1
|
Tan J, McLoone JK, Wakefield CE, Nassar N, Cohn RJ, Signorelli C. Neuroblastoma survivors' self-reported late effects, quality of life, health-care use, and risk perceptions. Palliat Support Care 2024; 22:296-305. [PMID: 37311662 DOI: 10.1017/s1478951523000615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND Survivors of childhood neuroblastoma are at risk of multiple treatment-related health problems (late effects), impacting their quality of life. While late effects and quality of life among Australia and New Zealand (ANZ) childhood cancer survivors have been reported, the outcomes of neuroblastoma survivors specifically have not been reported, limiting critical information to inform treatment and care. METHODS Young neuroblastoma survivors or their parents (as proxy for survivors <16 years) were invited to complete a survey and optional telephone interview. Survivors' late effects, risk perceptions, health-care use, and health-related quality of life were surveyed and analyzed using descriptive statistics and linear regression analyses. In-depth interviews explored participants' experiences, knowledge, and perception of late effects and information needs. Thematic content analysis was used to summarize the data. RESULTS Thirty-nine neuroblastoma survivors or parents completed questionnaires (median age = 16 years, 39% male), with 13 also completing interviews. Thirty-two participants (82%) reported experiencing at least 1 late effect, most commonly dental problems (56%), vision/hearing problems (47%), and fatigue (44%). Participants reported high overall quality of life (index = 0.9, range = 0.2-1.0); however, more participants experienced anxiety/depression compared to the population norm (50% met criteria versus 25%, χ2 = 13, p < 0.001). Approximately half of participants (53%) believed they were at risk of developing further late effects. Qualitatively, participants reported knowledge gaps in understanding their risk of developing late effects. CONCLUSION Many neuroblastoma survivors appear to experience late effects, anxiety/depression and have unmet cancer-related information needs. This study highlights important areas for intervention to reduce the impact of neuroblastoma and its treatment in childhood and young adulthood.
Collapse
Affiliation(s)
- Jessica Tan
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Randwick Clinical Campus, UNSW Sydney, Sydney, NSW, Australia
| | - Jordana K McLoone
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Randwick Clinical Campus, UNSW Sydney, Sydney, NSW, Australia
| | - Claire E Wakefield
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Randwick Clinical Campus, UNSW Sydney, Sydney, NSW, Australia
| | - Natasha Nassar
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Richard J Cohn
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Randwick Clinical Campus, UNSW Sydney, Sydney, NSW, Australia
| | - Christina Signorelli
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Randwick Clinical Campus, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
2
|
Gonzalez Malagon SG, Liu KJ. Linking neural crest development to neuroblastoma pathology. Development 2022; 149:276149. [DOI: 10.1242/dev.200331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Although rare, childhood (paediatric) cancers are a major cause of death in young children. Unlike many adult cancers, paediatric cancers, such as neuroblastoma (NB), are developmental diseases that rarely show genetic predispositions. NB is the most common extracranial solid tumour in children, accounting for ∼15% of paediatric cancer deaths. This heterogeneous cancer arises from undifferentiated neural crest-derived progenitor cells. As neural crest cells are multipotent and migratory, they are often considered the embryonic paradigm of cancer stem cells. However, very little is known about the events that trigger tumour initiation and progression. Here, we discuss recent insights into sympathoadrenal lineage specification, as well as genetic factors associated with NB. With this in mind, we consider the molecular underpinnings of NB in the context of developmental trajectories of the neural crest lineage. This allows us to compare distinct subtypes of the disease and gene-function interactions during sensitive phases of neural crest development.
Collapse
Affiliation(s)
- Sandra Guadalupe Gonzalez Malagon
- Biomedical Research Institute, Foundation for Research and Technology, University of Ioannina Campus 1 , 45115 Ioannina , Greece
- School of Health Sciences and Institute of Biosciences, University Research Centre, University of Ioannina 2 Department of Biological Applications and Technology , , 45110 Ioannina , Greece
| | - Karen J. Liu
- Centre for Craniofacial and Regenerative Biology, King's College London 3 , London SE1 9RT , UK
| |
Collapse
|
3
|
Usmani N, Deyell RJ, Portwine C, Rafael MS, Moorehead PC, Shammas A, Vali R, Farfan M, Vanniyasingam T, Morgenstern DA, Irwin MS. Residual meta-iodobenzyl guanidine (MIBG) positivity following therapy for metastatic neuroblastoma: Patient characteristics, imaging, and outcome. Pediatr Blood Cancer 2021; 68:e29289. [PMID: 34411405 DOI: 10.1002/pbc.29289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 07/18/2021] [Accepted: 07/26/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Meta-iodobenzylguanidine(MIBG) scans are used to detect neuroblastoma metastatic lesions at diagnosis and during posttreatment surveillance. MIBG positivity following induction chemotherapy correlates with poor outcome; however, there are reports of patients with progression-free survival despite MIBG positivity at the end of therapy. The factors distinguishing these survivors from patients who progress or relapse are unclear. FDG-positron-emission tomography (PET) scans can also detect metastatic lesions at diagnosis; however, their role in posttherapy surveillance is less well studied. METHODS We performed a retrospective analysis of International Neuroblastoma Staging System (INSS) stage 4 patients to identify those with residual MIBG-avid metastatic lesions on end-of-therapy scans without prior progression. Data collected included age, disease sites, histopathology, biomarkers, treatment, imaging studies, and response. RESULTS Eleven of 265 patients met inclusion criteria. At diagnosis three of 11 patients were classified as intermediate and eight of 11 high risk; nine of 11 had documented marrow involvement. Histologic classification was favorable for four of 10 and MYCN amplification was detected in zero of 11 cases. The median time with persistent MIBG positivity following treatment was 1.5 years. Seven patients had at least one PET scan with low or background activity. Biopsies of three of three MIBG-avid residual lesions showed differentiation. All patients remain alive with no disease progression at a median of 4.0 years since end of therapy. CONCLUSION Persistently MIBG-avid metastatic lesions in subsets of patients following completion of therapy may not represent active disease that will progress. Further studies are needed to determine whether MYCN status or other biomarkers, and/or PET scans, may help identify patients with residual inactive MIBG lesions who require no further therapy.
Collapse
Affiliation(s)
- Nida Usmani
- Department of Pediatrics, Division of Hematology and Oncology, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Rebecca J Deyell
- Department of Pediatrics, Division of Hematology/Oncology/BMT, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Carol Portwine
- Department of Pediatrics, Division of Hematology and Oncology, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Margarida Simao Rafael
- Department of Pediatrics, Division of Hematology and Oncology, Janeway Children's Health and Rehabilitation Centre, and Memorial University, St. John's, Newfoundland and Labrador, Canada
| | - Paul C Moorehead
- Department of Pediatrics, Division of Hematology and Oncology, Janeway Children's Health and Rehabilitation Centre, and Memorial University, St. John's, Newfoundland and Labrador, Canada
| | - Amer Shammas
- Department of Diagnostic Imaging, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Reza Vali
- Department of Diagnostic Imaging, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Mateo Farfan
- Department of Pediatrics, Division of Hematology/Oncology/BMT, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Daniel A Morgenstern
- Department of Pediatrics, Division of Hematology & Oncology, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Meredith S Irwin
- Department of Pediatrics, Division of Hematology & Oncology, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Butler E, Ludwig K, Pacenta HL, Klesse LJ, Watt TC, Laetsch TW. Recent progress in the treatment of cancer in children. CA Cancer J Clin 2021; 71:315-332. [PMID: 33793968 DOI: 10.3322/caac.21665] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Although significant improvements have been made in the outcomes of children with cancer, the pace of improvement has slowed in recent years as the limits of therapy intensification may have been reached for many pediatric cancers. Furthermore, with increasing numbers of pediatric cancer survivors, the long-term side effects of treatment have become increasingly apparent. Therefore, attention has shifted to the use of molecularly targeted agents and immunotherapies to improve the outcomes of children who are not cured by traditional cytotoxic chemotherapies and to decrease exposure to cytotoxic chemotherapy and reduce late effects. This review describes the recent progress in the treatment of children with cancer, focusing in particular on diseases in which targeted and immunotherapeutic agents have made an impact.
Collapse
Affiliation(s)
- Erin Butler
- Department of Pediatrics and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center and Children's Health, Dallas, Texas
| | - Kathleen Ludwig
- Department of Pediatrics and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center and Children's Health, Dallas, Texas
| | - Holly L Pacenta
- Division of Hematology and Oncology, Cook Children's Medical Center, Fort Worth, Texas
| | - Laura J Klesse
- Department of Pediatrics and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center and Children's Health, Dallas, Texas
| | - Tanya C Watt
- Department of Pediatrics and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center and Children's Health, Dallas, Texas
| | - Theodore W Laetsch
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
5
|
Long-term follow-up of high-risk neuroblastoma survivors treated with high-dose chemotherapy and stem cell transplantation rescue. Bone Marrow Transplant 2021; 56:1984-1997. [PMID: 33824435 DOI: 10.1038/s41409-021-01258-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 02/08/2021] [Accepted: 02/23/2021] [Indexed: 11/08/2022]
Abstract
Intensive treatments including high-dose chemotherapy (HDC) with autologous stem cell rescue have improved high-risk neuroblastoma (HRNB) survival. We report the long-term health status of 145 HRNB survivors, alive and disease-free 5 years post HDC. Median follow-up was 15 years (range = 5-34). Six patients experienced late relapses, 11 developed second malignant neoplasms (SMNs), and 9 died. Event-free and overall survivals 20 years post HDC were 82% (95% CI = 70%-90%) and 89% (78%-95%), respectively. Compared with the French general population, the standardized mortality ratio was 19 (95% CI = 8.7-36.1; p < 0.0001) and the absolute excess risk was 37.6 (19.2-73.5). Late effects were observed in 135/145 patients (median = 3 events/patient); 103 had at least one severe event. SMNs arose at a median of 20 years post HDC and included carcinoma (n = 5), sarcoma (2), acute myeloid leukemia (2), melanoma (1), and malignant glioma (1). Non-oncologic health events included dental maldevelopment (60%), severe hearing loss (20% cumulative probability at 15 years), hepatic focal nodular hyperplasia (14%), thyroid (11%), cardiac (8%), and renal (7%) diseases and growth retardation (height-for-age z-score ≤ -2 for 21%). Gonadal insufficiency was near-universal after busulfan (40/43 females, 33/35 males). Severe late effects are frequent and progressive in HRNB survivors needing systematic very long-term follow-up.
Collapse
|
6
|
Streefkerk N, Fioole LCE, Beijer JGM, Feijen ELAM, Teepen JC, Winther JF, Ronckers CM, Loonen JJ, van Dulmen-den Broeder E, Skinner R, Hudson MM, Tissing WJE, Korevaar JC, Mulder RL, Kremer LCM. Large variation in assessment and outcome definitions to describe the burden of long-term morbidity in childhood cancer survivors: A systematic review. Pediatr Blood Cancer 2020; 67:e28611. [PMID: 32881287 DOI: 10.1002/pbc.28611] [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] [Received: 03/14/2020] [Revised: 06/14/2020] [Accepted: 07/01/2020] [Indexed: 12/16/2022]
Abstract
We systematically reviewed outcome assessment methods, outcome classification, and severity grading of reported outcomes in studies investigating the burden of physical long-term morbidity in childhood cancer survivors (CCS). A MEDLINE and EMBASE search identified 56 studies reporting on three or more types of health conditions in 5-year CCS, for which information was extracted on outcome types and classification, methods of outcome ascertainment, and severity grading. There was substantial variability in classification and types of health conditions reported and in methods of outcome ascertainment. Only 59% of the included studies applied severity grading, mainly the common terminology criteria of adverse events. This large variation in assessment and definition of the burden of physical long-term morbidity in CCS challenges interpretation, comparison, and pooling data across studies. Global collaboration is needed to standardize assessments and harmonize definitions of long-term physical morbidity and associated outcomes in childhood cancer survivorship research.
Collapse
Affiliation(s)
- Nina Streefkerk
- Department of Pediatric Oncology and Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Lisanne C E Fioole
- Department of Pediatric Oncology and Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Josien G M Beijer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Elizabeth Lieke A M Feijen
- Department of Pediatric Oncology and Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jop C Teepen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jeanette F Winther
- Department of Clinical Sciences, Pediatric Oncology and Hematology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Cecile M Ronckers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jaqueline J Loonen
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eline van Dulmen-den Broeder
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Pediatric Oncology/Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Rod Skinner
- Department of Paediatric and Adolescent Haematology and Oncology and Children's BMT Unit, Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, and Northern Institute of Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Melissa M Hudson
- Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Wim J E Tissing
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Pediatric Oncology/Hematology, Beatrix Children's Hospital/University of Groningen/University Medical Center Groningen, Groningen, The Netherlands
| | - Joke C Korevaar
- Netherlands Institute for Health Services Research, Utrecht, The Netherlands
| | - Renée L Mulder
- Department of Pediatric Oncology and Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Leontine C M Kremer
- Department of Pediatric Oncology and Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
7
|
Park JR, Kreissman SG, London WB, Naranjo A, Cohn SL, Hogarty MD, Tenney SC, Haas-Kogan D, Shaw PJ, Kraveka JM, Roberts SS, Geiger JD, Doski JJ, Voss SD, Maris JM, Grupp SA, Diller L. Effect of Tandem Autologous Stem Cell Transplant vs Single Transplant on Event-Free Survival in Patients With High-Risk Neuroblastoma: A Randomized Clinical Trial. JAMA 2019; 322:746-755. [PMID: 31454045 PMCID: PMC6714031 DOI: 10.1001/jama.2019.11642] [Citation(s) in RCA: 213] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
IMPORTANCE Induction chemotherapy followed by high-dose therapy with autologous stem cell transplant and subsequent antidisialoganglioside antibody immunotherapy is standard of care for patients with high-risk neuroblastoma, but survival rate among these patients remains low. OBJECTIVE To determine if tandem autologous transplant improves event-free survival (EFS) compared with single transplant. DESIGN, SETTING, AND PARTICIPANTS Patients were enrolled in this randomized clinical trial from November 2007 to February 2012 at 142 Children's Oncology Group centers in the United States, Canada, Switzerland, Australia, and New Zealand. A total of 652 eligible patients aged 30 years or younger with protocol-defined high-risk neuroblastoma were enrolled and 355 were randomized. The final date of follow-up was June 29, 2017, and the data analyses cut-off date was June 30, 2017. INTERVENTIONS Patients were randomized to receive tandem transplant with thiotepa/cyclophosphamide followed by dose-reduced carboplatin/etoposide/melphalan (n = 176) or single transplant with carboplatin/etoposide/melphalan (n = 179). MAIN OUTCOMES AND MEASURES The primary outcome was EFS from randomization to the occurrence of the first event (relapse, progression, secondary malignancy, or death from any cause). The study was designed to test the 1-sided hypothesis of superiority of tandem transplant compared with single transplant. RESULTS Among the 652 eligible patients enrolled, 297 did not undergo randomization because they were nonrandomly assigned (n = 27), ineligible for randomization (n = 62), had no therapy (n = 1), or because of physician/parent preference (n = 207). Among 355 patients randomized (median diagnosis age, 36.1 months; 152 [42.8%] female), 297 patients (83.7%) completed the study and 21 (5.9%) were lost to follow-up after completing protocol therapy. Three-year EFS from the time of randomization was 61.6% (95% CI, 54.3%-68.9%) in the tandem transplant group and 48.4% (95% CI, 41.0%-55.7%) in the single transplant group (1-sided log-rank P=.006). The median (range) duration of follow-up after randomization for 181 patients without an event was 5.6 (0.6-8.9) years. The most common significant toxicities following tandem vs single transplant were mucosal (11.7% vs 15.4%) and infectious (17.9% vs 18.3%). CONCLUSIONS AND RELEVANCE Among patients aged 30 years or younger with high-risk neuroblastoma, tandem transplant resulted in a significantly better EFS than single transplant. However, because of the low randomization rate, the findings may not be representative of all patients with high-risk neuroblastoma. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT00567567.
Collapse
Affiliation(s)
- Julie R. Park
- Department of Pediatrics, Seattle Children’s Hospital, Seattle, Washington
- University of Washington, Seattle
| | - Susan G. Kreissman
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina
| | - Wendy B. London
- Department of Pediatrics, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Arlene Naranjo
- Department of Biostatistics, University of Florida, Children’s Oncology Group Statistics and Data Center, Gainesville
| | | | - Michael D. Hogarty
- Department of Pediatrics Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Sheena C. Tenney
- Department of Biostatistics, University of Florida, Children’s Oncology Group Statistics and Data Center, Gainesville
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Dana Farber/Brigham and Women’s Cancer Center and Harvard Medical School, Boston, Massachusetts
| | - Peter John Shaw
- Bone Marrow Transplant, Children's Hospital at Westmead, Sydney, Australia
| | | | - Stephen S. Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - James Duncan Geiger
- Section of Pediatric Surgery, CS Mott Children’s Hospital, Michigan Medicine, Ann Arbor
| | - John J. Doski
- Departments of Surgery and Pediatrics, UT Health San Antonio, San Antonio, Texas
| | - Stephan D. Voss
- Department of Radiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - John M. Maris
- Department of Pediatrics Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Stephan A. Grupp
- Department of Pediatrics Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Lisa Diller
- Department of Pediatrics, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
8
|
Delgado J, Jaramillo D, Chauvin NA, Guo M, Stratton MS, Sweeney HE, Barrera CA, Mostoufi-Moab S. Evaluating growth failure with diffusion tensor imaging in pediatric survivors of high-risk neuroblastoma treated with high-dose cis-retinoic acid. Pediatr Radiol 2019; 49:1056-1065. [PMID: 31055614 PMCID: PMC6599475 DOI: 10.1007/s00247-019-04409-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/09/2019] [Accepted: 04/11/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND The survival of patients with high-risk neuroblastoma has increased with multimodal therapy, but most survivors demonstrate growth failure. OBJECTIVE To assess physeal abnormalities in children with high-risk neuroblastoma in comparison to normal controls by using diffusion tensor imaging (DTI) of the distal femoral physis and adjacent metaphysis. MATERIALS AND METHODS We prospectively obtained physeal DTI at 3.0 T in 20 subjects (mean age: 12.4 years, 7 females) with high-risk neuroblastoma treated with high-dose cis-retinoic acid, and 20 age- and gender-matched controls. We compared fractional anisotropy (FA), normalized tract volume (cm3/cm2) and tract concentration (tracts/cm2) between the groups, in relation to height Z-score and response to growth hormone therapy. Tractography images were evaluated qualitatively. RESULTS DTI parameters were significantly lower in high-risk neuroblastoma survivors compared to controls (P<0.01), particularly if the patients were exposed to both cis-retinoic acid and total body irradiation (P<0.05). For survivors and controls, DTI values were respectively [mean ± standard deviation]: tract concentration (tracts/cm2), 23.2±14.7 and 36.7±10.5; normalized tract volume (cm3/cm2), 0.44±0.27 and 0.70±0.21, and FA, 0.22±0.05 and 0.26±0.02. High-risk neuroblastoma survivors responding to growth hormone compared to non-responders had higher FA (0.25±0.04 and 0.18±0.03, respectively, P=0.02), and tract concentration (tracts/cm2) (31.4±13.7 and 14.8±7.9, respectively, P<0.05). FA, normalized tract volume and tract concentration were linearly related to height Z-score (R2>0.31; P<0.001). Qualitatively, tracts were nearly absent in all non-responders to growth hormone and abundant in all responders (P=0.02). CONCLUSION DTI shows physeal abnormalities that correlate with short stature in high-risk neuroblastoma survivors and demonstrates response to growth hormone treatment.
Collapse
Affiliation(s)
- Jorge Delgado
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Diego Jaramillo
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Nancy A. Chauvin
- Department of Radiology, Penn State Medical Center, Hershey, PA, USA
| | - Michelle Guo
- Division of Oncology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Perelman School of Medicine, Philadelphia, PA, USA
| | - Mackenzie S. Stratton
- Division of Oncology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Perelman School of Medicine, Philadelphia, PA, USA
| | - Hannah E. Sweeney
- Division of Oncology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Perelman School of Medicine, Philadelphia, PA, USA
| | - Christian A. Barrera
- Department of Radiology, The Children’s Hospital of Philadelphia, Perelman School of Medicine, Philadelphia, PA, USA
| | - Sogol Mostoufi-Moab
- Division of Oncology, Department of Pediatrics, The Children's Hospital of Philadelphia, Perelman School of Medicine, Philadelphia, PA, USA. .,Division of Endocrinology, Department of Pediatrics, The Children's Hospital of Philadelphia, Perelman School of Medicine, 2716 South St., 14th Floor, Philadelphia, PA, 19146, USA.
| |
Collapse
|
9
|
Moreno L, Vaidya SJ, Schrey D, Pinkerton CR, Lewis IJ, Kearns PR, Machin D, Pearson ADJ. Long-term analysis of children with metastatic neuroblastoma treated in the ENSG5 randomised clinical trial. Pediatr Blood Cancer 2019; 66:e27565. [PMID: 30516328 DOI: 10.1002/pbc.27565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/24/2018] [Accepted: 10/30/2018] [Indexed: 11/11/2022]
Abstract
BACKGROUND The European Neuroblastoma Study Group 5 (ENSG5) trial showed that time-intensive "rapid" induction chemotherapy (COJEC) was superior to "standard" 3-weekly chemotherapy for children with high-risk metastatic neuroblastoma. Long-term outcomes of the ENSG5 trial were analysed. PROCEDURE Patients with metastatic neuroblastoma aged ≥12 months were randomly assigned to "standard" or "rapid" induction, receiving the same chemotherapy drugs and doses. Event-free survival (EFS) and overall survival (OS) were analysed and prognostic factors evaluated. Amongst patients surviving >5 years, a population of children with persistent metastatic disease after the end of treatment was identified and described. RESULTS Ten-year EFS was 18.2% (95% confidence interval: 12.2-25.2) for the "standard" arm and 26.8% (19.5-34.7) for the "rapid" arm (hazard ratio [HR] 0.85, P = 0.28). Ten-year OS for the "standard" arm was 19.7% (13.4-26.8) and 28.3% (20.8-36.2) for the "rapid arm" (HR 0.83, P = 0.19). There was a trend for worse EFS and OS for patients having MYCN amplification (HR 1.37 and 1.40, respectively) and those with partial and mixed response to induction (HR 1.69 and 1.75 for EFS and 1.66 and 2.00 for OS, respectively). Among 69 patients who survived >5 years, six had persistent metastatic disease after the end of treatment. CONCLUSION The benefit of the "rapid" induction regimen seems to be maintained in the long term, although the small number of survivors could justify the lack of statistical significance. MYCN amplification and poor metastatic response to induction could be associated with worse outcomes. A small group of patients with persistent metastatic disease that survived long term has been described.
Collapse
Affiliation(s)
- Lucas Moreno
- Clinical Trials Unit, Department of, Paediatric Haematology, Oncology and SCT, Hospital Infantil Universitario Niño Jesus, Madrid, Spain
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Sucheta J Vaidya
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Dominik Schrey
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | | | - Ian J Lewis
- Leeds Community Healthcare NHS Trust, Leeds, United Kingdom
| | - Pamela R Kearns
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - David Machin
- Department of Cancer Studies, Clinical Sciences Building, Leicester Royal Infirmary, University of Leicester, Leicester, United Kingdom
| | - Andrew D J Pearson
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom (retired)
| |
Collapse
|
10
|
Larsson K, Kock A, Kogner P, Jakobsson PJ. Targeting the COX/mPGES-1/PGE 2 Pathway in Neuroblastoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1161:89-100. [PMID: 31562624 DOI: 10.1007/978-3-030-21735-8_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The importance of prostaglandin E2 in cancer progression is well established, but research on its role in cancer has so far mostly been focused on epithelial cancer in adults while the knowledge about the contribution of prostaglandin E2 to childhood malignancies is limited. Neuroblastoma, an extracranial solid tumor of the sympathetic nervous system, mainly affects young children. Patients with tumors classified as high-risk have poor survival despite receiving intensive treatment, illustrating a need for new treatments complimenting existing ones. The basis of neuroblastoma treatment e.g. chemotherapy and radiation therapy, target the proliferating genetically unstable tumor cells leading to treatment resistance and relapses. The tumor microenvironment is an avenue, still to a great extent, unexplored and lacking effective targeted therapies. Cancer-associated fibroblasts is the main source of prostaglandin E2 in neuroblastoma contributing to angiogenesis, immunosuppression and tumor growth. Prostaglandin E2 is formed from its precursor arachidonic acid in a two-step enzymatic reaction. Arachidonic acid is first converted by cyclooxygenases into prostaglandin H2 and then further converted by microsomal prostaglandin E synthase-1 into prostaglandin E2. We believe targeting of microsomal prostaglandin E synthase-1 in cancer-associated fibroblasts will be an effective future therapeutic strategy in fighting neuroblastoma.
Collapse
Affiliation(s)
- Karin Larsson
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden.
| | - Anna Kock
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Nguyen F, Alferiev I, Guan P, Guerrero DT, Kolla V, Moorthy GS, Chorny M, Brodeur GM. Enhanced Intratumoral Delivery of SN38 as a Tocopherol Oxyacetate Prodrug Using Nanoparticles in a Neuroblastoma Xenograft Model. Clin Cancer Res 2018. [PMID: 29514842 DOI: 10.1158/1078-0432.ccr-17-3811] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Purpose: Currently, <50% of high-risk pediatric solid tumors like neuroblastoma can be cured, and many survivors experience serious or life-threatening toxicities, so more effective, less toxic therapy is needed. One approach is to target drugs to tumors using nanoparticles, which take advantage of the enhanced permeability of tumor vasculature.Experimental Design: SN38, the active metabolite of irinotecan (CPT-11), is a potent therapeutic agent that is readily encapsulated in polymeric nanoparticles. Tocopherol oxyacetate (TOA) is a hydrophobic mitocan that was linked to SN38 to significantly increase hydrophobicity and enhance nanoparticle retention. We treated neuroblastomas with SN38-TOA nanoparticles and compared the efficacy with the parent prodrug CPT-11 using a mouse xenograft model.Results: Nanoparticle treatment induced prolonged event-free survival (EFS) in most mice, compared with CPT-11. This was shown for both SH-SY5Y and IMR-32 neuroblastoma xenografts. Enhanced efficacy was likely due to increased and sustained drug levels of SN38 in the tumor compared with conventional CPT-11 delivery. Interestingly, when recurrent CPT-11-treated tumors were re-treated with SN38-TOA nanoparticles, the tumors transformed from undifferentiated neuroblastomas to maturing ganglioneuroblastomas. Furthermore, these tumors were infiltrated with Schwann cells of mouse origin, which may have contributed to the differentiated histology.Conclusions: Nanoparticle delivery of SN38-TOA produced increased drug delivery and prolonged EFS compared to conventional delivery of CPT-11. Also, lower total dose and drug entrapment in nanoparticles during circulation should decrease toxicity. We propose that nanoparticle-based delivery of a rationally designed prodrug is an attractive approach to enhance chemotherapeutic efficacy in pediatric and adult tumors. Clin Cancer Res; 24(11); 2585-93. ©2018 AACR.
Collapse
Affiliation(s)
- Ferro Nguyen
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ivan Alferiev
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Peng Guan
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - David T Guerrero
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Venkatadri Kolla
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ganesh S Moorthy
- Department of Anesthesiology and Critical Care, University of Pennsylvania/Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Michael Chorny
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Garrett M Brodeur
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.
| |
Collapse
|
12
|
Adams M, Traunecker H, Doull I, Cox R. Bronchiectasis following treatment for high-risk neuroblastoma: A case series. Pediatr Blood Cancer 2017; 64. [PMID: 28296062 DOI: 10.1002/pbc.26509] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 02/07/2017] [Accepted: 02/11/2017] [Indexed: 11/07/2022]
Abstract
High-risk (HR) neuroblastoma remains a very challenging disease to treat and long-term cure is only possible with intensive, multimodal treatment including chemotherapy, high-dose therapy, radiotherapy, surgery, and immunotherapy. As a result, treatment-related morbidity and late effects are common in survivors. This report outlines a case series of six patients who developed a chronic productive cough following treatment for HR neuroblastoma. High-resolution computed tomography scanning confirmed the diagnosis of bronchiectasis. Two of the patients who have undergone immunological testing demonstrate hypogammaglobulinaemia and impaired vaccine response. Persistent cough in patients treated for neuroblastoma warrants investigation and consideration of immunological referral.
Collapse
Affiliation(s)
- Madeleine Adams
- Department of Paediatric Oncology, Children's Hospital for Wales, Heath Park, Cardiff, UK
| | - Heidi Traunecker
- Department of Paediatric Oncology, Children's Hospital for Wales, Cardiff, UK
| | - Iolo Doull
- Department of Paediatric Respiratory Medicine, Children's Hospital for Wales, Cardiff, UK
| | - Rachel Cox
- Department of Paediatric Oncology, Bristol Children's Hospital, Bristol, UK
| |
Collapse
|
13
|
Moreno L, Caron H, Geoerger B, Eggert A, Schleiermacher G, Brock P, Valteau-Couanet D, Chesler L, Schulte JH, De Preter K, Molenaar J, Schramm A, Eilers M, Van Maerken T, Johnsen JI, Garrett M, George SL, Tweddle DA, Kogner P, Berthold F, Koster J, Barone G, Tucker ER, Marshall L, Herold R, Sterba J, Norga K, Vassal G, Pearson AD. Accelerating drug development for neuroblastoma - New Drug Development Strategy: an Innovative Therapies for Children with Cancer, European Network for Cancer Research in Children and Adolescents and International Society of Paediatric Oncology Europe Neuroblastoma project. Expert Opin Drug Discov 2017; 12:801-811. [PMID: 28604107 DOI: 10.1080/17460441.2017.1340269] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Neuroblastoma, the commonest paediatric extra-cranial tumour, remains a leading cause of death from cancer in children. There is an urgent need to develop new drugs to improve cure rates and reduce long-term toxicity and to incorporate molecularly targeted therapies into treatment. Many potential drugs are becoming available, but have to be prioritised for clinical trials due to the relatively small numbers of patients. Areas covered: The current drug development model has been slow, associated with significant attrition, and few new drugs have been developed for neuroblastoma. The Neuroblastoma New Drug Development Strategy (NDDS) has: 1) established a group with expertise in drug development; 2) prioritised targets and drugs according to tumour biology (target expression, dependency, pre-clinical data; potential combinations; biomarkers), identifying as priority targets ALK, MEK, CDK4/6, MDM2, MYCN (druggable by BET bromodomain, aurora kinase, mTORC1/2) BIRC5 and checkpoint kinase 1; 3) promoted clinical trials with target-prioritised drugs. Drugs showing activity can be rapidly transitioned via parallel randomised trials into front-line studies. Expert opinion: The Neuroblastoma NDDS is based on the premise that optimal drug development is reliant on knowledge of tumour biology and prioritisation. This approach will accelerate neuroblastoma drug development and other poor prognosis childhood malignancies.
Collapse
Affiliation(s)
- Lucas Moreno
- a Paediatric Phase I-II Clinical Trials Unit, Paediatric Haematology & Oncology , Hospital Niño Jesus , Madrid , Spain
- b Instituto de Investigación Sanitaria La Princesa , Madrid , Spain
- c Paediatric Drug Development, Children and Young People's Unit , Royal Marsden Hospital , London , UK
| | - Hubert Caron
- d Emma Children's Hospital , Amsterdam , Netherlands
- e Hoffman-La Roche , Basel , Switzerland
| | - Birgit Geoerger
- f Department of Paediatric and Adolescent Oncology , Institut Gustave Roussy , Villejuif , France
| | - Angelika Eggert
- g Department of Pediatric Oncology and Hematology , Charite University Hospital , Berlin , Germany
| | - Gudrun Schleiermacher
- h Department of Paediatric, Adolescents and Young Adults Oncology and INSERM U830 , Institut Curie , Paris , France
| | - Penelope Brock
- i Department Paediatric Oncology , Great Ormond Street Hospital , London , UK
| | | | - Louis Chesler
- c Paediatric Drug Development, Children and Young People's Unit , Royal Marsden Hospital , London , UK
- j Division of Clinical Studies , Institute of Cancer Research , London , UK
| | - Johannes H Schulte
- g Department of Pediatric Oncology and Hematology , Charite University Hospital , Berlin , Germany
| | | | - Jan Molenaar
- l Princess Maxima Center for Pediatric Oncology , University of Amsterdam , Amsterdam , Netherlands
| | - Alexander Schramm
- m Department of Pediatric Oncology , University of Essen , Essen , Germany
| | - Martin Eilers
- n Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter , University of Wurzburg , Wurzburg , Germany
| | - Tom Van Maerken
- k Centre for Medical Genetic , Ghent University , Ghent , Belgium
| | - John Inge Johnsen
- o Department of Women's and Children's Health , Karolinska Institute , Stockholm , Sweden
| | | | - Sally L George
- c Paediatric Drug Development, Children and Young People's Unit , Royal Marsden Hospital , London , UK
- j Division of Clinical Studies , Institute of Cancer Research , London , UK
| | - Deborah A Tweddle
- q Wolfson Childhood Cancer Research Centre , Newcastle University , Newcastle , UK
| | - Per Kogner
- o Department of Women's and Children's Health , Karolinska Institute , Stockholm , Sweden
| | - Frank Berthold
- r Department of Pediatric Oncology and Hematology , University of Cologne , Cologne , Germany
| | - Jan Koster
- l Princess Maxima Center for Pediatric Oncology , University of Amsterdam , Amsterdam , Netherlands
| | - Giuseppe Barone
- c Paediatric Drug Development, Children and Young People's Unit , Royal Marsden Hospital , London , UK
- j Division of Clinical Studies , Institute of Cancer Research , London , UK
| | - Elizabeth R Tucker
- c Paediatric Drug Development, Children and Young People's Unit , Royal Marsden Hospital , London , UK
- j Division of Clinical Studies , Institute of Cancer Research , London , UK
| | - Lynley Marshall
- c Paediatric Drug Development, Children and Young People's Unit , Royal Marsden Hospital , London , UK
- j Division of Clinical Studies , Institute of Cancer Research , London , UK
| | | | - Jaroslav Sterba
- t Masaryk University, University Hospital , Brno , Czech Republic
- u Department of Pediatric Oncology , International Clinical Research Center, St. Anne's University Hospital , Brno , Czech Republic
- v RECAMO, Masaryk Memorial Cancer Centre , Brno , Czech Republic
| | - Koen Norga
- w Pediatric Hematology/Oncology Unit , Antwerp University Hospital , Antwerp , Belgium
| | - Gilles Vassal
- x Department of Clinical Research, Gustave Roussy , Paris-Sud University , Paris , France
| | - Andrew Dj Pearson
- c Paediatric Drug Development, Children and Young People's Unit , Royal Marsden Hospital , London , UK
- j Division of Clinical Studies , Institute of Cancer Research , London , UK
| |
Collapse
|
14
|
Chemaitilly W, Cohen LE. DIAGNOSIS OF ENDOCRINE DISEASE: Endocrine late-effects of childhood cancer and its treatments. Eur J Endocrinol 2017; 176:R183-R203. [PMID: 28153840 DOI: 10.1530/eje-17-0054] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 02/01/2017] [Indexed: 12/12/2022]
Abstract
Endocrine complications are frequently observed in childhood cancer survivors (CCS). One of two CCS will experience at least one endocrine complication during the course of his/her lifespan, most commonly as a late-effect of cancer treatments, especially radiotherapy and alkylating agent chemotherapy. Endocrine late-effects include impairments of the hypothalamus/pituitary, thyroid and gonads, as well as decreased bone mineral density and metabolic derangements leading to obesity and/or diabetes mellitus. A systematic approach where CCS are screened for endocrine late-effects based on their cancer history and treatment exposures may improve health outcomes by allowing the early diagnosis and treatment of these complications.
Collapse
Affiliation(s)
- Wassim Chemaitilly
- Departments of Pediatric Medicine-Division of Endocrinology
- Departments of Epidemiology and Cancer ControlSt Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Laurie E Cohen
- Departments of Epidemiology and Cancer ControlSt Jude Children's Research Hospital, Memphis, Tennessee, USA
- Division of EndocrinologyBoston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Whittle SB, Smith V, Doherty E, Zhao S, McCarty S, Zage PE. Overview and recent advances in the treatment of neuroblastoma. Expert Rev Anticancer Ther 2017; 17:369-386. [PMID: 28142287 DOI: 10.1080/14737140.2017.1285230] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Children with neuroblastoma have widely divergent outcomes, ranging from cure in >90% of patients with low risk disease to <50% for those with high risk disease. Recent research has shed light on the biology of neuroblastoma, allowing for more accurate risk stratification and treatment reduction in many cases, although newer treatment strategies for children with high-risk and relapsed neuroblastoma are needed to improve outcomes. Areas covered: Neuroblastoma epidemiology, diagnosis, risk stratification, and recent advances in treatment of both newly diagnosed and relapsed neuroblastoma. Expert commentary: The identification of newer tumor targets and of novel cell-mediated immunotherapy agents may lead to novel therapeutic approaches, and clinical trials for regimens designed to target individual genetic aberrations in tumors are underway. A combination of therapeutic modalities will likely be required to improve survival and cure rates for patients with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Sarah B Whittle
- a Department of Pediatrics, Section of Hematology-Oncology , Texas Children's Cancer and Hematology Centers, Baylor College of Medicine , Houston , TX , USA
| | - Valeria Smith
- a Department of Pediatrics, Section of Hematology-Oncology , Texas Children's Cancer and Hematology Centers, Baylor College of Medicine , Houston , TX , USA
| | - Erin Doherty
- a Department of Pediatrics, Section of Hematology-Oncology , Texas Children's Cancer and Hematology Centers, Baylor College of Medicine , Houston , TX , USA
| | - Sibo Zhao
- a Department of Pediatrics, Section of Hematology-Oncology , Texas Children's Cancer and Hematology Centers, Baylor College of Medicine , Houston , TX , USA
| | - Scott McCarty
- b Department of Pediatrics, Division of Hematology-Oncology , University of California San Diego, La Jolla, CA and Peckham Center for Cancer and Blood Disorders, Rady Children's Hospital , San Diego , CA , USA
| | - Peter E Zage
- b Department of Pediatrics, Division of Hematology-Oncology , University of California San Diego, La Jolla, CA and Peckham Center for Cancer and Blood Disorders, Rady Children's Hospital , San Diego , CA , USA
| |
Collapse
|
16
|
Speleman F, Park JR, Henderson TO. Neuroblastoma: A Tough Nut to Crack. Am Soc Clin Oncol Educ Book 2017; 35:e548-57. [PMID: 27249766 DOI: 10.1200/edbk_159169] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neuroblastoma, an embryonal tumor arising from neural crest-derived progenitor cells, is the most common solid tumor in childhood, with more than 700 cases diagnosed per year in the United States. In the past several decades, significant advances have been made in the treatment of neuroblastoma. Treatment advances reflect improved understanding of the biology of neuroblastoma. Although amplification of MYCN was discovered in the early 1980s, our understanding of neuroblastoma oncogenesis has advanced in the last decade as a result of high-throughput genomic analysis, exome and whole-genome sequencing, genome-wide association studies, and synthetic lethal drug screens. Our refined understanding of neuroblastoma biology and genetics is reflected in improved prognostic stratification and appropriate tailoring of therapy in recent clinical trials. Moreover, for high-risk neuroblastoma, a disease that was uniformly fatal 3 decades ago, recent clinical trials incorporating autologous hematopoietic transplant and immunotherapy utilizing anti-GD2 antibody plus cytokines have shown improved event-free and overall survival. These advances have resulted in a growing population of long-term survivors of neuroblastoma. Examination of the late effects and second malignant neoplasms (SMNs) in both older generations of survivors and more recently treated survivors will inform both design of future trials and surveillance guidelines for long-term follow-up. As a consequence of advances in understanding of the biology of neuroblastoma, successful clinical trials, and refined understanding of the late effects and SMNs of survivors, the promise of precision medicine is becoming a reality for patients with neuroblastoma.
Collapse
Affiliation(s)
- Frank Speleman
- From the Center for Medical Genetics Ghent, Cancer Research Institute Ghent, Ghent, Belgium; Seattle Children's Hospital, Seattle, WA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA; University of Chicago Comer Children's Hospital, Chicago, IL
| | - Julie R Park
- From the Center for Medical Genetics Ghent, Cancer Research Institute Ghent, Ghent, Belgium; Seattle Children's Hospital, Seattle, WA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA; University of Chicago Comer Children's Hospital, Chicago, IL
| | - Tara O Henderson
- From the Center for Medical Genetics Ghent, Cancer Research Institute Ghent, Ghent, Belgium; Seattle Children's Hospital, Seattle, WA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA; University of Chicago Comer Children's Hospital, Chicago, IL
| |
Collapse
|
17
|
Luksch R, Castellani MR, Collini P, De Bernardi B, Conte M, Gambini C, Gandola L, Garaventa A, Biasoni D, Podda M, Sementa AR, Gatta G, Tonini GP. Neuroblastoma (Peripheral neuroblastic tumours). Crit Rev Oncol Hematol 2016; 107:163-181. [PMID: 27823645 DOI: 10.1016/j.critrevonc.2016.10.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 09/05/2016] [Accepted: 10/03/2016] [Indexed: 02/07/2023] Open
Abstract
Peripheral neuroblastic tumours (PNTs), a family of tumours arising in the embryonal remnants of the sympathetic nervous system, account for 7-10% of all tumours in children. In two-thirds of cases, PNTs originate in the adrenal glands or the retroperitoneal ganglia. At least one third present metastases at onset, with bone and bone marrow being the most frequent metastatic sites. Disease extension, MYCN oncogene status and age are the most relevant prognostic factors, and their influence on outcome have been considered in the design of the recent treatment protocols. Consequently, the probability of cure has increased significantly in the last two decades. In children with localised operable disease, surgical resection alone is usually a sufficient treatment, with 3-year event-free survival (EFS) being greater than 85%. For locally advanced disease, primary chemotherapy followed by surgery and/or radiotherapy yields an EFS of around 75%. The greatest problem is posed by children with metastatic disease or amplified MYCN gene, who continue to do badly despite intensive treatments. Ongoing trials are exploring the efficacy of new drugs and novel immunological approaches in order to save a greater number of these patients.
Collapse
Affiliation(s)
- Roberto Luksch
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | | | - Paola Collini
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Massimo Conte
- Giannina Gaslini Children's Research Hospital, Genoa, Italy
| | | | - Lorenza Gandola
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Davide Biasoni
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marta Podda
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Gemma Gatta
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Gian Paolo Tonini
- Neuroblastoma Laboratory, Paediatric Research Institute, Padua, Italy
| |
Collapse
|
18
|
Shiokawa N, Okamoto Y, Kodama Y, Nishikawa T, Tanabe T, Mukai M, Yoshioka T, Kawano Y. Conservative treatment of massive hemothorax in a girl with neuroblastoma. Pediatr Int 2016; 58:1090-1092. [PMID: 27804245 DOI: 10.1111/ped.13094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/10/2016] [Accepted: 07/13/2016] [Indexed: 11/26/2022]
Abstract
We report the case of a 1-year-old girl with stage 4 neuroblastoma who developed massive hemothorax due to tumor invasion before treatment. She presented with tachypnea, worsening anemia, and oxygen desaturation. Hemothorax was diagnosed based on chest radiography, ultrasonography, and diagnostic thoracic puncture results. High neuron-specific enolase, vanillylmandelic acid, and homovanillic acid as well as computed tomography strongly supported a diagnosis of neuroblastoma. Chemotherapy along with intermittent puncture drainage, oxygen, and blood transfusion reduced the accumulated blood, and hemothorax disappeared within 1 week. Thus, it is possible to avoid invasive treatment for massive hemothorax by initiating chemotherapy for chemosensitive solid tumors, including neuroblastoma.
Collapse
Affiliation(s)
- Naohiro Shiokawa
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima City, Kagoshima, Japan
| | - Yasuhiro Okamoto
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima City, Kagoshima, Japan.
| | - Yuichi Kodama
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima City, Kagoshima, Japan
| | - Takuro Nishikawa
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima City, Kagoshima, Japan
| | - Takayuki Tanabe
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima City, Kagoshima, Japan
| | - Motoi Mukai
- Department of Pediatric Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima City, Kagoshima, Japan
| | - Takako Yoshioka
- Department of Pathology, Kagoshima University Medical and Dental Hospital, Kagoshima City, Kagoshima, Japan.,Department of Pathology, National Center of Child Health and Development, Setagaya, Tokyo, Japan
| | - Yoshifumi Kawano
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima City, Kagoshima, Japan
| |
Collapse
|
19
|
Huibregtse KE, Vo KT, DuBois SG, Fetzko S, Neuhaus J, Batra V, Maris JM, Weiss B, Marachelian A, Yanik GA, Matthay KK. Incidence and risk factors for secondary malignancy in patients with neuroblastoma after treatment with (131)I-metaiodobenzylguanidine. Eur J Cancer 2016; 66:144-52. [PMID: 27573428 DOI: 10.1016/j.ejca.2016.07.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/19/2016] [Accepted: 07/15/2016] [Indexed: 01/22/2023]
Abstract
Several reports of second malignant neoplasm (SMN) in patients with relapsed neuroblastoma after treatment with (131)I-MIBG suggest the possibility of increased risk. Incidence of and risk factors for SMN after (131)I-MIBG have not been defined. This is a multi-institutional retrospective review of patients with neuroblastoma treated with (131)I-MIBG therapy. A competing risk approach was used to calculate the cumulative incidence of SMN from time of first exposure to (131)I-MIBG. A competing risk regression was used to identify potential risk factors for SMN. The analytical cohort included 644 patients treated with (131)I-MIBG. The cumulative incidence of SMN was 7.6% (95% confidence interval [CI], 4.4-13.0%) and 14.3% (95% CI, 8.3-23.9%) at 5 and 10 years from first (131)I-MIBG, respectively. No increase in SMN risk was found with increased number of (131)I-MIBG treatments or higher cumulative activity per kilogram of (131)I-MIBG received (p = 0.72 and p = 0.84, respectively). Thirteen of the 19 reported SMN were haematologic. In a multivariate analysis controlling for variables with p < 0.1 (stage, age at first (131)I-MIBG, bone disease, disease status at time of first (131)I-MIBG), patients with relapsed/progressive disease had significantly lower risk of SMN (subdistribution hazard ratio 0.3, 95% CI, 0.1-0.8, p = 0.023) compared to patients with persistent/refractory neuroblastoma. The cumulative risk of SMN after (131)I-MIBG therapy for patients with relapsed or refractory neuroblastoma is similar to the greatest published incidence for high-risk neuroblastoma after myeloablative therapy, with no dose-dependent increase. As the number of patients treated and length of follow-up time increase, it will be important to reassess this risk.
Collapse
Affiliation(s)
- Kelly E Huibregtse
- University of California San Francisco Benioff Children's Hospital, USA.
| | - Kieuhoa T Vo
- Department of Pediatrics, University of California San Francisco Benioff Children's Hospital, USA.
| | - Steven G DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, USA.
| | - Stephanie Fetzko
- Department of Pediatrics, Baylor University Medical Center, USA.
| | - John Neuhaus
- University of California San Francisco Benioff Children's Hospital, Department of Biostatistics, USA.
| | - Vandana Batra
- Children's Hospital of Philadelphia, Department of Pediatric Oncology, USA.
| | - John M Maris
- Children's Hospital of Philadelphia, Department of Pediatric Oncology, USA.
| | - Brian Weiss
- Cincinnati Children's Hospital Medical Center, Division of Pediatric Oncology, USA.
| | - Araz Marachelian
- Children's Hospital of Los Angeles, New Approaches to Neuroblastoma Research, USA.
| | - Greg A Yanik
- Department of Pediatrics, University of Michigan Medical Center, USA.
| | - Katherine K Matthay
- Department of Pediatrics, University of California San Francisco Benioff Children's Hospital, USA.
| |
Collapse
|
20
|
Ploessl C, Pan A, Maples KT, Lowe DK. Dinutuximab: An Anti-GD2 Monoclonal Antibody for High-Risk Neuroblastoma. Ann Pharmacother 2016; 50:416-22. [PMID: 26917818 DOI: 10.1177/1060028016632013] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE To review the pharmacology, pharmacokinetics, efficacy, safety, dosage and administration, and formulary considerations for dinutuximab. DATA SOURCES MEDLINE was searched (1964 to January 2016) using the terms ch14.18, dinutuximab, immunotherapy, and neuroblastoma. Other information was identified from package insert, Biologics License Application, abstracts, news releases, and ClinicalTrials.gov. STUDY SELECTION AND DATA EXTRACTION Identified English-language articles were reviewed. Selected studies included phase I through III. DATA SYNTHESIS High-risk neuroblastoma is primarily a childhood cancer with 5-year survival rates of 40% to 50%. Treatment for high-risk neuroblastoma includes induction chemotherapy, surgery, myeloablative chemotherapy with autologous hematopoietic stem cell transplant, and radiation therapy. For patients achieving clinical remission, limited treatments exist for preventing relapse. Dinutuximab is a chimeric, human-murine, anti-GD2 monoclonal antibody approved in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF), aldesleukin (interleukin-2 [IL-2]), and isotretinoin (13-cis-retinoic acid [RA]) for maintenance treatment of pediatric patients with high-risk neuroblastoma who achieve at least a partial response to first-line multiagent, multimodality therapy. In phase III trials, dinutuximab increased 2-year event-free survival and overall survival when compared to standard treatment. Severe adverse effects of dinutuximab include pain, hypersensitivity reactions, capillary leak syndrome, and hypotension. CONCLUSIONS Dinutuximab is the first anti-GD2 monoclonal antibody approved in combination with GM-CSF, IL-2, and RA for maintenance treatment of pediatric patients with high-risk neuroblastoma who achieve at least a partial response to first-line multiagent, multimodality therapy. Ongoing research will determine if dinutuximab could be used earlier in treatment, in nonresponders to initial therapies, in combination with chemotherapy, or in other cancers.
Collapse
Affiliation(s)
- Cady Ploessl
- Virginia Commonwealth University Health System/Medical College of Virginia Hospitals, Richmond, VA, USA
| | - Alice Pan
- Virginia Commonwealth University Health System/Medical College of Virginia Hospitals, Richmond, VA, USA
| | - Kathryn T Maples
- Virginia Commonwealth University Health System/Medical College of Virginia Hospitals, Richmond, VA, USA
| | - Denise K Lowe
- Virginia Commonwealth University Health System/Medical College of Virginia Hospitals, Richmond, VA, USA
| |
Collapse
|
21
|
Pentek F, Schulte JH, Schweiger B, Metzelder M, Schündeln MM. Development of Port-Site Metastases Following Thoracoscopic Resection of a Neuroblastoma. Pediatr Blood Cancer 2016. [PMID: 26206749 DOI: 10.1002/pbc.25677] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We report a 26-month-old female who developed port-site metastases of a neuroblastoma following minimally invasive thoracoscopic interventions. After diagnosis of an intrathoracic low-risk neuroblastoma and 6 months of observation, she developed respiratory problems. She subsequently underwent total resection of a locally progressive tumor via thoracoscopy. Six months later, she developed local relapse and subcutaneous metastases within the thoracic wall. These port-site metastases were most likely iatrogenic. After excision of metastases, the residual tumor responded well to salvage chemotherapy. The patient has remained in remission for over 4 years.
Collapse
Affiliation(s)
- Falk Pentek
- Department of Pediatric Hematology and Oncology, Kinderklinik III, Universitätsklinikum-Essen and the University of Duisburg-Essen, Essen, Germany
| | - Johannes H Schulte
- Department of Pediatric Hematology and Oncology, Kinderklinik III, Universitätsklinikum-Essen and the University of Duisburg-Essen, Essen, Germany.,Charité Universitätsmedizin, Department of Pediatrics, Division of Oncology and Hematology, Berlin, Germany.,German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,Translational Neuro-Oncology, West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Bernd Schweiger
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, Universitätsklinikum-Essen and the University of Duisburg-Essen, Essen, Germany
| | - Martin Metzelder
- Division of Pediatric Surgery, Universitätsklinikum-Essen and the University of Duisburg-Essen, Essen, Germany.,Department of Pediatric Surgery, Medical University Vienna, Vienna, Austria
| | - Michael M Schündeln
- Department of Pediatric Hematology and Oncology, Kinderklinik III, Universitätsklinikum-Essen and the University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
22
|
Cochlear Implantation for Profound Hearing Loss After Multimodal Treatment for Neuroblastoma in Children. Clin Exp Otorhinolaryngol 2015; 8:329-34. [PMID: 26622949 PMCID: PMC4661246 DOI: 10.3342/ceo.2015.8.4.329] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/12/2014] [Accepted: 11/17/2014] [Indexed: 11/13/2022] Open
Abstract
Objectives Neuroblastoma (NBL) predominantly affects children under 5 years of age. Through multimodal therapy, including chemotherapy, radiotherapy, surgery, and peripheral blood stem cell transplantation, the survival rate in patients with NBL have improved while treatment-related complications have also increased. Treatment-related ototoxicity, mainly from cisplatin, can result in profound hearing loss requiring cochlear implantation (CI). We analyzed the effectiveness and hearing preservation of CI recipients who had treated with multimodal therapy due to NBL. Methods Patients who received multimodal therapy for NBL and subsequent CIs were enrolled. A detailed review of the perioperative hearing test, speech evaluation, and posttreatment complications was conducted. Speech performance was analyzed using the category of auditory performance (CAP) score and the postoperative hearing preservation of low frequencies was also compared. Patients who were candidates for electro-acoustic stimulation (EAS) used an EAS electrode for low frequency hearing preservation. Results Three patients were identified and all patients showed improvement of speech performance after CI. The average of CAP score improved from 4.3 preoperatively to 5.8 at 1 year postoperatively. Two patients who were fitted with the Flex electrode showed complete hearing preservation and the preserved hearing was maintained over 1 year. The one remaining patient was given the standard CI-512 electrode and showed partial hearing preservation. Conclusion Patients with profound hearing loss resulting from NBL multimodal therapy can be good candidates for CI, especially for EAS. A soft surgical technique as well as a specifically designed electrode should be applied to this specific population during the CI operation in order to preserve residual hearing and achieve better outcomes.
Collapse
|
23
|
Liu QL, Liang QL, Ou WT, Li ZY, Zhang XN. Surgical resection and post-operative radiotherapy in an adult renal neuroblastoma patient with multiple bone and joint metastases: A case report. Oncol Lett 2015; 9:2591-2594. [PMID: 26137112 DOI: 10.3892/ol.2015.3109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 01/09/2015] [Indexed: 12/28/2022] Open
Abstract
Renal neuroblastoma is uncommon, particularly in adults, with only a few cases having been reported in studies published in the English language. The incidence is only 0.12 cases/1 million individuals in those aged >20 years. Studies of the pathogenesis, biological characteristics, treatment and prognosis of renal neuroblastoma are limited due to this low incidence. The present study reports the case of a 22-year-old adult female who was diagnosed with a left renal neuroblastoma by computed tomography (CT), bone scan and pathological examination. The patient underwent a left nephroureterectomy, ipsilateral lymph node dissection and post-operative radiotherapy, prior to discharge 60 days after admittance. At the nine-month follow-up examination, the patient showed no evidence of recurrence, progression or metastatic disease on the CT scans of the chest, abdomen and pelvis. Renal neuroblastoma is extremely uncommon in adults. The diagnosis and treatment of renal neuroblastoma is complicated by the overall low incidence, lack of specific treatment guidelines, advanced disease state due to late presentation, and its associated co-morbidities. Further study of the pathogenesis, biological and clinical characteristics, and treatment of renal neuroblastoma is required to provide an optimal treatment for patients and to improve the patient's quality of life.
Collapse
Affiliation(s)
- Qiu-Long Liu
- Department of Oncology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Qi-Lian Liang
- Department of Oncology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Wen-Ting Ou
- Department of Oncology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Zhou-Yu Li
- Department of Radiotherapy, Cancer Center of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Xiang-Ning Zhang
- Department of Pathophysiology, School of Basic Medical Science, Guangdong Medical College, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
24
|
Bassiri H, Benavides A, Haber M, Gilmour SK, Norris MD, Hogarty MD. Translational development of difluoromethylornithine (DFMO) for the treatment of neuroblastoma. Transl Pediatr 2015; 4:226-38. [PMID: 26835380 PMCID: PMC4729051 DOI: 10.3978/j.issn.2224-4336.2015.04.06] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/08/2015] [Indexed: 01/01/2023] Open
Abstract
Neuroblastoma is a childhood tumor in which MYC oncogenes are commonly activated to drive tumor progression. Survival for children with high-risk neuroblastoma remains poor despite treatment that incorporates high-dose chemotherapy, stem cell support, surgery, radiation therapy and immunotherapy. More effective and less toxic treatments are sought and one approach under clinical development involves re-purposing the anti-protozoan drug difluoromethylornithine (DFMO; Eflornithine) as a neuroblastoma therapeutic. DFMO is an irreversible inhibitor of ornithine decarboxylase (Odc), a MYC target gene, bona fide oncogene, and the rate-limiting enzyme in polyamine synthesis. DFMO is approved for the treatment of Trypanosoma brucei gambiense encephalitis ("African sleeping sickness") since polyamines are essential for the proliferation of these protozoa. However, polyamines are also critical for mammalian cell proliferation and the finding that MYC coordinately regulates all aspects of polyamine metabolism suggests polyamines may be required to support cancer promotion by MYC. Pre-emptive blockade of polyamine synthesis is sufficient to block tumor initiation in an otherwise fully penetrant transgenic mouse model of neuroblastoma driven by MYCN, underscoring the necessity of polyamines in this process. Moreover, polyamine depletion regimens exert potent anti-tumor activity in pre-clinical models of established neuroblastoma as well, in combination with numerous chemotherapeutic agents and even in tumors with unfavorable genetic features such as MYCN, ALK or TP53 mutation. This has led to the testing of DFMO in clinical trials for children with neuroblastoma. Current trial designs include testing lower dose DFMO alone (2,000 mg/m(2)/day) starting at the completion of standard therapy, or higher doses combined with chemotherapy (up to 9,000 mg/m(2)/day) for patients with relapsed disease that has progressed. In this review we will discuss important considerations for the future design of DFMO-based clinical trials for neuroblastoma, focusing on the need to better define the principal mechanisms of anti-tumor activity for polyamine depletion regimens. Putative DFMO activities that are both cancer cell intrinsic (targeting the principal oncogenic driver, MYC) and cancer cell extrinsic (altering the tumor microenvironment to support anti-tumor immunity) will be discussed. Understanding the mechanisms of DFMO activity are critical in determining how it might be best leveraged in upcoming clinical trials. This mechanistic approach also provides a platform by which iterative pre-clinical testing using translational tumor models may complement our clinical approaches.
Collapse
|
25
|
Kushner BH, Roberts SS, Friedman DN, Kuk D, Ostrovnaya I, Modak S, Kramer K, Basu EM, Cheung NKV. Osteochondroma in long-term survivors of high-risk neuroblastoma. Cancer 2015; 121:2090-6. [PMID: 25728463 PMCID: PMC4970322 DOI: 10.1002/cncr.29316] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 01/15/2015] [Accepted: 01/20/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Osteochondromas are benign bony protrusions that can be spontaneous or associated with radiotherapy (RT). Current treatment of high-risk neuroblastoma includes dose-intensive chemotherapy, local RT, an anti-GD2 monoclonal antibody (MoAb), and isotretinoin. Late effects are emerging. METHODS The authors examined osteochondromas in 362 patients who were aged <10 years when diagnosed with neuroblastoma, had received a MoAb plus isotretinoin since 2000, and had survived >24 months from the time of the first dose of the MoAb. The incidence rate of osteochondroma was determined using the competing risks approach, in which the primary event was osteochondroma calculated from the date of neuroblastoma diagnosis and the competing event was death without osteochondroma. RESULTS A total of 21 osteochondroma cases were found among 14 patients who were aged 5.7 to 15.3 years (median, 10.4 years) and 3.1 to 11.2 years (median, 8.2 years) from the time of neuroblastoma diagnosis. The cumulative incidence rate was 0.6% at 5 years and 4.9% at 10 years from the neuroblastoma diagnosis. Nine osteochondromas were revealed incidentally during assessments of neuroblastoma disease status or bone age. Thirteen osteochondromas were detected outside RT portals and had characteristics of spontaneous forms. Complications were limited to pain necessitating surgical resection in 3 patients, but follow-up was short at 0.3 to 7.7 years (median, 3.5 years). CONCLUSIONS Osteochondromas in long-term survivors of neuroblastoma should be expected because these benign growths can be related to RT and these patients undergo radiologic studies over years, are monitored for late toxicities through and beyond adolescence, and receive special attention (because of concerns about disease recurrence) if they develop a bony protuberance. A pathogenic role for chemotherapy, anti-GD2 MoAbs, or isotretinoin remains speculative.
Collapse
Affiliation(s)
| | | | | | - Deborah Kuk
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Shakeel Modak
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kim Kramer
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ellen M. Basu
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | |
Collapse
|
26
|
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
|
27
|
Long-term side effects of radiotherapy for pediatric localized neuroblastoma : results from clinical trials NB90 and NB94. Strahlenther Onkol 2015; 191:604-12. [PMID: 25896312 DOI: 10.1007/s00066-015-0837-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/25/2015] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Neuroblastoma (NB) is the most frequent indication for extracranial pediatric radiotherapy. As long-term survival of high-risk localized NB has greatly improved, we reviewed treatment-related late toxicities in pediatric patients who received postoperative radiotherapy (RT) for localized NB within two French prospective clinical trials: NB90 and NB94. PATIENTS AND METHODS From 1990-2000, 610 children were enrolled. Among these, 35 were treated with induction chemotherapy, surgery, and RT. The recommended RT dose was 24 Gy at ≤ 2 years, 34 Gy at > 2 years, ± a 5 Gy boost in both age groups. RESULTS The 22 patients still alive after 5 years were analyzed. The median follow-up time was 14 years (range 5-21 years). Late effects after therapy occurred in 73 % of patients (16/22), within the RT field for 50 % (11/22). The most frequent in-field effects were musculoskeletal abnormalities (n = 7) that occurred only with doses > 31 Gy/1.5 Gy fraction (p = 0.037). Other effects were endocrine in 3 patients and second malignancies in 2 patients. Four patients presented with multiple in-field late effects only with doses > 31 Gy. CONCLUSION After a median follow-up of 14 years, late effects with multimodality treatment were frequent. The most frequent effects were musculoskeletal abnormalities and the threshold for their occurrence was 31 Gy.
Collapse
|
28
|
Abstract
Neuroblastoma (NB) is the third most common pediatric cancer. Although NB accounts for 7% of pediatric malignancies, it is responsible for more than 10% of childhood cancer-related mortality. Prognosis and treatment are determined by clinical and biological risk factors. Estimated 5-year survival rates for patients with non-high-risk and high-risk NB are more than 90% and less than 50%, respectively. Recent clinical trials have continued to reduce therapy for patients with non-high-risk NB, including the most favorable subsets who are often followed with observation approaches. In contrast, high-risk patients are treated aggressively with chemotherapy, radiation, surgery, and myeloablative and immunotherapies.
Collapse
|
29
|
|
30
|
Kyle UG, Shekerdemian LS, Coss-Bu JA. Growth failure and nutrition considerations in chronic childhood wasting diseases. Nutr Clin Pract 2014; 30:227-38. [PMID: 25378356 DOI: 10.1177/0884533614555234] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Growth failure is a common problem in many children with chronic diseases. This article is an overview of the most common causes of growth failure/growth retardation that affect children with a number of chronic diseases. We also briefly review the nutrition considerations and treatment goals. Growth failure is multifactorial in children with chronic conditions, including patients with cystic fibrosis, chronic kidney disease, chronic liver disease, congenital heart disease, human immunodeficiency virus, inflammatory bowel disease, short bowel syndrome, and muscular dystrophies. Important contributory factors to growth failure include increased energy needs, increased energy loss, malabsorption, decreased energy intake, anorexia, pain, vomiting, intestinal obstruction, and inflammatory cytokines. Various metabolic and pathologic abnormalities that are characteristic of chronic diseases further lead to significant malnutrition and growth failure. In addition to treating disease-specific abnormalities, treatment should address the energy and protein deficits, including vitamin and mineral supplements to correct deficiencies, correct metabolic and endocrinologic abnormalities, and include long-term monitoring of weight and growth. Individualized, age-appropriate nutrition intervention will minimize the malnutrition and growth failure seen in children with chronic diseases.
Collapse
Affiliation(s)
- Ursula G Kyle
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas
| | - Lara S Shekerdemian
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas
| | - Jorge A Coss-Bu
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas
| |
Collapse
|
31
|
Moreno L, Marshall LV, Pearson ADJ. At the frontier of progress for paediatric oncology: the neuroblastoma paradigm. Br Med Bull 2013; 108:173-88. [PMID: 24211816 DOI: 10.1093/bmb/ldt033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Neuroblastoma is one of the commonest and deadliest forms of childhood cancer and major initiatives are ongoing to improve the outcome of these patients. SOURCES OF DATA Data for this review were obtained from PubMed and abstracts from the American Society of Clinical Oncology and Advances in Neuroblastoma Research. AREAS OF AGREEMENT Collaborative clinical trials have led to major improvements in treatment outcomes for low and intermediate risk neuroblastoma, and international initiatives such as the International Neuroblastoma Risk Group have produced a very refined risk stratification incorporating clinical and biological risk factors. AREAS OF CONTROVERSY Despite many efforts, the outcome for high-risk neuroblastoma is still poor and the only new strategy incorporated into frontline treatment is anti-GD2 immunotherapy. It is unclear how new drugs targeting specific molecular aberrations will be incorporated. GROWING POINTS Genomic characterization and drug development have undergone major advances in the last 5 years leading to a much deeper understanding of tumour biology as well as active biomarker-driven preclinical and clinical research on new molecules that will hopefully progress faster and more efficiently into frontline combination treatment strategies. AREAS TIMELY FOR DEVELOPING RESEARCH Significant effort remains to be done in integrating the different new strategies, combining new molecularly targeted agents to maximize therapeutic benefit and incorporate immunotherapy together with targeted therapies.
Collapse
Affiliation(s)
- Lucas Moreno
- Paediatric Drug Development Team, Di visions of Cancer Therapeutics and Clinical Studies, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | | | | |
Collapse
|