWGermany50937
| | - Doreen A Kahangire
- University of BirminghamBirmingham and Black Country NIHR CLAHRCSchool of Health and population, Public Health BuildingCollege of Medical and Dental SciencesBirminghamWest MidlandsUKB15 2TT
| | - Elvira C van Dalen
- Emma Children's Hospital/Academic Medical CenterDepartment of Paediatric OncologyPO Box 22660 (room H4‐139)AmsterdamNetherlands1100 DD
| | - Frank Berthold
- Children's Hospital, University of ColognePediatric Oncology and HematologyKerpener Strasse 62CologneGermany50937
| | | |
Collapse
|
6
|
Ameis HM, Drenckhan A, Freytag M, Izbicki JR, Supuran CT, Reinshagen K, Holland-Cunz S, Gros SJ. Carbonic anhydrase IX correlates with survival and is a potential therapeutic target for neuroblastoma. J Enzyme Inhib Med Chem 2015; 31:404-9. [PMID: 25884234 DOI: 10.3109/14756366.2015.1029471] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Carbonic anhydrase IX (CAIX) is involved in pathological processes including tumorgenicity, metastases and poor survival in solid tumors. Twenty-two neuroblastoma samples of patients who were surgically treated at the University Medical Center Hamburg-Eppendorf were evaluated immunohistochemically for expression of CAIX. Results were correlated with clinical parameters and outcome. Neuroblastoma Kelly and SH-EP-Tet-21/N cells were examined for CAIX expression and inhibited with specific inhibitors, FC5-207A and FC8-325A. 32% of neuroblastoma tumors expressed CAIX. This was significantly associated with poorer survival. Kelly and SH-EP-Tet-21/N cells showed a major increase of CAIX RNA under hypoxic conditions. Proliferation of Kelly cells was significantly decreased by CAIX inhibitors, FC5-207A and FC8-325A, while proliferation of SH-EP-Tet-21/N cells was only significantly affected by FC8-325A. CAIX is a potent biomarker that predicts survival in neuroblastoma patients. CAIX-targeted therapy in neuroblastoma cell lines is highly effective and strengthens the potential of CAIX as a clinical therapeutic target in a selected patient collective.
Collapse
Affiliation(s)
- Helen M Ameis
- a Department of Pediatric Surgery , University Medical Center Hamburg-Eppendorf/Altona Children's Hospital , Hamburg , Germany
| | - Astrid Drenckhan
- b Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Morton Freytag
- b Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Jakob R Izbicki
- b Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Claudiu T Supuran
- c Department Neurofarba , Sezione di Scienze farmaceutiche, University of Florence , Florence , Italy , and
| | - Konrad Reinshagen
- a Department of Pediatric Surgery , University Medical Center Hamburg-Eppendorf/Altona Children's Hospital , Hamburg , Germany
| | - Stefan Holland-Cunz
- d Department of Pediatric Surgery , University Children's Hospital of Basel , Basel , Switzerland
| | - Stephanie J Gros
- b Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany .,d Department of Pediatric Surgery , University Children's Hospital of Basel , Basel , Switzerland
| |
Collapse
|
7
|
Ameis HM, Drenckhan A, von Loga K, Escherich G, Wenke K, Izbicki JR, Reinshagen K, Gros SJ. PGK1 as predictor of CXCR4 expression, bone marrow metastases and survival in neuroblastoma. PLoS One 2013; 8:e83701. [PMID: 24376734 PMCID: PMC3869792 DOI: 10.1371/journal.pone.0083701] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 11/06/2013] [Indexed: 12/03/2022] Open
Abstract
Background and Aim A close relationship between phosphoglycerate kinase 1 (PGK1) and the CXCR4/SDF1 axis (chemokine receptor 4/stromal cell derived factor 1) has been shown for several cancers. However, the role of PGK1 has not been investigated for neuroblastoma, and PGK1 might be a therapeutic target for this tumor entity. The aim of the current study was to evaluate the role of PGK1 expression in neuroblastoma patients, to determine the impact of PGK1 expression levels on survival, and to correlate PGK1 expression with CXCR4 expression and bone marrow dissemination. Materials and Methods Samples from 22 patients with neuroblastoma that were surgically treated at the University Medical Center Hamburg-Eppendorf were evaluated for expression of PGK1 and CXCR4 using immunohistochemistry. Results were correlated with clinical parameters, metastases and outcome of patients. Immunocytochemistry, proliferation and expression analysis of CXCR4 and PGK1 were performed in neuroblastoma cell lines. Results PGK1 is expressed in neuroblastoma cells. PGK1 expression is significantly positively correlated with CXCR4 expression and tumor dissemination to the bone marrow. Moreover the expression of PGK1 is significantly associated with a negative impact on survival in patients with neuroblastoma. PGK1 is downregulated by inhibition of CXCR4 in neuroblastoma cells. Conclusion PGK1 appears to play an important role for neuroblastoma, predicting survival and tumor dissemination. Further in vivo studies outstanding, it is a candidate target for novel therapeutic strategies.
Collapse
Affiliation(s)
- Helen M. Ameis
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altona Children's Hospital, Hamburg, Germany
| | - Astrid Drenckhan
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina von Loga
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele Escherich
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Wenke
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altona Children's Hospital, Hamburg, Germany
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf/Altona Children's Hospital, Hamburg, Germany
| | - Stephanie J. Gros
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
8
|
Balza E, Carnemolla B, Mortara L, Castellani P, Soncini D, Accolla RS, Borsi L. Therapy-induced antitumor vaccination in neuroblastomas by the combined targeting of IL-2 and TNFalpha. Int J Cancer 2010; 127:101-10. [PMID: 19877124 DOI: 10.1002/ijc.25018] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
L19-IL2 and L19TNFalpha are fusion proteins composed of L19(scFv), specific for the angiogenesis-associated ED-B containing fibronectin isoform and IL-2 or TNFalpha. Because of the tumor targeting properties of L19, IL-2 and TNFalpha concentrate at therapeutic doses at the tumor vascular level. To evaluate the therapeutic effects of L19-IL2 and L19mTNFalpha in neuroblastoma (NB)-bearing mice, A/J mice bearing Neuro2A or NIE115 NB were systemically treated with L19-IL2 and L19mTNFalpha, alone or in combination protocols. Seventy percent of Neuro2A- and 30% of NIE115-bearing mice were cured by the combined treatment with L19-IL2 and L19mTNFalpha, and further rejected a homologous tumor challenge, indicating specific antitumor immune memory. The immunological bases of tumor cure and rejection were studied. A highly efficient priming of CD4(+) T helper cells and CD8(+) CTL effectors was generated, paralleled by massive infiltration in the tumor tissue of CD4(+) and CD8(+) T cells at day 16 after tumor cell implantation, when, after therapy, tumor volume was drastically reduced and tumor necrosis reached about 80%. The curative treatment resulted in a long-lasting antitumor immune memory, accompanied by a mixed Th1/Th2 type of response. Concluding, L19-IL2 and L19mTNFalpha efficiently cooperate in determining a high percentage of NB cure that, in our experimental models, is strongly associated to the generation of adaptive immunity involving CD4(+) and CD8(+) T cells.
Collapse
Affiliation(s)
- Enrica Balza
- Cell Biology Laboratory, Department of Translational Oncology, Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | | | | | | | | | | | | |
Collapse
|
9
|
García-Castro J, Alemany R, Cascalló M, Martínez-Quintanilla J, Arriero MDM, Lassaletta A, Madero L, Ramírez M. Treatment of metastatic neuroblastoma with systemic oncolytic virotherapy delivered by autologous mesenchymal stem cells: an exploratory study. Cancer Gene Ther 2010; 17:476-83. [PMID: 20168350 DOI: 10.1038/cgt.2010.4] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Treatment of metastatic tumors with engineered adenoviruses that replicate selectively in tumor cells is a new therapeutic approach in cancer. Systemic administration of these oncolytic adenoviruses lack metastatic targeting ability. The tumor stroma engrafting property of intravenously injected mesenchymal stem cells (MSCs) may allow the use of MSCs as cellular vehicles for targeted delivery. In this work, we study the safety and the efficacy of infusing autologous MSCs infected with ICOVIR-5, a new oncolytic adenovirus, for treating metastatic neuroblastoma. Four children with metastatic neuroblastoma refractory to front-line therapies received several doses of autologous MSCs carrying ICOVIR-5, under an approved preliminary study. The tolerance to the treatment was excellent. A complete clinical response was documented in one case, and the child is in complete remission 3 years after this therapy. We postulate that MSCs can deliver oncolytic adenoviruses to metastatic tumors with very low systemic toxicity and with beneficial antitumor effects.
Collapse
Affiliation(s)
- J García-Castro
- Servicio de Oncohematología y Trasplante, Hospital Universitario Niño Jesús, Avenida Menéndez Pelayo 65, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Characterization of highly stable liposomal and immunoliposomal formulations of vincristine and vinblastine. Cancer Chemother Pharmacol 2009; 64:741-51. [PMID: 19184019 PMCID: PMC2717390 DOI: 10.1007/s00280-008-0923-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Accepted: 12/30/2008] [Indexed: 01/15/2023]
Abstract
Purpose Liposome and immunoliposome formulations of two vinca alkaloids, vincristine and vinblastine, were prepared using intraliposomal triethylammonium sucroseoctasulfate and examined for their ability to stabilize the drug for targeted drug delivery in vivo. Methods The pharmacokinetics of both the encapsulated drug (vincristine or vinblastine) and liposomal carrier were examined in Sprague Dawley rats, and the in vivo drug release rates determined. Anti-HER2 immunoliposomal vincristine was prepared from a human anti-HER2/neu scFv and studied for targeted cytotoxic activity in cell culture, and antitumor efficacy in vivo. Results Nanoliposome formulations of vincristine and vinblastine demonstrated similar pharmacokinetic profiles for the liposomal carrier, but increased clearance for liposome encapsulated vinblastine (t1/2 = 9.7 h) relative to vincristine (t1/2 = 18.5 h). Immunoliposome formulations of vincristine targeted to HER2 using an anti-HER2 scFv antibody fragment displayed a marked enhancement in cytotoxicity when compared to non-targeted liposomal vincristine control; 63- or 253-fold for BT474 and SKBR3 breast cancer cells, respectively. Target-specific activity was also demonstrated in HER2-overexpressing human tumor xenografts, where the HER2-targeted formulation was significantly more efficacious than either free vincristine or non-targeted liposomal vincristine. Conclusions These results demonstrate that active targeting of solid tumors with liposomal formulations of vincristine is possible when the resulting immunoliposomes are sufficiently stabilized.
Collapse
|
11
|
|
12
|
Pearson ADJ, Pinkerton CR, Lewis IJ, Imeson J, Ellershaw C, Machin D. High-dose rapid and standard induction chemotherapy for patients aged over 1 year with stage 4 neuroblastoma: a randomised trial. Lancet Oncol 2008; 9:247-56. [PMID: 18308250 DOI: 10.1016/s1470-2045(08)70069-x] [Citation(s) in RCA: 258] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND The current standard treatment for patients with high-risk neuroblastoma includes initial induction chemotherapy with a 21-day interval between induction treatments. We aimed to assess whether an intensive chemotherapy protocol that had a 10-day interval between treatments would improve event-free survival (EFS) in patients aged 1 year or over with high-risk neuroblastoma. METHODS Between Oct 30, 1990, and March 18, 1999, patients with stage 4 neuroblastoma who had not received previous chemotherapy were enrolled from 29 centres in Europe. Patients were randomly assigned to rapid treatment (cisplatin [C], vincristine [O], carboplatin [J], etoposide [E], and cyclophosphamide [C], known as COJEC) or standard treatment (vincristine [O], cisplatin [P], etoposide [E], and cyclophosphamide [C], ie, OPEC, alternated with vincristine [O], carboplatin [J], etoposide [E], and cyclophosphamide [C], ie, OJEC). Both regimens used the same total cumulative doses of each drug (except vincristine), but the dose intensity of the rapid regimen was 1.8-times higher than that of the standard regimen. The standard regimen was given every 21 days if patients showed haematological recovery, whereas the rapid regimen was given every 10 days irrespective of haematological recovery. Response to chemotherapy was assessed according to the conventional International Neuroblastoma Response Criteria (INRC). In responders, surgical excision of the primary tumour was attempted, followed by myeloablation (with 200 mg/m2 of melphalan) and haemopoietic stem-cell rescue. Primary endpoints were 3-year, 5-year, and 10-year EFS. Data were analysed by intention to treat. This trial is registered on the clinical trials site of the US National Cancer Institute website, number NCT00365755, and also as EU-20592 and CCLG-NB-1990-11. FINDINGS 262 patients, of median age 2.95 years (range 1.03-20.97), were randomly assigned-132 patients to standard and 130 patients to rapid treatment. 111 patients in the standard group and 109 patients in the rapid group completed chemotherapy. Chemotherapy doses were recorded for 123 patients in the standard group and 126 patients in the rapid group. 97 of 123 (79%) patients in the standard group and 84 of 126 (67%) patients in the rapid group received at least 90% of the scheduled chemotherapy, and the relative dose intensity was 1.94 compared with the standard regimen. 3-year EFS was 24.2% for patients in the standard group and 31.0% for those in the rapid group (hazard ratio [HR] 0.86 [95% CI 0.66-1.14], p=0.30. 5-year EFS was 18.2% in the standard group and 30.2% in the rapid group, representing a difference of 12.0% (1.8 to 22.3), p=0.022. 10-year EFS was 18.2% in the standard group and 27.1% in the rapid group, representing a difference of 8.9% (-1.2 to 19.0), p=0.085. Myeloablation was given a median of 55 days earlier in patients assigned rapid treatment than those assigned standard treatment. Infective complications (numbers of patients with febrile neutropenia and septicaemia, and if given, time on antibiotic and antifungal treatment) and time in hospital were greater with rapid treatment. Occurrence of fungal infection was the same in both regimens. INTERPRETATION Dose intensity can be increased with a rapid induction regimen in patients with high-risk neuroblastoma. There was no significant difference in OS between the rapid and standard regimens at 5 years and 10 years. However, an increasing difference in EFS after 3 years suggests that the efficacy of the rapid regimen is better than the standard regimen. A rapid induction regimen enables myeloablation to be given much earlier, which might contribute to a better outcome.
Collapse
Affiliation(s)
- Andrew D J Pearson
- Children's Department, Institute of Cancer Research, Royal Marsden Hospital, Sutton, Surrey, UK.
| | | | | | | | | | | |
Collapse
|
13
|
Barker SE, Grosse SM, Siapati EK, Kritz A, Kinnon C, Thrasher AJ, Hart SL. Immunotherapy for neuroblastoma using syngeneic fibroblasts transfected with IL-2 and IL-12. Br J Cancer 2007; 97:210-7. [PMID: 17595664 PMCID: PMC2360308 DOI: 10.1038/sj.bjc.6603857] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/17/2007] [Accepted: 05/24/2007] [Indexed: 11/30/2022] Open
Abstract
Cytokine-modified tumour cells have been used in clinical trials for immunotherapy of neuroblastoma, but primary tumour cells from surgical biopsies are difficult to culture. Autologous fibroblasts, however, are straightforward to manipulate in culture and easy to transfect using nonviral or viral vectors. Here we have compared the antitumour effect of fibroblasts and tumour cells transfected ex vivo to coexpress interleukin-2 (IL-2) and IL-12 in a syngeneic mouse model of neuroblastoma. Coinjection of cytokine-modified fibroblasts with Neuro-2A tumour cells abolished their in vivo tumorigenicity. Treatment of established tumours with three intratumoral doses of transfected fibroblasts showed a significant therapeutic effect with reduced growth or complete eradication of tumours in 90% of mice, associated with extensive leukocyte infiltration. Splenocytes recovered from vaccinated mice showed enhanced IL-2 production following Neuro-2A coculture, and increased cytotoxicity against Neuro-2A targets compared with controls. Furthermore, 100% of the tumour-free mice exhibited immune memory against tumour cells when rechallenged three months later. The potency of transfected fibroblasts was equivalent to that of tumour cells in all experiments. We conclude that syngeneic fibroblasts cotransfected with IL-2 and IL-12 mediate therapeutic effects against established disease, and are capable of generating immunological memory. Furthermore, as they are easier to recover and manipulate than autologous tumour cells, fibroblasts provide an attractive alternative immunotherapeutic strategy for the treatment of neuroblastoma.
Collapse
Affiliation(s)
- S E Barker
- Molecular Immunology Unit, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - S M Grosse
- Molecular Immunology Unit, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - E K Siapati
- Molecular Immunology Unit, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - A Kritz
- Molecular Immunology Unit, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - C Kinnon
- Molecular Immunology Unit, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - A J Thrasher
- Molecular Immunology Unit, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - S L Hart
- Molecular Immunology Unit, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
14
|
Luksch R, Podda M, Gandola L, Polastri D, Piva L, Castellani R, Collini P, Massimino M, Cefalo G, Terenziani M, Ferrari A, Casanova M, Spreafico F, Meazza C, Bozzi F, Marchianò A, Ravagnani F, Fossati-Bellani F. Stage 4 neuroblastoma: sequential hemi-body irradiation or high-dose chemotherapy plus autologous haemopoietic stem cell transplantation to consolidate primary treatment. Br J Cancer 2005; 92:1984-8. [PMID: 15900298 PMCID: PMC2361794 DOI: 10.1038/sj.bjc.6602615] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The aim of the present study was to evaluate the effectiveness of two consecutive nonrandomised treatment programs applied between 1989 and 1999 at the Istituto Nazionale Tumori of Milan in an unselected cohort of 59 children over the age of one with stage 4 neuroblastoma. Both treatment programs consisted of two phases, the induction of the remission phase and the consolidation phase. The induction of the remission phase consisted of intensive chemotherapy, and remained the same throughout the study period. The consolidation phase consisted of sequential hemi-body irradiation (HBI) (10 Gy per session, 6 weeks apart) in the first period (1988–June 1994) and sequential high-dose cyclophosphamide, etoposide, mitoxantrone+L-PAM and autologous haemopoietic stem cell transplantation in the second (July 1994–1999). Intention-to-treat analysis revealed a significantly better outcome for patients treated with the second program, the 5-year event-free survival probability being 0.12 for program 1 and 0.31 for program 2 (P=0.03). This finding led us to conclude that sequential HBI is useless as consolidation treatment. The high-dose chemotherapy adopted in the second program enabled a proportion of patients to obtain long-term survival but, since the clinical results remain unsatisfactory, new treatment strategies are warranted.
Collapse
Affiliation(s)
- R Luksch
- Unità di Pediatria, Istituto Nazionale Tumori di Milano, Via Venezian, 1-20133 Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Affiliation(s)
- Victoria Castel
- Pediatric Oncology Unit, Hospital Infantil La Fe, Avda. Campanar 21, 48009 Valencia, Spain.
| | | | | | | | | |
Collapse
|
16
|
Goldberg-Bittman L, Sagi-Assif O, Meshel T, Nevo I, Levy-Nissenbaum O, Yron I, Witz IP, Ben-Baruch A. Cellular characteristics of neuroblastoma cells: regulation by the ELR--CXC chemokine CXCL10 and expression of a CXCR3-like receptor. Cytokine 2004; 29:105-17. [PMID: 15613278 DOI: 10.1016/j.cyto.2004.10.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2004] [Revised: 10/04/2004] [Accepted: 10/05/2004] [Indexed: 11/20/2022]
Abstract
Bone marrow stroma cells secrete the chemokine CXCL12 that may support bone marrow metastasis formation by neuroblastoma cells. The present study demonstrates that bone marrow stroma cell lines also secrete CXCL10, a chemokine that was shown in the past to have anti-malignancy functions. A receptor recognized by antibodies against CXCR3 was shown to be expressed by six neuroblastoma cell lines. Further detailed analysis was performed on the NUB6 and SK-NMC neuroblastoma cells, showing that CXCL10 induced potent Erk phosphorylation in a G(alpha)i-dependent manner. The role of a CXCR3-like receptor in Erk phosphorylation was substantiated by the ability of CXCL11, another potent CXCR3 ligand, to induce Erk phosphorylation in the NUB6 and SK-NMC cells. Further characterization of CXCL10 activities indicated that CXCL10 partly inhibited the growth of the NUB6 and SK-NMC cells. Both NUB6 and SK-NMC cells did not migrate to CXCL10, although their migratory machinery was intact, as evidenced by their migration to bone marrow constituents. Altogether, these results suggest that CXCL10 interacts with a CXCR3-like receptor in neuroblastoma cell lines, raising the possibility that following the homing of the tumor cells to the bone marrow (through a CXCL10-independent mechanism), CXCL10 may partly inhibit neuroblastoma cell growth at this site.
Collapse
Affiliation(s)
- Lilach Goldberg-Bittman
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Wei JS, Greer BT, Westermann F, Steinberg SM, Son CG, Chen QR, Whiteford CC, Bilke S, Krasnoselsky AL, Cenacchi N, Catchpoole D, Berthold F, Schwab M, Khan J. Prediction of clinical outcome using gene expression profiling and artificial neural networks for patients with neuroblastoma. Cancer Res 2004; 64:6883-91. [PMID: 15466177 PMCID: PMC1298184 DOI: 10.1158/0008-5472.can-04-0695] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Currently, patients with neuroblastoma are classified into risk groups (e.g., according to the Children's Oncology Group risk-stratification) to guide physicians in the choice of the most appropriate therapy. Despite this careful stratification, the survival rate for patients with high-risk neuroblastoma remains <30%, and it is not possible to predict which of these high-risk patients will survive or succumb to the disease. Therefore, we have performed gene expression profiling using cDNA microarrays containing 42,578 clones and used artificial neural networks to develop an accurate predictor of survival for each individual patient with neuroblastoma. Using principal component analysis we found that neuroblastoma tumors exhibited inherent prognostic specific gene expression profiles. Subsequent artificial neural network-based prognosis prediction using expression levels of all 37,920 good-quality clones achieved 88% accuracy. Moreover, using an artificial neural network-based gene minimization strategy in a separate analysis we identified 19 genes, including 2 prognostic markers reported previously, MYCN and CD44, which correctly predicted outcome for 98% of these patients. In addition, these 19 predictor genes were able to additionally partition Children's Oncology Group-stratified high-risk patients into two subgroups according to their survival status (P = 0.0005). Our findings provide evidence of a gene expression signature that can predict prognosis independent of currently known risk factors and could assist physicians in the individual management of patients with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Jun S. Wei
- Advanced Technology Center, Oncogenomics Section, Pediatric Oncology Branch, National Cancer Institute, NIH, Gaithersburg, Maryland
| | - Braden T. Greer
- Advanced Technology Center, Oncogenomics Section, Pediatric Oncology Branch, National Cancer Institute, NIH, Gaithersburg, Maryland
| | - Frank Westermann
- Department of Tumour Genetics–B030, German Cancer Research Center, Heidelberg, Germany
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Chang-Gue Son
- Advanced Technology Center, Oncogenomics Section, Pediatric Oncology Branch, National Cancer Institute, NIH, Gaithersburg, Maryland
- Department of Internal Medicine, College of Oriental Medicine, Daejeon University, Daejeon, Korea
| | - Qing-Rong Chen
- Advanced Technology Center, Oncogenomics Section, Pediatric Oncology Branch, National Cancer Institute, NIH, Gaithersburg, Maryland
| | - Craig C. Whiteford
- Advanced Technology Center, Oncogenomics Section, Pediatric Oncology Branch, National Cancer Institute, NIH, Gaithersburg, Maryland
| | - Sven Bilke
- Advanced Technology Center, Oncogenomics Section, Pediatric Oncology Branch, National Cancer Institute, NIH, Gaithersburg, Maryland
| | - Alexei L. Krasnoselsky
- Advanced Technology Center, Oncogenomics Section, Pediatric Oncology Branch, National Cancer Institute, NIH, Gaithersburg, Maryland
| | - Nicola Cenacchi
- Advanced Technology Center, Oncogenomics Section, Pediatric Oncology Branch, National Cancer Institute, NIH, Gaithersburg, Maryland
| | - Daniel Catchpoole
- Tumour Bank, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia; and
| | - Frank Berthold
- Department of Pediatrics, Klinik für Kinderheilkunde der Universität zu Köln, Köln, Germany
| | - Manfred Schwab
- Department of Tumour Genetics–B030, German Cancer Research Center, Heidelberg, Germany
| | - Javed Khan
- Advanced Technology Center, Oncogenomics Section, Pediatric Oncology Branch, National Cancer Institute, NIH, Gaithersburg, Maryland
- Requests for reprints: Javed Khan, Pediatric Oncology Branch, Oncogenomics Section, National Cancer Institute, 8717 Government Circle, Gaithersburg, MD 20877. Phone: 301-435-2937; Fax: 301-480-0314 or 301-402-3134; E-mail:
| |
Collapse
|
18
|
Lindskog M, Spenger C, Jarvet J, Gräslund A, Kogner P. Predicting Resistance or Response to Chemotherapy by Proton Magnetic Resonance Spectroscopy in Neuroblastoma. J Natl Cancer Inst 2004; 96:1457-66. [PMID: 15467035 DOI: 10.1093/jnci/djh273] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND We previously showed that proton magnetic resonance spectroscopy (1H-MRS) enables estimation of neuroblastoma tumor viability. Here we investigated if 1H-MRS can predict response or resistance to chemotherapy in neuroblastoma. METHODS Neuroblastoma cell lines with various drug sensitivities were treated with cytotoxic drugs (cisplatin, etoposide, and irinotecan) and examined by 1H-MRS. Viability was assessed by trypan blue staining and the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. Nude rats carrying drug-sensitive or drug-resistant neuroblastoma xenografts were treated for 4 days with irinotecan (n = 11) or saline (n = 11) and were examined with 1H-MRS at 4.7 T before and during treatment. The Wilcoxon matched-pairs test was used to test statistical significance of difference within treatment groups. Independent groups were compared using the Mann-Whitney U test. Correlation was assessed with Spearman's rank correlation. All statistical tests were two-sided. RESULTS Cytotoxic drug treatment of drug-sensitive SH-SY5Y neuroblastoma cells resulted in increased methylene and polyunsaturated fatty acid resonances and decreased choline resonance. The methylene/choline ratio correlated with cell death (r(s) = .94, P<.001) and was increased in cisplatin-treated drug-sensitive (SH-SY5Y, IMR-32) but not drug-resistant [SK-N-BE2, SK-N-FI, SK-N-AS] cell lines. No changes were observed in SK-N-BE2 cells treated with irinotecan or cisplatin, whereas circumvention of the resistance by arsenic trioxide treatment led to lipid accumulation and choline depletion. Irinotecan therapy of rats carrying drug-sensitive xenografts caused the methylene/choline ratio of tumors to increase eightfold after 3 days (95% confidence interval [CI] = fivefold to 12-fold; P = .005 compared with pretreatment spectra at day 0) and caused tumors to regress statistically significantly on day 10 compared with pretreatment volume on day 0 (difference = -60%, 95% CI = -12% to -100%, n = 6; P = .012). The methylene/choline ratio of nonregressing drug-resistant xenografts was unaffected. No differences were observed after saline treatment. CONCLUSIONS Response or resistance to chemotherapy is accurately predicted by 1H-MRS in experimental neuroblastoma models in vivo.
Collapse
Affiliation(s)
- Magnus Lindskog
- Childhood Cancer Research Unit, Department of Woman and Child Health, Karolinska Institutet, Karolinska Hospital, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
19
|
Vitali R, Cesi V, Nicotra MR, McDowell HP, Donfrancesco A, Mannarino O, Natali PG, Raschellà G, Dominici C. c-Kit is preferentially expressed in MYCN-amplified neuroblastoma and its effect on cell proliferation is inhibited in vitro by STI-571. Int J Cancer 2003; 106:147-52. [PMID: 12800187 DOI: 10.1002/ijc.11187] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Coexpression for c-Kit receptor and its ligand stem cell factor (SCF) has been described in neuroblastoma (NB) cell lines and tumors, suggesting the existence of an autocrine loop modulating tumor growth. We evaluated c-Kit and SCF expression by immunohistochemistry in a series of 75 primary newly diagnosed neuroblastic tumors. Immunostaining for c-Kit was found in 10/75 and for SCF in 17/75, with 5/10 c-Kit-positive tumors also expressing SCF. For both, c-Kit and SCF staining were predominantly found in the most aggressive subset of tumors, i.e., those amplified for MYCN: c-Kit was detected in 8/14 amplified vs. 2/61 single copy (p<0.001), and SCF in 9/14 amplified vs. 8/61 single copy tumors (p<0.001). Furthermore, the association of c-Kit expression with advanced stage (3 or 4) (p=0.001) and of SCF expression with adrenal primary (p=0.03) was substantiated. The in vitro activity of the tyrosine kinase inhibitor STI-571 (imatinib mesylate, Gleevec, Glivec) on NB cell lines positive or negative for c-Kit was also assessed. When cells were grown in 10% fetal calf serum, the 4 c-Kit-positive cell lines tested were sensitive to STI-571 growth inhibition to a different extent (ranging from 30 to 80%); also the c-Kit-negative cell line GI-CA-N was slightly affected, suggesting that other STI-571 targets operate in regulating NB proliferation. In addition, c-Kit-positive cell lines SK-N-BE2(c) and HTLA230, grown in SCF only, remained sensitive (40 and 70% of growth inhibition, respectively), while, in the same conditions, proliferation of the c-Kit-negative cell line GI-CA-N was not affected. Immunoprecipitation of c-Kit from cell lysates of SK-N-BE2(c) and HTLA230 cells grown in SCF and subsequent western blot analysis of the immunoprecipitates revealed a sharp decrease of c-Kit phosphorylation after STI-571 treatment. These data demonstrate that both c-Kit and SCF are preferentially expressed in vivo in the most aggressive neuroblastic tumors and that their signaling is active in promoting in vitro NB cell proliferation that can be selectively inhibited by treatment with STI-571.
Collapse
Affiliation(s)
- Roberta Vitali
- Bambino Gesù Children's Hospital, Laboratory of Oncology, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Our understanding of the clinical and cellular pharmacology of drugs commonly used in the treatment of childhood cancer have increased greatly over the past two decades. However, with the exception of childhood acute lymphoblastic leukaemia (ALL), our current knowledge of factors such as inter-patient pharmacokinetic variability and cellular determinants of chemosensitivity has not been utilized in the design of integrated clinical studies. Recent pre-clinical and clinical evaluation of the topoisomerase I inhibitors topotecan and irinotecan has highlighted the potential importance of pharmacological factors in their effectiveness as cytotoxics. In this review, the clinical and cellular pharmacology of vincristine, actinomycin D, doxorubicin, cyclophosphamide, ifosfamide, cisplatin, carboplatin and etoposide will be discussed in relation to the major paediatric solid tumours. For each disease type, knowledge of the clinical and cellular pharmacology of a candidate drug will be related to pharmacodynamic responses such as response, toxicity and prognosis. For diseases such as Wilms' tumour, osteogenic sarcoma and Ewing's tumour, histological response to initial induction chemotherapy is of prognostic significance, and the depth of response is increasingly recognised as an important determinant of prognosis for high-risk neuroblastoma. For several of these tumour types, the dose-intensity of chemotherapy may be an important variable in determining prognosis. However the relationship between pharmacokinetic variability, cellular pharmacology and the major determinants of chemosensitivity to those drugs employed in first line therapy is unknown. The study of these relationships, by means of up front window studies in children who present with high-risk disease, may be as important as the introduction of new agents. Indeed, the optimisation of current therapies may be required to allow a fully informed selection of those children for whom novel therapies are truly needed. Funding and international collaboration at the clinical and scientific level would be required to achieve these aims.
Collapse
Affiliation(s)
- E J Estlin
- Department of Paediatric Oncology, Royal Manchester Children's Hospital, Pendlebury, Manchester, M27 4HA, UK.
| | | |
Collapse
|
21
|
Siapati KE, Barker S, Kinnon C, Michalski A, Anderson R, Brickell P, Thrasher AJ, Hart SL. Improved antitumour immunity in murine neuroblastoma using a combination of IL-2 and IL-12. Br J Cancer 2003; 88:1641-8. [PMID: 12771934 PMCID: PMC2377114 DOI: 10.1038/sj.bjc.6600928] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Neuroblastoma immunotherapy using cytokine-modified tumour cells has been tested in clinical trials. However, because of the complex nature of antitumour immune responses, a number of therapies may be required for complete tumour eradication and generation of systemic immunity. We report here the improved antitumour effect of two cytokines, interleukin-2 (IL-2) and interleukin-12 (IL-12), when coexpressed by neuroblastoma cell lines. Initially, transfection of human and mouse neuroblastoma cell lines resulted in high expression levels of biologically active IL-2 and IL-12 in vitro. These cytokines when expressed by transfected Neuro-2A cells completely abolished their in vivo tumorigenicity in a syngeneic neuroblastoma model. Vaccination of established tumours with IL-12-producing cells exhibited a clear effect with reduced tumour growth in the presence of IL-2. In vivo depletion studies showed that CD4(+) and CD8(+) T cells mediate the response against cytokine-producing cells. These results suggest that IL-2 and IL-12, when cotransfected in tumour cells, are effective against established disease and provide a promising immunotherapeutic approach for the treatment of neuroblastoma.
Collapse
Affiliation(s)
- K E Siapati
- Molecular Immunology Unit, Institute of Child Health, London, UK
| | - S Barker
- Molecular Immunology Unit, Institute of Child Health, London, UK
| | - C Kinnon
- Molecular Immunology Unit, Institute of Child Health, London, UK
| | - A Michalski
- Oncology and Haematology Department, Great Ormond Street Hospital for Children NHS Trust, London, UK
| | - R Anderson
- Department of Haematology, Royal Free Hospital School of Medicine, London, UK
| | - P Brickell
- Oncology and Haematology Department, Great Ormond Street Hospital for Children NHS Trust, London, UK
| | - A J Thrasher
- Molecular Immunology Unit, Institute of Child Health, London, UK
| | - S L Hart
- Molecular Immunology Unit, Institute of Child Health, London, UK
- Molecular Immunology Unit, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK. E-mail:
| |
Collapse
|
22
|
Frappaz D, Perol D, Michon J, Berger C, Coze C, Bernard JL, Zucker JM, Philip T. The LMCE5 unselected cohort of 25 children consecutively diagnosed with untreated stage 4 neuroblastoma over 1 year at diagnosis. Br J Cancer 2002; 87:1197-203. [PMID: 12439705 PMCID: PMC2408899 DOI: 10.1038/sj.bjc.6600627] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2002] [Revised: 08/27/2002] [Accepted: 09/08/2002] [Indexed: 01/21/2023] Open
Abstract
The Lyon-Marseille-Curie-Est (LMCE) of France cooperative group has previously reported successive series of unselected stage four children older than 1 year at diagnosis with metastatic neuroblastoma (LMCE 1 and 3). The goal of LMCE 5 study was to increase progression free survival rate as compared to LMCE 1 and 3. Based on improvements reported with post induction chemotherapy, the LMCE 5 used post induction for all children, but omitted total body irradiation and immunomagnetic purging in megatherapy regimen for all children. Twenty-five sequentially diagnosed children received an induction regimen which compared with previous induction included an increased dose of etoposide and cyclophosphamide, delivered similar dose of cisplatinum, and deleted doxorubicin and vincristin. After surgery treatment was stratified based on response and eligible children received etoposide carboplatin (LMCE 5A : n=10)+/-doxorubicin (LMCE 5B-C n=13) followed by megatherapy (melphalan without total body irradiation and unpurged peripheral blood stem cell rescue). The increase in drug doses during induction did not improve remission rate. The progression free survival at 6 years is 8%. It is significantly worse than LMCE 3, and equivalent to LMCE 1 study though toxic death rate has decreased with increasing experience. Failure to improve the response rate during induction and reducing the megatherapy regimen may be the main factors in this disappointing result. Modified strategies for induction, non toxic alternative to total body irradiation, and post megatherapy regimen should be developed.
Collapse
Affiliation(s)
- D Frappaz
- Department of Pediatrics, Centre Léon Bérard, 28 rue Laënnec, Lyon, 69373, France.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Meczes EL, Pearson ADJ, Austin CA, Tilby MJ. Schedule-dependent response of neuroblastoma cell lines to combinations of etoposide and cisplatin. Br J Cancer 2002; 86:485-9. [PMID: 11875719 PMCID: PMC2375204 DOI: 10.1038/sj.bjc.6600060] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2001] [Revised: 10/26/2001] [Accepted: 11/01/2001] [Indexed: 12/20/2022] Open
Abstract
The growth inhibitory effects of cisplatin and etoposide on neuroblastoma cell lines were investigated in several scheduled combinations. Results were analyzed using median effect and combination index analyses. In all schedules in which cisplatin was administered prior to etoposide a synergistic effect was observed. Conversely, an antagonistic effect was seen in all schedules where etoposide was administered before cisplatin.
Collapse
Affiliation(s)
- E L Meczes
- Paediatric Oncology Laboratory, Cancer Research Unit, Catherine Cookson Building, The Medical School, University of Newcastle upon Tyne, Newcastle upon Tyne NE2 4HH UK
| | | | | | | |
Collapse
|
24
|
Geminder H, Sagi-Assif O, Goldberg L, Meshel T, Rechavi G, Witz IP, Ben-Baruch A. A possible role for CXCR4 and its ligand, the CXC chemokine stromal cell-derived factor-1, in the development of bone marrow metastases in neuroblastoma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:4747-57. [PMID: 11591806 DOI: 10.4049/jimmunol.167.8.4747] [Citation(s) in RCA: 288] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The homing of hemopoietic stem cells to the bone marrow is mediated by specific interactions occurring between CXCR4, which is expressed on hemopoietic stem cells, and its ligand, stromal cell-derived factor-1 (SDF-1), a CXC chemokine secreted by bone marrow stromal cells. In the present study we evaluated the possibility that neuroblastoma cells use a mechanism similar to that used by hemopoietic stem cells to home to the bone marrow and adhere to bone marrow stromal cells. Our study suggests that CXCR4 expression may be a general characteristic of neuroblastoma cells. SH-SY5Y neuroblastoma cells express not only CXCR4, but also its ligand, SDF-1. CXCR4 expression on SH-SY5Y neuroblastoma cells is tightly regulated by tumor cell-derived SDF-1, as demonstrated by the ability of neutralizing Abs against human SDF-1alpha to up-regulate CXCR4 expression on the tumor cells. The reduction in CXCR4 expression following short term exposure to recombinant human SDF-1alpha can be recovered as a result of de novo receptor synthesis. Recombinant human SDF-1alpha induces the migration of CXCR4-expressing SH-SY5Y neuroblastoma cells in CXCR4- and heterotrimeric G protein-dependent manners. Furthermore, SH-SY5Y cells interact at multiple levels with bone marrow components, as evidenced by the fact that bone marrow-derived constituents promote SH-SY5Y cell migration, adhesion to bone marrow stromal cells, and proliferation. These results suggest that SH-SY5Y neuroblastoma cells are equipped with adequate machinery to support their homing to the bone marrow. Therefore, the ability of neuroblastoma tumors to preferentially form metastases in the bone marrow may be influenced by a set of complex CXCR4-SDF-1 interactions.
Collapse
Affiliation(s)
- H Geminder
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences and The Ela Kodesz Institute for Research on Cancer Development and Prevention, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | |
Collapse
|