1
|
Seo ES, Lee JW, Cho HW, Ju HY, Cho YS, Lee S, Moon SH, Yoo KH, Lim DH, Sung KW. Response-adapted consolidation therapy strategy for patients with metastatic high-risk neuroblastoma: Results of the SMC NB-2014 study. Pediatr Blood Cancer 2024; 71:e31173. [PMID: 38965702 DOI: 10.1002/pbc.31173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/28/2024] [Accepted: 06/16/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Tandem high-dose chemotherapy and autologous stem cell transplantation (HDCT/auto-SCT) and incorporation of 131I-metaiodobenzylguanidine (131I-MIBG) treatment have shown positive outcomes in high-risk neuroblastoma. However, more optimized treatment strategies are still needed. PROCEDURE The NB-2014 study was a nonrandomized, prospective trial that examined survival outcomes in metastatic high-risk neuroblastoma patients using response-adapted consolidation therapy. We used post-induction residual 123I-MIBG status at metastatic sites as a treatment response marker. Patients achieving complete resolution of MIBG uptake at metastatic sites underwent a reduced first HDCT/auto-SCT with a 20% dose reduction in HDCT. After the first HDCT/auto-SCT, patients with remaining MIBG uptake received dose-escalated (18 mCi/kg) 131I-MIBG treatment. In contrast, those with complete resolution of MIBG at metastatic sites received a standard dose (12 mCi/kg) of 131I-MIBG. We compared survival and toxicity outcomes with a historical control group from the NB-2009. RESULTS Of 65 patients treated, 63% achieved complete resolution of MIBG uptake at metastatic sites following induction chemotherapy, while 29% of patients still had MIBG uptake at metastatic sites after the first HDCT/auto-SCT. The 3-year event-free survival (EFS) and overall survival (OS) rates were 68.2% ± 6.0% and 86.5% ± 4.5%, respectively. Compared to NB-2009, EFS was similar (p = .855); however, NB-2014 had a higher OS (p = .031), a lower cumulative incidence of treatment-related mortality (p = .036), and fewer acute and late toxicities. CONCLUSIONS Our results suggest that response-adaptive consolidation therapy based on chemotherapy response at metastatic sites facilitates better treatment tailoring, and appears promising for patients with metastatic high-risk neuroblastoma.
Collapse
Affiliation(s)
- Eun Seop Seo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hee Young Ju
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Seok Cho
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sanghoon Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Liu KX, Shaaban SG, Chen JJ, Bagatell R, Lerman BJ, Catalano PJ, DuBois SG, Shusterman S, Ioakeim-Ioannidou M, Yock TI, Shamberger RC, Mattei P, Vu L, Elhalawani H, Dusenbery KE, Vo KT, Huang MS, Friedmann AM, Diller LR, Marcus KJ, MacDonald SM, Terezakis SA, Braunstein SE, Hill-Kayser CE, Haas-Kogan DA. Patterns of recurrence after radiotherapy for high-risk neuroblastoma: Implications for radiation dose and field. Radiother Oncol 2024; 198:110384. [PMID: 38880415 DOI: 10.1016/j.radonc.2024.110384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/29/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Prognosis for patients with high-risk neuroblastoma (HR-NBL) is guarded despite aggressive therapy, and few studies have characterized outcomes after radiotherapy in relation to radiation treatment fields. METHODS Multi-institutional retrospective cohort of 293 patients with HR-NBL who received autologous stem cell transplant (ASCT) and EBRT between 1997-2021. LRR was defined as recurrence at the primary site or within one nodal echelon beyond disease present at diagnosis. Follow-up was defined from the end of EBRT. Event-free survival (EFS) and OS were analyzed by Kaplan-Meier method. Cumulative incidence of locoregional progression (CILP) was analyzed using competing risks of distant-only relapse and death with Gray's test. RESULTS Median follow-up was 7.0 years (range: 0.01-22.4). Five-year CILP, EFS, and OS were 11.9 %, 65.2 %, and 77.5 %, respectively. Of the 31 patients with LRR and imaging review, 15 (48.4 %) had in-field recurrences (>12 Gy), 6 (19.4 %) had marginal failures (≤12 Gy), and 10 (32.3 %) had both in-field and marginal recurrences. No patients receiving total body irradiation (12 Gy) experienced marginal-only failures (p = 0.069). On multivariable analyses, MYCN amplification had higher risk of LRR (HR: 2.42, 95 % CI: 1.06-5.50, p = 0.035) and post-consolidation isotretinoin and anti-GD2 antibody therapy (HR: 0.42, 95 % CI: 0.19-0.94, p = 0.035) had lower risk of LRR. CONCLUSIONS Despite EBRT, LRR remains a contributor to treatment failure in HR-NBL with approximately half of LRRs including a component of marginal failure. Future prospective studies are needed to explore whether radiation fields and doses should be defined based on molecular features such as MYCN amplification, and/or response to chemotherapy.
Collapse
Affiliation(s)
- Kevin X Liu
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sherif G Shaaban
- Department of Radiation Oncology & Molecular Radiation Sciences, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Jie Jane Chen
- Department of Radiation Oncology, University of California at San Francisco, UCSF Benioff Children's Hospital, San Francisco, CA, USA; Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rochelle Bagatell
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin J Lerman
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pediatrics, UCSF Benioff Children's Hospital and UCSF School of Medicine, San Francisco, CA, USA
| | - Paul J Catalano
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, and Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Steven G DuBois
- Department of Pediatrics, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Suzanne Shusterman
- Department of Pediatrics, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Myrsini Ioakeim-Ioannidou
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Torunn I Yock
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert C Shamberger
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter Mattei
- Department of Surgery, University of Pennsylvania and the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lan Vu
- Department of Surgery, UCSF Benioff Children's Hospital and UCSF School of Medicine, San Francisco, CA, USA
| | - Hesham Elhalawani
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn E Dusenbery
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, USA
| | - Kieuhoa T Vo
- Department of Pediatrics, UCSF Benioff Children's Hospital and UCSF School of Medicine, San Francisco, CA, USA
| | - Mary S Huang
- Department of Pediatric Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Alison M Friedmann
- Department of Pediatric Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Lisa R Diller
- Department of Pediatrics, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Karen J Marcus
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shannon M MacDonald
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Steve E Braunstein
- Department of Radiation Oncology, University of California at San Francisco, UCSF Benioff Children's Hospital, San Francisco, CA, USA
| | - Christine E Hill-Kayser
- Department of Radiation Oncology, University of Pennsylvania and the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Daphne A Haas-Kogan
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Kitko CL, Bollard CM, Cairo MS, Chewning J, Fry TJ, Pulsipher MA, Shenoy S, Wall DA, Levine JE. Children's Oncology Group's 2023 blueprint for research: Cellular therapy and stem cell transplantation. Pediatr Blood Cancer 2023; 70 Suppl 6:e30577. [PMID: 37480158 PMCID: PMC10527977 DOI: 10.1002/pbc.30577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/23/2023]
Abstract
Since the publication of the last Cellular Therapy and Stem Cell Transplant blueprint in 2013, Children's Oncology Group cellular therapy-based trials advanced the field and created new standards of care across a wide spectrum of pediatric cancer diagnoses. Key findings include that tandem autologous transplant improved survival for patients with neuroblastoma and atypical teratoid/rhabdoid brain tumors, one umbilical cord blood (UCB) donor was safer than two UCB donors, killer immunoglobulin receptor (KIR) mismatched donors did not improve survival for pediatric acute myeloid leukemia when in vivo T-cell depletion is used, and the depth of remission as measured by next-generation sequencing-based minimal residual disease assessment pretransplant was the best predictor of relapse for acute lymphoblastic leukemia. Plans for the next decade include optimizing donor selection for transplants for acute leukemia/myelodysplastic syndrome, using novel engineered cellular therapies to target a wide array of malignancies, and developing better treatments for cellular therapy toxicities such as viral infections and graft-vs-host disease.
Collapse
Affiliation(s)
- Carrie L. Kitko
- Pediatric Stem Cell Transplant Program, Vanderbilt University Medical Center, Nashville, TN
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
- GW Cancer Center, George Washington University, Washington, DC
- Division of Blood and Marrow Transplantation, Children’s National Hospital, Washington, DC
| | - Mitchell S. Cairo
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Maria Fareri Children's Hospital, Westchester Medical Center, New York Medical College, Valhalla, New York, NY
| | - Joseph Chewning
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL
| | - Terry J. Fry
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO
| | - Michael A. Pulsipher
- Division of Hematology and Oncology, Intermountain Primary Children’s Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine, Salt Lake City, UT
| | - Shalini Shenoy
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Washington University, St Louis, MO
| | - Donna A. Wall
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Canada
| | - John E. Levine
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
4
|
Streby KA, Parisi MT, Shulkin BL, LaBarre B, Bagatell R, Diller L, Grupp SA, Matthay KK, Voss SD, Yu AL, London WB, Park JR, Yanik GA, Naranjo A. Impact of diagnostic and end-of-induction Curie scores with tandem high-dose chemotherapy and autologous transplants for metastatic high-risk neuroblastoma: A report from the Children's Oncology Group. Pediatr Blood Cancer 2023; 70:e30418. [PMID: 37199022 PMCID: PMC10511015 DOI: 10.1002/pbc.30418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/12/2023] [Accepted: 04/26/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Diagnostic mIBG (meta-iodobenzylguanidine) scans are an integral component of response assessment in children with high-risk neuroblastoma. The role of end-of-induction (EOI) Curie scores (CS) was previously described in patients undergoing a single course of high-dose chemotherapy (HDC) and autologous hematopoietic cell transplant (AHCT) as consolidation therapy. OBJECTIVE We now examine the prognostic significance of CS in patients randomized to tandem HDC and AHCT on the Children's Oncology Group (COG) trial ANBL0532. STUDY DESIGN A retrospective analysis of mIBG scans obtained from patients enrolled in COG ANBL0532 was performed. Evaluable patients had mIBG-avid, International Neuroblastoma Staging System (INSS) stage 4 disease, did not progress during induction therapy, consented to consolidation randomization, and received either single or tandem HDC (n = 80). Optimal CS cut points maximized the outcome difference (≤CS vs. >CS cut-off) according to the Youden index. RESULTS For recipients of tandem HDC, the optimal cut point at diagnosis was CS = 12, with superior event-free survival (EFS) from study enrollment for patients with CS ≤ 12 (3-year EFS 74.2% ± 7.9%) versus CS > 12 (59.2% ± 7.1%) (p = .002). At EOI, the optimal cut point was CS = 0, with superior EOI EFS for patients with CS = 0 (72.9% ± 6.4%) versus CS > 0 (46.5% ± 9.1%) (p = .002). CONCLUSION In the setting of tandem transplantation for children with high-risk neuroblastoma, CS at diagnosis and EOI may identify a more favorable patient group. Patients treated with tandem HDC who exhibited a CS ≤ 12 at diagnosis or CS = 0 at EOI had superior EFS compared to those with CS above these cut points.
Collapse
Affiliation(s)
- Keri A. Streby
- Division of Hematology/Oncology/BMT, Department of Pediatrics, Nationwide Children’s Hospital/The Ohio State University, Columbus, Ohio
| | - Marguerite T. Parisi
- Department of Radiology, Seattle Children’s Hospital/University of Washington School of Medicine, Seattle, Washington
- Department of Pediatrics, Seattle Children’s Hospital/University of Washington School of Medicine, Seattle, Washington
| | - Barry L. Shulkin
- Department of Radiological Sciences, St. Jude Children’s Research Hospital, Adjunct Professor of Radiology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Brian LaBarre
- Children’s Oncology Group Statistics & Data Center, Department of Biostatistics, University of Florida, Gainesville, Florida
| | - Rochelle Bagatell
- Department of Pediatrics, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lisa Diller
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Stephan A. Grupp
- Department of Pediatrics, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania
| | - Katherine K. Matthay
- Department of Pediatrics, University of California San Francisco School of Medicine, San Francisco, California
| | - Stephan D. Voss
- Department of Radiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alice L. Yu
- University of California in San Diego, San Diego, California
- Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Wendy B. London
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Julie R. Park
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Gregory A. Yanik
- Department of Pediatrics, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Arlene Naranjo
- Children’s Oncology Group Statistics & Data Center, Department of Biostatistics, University of Florida, Gainesville, Florida
| |
Collapse
|
5
|
Ash S, Askenasy N. Immunotherapy for neuroblastoma by hematopoietic cell transplantation and post-transplant immunomodulation. Crit Rev Oncol Hematol 2023; 185:103956. [PMID: 36893946 DOI: 10.1016/j.critrevonc.2023.103956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/14/2022] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Neuroblastoma represents a relatively common childhood tumor that imposes therapeutic difficulties. High risk neuroblastoma patients have poor prognosis, display limited response to radiochemotherapy and may be treated by hematopoietic cell transplantation. Allogeneic and haploidentical transplants have the distinct advantage of reinstitution of immune surveillance, reinforced by antigenic barriers. The key factors favorable to ignition of potent anti-tumor reactions are transition to adaptive immunity, recovery from lymphopenia and removal of inhibitory signals that inactivate immune cells at the local and systemic levels. Post-transplant immunomodulation may further foster anti-tumor reactivity, with positive but transient impact of infusions of lymphocytes and natural killer cells both from the donor, the recipient or third party. The most promising approaches include introduction of antigen-presenting cells in early post-transplant stages and neutralization of inhibitory signals. Further studies will likely shed light on the nature and actions of suppressor factors within tumor stroma and at the systemic level.
Collapse
Affiliation(s)
- Shifra Ash
- Department of Pediatric Hematology-Oncology, Rambam Medical Center, Haifa, Israel; Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Nadir Askenasy
- Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
6
|
Bird N, Scobie N, Palmer A, Ludwinski D. To transplant, or not to transplant? That is the question. A patient advocate evaluation of autologous stem cell transplant in neuroblastoma. Pediatr Blood Cancer 2022; 69:e29663. [PMID: 35373890 DOI: 10.1002/pbc.29663] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/12/2022] [Accepted: 02/27/2022] [Indexed: 12/27/2022]
Abstract
High-dose chemotherapy with autologous stem cell transplant (ASCT) has been a mainstay of high-risk neuroblastoma treatment for several decades, demonstrating improvements in event-free survival but with risks of serious or even life-threatening acute toxicities, severe long-term adverse health effects for survivors, and ongoing contention regarding overall survival benefit. The merits of ASCT in the modern era of immunotherapy are a source of debate among parents, advocates, and some physicians. Here we examine evidence for and against ASCT, explore parent attitudes and their turmoil over decision-making, and strongly encourage international research consortia to develop a coordinated strategy to accelerate progress toward a future that avoids the routine use of ASCT in high-risk neuroblastoma.
Collapse
|
7
|
Wei Z, Li J, Jin Y, Liu Y, Wang P, Cao Y, Zhao Q. The application and value of radiotherapy at the primary site in patients with high-risk neuroblastoma. Br J Radiol 2022; 95:20211086. [PMID: 35312349 PMCID: PMC10996409 DOI: 10.1259/bjr.20211086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/22/2022] [Accepted: 03/14/2022] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To retrospectively analyze radiotherapy (RT) regimens for patients with high-risk neuroblastoma (HRNB) at the primary site after surgery, and to further analyze the characteristics of patients who would benefit more from RT. METHODS 98 pediatric patients with HRNB were analyzed for local control (LC), RT dose, extent of excision and prognostic factors. Among them, 69 children received RT. RESULTS The 3 year LC rates were 96.9 and 62.1% (p < 0.001) in the RT and non-RT groups, respectively. In the non-RT group, LC was better in patients with complete macroscopic resection (CME) than in those with incomplete macroscopic resection (IME) (p = 0.026), while in the RT group, no significant difference in LC was found (p = 0.985). Among patients with IME, the LC was 100% in patients with RT doses >= 36 Gy and 66.7% in patients with doses <36 Gy. CONCLUSION RT is valuable, provides patients with excellent LC, and is safe in the short term. RT had a complementary therapeutic effect on incompletely resected tumors, thus bringing their LC to the level of patients with CME. For patients with IME, RT at a dose of not less than 36 Gy may improve LC. ADVANCES IN KNOWLEDGE This study analysed the role of radiotherapy in HRNB, investigated the dose of RT depending on the degree of resection, and explored the characteristics of patients who would benefit more from RT.
Collapse
Affiliation(s)
- Zixuan Wei
- Department of Pediatric Oncology, Tianjin Medical University
Cancer Institute and Hospital, National Clinical Research Center for
Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin,
Tianjin’s Clinical Research Center for Cancer,
Tianjin, China
| | - Jie Li
- Department of Pediatric Oncology, Tianjin Medical University
Cancer Institute and Hospital, National Clinical Research Center for
Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin,
Tianjin’s Clinical Research Center for Cancer,
Tianjin, China
| | - Yan Jin
- Department of Pediatric Oncology, Tianjin Medical University
Cancer Institute and Hospital, National Clinical Research Center for
Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin,
Tianjin’s Clinical Research Center for Cancer,
Tianjin, China
| | - Yun Liu
- Department of Pediatric Oncology, Tianjin Medical University
Cancer Institute and Hospital, National Clinical Research Center for
Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin,
Tianjin’s Clinical Research Center for Cancer,
Tianjin, China
| | - Peiguo Wang
- Department of Radiotherapy, Tianjin Medical University Cancer
Institute and Hospital, National Clinical Research Center for Cancer,
Key Laboratory of Cancer Prevention and Therapy of Tianjin,
Tianjin’s Clinical Research Center for Cancer,
Tianjin, China
| | - Yanna Cao
- Department of Pediatric Oncology, Tianjin Medical University
Cancer Institute and Hospital, National Clinical Research Center for
Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin,
Tianjin’s Clinical Research Center for Cancer,
Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University
Cancer Institute and Hospital, National Clinical Research Center for
Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin,
Tianjin’s Clinical Research Center for Cancer,
Tianjin, China
| |
Collapse
|
8
|
Carcamo B, Francia G. Cyclic Metronomic Chemotherapy for Pediatric Tumors: Six Case Reports and a Review of the Literature. J Clin Med 2022; 11:jcm11102849. [PMID: 35628975 PMCID: PMC9144744 DOI: 10.3390/jcm11102849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/06/2022] [Accepted: 05/13/2022] [Indexed: 12/03/2022] Open
Abstract
We report a retrospective case series of six Hispanic children with tumors treated with metronomic chemotherapy. The six cases comprised one rhabdoid tumor of the kidney, one ependymoma, two medulloblastomas, one neuroblastoma, and a type II neurocytoma of the spine. Treatment included oral cyclophosphamide daily for 21 days alternating with oral etoposide daily for 21 days in a backbone of daily valproic acid and celecoxib. In one case, celecoxib was substituted with sulindac. Of the six patients, three showed complete responses, and all patients showed some response to metronomic therapy with only minor hematologic toxicity. One patient had hemorrhagic gastritis likely associated with NSAIDs while off prophylactic antacids. These data add to a growing body of evidence suggesting that continuous doses of valproic acid and celecoxib coupled with alternating metronomic chemotherapy of agents such as etoposide and cyclophosphamide can produce responses in pediatric tumors relapsing to conventional dose chemotherapy.
Collapse
Affiliation(s)
- Benjamin Carcamo
- Department of Pediatric Hematology Oncology, El Paso Children’s Hospital, El Paso, TX 79905, USA
- Department of Pediatrics, Texas Tech University Health Science Center, El Paso, TX 79430, USA
- Correspondence: (B.C.); (G.F.); Tel.: +1-915-479-8970 (B.C.); +1-915-747-8025 (G.F.); Fax: +1-915-242-8437 (B.C.); +1-915-747-5808 (G.F.)
| | - Giulio Francia
- Border Biomedical Research Center, University of Texas at El Paso (UTEP), El Paso, TX 79968, USA
- Correspondence: (B.C.); (G.F.); Tel.: +1-915-479-8970 (B.C.); +1-915-747-8025 (G.F.); Fax: +1-915-242-8437 (B.C.); +1-915-747-5808 (G.F.)
| |
Collapse
|
9
|
Hara J, Nitani C, Shichino H, Kuroda T, Hishiki T, Soejima T, Mori T, Matsumoto K, Sasahara Y, Iehara T, Miyamura T, Kosaka Y, Takimoto T, Nakagawara A, Tajiri T. Outcome of children with relapsed high-risk neuroblastoma in Japan and analysis of the role of allogeneic hematopoietic stem cell transplantation. Jpn J Clin Oncol 2022; 52:486-492. [PMID: 35137156 DOI: 10.1093/jjco/hyac007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In Japan, allogeneic hematopoietic stem cell transplantation is widely performed for recurrent neuroblastomas. This retrospective study aimed to investigate the prognosis of recurrent neuroblastoma in Japan and explore the effectiveness of allogeneic hematopoietic stem cell transplantation. METHODS Clinical characteristics and data on the treatment of patients with high-risk neuroblastoma who experienced first progression between 2003 and 2010 after attaining complete remission or partial remission were collected from hospitals participating in the Japanese Neuroblastoma Research Group. RESULTS Data from 61 patients who fulfilled these criteria were collected. The median interval from disease onset to first progression was 19 months (range, 7-65 months), whereas the median observation time of the surviving patients was 18 months (range, 1-69 months). All patients were treated with chemotherapy, where 22 and 3 patients received allogeneic and autologous hematopoietic stem cell transplantation, respectively. Seven patients were alive in second complete remission, and 39 died, including two in complete remission. The 3-year progression-free survival and overall survival rates were 15.3% (SE: 6.1%) and 16.9% (SE: 6.5%), respectively. For patients with allogeneic hematopoietic stem cell transplantation, the 3-year progression-free survival and overall survival were 28.3% (standard error, 12.0%) and 24.3% (standard error, 11.5%), respectively, and for patients without allogeneic hematopoietic stem cell transplantation, the 3-year progression-free survival and overall survival were 6.0% (standard error 5.5%) and 12.0% (standard error 7.6%), respectively. The duration of initial remission (≥ 18 months) and implementation of allogeneic hematopoietic stem cell transplantation were independently predictive of progression-free survival (P = 0.002 and P = 0.017), whereas for overall survival, only allogeneic hematopoietic stem cell transplantation was predictive (P = 0.012). CONCLUSION Although allogeneic hematopoietic stem cell transplantation contributed to some improvement in prognosis, it was insufficient.
Collapse
Affiliation(s)
- Junichi Hara
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Chika Nitani
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Hiroyuki Shichino
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Tatsuo Kuroda
- Department of Pediatric Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Tomoro Hishiki
- Department of Pediatric Surgery, Chiba University, Chiba, Japan
| | | | - Tetsuya Mori
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kimikazu Matsumoto
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Yoji Sasahara
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoko Iehara
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Takako Miyamura
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Tetsuya Takimoto
- Department of Childhood Cancer Data Management, National Center for Child Health and Development, Tokyo, Japan
| | | | - Tatsuro Tajiri
- Department of Pediatric Surgery, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan.,Department of Pediatric Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
10
|
Yan J, Jie L, Jiaxing Y, Yanna C, Zhanglin L, Zhongyuan L, Daowei W, Guangzong Z, Benfu Z, Jie Y, Qiang Z. Analysis of the efficacy of autologous peripheral blood stem cell transplantation in high-risk neuroblastoma. Int J Med Sci 2022; 19:1715-1723. [PMID: 36237985 PMCID: PMC9553861 DOI: 10.7150/ijms.76305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 09/23/2022] [Indexed: 12/03/2022] Open
Abstract
Objective: This study aimed to analyze the efficacy of autologous peripheral blood stem cell transplantation for high-risk neuroblastoma in China. Methods: The data of 90 high-risk neuroblastoma patients treated with the CCCG-NB 2015 regimen were reviewed. The baseline clinicopathological characteristics and prognosis were analyzed and compared. In addition, the prognoses of tandem autologous stem cell transplantation and single autologous stem cell transplantation groups were compared. Results: The results of survival analysis showed that autologous peripheral blood stem cell transplantation based on this pretreatment regimen significantly improved the prognosis of children in the high-risk group. The 3-year event-free survival (EFS) and overall survival (OS) rates for the transplantation group and the nontransplantation group were 65.5% vs. 41.3% (p=0.023) and 77.1% vs. 57.9% (p=0.03), respectively. There was no difference in the distribution of baseline clinical case characteristics between the single transplantation group and the tandem transplantation group (p>0.05), and there was no significant difference in EFS and OS between the two groups (p>0.05). Conclusion: Based on this pretreatment programme, autologous peripheral blood stem cell transplantation is safe and tolerable and significantly improves the prognosis of children in the high-risk group. The value of tandem autologous stem cell transplantation is worthy of further discussion, which should consider various aspects such as the transplantation medication regimen and the patient's state.
Collapse
Affiliation(s)
- Jin Yan
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention & Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Li Jie
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention & Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yang Jiaxing
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention & Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Cao Yanna
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention & Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Li Zhanglin
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention & Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Li Zhongyuan
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention & Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Wang Daowei
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention & Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhao Guangzong
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention & Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhong Benfu
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention & Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yan Jie
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention & Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhao Qiang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention & Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
11
|
Yamazaki F, Yamasaki K, Kiyotani C, Hashii Y, Shioda Y, Hara J, Matsumoto K. Thiotepa-melphalan myeloablative therapy for high-risk neuroblastoma. Pediatr Blood Cancer 2021; 68:e28896. [PMID: 33788375 DOI: 10.1002/pbc.28896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 12/02/2020] [Accepted: 12/26/2020] [Indexed: 01/30/2023]
Abstract
BACKGROUND Appropriate high-dose chemotherapy (HDC) for high-risk neuroblastoma has not yet been established. In Japan, a unique HDC regimen that comprises two cycles of a total of 800 mg/m2 of thiotepa and a total of 280 mg/m2 of melphalan is widely utilized. METHODS To evaluate the safety and efficacy of this thiotepa-melphalan high-dose therapy for high-risk neuroblastoma, we reviewed the medical records of 41 patients with high-risk neuroblastoma who underwent this regimen followed by autologous peripheral blood stem cell rescue between 2002 and 2012. MYCN-amplified high-risk neuroblastomas were observed in 23 patients. All patients underwent intensive multidrug induction chemotherapy, but none underwent anti-GD2 antibody immunotherapy. The primary tumor was resected at the adequate time point. RESULTS The median follow-up duration for living patients was 9.2 years (range 5.5-14.0 years). The 5-year event-free survival (EFS) and overall survival from treatment initiation were 41.5 ± 7.7% and 56.1 ± 7.8%, respectively. The 5-year EFS of MYCN-amplified high-risk neuroblastoma patients was 60.9 ± 10.2%, which was significantly superior compared with those with MYCN-nonamplified high-risk neuroblastoma (16.7 ± 8.8%; p < .001). MYCN amplification was the most favorable prognostic factor for EFS (hazard ratio = 0.29; 95% confidence interval = 0.12-0.66). Of the 41 patients, three died because of regimen-related toxicity (infection, n = 2; microangiopathy, n = 1). CONCLUSION The thiotepa-melphalan high-dose therapy with thiotepa and melphalan may be effective for high-risk neuroblastoma. However, this regimen is toxic and warrants special attention in clinical practice.
Collapse
Affiliation(s)
- Fumito Yamazaki
- National Center for Child Health and Development, Children's Cancer Center, Tokyo, Japan.,Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Kai Yamasaki
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Chikako Kiyotani
- National Center for Child Health and Development, Children's Cancer Center, Tokyo, Japan
| | - Yoshiko Hashii
- Department of Pediatrics, Osaka University Hospital, Osaka, Japan
| | - Yoko Shioda
- National Center for Child Health and Development, Children's Cancer Center, Tokyo, Japan
| | - Junichi Hara
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Kimikazu Matsumoto
- National Center for Child Health and Development, Children's Cancer Center, Tokyo, Japan
| |
Collapse
|
12
|
Grèze V, Rouel N, Rochette E, Merlin E, Halle P, Plantaz D, Deméocq F, Kanold J. Peripheral blood stem cell collection in children with extremely low body weight (≤8 kg). What have we learned over the past 25 years and where are the limits? J Clin Apher 2020; 36:322-331. [PMID: 33382142 DOI: 10.1002/jca.21863] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/23/2020] [Accepted: 12/02/2020] [Indexed: 11/10/2022]
Abstract
Hematopoietic progenitor cells-apheresis (HPC-A) collection is now a routine procedure for autologous hematopoietic stem cell transplantation. Here we present our 25 years' experience of HPC-A collection in children weighing 8 kg or less, with a focus on the evolution of our standard operating procedures, and the safety limits for these young patients, in the Pediatric Apheresis Unit of Clermont-Ferrand University Hospital (France). Fifteen children weighing 8 kg or less underwent 26 HPC-A collections over 25 years. Median CD34+ cell yield by leukapheresis was 4.4 106 /kg. No procedure-related complications were encountered during or after the collection. No patient had profound thrombocytopenia or anemia that needed post-collection transfusions. Our experience in pediatric oncology patients who underwent HPC-A collections shows that this procedure can be performed even in the smallest of children with no increase in toxicity provided all precautions are taken to ensure that the procedure is carried out under the ideal conditions.
Collapse
Affiliation(s)
- Victoria Grèze
- CHU Clermont-Ferrand, Service Hématologie Oncologie Pédiatrique, Hôpital Estaing, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, CIC 1405, Unité CRECHE, Clermont-Ferrand, France.,CHU Clermont-Ferrand, Centre de Biothérapie d'Auvergne, Clermont-Ferrand, France
| | - Nadège Rouel
- CHU Clermont-Ferrand, Service Hématologie Oncologie Pédiatrique, Hôpital Estaing, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, CIC 1405, Unité CRECHE, Clermont-Ferrand, France
| | - Emmanuelle Rochette
- CHU Clermont-Ferrand, Service Hématologie Oncologie Pédiatrique, Hôpital Estaing, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, CIC 1405, Unité CRECHE, Clermont-Ferrand, France
| | - Etienne Merlin
- Université Clermont Auvergne, INSERM, CIC 1405, Unité CRECHE, Clermont-Ferrand, France.,CHU Clermont-Ferrand, Centre de Biothérapie d'Auvergne, Clermont-Ferrand, France.,Université Clermont Auvergne, INRA, UMR 1019 UNH, ECREIN, Clermont-Ferrand, France
| | - Pascale Halle
- CHU Clermont-Ferrand, Service Hématologie Oncologie Pédiatrique, Hôpital Estaing, Clermont-Ferrand, France
| | - Dominique Plantaz
- CHU Grenoble, département de pédiatrie, Hôpital couple-enfant, Grenoble, France
| | - François Deméocq
- Université Clermont Auvergne, INRA, UMR 1019 UNH, ECREIN, Clermont-Ferrand, France
| | - Justyna Kanold
- CHU Clermont-Ferrand, Service Hématologie Oncologie Pédiatrique, Hôpital Estaing, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, CIC 1405, Unité CRECHE, Clermont-Ferrand, France.,CHU Clermont-Ferrand, Centre de Biothérapie d'Auvergne, Clermont-Ferrand, France.,Université Clermont Auvergne, INRA, UMR 1019 UNH, ECREIN, Clermont-Ferrand, France
| |
Collapse
|
13
|
Kato S, Kubota Y, Watanabe K, Hogetsu K, Arakawa Y, Koh K, Takita J, Hiwatari M. Tandem high-dose chemotherapy with autologous stem cell rescue for stage M high-risk neuroblastoma: Experience using melphalan/etoposide/carboplatin and busulfan/melphalan regimens. Pediatr Transplant 2020; 24:e13772. [PMID: 32543778 DOI: 10.1111/petr.13772] [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: 09/23/2019] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 11/30/2022]
Abstract
The efficacy of tandem HDCT against high-risk neuroblastoma has been reported; however, an optimal regimen remains to be established. In this paper, we report our experience using tandem HDCT comprising the MEC and BuMel regimens in patients with high-risk neuroblastoma. We retrospectively analyzed four patients with stage M high-risk neuroblastoma who received HDCT with MEC followed by BuMel combined with autologous stem cell rescue. Although none of their metastatic lesions had disappeared after induction chemotherapy, three patients showed a CR after tandem HDCT. Gastrointestinal mucosal injuries and renal dysfunction were observed as non-hematologic adverse events of grade 3 or higher. Gastrointestinal mucosal injuries were observed in all four patients following the first HDCT and in one patient following the second HDCT and were treated with parenteral nutrition and analgesics. One patient experienced renal dysfunction during the first HDCT, which was alleviated by sufficient hydration and diuretics and resulted in the reduction of melphalan dosage for the second HDCT. SOS was not observed in any patient. The HDCT regimens examined in this study were observed to be feasible and did not result in any life-threatening adverse events. Our findings indicate that tandem HDCT comprising MEC and BuMel is a potentially effective regimen for patients with high-risk neuroblastoma, including for those who respond poorly to induction chemotherapy, although additional studies in a larger population should be conducted to verify any long-term outcomes and toxicity.
Collapse
Affiliation(s)
- Shota Kato
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuo Kubota
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kentaro Watanabe
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Keita Hogetsu
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuki Arakawa
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Junko Takita
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mitsuteru Hiwatari
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Cell Therapy and Transplantation Medicine, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
14
|
Aravindan N, Somasundaram DB, Herman TS, Aravindan S. Significance of hematopoietic surface antigen CD34 in neuroblastoma prognosis and the genetic landscape of CD34-expressing neuroblastoma CSCs. Cell Biol Toxicol 2020; 37:461-478. [PMID: 32979173 DOI: 10.1007/s10565-020-09557-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022]
Abstract
High-risk neuroblastoma (HR-NB) is branded with hematogenous metastasis, relapses, and dismal long-term survival. Intensification of consolidation therapy with tandem/triple autologous stem cell (SC) rescue (with bone marrow [BM]/peripheral blood [PB] CD34+ selection) after myeloablative chemotherapy has improved long-term survival. However, the benefit is limited by the indication of NB cells in CD34+ PBSCs, CD34 expression in NB cells, and the risk of reinfusing NB cancer stem cells (NB CSCs) that could lead to post-transplant relapse. We investigated the association of CD34 surface expression (92 patients) with NB evolution/clinical outcomes. CD34 gene-level status in NB was assessed through RNA-Seq data mining (18 cohorts, n, 3324). Genetic landscape of CD34-expressing NB CSCs (CD133+CD34+) was compared with CD34- CSCs (CD133+CD34-). RNA-seq data revealed equivocal association patterns of CD34 expression with patient survival. Our immunohistochemistry data revealed definite, but rare (mean, 0.73%; range 0.00-7.87%; median, 0.20%) CD34 positivity in NB. CD34+ significantly associated with MYCN amplification (p, 0.003), advanced disease stage (p, 0.016), and progressive disease (PD, p < 0.0009) after clinical therapy. A general high-is-worse tendency was observed in patients with relapsed disease. High CD34+ correlated with poor survival in patients with N-MYC-amplified HR-NB. Gene expression analysis of CD34+-NB CSCs identified significant up (4631) and downmodulation (4678) of genes compared with NB CSCs that lack CD34. IPA recognized the modulation of crucial signaling elements (EMT, stemness maintenance, differentiation, inflammation, clonal expansion, drug resistance, metastasis) that orchestrate NB disease evolution in CD34+ CSCs compared with CD34- CSCs. While the function of CD34 in NB evolution requires further in-depth investigation, careful consideration should be exercised for autologous stem cell rescue with CD34+ selection in NB patients. Graphical abstract.
Collapse
Affiliation(s)
- Natarajan Aravindan
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, BMSB 737, 940 Stanton L. Young Boulevard, Oklahoma City, OK, 73104, USA. .,Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA. .,Department of Anesthesiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Dinesh Babu Somasundaram
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, BMSB 737, 940 Stanton L. Young Boulevard, Oklahoma City, OK, 73104, USA
| | - Terence S Herman
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, BMSB 737, 940 Stanton L. Young Boulevard, Oklahoma City, OK, 73104, USA.,Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
| | | |
Collapse
|
15
|
Becherucci V, Ermini S, Piccini L, Bisin S, Gori V, Gentile F, Ceccantini R, De Rienzo E, Bindi B, Pavan P, Cunial V, Allegro E, Brugnolo F, Maggio D, Calzolari D, Maccarelli E, Galli S, Muricci S, Berchielli M, Tintori V, Sardi I, Bambi F. An alternative procedure to leukapheresis for peripheral hematopoietic progenitor cell collection in very-low-weight children: A single pediatric center experience. J Clin Apher 2020; 35:406-412. [PMID: 32710805 DOI: 10.1002/jca.21813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/27/2020] [Accepted: 06/22/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND PBSC collection using a blood cell separator in very low weight patients can be frequently complicated by severe adverse effects and technical difficulties. MATERIAL AND METHODS From March 2013 to January 2017, 14 PBSC collections were performed in 12 children weighing less than 10 kg, affected by different solid tumours. PBSC collection was performed with a "homemade" aseptically assembled circuit. The circuit is composed by a 150 mL collection bag connected with a 4 stopcock ramp, perfused with ACD. This circuit allows collection of a specific total blood amount from CVC, depending on CD34+ /kg target. RESULTS Mean CD34+ cell performance per collection was 9.3 × 106 /kg. Tolerance to the procedure was very good as none of the patients experienced complications, with the exception of a patient who showed mild cyanosis and pallor after collection. Moreover, no bleeding or thrombotic complications have been observed. To date, 16 PBSC reinfusions have been performed in 7 children with a mean CD34+ cells viability of 98.1% ± 2.7 and mean WBC viability of 57% ± 10. Cell recovery after thawing was 87% ± 10.8. A rapid graft intake for both neutrophils and platelets, between day 7 and 20 after reinfusion was observed. DISCUSSION The procedure of total blood collection without the use of a cell separator is feasible and allows a good PBSC collection without significant side effects in very low-weight children. Moreover, this method could represent a valid and safe alternative to leukapheresis in patients where classic procedure could be difficult to apply.
Collapse
Affiliation(s)
- Valentina Becherucci
- Immunohematology, Transfusion Medicine and Laboratory, Meyer Children's Hospital, Florence, Italy
| | - Stefano Ermini
- Stem cell collection and Therapeutic Apheresis unit, Meyer Children's Hospital, Florence, Italy
| | - Luisa Piccini
- Immunohematology, Transfusion Medicine and Laboratory, Meyer Children's Hospital, Florence, Italy
| | - Silvia Bisin
- Immunohematology, Transfusion Medicine and Laboratory, Meyer Children's Hospital, Florence, Italy
| | - Valentina Gori
- Immunohematology, Transfusion Medicine and Laboratory, Meyer Children's Hospital, Florence, Italy
| | - Francesca Gentile
- Immunohematology, Transfusion Medicine and Laboratory, Meyer Children's Hospital, Florence, Italy
| | - Riccardo Ceccantini
- Immunohematology, Transfusion Medicine and Laboratory, Meyer Children's Hospital, Florence, Italy
| | - Elena De Rienzo
- Immunohematology, Transfusion Medicine and Laboratory, Meyer Children's Hospital, Florence, Italy
| | - Barbara Bindi
- Immunohematology, Transfusion Medicine and Laboratory, Meyer Children's Hospital, Florence, Italy
| | - Paola Pavan
- Immunohematology, Transfusion Medicine and Laboratory, Meyer Children's Hospital, Florence, Italy
| | - Vanessa Cunial
- Immunohematology, Transfusion Medicine and Laboratory, Meyer Children's Hospital, Florence, Italy
| | - Elisa Allegro
- Stem cell collection and Therapeutic Apheresis unit, Meyer Children's Hospital, Florence, Italy
| | - Francesca Brugnolo
- Immunohematology, Transfusion Medicine and Laboratory, Meyer Children's Hospital, Florence, Italy
| | - Daniela Maggio
- Stem cell collection and Therapeutic Apheresis unit, Meyer Children's Hospital, Florence, Italy
| | - Daniela Calzolari
- Stem cell collection and Therapeutic Apheresis unit, Meyer Children's Hospital, Florence, Italy
| | - Erika Maccarelli
- Stem cell collection and Therapeutic Apheresis unit, Meyer Children's Hospital, Florence, Italy
| | - Silvia Galli
- Stem cell collection and Therapeutic Apheresis unit, Meyer Children's Hospital, Florence, Italy
| | - Sonia Muricci
- Stem cell collection and Therapeutic Apheresis unit, Meyer Children's Hospital, Florence, Italy
| | - Marco Berchielli
- Stem cell collection and Therapeutic Apheresis unit, Meyer Children's Hospital, Florence, Italy
| | - Veronica Tintori
- Transplantation Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| | - Iacopo Sardi
- Neuro-oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| | - Franco Bambi
- Immunohematology, Transfusion Medicine and Laboratory, Meyer Children's Hospital, Florence, Italy
| |
Collapse
|
16
|
Tolbert VP, Dvorak CC, Golden C, Vissa M, El-Haj N, Perwad F, Matthay KK, Vo KT. Risk Factors for Transplant-Associated Thrombotic Microangiopathy after Autologous Hematopoietic Cell Transplant in High-Risk Neuroblastoma. Biol Blood Marrow Transplant 2019; 25:2031-2039. [PMID: 31199983 PMCID: PMC9161973 DOI: 10.1016/j.bbmt.2019.06.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/21/2019] [Accepted: 06/05/2019] [Indexed: 12/12/2022]
Abstract
High-risk neuroblastoma has a poor prognosis, and research studies have shown that increasing the intensity of therapy improves outcomes. Autologous hematopoietic cell transplant (aHCT) as consolidation therapy confers a significant survival advantage but is accompanied by significant morbidity. Transplant-associated thrombotic microangiopathy (TA-TMA) is a life-threatening complication caused by endothelial injury that often leads to hemolytic anemia, microthrombotic platelet consumption, and renal injury. Here we investigated the incidence, potential risk factors, and sequelae of TA-TMA in patients with high-risk neuroblastoma. We conducted a retrospective chart review of all patients (n = 141) with neuroblastoma in our institutions who underwent aHCT from 2000 to 2017. Ten patients (7%) developed TA-TMA. The patients in the TA-TMA group were similar to the rest of the subjects in demographics, disease burden, prior therapies, renal function, and timing of transplant. The type of conditioning regimen was the only statistically significant pretransplant variable (P < .001). Six of 15 patients (40%) intended to receive tandem transplants (cyclophosphamide/thiotepa and then carboplatin/etoposide/melphalan (CEM)), 4 of 68 patients (6%) who received conditioning with single CEM, and none of the 56 patients who received busulfan/melphalan were diagnosed with TA-TMA. Patients with TA-TMA were more likely to require intensive care unit transfer, have a longer length of stay in the hospital, and experience a delay or change in their subsequent therapy. In our cohort overall, patients with a delay in therapy after transplant appeared to have a worse overall survival, although the difference was not statistically significant. Because of this high incidence and significant morbidity, we have implemented standardized screening for TA-TMA during and after transplant. We anticipate that screening will lead to earlier intervention and decreased severity of disease.
Collapse
Affiliation(s)
- Vanessa P Tolbert
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children's Hospital, San Francisco, California
| | - Christopher C Dvorak
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children's Hospital, San Francisco, California
| | - Carla Golden
- Division of Hematology/Oncology, University of California San Francisco Benioff Children's Hospital, Oakland, California
| | - Madhav Vissa
- Division of Hematology/Oncology, University of California San Francisco Benioff Children's Hospital, Oakland, California
| | - Nura El-Haj
- Division of Hematology/Oncology, University of California San Francisco Benioff Children's Hospital, Oakland, California
| | - Farzana Perwad
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children's Hospital, San Francisco, California
| | - Katherine K Matthay
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children's Hospital, San Francisco, California
| | - Kieuhoa T Vo
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children's Hospital, San Francisco, California.
| |
Collapse
|
17
|
Jo JH, Ahn SD, Koh M, Kim JH, Lee SW, Song SY, Yoon SM, Kim YS, Kim SS, Park JH, Jung J, Choi EK. Patterns of recurrence after radiation therapy for high-risk neuroblastoma. Radiat Oncol J 2019; 37:224-231. [PMID: 31591871 PMCID: PMC6790795 DOI: 10.3857/roj.2019.00353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/16/2019] [Indexed: 12/28/2022] Open
Abstract
Purpose To investigate the patterns of recurrence in patients with neuroblastoma treated with radiation therapy to the primary tumor site. Materials and Methods We retrospectively analyzed patients with high-risk neuroblastoma managed with definitive treatment with radiation therapy to the primary tumor site between January 2003 and June 2017. These patients underwent three-dimensional conformal radiation therapy or intensity-modulated radiation therapy. A total of 14–36 Gy was delivered to the planning target volume, which included the primary tumor bed and the selected metastatic site. The disease stage was determined according to the International Neuroblastoma Staging System (INSS). We evaluated the recurrence pattern (i.e., local or systemic), progression-free survival, and overall survival. Results A total of 40 patients with high-risk neuroblastoma were included in this study. The median patient age was 4 years (range, 1 to 11 years). Thirty patients (75%) had INSS stage 4 neuroblastoma. At the median follow-up of 58 months, there were 6 cases of local recurrence and 10 cases of systemic recurrence. Among the 6 local failure cases, 4 relapsed adjacent to the radiation field. The other 2 relapsed in the radiation field (i.e., para-aortic and retroperitoneal areas). The main sites of distant metastasis were the bone, lymph nodes, and bone marrow. The 5-year progression-free survival was 70.9% and the 5-year overall survival was 74.3%. Conclusion Radiation therapy directed at the primary tumor site provides good local control. It seems to be adequate for disease control in patients with high-risk neuroblastoma after chemotherapy and surgical resection.
Collapse
Affiliation(s)
- Ji Hwan Jo
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung Do Ahn
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Minji Koh
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jong Hoon Kim
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang-Wook Lee
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Si Yeol Song
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang Min Yoon
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Young Seok Kim
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Su Ssan Kim
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jin-Hong Park
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jinhong Jung
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun Kyung Choi
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
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
|
19
|
Bagatell R, Irwin MS. Tandem Transplant for High-Risk Neuroblastoma: Next Steps in the Era of Precision Medicine. JAMA 2019; 322:729-731. [PMID: 31454025 DOI: 10.1001/jama.2019.11641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Rochelle Bagatell
- Perelman School of Medicine, Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia
| | - Meredith S Irwin
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Peinemann F, van Dalen EC, Enk H, Tytgat GAM. Anti-GD2 antibody-containing immunotherapy postconsolidation therapy for people with high-risk neuroblastoma treated with autologous haematopoietic stem cell transplantation. Cochrane Database Syst Rev 2019; 4:CD012442. [PMID: 31016728 PMCID: PMC6479178 DOI: 10.1002/14651858.cd012442.pub2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Neuroblastoma is a rare malignant disease that primarily affects children. The tumours mainly develop in the adrenal medullary tissue, and an abdominal mass is the most common presentation. High-risk disease is characterised by metastasis and other primary tumour characteristics resulting in increased risk for an adverse outcome. The GD2 carbohydrate antigen is expressed on the cell surface of neuroblastoma tumour cells and is thus a promising target for anti-GD2 antibody-containing immunotherapy. OBJECTIVES To assess the efficacy of anti-GD2 antibody-containing postconsolidation immunotherapy after high-dose chemotherapy (HDCT) and autologous haematopoietic stem cell transplantation (HSCT) compared to standard therapy after HDCT and autologous HSCT in people with high-risk neuroblastoma. Our primary outcomes were overall survival and treatment-related mortality. Our secondary outcomes were progression-free survival, event-free survival, early toxicity, late non-haematological toxicity, and health-related quality of life. SEARCH METHODS We searched the electronic databases CENTRAL (2018, Issue 9), MEDLINE (PubMed), and Embase (Ovid) on 20 September 2018. We searched trial registries and conference proceedings on 28 October 2018. Further searches included reference lists of recent reviews and relevant articles as well as contacting experts in the field. There were no limits on publication year or language. SELECTION CRITERIA Randomised controlled trials evaluating anti-GD2 antibody-containing immunotherapy after HDCT and autologous HSCT in people with high-risk neuroblastoma. DATA COLLECTION AND ANALYSIS Two review authors independently performed study selection, abstracted data on study and participant characteristics, and assessed risk of bias and GRADE. Any differences were resolved by discussion, with third-party arbitration unnecessary. We performed analyses according to the guidelines of the Cochrane Handbook for Systematic Reviews of Interventions. We used the five GRADE considerations, that is study limitations, consistency of effect, imprecision, indirectness, and publication bias, to judge the quality of the evidence. MAIN RESULTS We identified one randomised controlled trial that included 226 people with high-risk neuroblastoma who were pre-treated with autologous HSCT. The study randomised 113 participants to receive immunotherapy including isotretinoin, granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-2, and ch14.18, a type of anti-GD2 antibody also known as dinutuximab. The study randomised another 113 participants to receive standard therapy including isotretinoin.The results on overall survival favoured the dinutuximab-containing immunotherapy group (hazard ratio (HR) 0.50, 95% confidence interval (CI) 0.31 to 0.80; P = 0.004). The results on event-free survival also favoured the dinutuximab-containing immunotherapy group (HR 0.61, 95% CI 0.41 to 0.92; P = 0.020). Randomised data on adverse events were not reported separately. The study did not report progression-free survival, late non-haematological toxicity, and health-related quality of life as separate endpoints. We graded the quality of the evidence as moderate. AUTHORS' CONCLUSIONS The evidence base favours dinutuximab-containing immunotherapy compared to standard therapy concerning overall survival and event-free survival in people with high-risk neuroblastoma pre-treated with autologous HSCT. Randomised data on adverse events are lacking, therefore more research is needed before definitive conclusions can be made regarding this outcome.
Collapse
Affiliation(s)
- Frank Peinemann
- Children's Hospital, University of ColognePediatric Oncology and HematologyKerpener Str. 62CologneGermany50937
| | - Elvira C van Dalen
- Princess Máxima Center for Pediatric OncologyHeidelberglaan 25UtrechtNetherlands3584 CS
| | - Heike Enk
- c/o Cochrane Childhood CancerAmsterdamNetherlands
| | - Godelieve AM Tytgat
- Princess Máxima Center for Pediatric OncologyHeidelberglaan 25UtrechtNetherlands3584 CS
| | | |
Collapse
|
21
|
Newman EA, Abdessalam S, Aldrink JH, Austin M, Heaton TE, Bruny J, Ehrlich P, Dasgupta R, Baertschiger RM, Lautz TB, Rhee DS, Langham MR, Malek MM, Meyers RL, Nathan JD, Weil BR, Polites S, Madonna MB. Update on neuroblastoma. J Pediatr Surg 2019; 54:383-389. [PMID: 30305231 DOI: 10.1016/j.jpedsurg.2018.09.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/09/2018] [Accepted: 09/14/2018] [Indexed: 12/15/2022]
Abstract
Neuroblastoma is an embryonic cancer arising from neural crest stem cells. This cancer is the most common malignancy in infants and the most common extracranial solid tumor in children. The clinical course may be highly variable with the possibility of spontaneous regression in the youngest patients and increased risk of aggressive disease in older children. Clinical heterogeneity is a consequence of the diverse biologic characteristics that determine patient risk and survival. This review will focus on current progress in neuroblastoma staging, risk stratification, and treatment strategies based on advancing knowledge in tumor biology and genetic characterization. TYPE OF STUDY: Review article. LEVEL OF EVIDENCE: Level II.
Collapse
Affiliation(s)
- Erika A Newman
- C.S Mott Children's Hospital, The University of Michigan Medicine, Ann Arbor, MI.
| | | | | | - Mary Austin
- Memorial Hermann Texas Medical Center, Houston, TX
| | - Todd E Heaton
- Memorial Sloan Kettering Cancer Center, NY, New York
| | | | - Peter Ehrlich
- C.S Mott Children's Hospital, The University of Michigan Medicine, Ann Arbor, MI
| | | | | | - Timothy B Lautz
- Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern University, Chicago, IL
| | | | - Max R Langham
- University of Tennessee Health Science Center and St. Jude Children's Research Hospital, Memphis, TN
| | - Marcus M Malek
- Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA
| | - Rebecka L Meyers
- Primary Children's Hospital, University of Utah, Salt Lake City, UT
| | | | - Brent R Weil
- Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA
| | | | - Mary Beth Madonna
- Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern University, Chicago, IL
| | | |
Collapse
|
22
|
Schechter T, Perez-Albuerne E, Lin TF, Irwin MS, Essa M, Desai AV, Frangoul H, Yanik G, Dupuis LL, Jacobsohn D, Kletzel M, Ranalli M, Soni S, Seif AE, Grupp S, Dvorak CC. Veno-occlusive disease after high-dose busulfan–melphalan in neuroblastoma. Bone Marrow Transplant 2018; 55:531-537. [DOI: 10.1038/s41409-018-0298-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 01/19/2023]
|
23
|
Neuroblastoma: clinical and biological approach to risk stratification and treatment. Cell Tissue Res 2018; 372:195-209. [PMID: 29572647 DOI: 10.1007/s00441-018-2821-2] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/28/2018] [Indexed: 01/15/2023]
Abstract
Neuroblastoma is the most common extra-cranial solid tumor of childhood and the most common in the first year of life. It is a unique malignancy in that infants often present with either localized or metastatic disease that can spontaneously regress without intervention while older children can succumb to the disease after months to years of arduous therapy. Given this wide range of outcomes, the International Neuroblastoma Risk Group was created to stratify patients based on presenting characteristics and tumor biology in order to guide intensity of treatment strategies. The goal has been to decrease therapy for low-risk patients to avoid long-term complications while augmenting and targeting therapies for high-risk patients to improve overall survival. The international risk stratification depends on age, stage, histology, MYCN gene amplification status, tumor cell ploidy and segmental chromosomal abnormalities. Treatment for asymptomatic low-risk patients with an estimated survival of > 98% is often observation or surgical resection alone, whereas intermediate-risk patients with an estimated survival of > 90% require moderate doses of response-adjusted chemotherapy along with resection. High-risk patients undergo multiple cycles of combination chemotherapy before surgery, followed by consolidation with myeloablative autologous hematopoietic stem cell transplantation and local radiation and finally immunotherapy with differentiation therapy as maintenance phase. With this approach, outcome for patients with neuroblastoma has improved, as the field continues to expand efforts in more targeted therapies for high-risk patients.
Collapse
|
24
|
|
25
|
Llosa NJ, Cooke KR, Chen AR, Gamper CJ, Klein OR, Zambidis ET, Luber B, Rosner G, Siegel N, Holuba MJ, Robey N, Hayashi M, Jones RJ, Fuchs E, Holdhoff M, Loeb DM, Symons HJ. Reduced-Intensity Haploidentical Bone Marrow Transplantation with Post-Transplant Cyclophosphamide for Solid Tumors in Pediatric and Young Adult Patients. Biol Blood Marrow Transplant 2017; 23:2127-2136. [PMID: 28807769 PMCID: PMC5986177 DOI: 10.1016/j.bbmt.2017.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/09/2017] [Indexed: 11/21/2022]
Abstract
High-risk, recurrent, or refractory solid tumors in pediatric, adolescent, and young adult (AYA) patients have an extremely poor prognosis despite current intensive treatment regimens. We piloted an allogeneic bone marrow transplant platform using reduced-intensity conditioning (RIC) and partially HLA-mismatched (haploidentical) related donors for this population of pediatric and AYA solid tumor patients. Sixteen patients received fludarabine, cyclophosphamide, melphalan, and low-dose total body irradiation RIC haploidentical BMT (haploBMT) followed by post-transplantation cyclophosphamide (PTCy), mycophenolate mofetil, and sirolimus. All assessable patients were full donor chimeras on day 30 with a median neutrophil recovery of 19 days and platelet recovery of 21 days. One patient (7%) exhibited secondary graft failure associated with concomitant infection. The median follow-up time was 15 months. Overall survival was 88%, 56%, and 21% at 6, 12, and 24 months, respectively. Median survival from transplant date was 14 months with a median progression-free survival 7 months. We observed limited graft-versus-host disease in 3 patients and nonrelapse mortality in 1 patient. We demonstrated that RIC haploBMT with PTCy is feasible and has acceptable toxicities in patients with incurable pediatric and AYA solid tumors; thus, this approach serves as a platform for post-transplant strategies to prevent relapse and optimize progression-free survival.
Collapse
Affiliation(s)
- Nicolas J Llosa
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland.
| | - Kenneth R Cooke
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Allen R Chen
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Christopher J Gamper
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Orly R Klein
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Elias T Zambidis
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Brandon Luber
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Gary Rosner
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Nicholas Siegel
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Mary Jo Holuba
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Nancy Robey
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Masanori Hayashi
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Richard J Jones
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Ephraim Fuchs
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Matthias Holdhoff
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - David M Loeb
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Heather J Symons
- Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| |
Collapse
|
26
|
Metrock LK, Qayed M, Simon D, Cash T, O'Connor MG, Johnson S, Esiashvili N, Katzenstein HM. Respiratory Difficulties in Children With Underlying Asthma During Immunotherapy for High-risk Neuroblastoma. J Pediatr Hematol Oncol 2017; 39:e450-e453. [PMID: 28121746 DOI: 10.1097/mph.0000000000000782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Treatment of high-risk neuroblastoma now includes antibody based antitumor immunotherapy as part of standard care. Although this therapy has resulted in dramatic improvements in survival, it is associated with significant side effects. Children with underlying respiratory issues, and in particular asthma, may be more susceptible to immunotherapy associated respiratory compromise and pulmonary complications. Early routine involvement of pulmonology care is warranted for these patients in an effort to allow maximal delivery of immunotherapy and minimize acute and long-term complications.
Collapse
Affiliation(s)
- Laura K Metrock
- *Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta †Division of Pediatric Pulmonology ∥Division of Radiation Oncology, Emory University, Atlanta, GA ‡Division of Pediatric Allergy, Immunology, and Pulmonology Medicine §Division of Pediatric Hematology/Oncology, Vanderbilt University School of Medicine, and Monroe Carell Jr Children's Hospital, Nashville, TN
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Kushner BH, LaQuaglia MP, Modak S, Wolden SL, Basu EM, Roberts SS, Kramer K, Yataghene K, Cheung IY, Cheung NKV. MYCN-amplified stage 2/3 neuroblastoma: excellent survival in the era of anti-G D2 immunotherapy. Oncotarget 2017; 8:95293-95302. [PMID: 29221128 PMCID: PMC5707022 DOI: 10.18632/oncotarget.20513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/07/2017] [Indexed: 11/25/2022] Open
Abstract
High-risk neuroblastoma (HR-NB) includes MYCN-amplified stage 2/3, but reports covering anti-GD2 immunotherapy, which recently became standard for HR-NB, do not provide details on this subset. We now report on all 20 MYCN-amplified stage 2/3 patients who received induction chemotherapy at our center during the era of consolidation with anti-GD2 antibody 3F8/ granulocyte-macrophage colony-stimulating factor (GM-CSF) (2000-2015). Early in this period, consolidation included autologous stem-cell transplantation (ASCT). Event-free survival (EFS) and overall survival (OS) were estimated using Kaplan-Meier analyses. With induction, 19/20 (95%) patients achieved complete/very good partial remission (CR/VGPR) but one had progressive disease with early death. One responder did not receive consolidation and died of relapse. Five-year post-diagnosis EFS/OS rates for all 20 patients were 72%/84%. The 18 CR/VGPR patients who received consolidation had EFS/OS 81%/94% at five years from starting 3F8/GM-CSF: 4/4 ASCT patients remained relapse-free, while 11/14 non-ASCT patients remained relapse-free and two of the three relapsed patients achieved 2nd CR (consolidated by retreatment with 3F8/GM-CSF) and remained in 2nd CR at 36+ and 95+ months post-relapse. The 14 non-ASCT patients had EFS/OS 73.5%/93% at five years from starting 3F8/GM-CSF. This subset appears to have a good prognosis with contemporary multi-modality therapy, possibly even without ASCT.
Collapse
Affiliation(s)
- Brian H Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael P LaQuaglia
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Suzanne L Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ellen M Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Karima Yataghene
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Irene Y Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
28
|
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
|
29
|
Li R, Polishchuk A, DuBois S, Hawkins R, Lee SW, Bagatell R, Shusterman S, Hill-Kayser C, Al-Sayegh H, Diller L, Haas-Kogan DA, Matthay KK, London WB, Marcus KJ. Patterns of Relapse in High-Risk Neuroblastoma Patients Treated With and Without Total Body Irradiation. Int J Radiat Oncol Biol Phys 2017; 97:270-277. [DOI: 10.1016/j.ijrobp.2016.10.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/06/2016] [Accepted: 10/31/2016] [Indexed: 11/27/2022]
|
30
|
Abstract
Neuroblastoma is the most common extracranial solid tumour occurring in childhood and has a diverse clinical presentation and course depending on the tumour biology. Unique features of these neuroendocrine tumours are the early age of onset, the high frequency of metastatic disease at diagnosis and the tendency for spontaneous regression of tumours in infancy. The most malignant tumours have amplification of the MYCN oncogene (encoding a transcription factor), which is usually associated with poor survival, even in localized disease. Although transgenic mouse models have shown that MYCN overexpression can be a tumour-initiating factor, many other cooperating genes and tumour suppressor genes are still under investigation and might also have a role in tumour development. Segmental chromosome alterations are frequent in neuroblastoma and are associated with worse outcome. The rare familial neuroblastomas are usually associated with germline mutations in ALK, which is mutated in 10-15% of primary tumours, and provides a potential therapeutic target. Risk-stratified therapy has facilitated the reduction of therapy for children with low-risk and intermediate-risk disease. Advances in therapy for patients with high-risk disease include intensive induction chemotherapy and myeloablative chemotherapy, followed by the treatment of minimal residual disease using differentiation therapy and immunotherapy; these have improved 5-year overall survival to 50%. Currently, new approaches targeting the noradrenaline transporter, genetic pathways and the tumour microenvironment hold promise for further improvements in survival and long-term quality of life.
Collapse
|
31
|
Kushner BH, Ostrovnaya I, Cheung IY, Kuk D, Modak S, Kramer K, Roberts SS, Basu EM, Yataghene K, Cheung NKV. Lack of survival advantage with autologous stem-cell transplantation in high-risk neuroblastoma consolidated by anti-GD2 immunotherapy and isotretinoin. Oncotarget 2016; 7:4155-66. [PMID: 26623730 PMCID: PMC4826196 DOI: 10.18632/oncotarget.6393] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/09/2015] [Indexed: 11/25/2022] Open
Abstract
Since 2003, high-risk neuroblastoma (HR-NB) patients at our center received anti-GD2 antibody 3F8/GM-CSF + isotretinoin – but not myeloablative therapy with autologous stem-cell transplantation (ASCT). Post-ASCT patients referred from elsewhere also received 3F8/GM-CSF + isotretinoin. We therefore accrued a study population of two groups treated during the same period and whose consolidative therapy, aside from ASCT, was identical. We analyzed patients enrolled in 1st complete/very good partial remission (CR/VGPR). Their event-free survival (EFS) and overall survival (OS) were calculated from study entry. Large study size allowed robust statistical analyses of key prognosticators including MYCN amplification, minimal residual disease (MRD), FCGR2A polymorphisms, and killer immunoglobulin-like receptor genotypes of natural killer cells. The 170 study patients included 60 enrolled following ASCT and 110 following conventional chemotherapy. The two cohorts had similar clinical and biological features. Five-year rates for ASCT and non-ASCT patients were, respectively: EFS 65% vs. 51% (p = .128), and OS 76% vs. 75% (p = .975). In multivariate analysis, ASCT was not prognostic and only MRD-negativity after two cycles of 3F8/GM-CSF correlated with significantly improved EFS and OS. Although a trend towards better EFS is seen with ASCT, OS is near identical. Cure rates may be similar, as close surveillance detects localized relapse and effective salvage treatments are applied. ASCT may not be needed to improve outcome when anti-GD2 immunotherapy is used for consolidation after dose-intensive conventional chemotherapy.
Collapse
Affiliation(s)
- Brian H Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Irene Y Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Deborah Kuk
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ellen M Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Karima Yataghene
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
32
|
Toxicities of busulfan/melphalan versus carboplatin/etoposide/melphalan for high-dose chemotherapy with stem cell rescue for high-risk neuroblastoma. Bone Marrow Transplant 2016; 51:1204-10. [DOI: 10.1038/bmt.2016.84] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 02/09/2016] [Accepted: 02/28/2016] [Indexed: 12/21/2022]
|
33
|
Mora J. Dinutuximab for the treatment of pediatric patients with high-risk neuroblastoma. Expert Rev Clin Pharmacol 2016; 9:647-53. [PMID: 26934530 DOI: 10.1586/17512433.2016.1160775] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Neuroblastoma (NB) is the most common extra cranial solid tumor of childhood, with 60% of patients presenting with high risk (HR) NB by means of clinical, pathological and biological features. The 5-year survival rate for HR-NB remains below 40%, with the majority of patients suffering relapse from chemorefractory tumor. Immunotherapy is the main strategy against minimal residual disease and clinical experience has mostly focused on monoclonal antibodies (MoAb) against the glycolipid disialoganglioside GD2. Three anti-GD2 antibodies have been tested in the clinic including murine 14G2a, human-mouse chimeric ch14.18 and 3F8. Anti-GD2 MoAb induces cellular cytoxicity against NB and is most effective when effector cells like natural killer cells, granulocytes and macrophages are amplified by cytokines. The combination of cytokines IL-2 and GM-CSF with the anti-GD2 MoAb ch14.18 (Dinutuximab) has shown a significant improvement in outcome for HR-NB. The FDA and EMA approved dinutuximab (Unituxin(R)) in 2015 for the treatment of patients with HR-NB who achieved at least a partial response after multimodality therapy.
Collapse
Affiliation(s)
- Jaume Mora
- a Department of Pediatric Onco-Hematology and Developmental Tumor Biology Laboratory , Hospital Sant Joan de Déu, Passeig Sant Joan de Déu , Barcelona , Spain
| |
Collapse
|
34
|
Pasqualini C, Dufour C, Goma G, Raquin MA, Lapierre V, Valteau-Couanet D. Tandem high-dose chemotherapy with thiotepa and busulfan–melphalan and autologous stem cell transplantation in very high-risk neuroblastoma patients. Bone Marrow Transplant 2015; 51:227-31. [DOI: 10.1038/bmt.2015.264] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/24/2015] [Accepted: 09/29/2015] [Indexed: 11/09/2022]
|
35
|
Garaventa A. High risk neuroblastoma: small steps towards cure. Pediatr Blood Cancer 2014; 61:964-5. [PMID: 24535945 DOI: 10.1002/pbc.24991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 01/17/2014] [Indexed: 01/27/2023]
|
36
|
Desai AV, Kavcic M, Huang YS, Herbst N, Fisher BT, Seif AE, Li Y, Hennessy S, Aplenc R, Bagatell R. Establishing a high-risk neuroblastoma cohort using the Pediatric Health Information System Database. Pediatr Blood Cancer 2014; 61:1129-1131. [PMID: 24616331 PMCID: PMC4323158 DOI: 10.1002/pbc.24930] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 12/13/2013] [Indexed: 11/11/2022]
Abstract
International Classification of Diseases, 9th Revision (ICD-9) code(s) for neuroblastoma do not exist, preventing identification of these patients in administrative databases. To overcome this challenge, a three-step algorithm, using ICD-9 codes, exclusion criteria, and manual review of chemotherapy billing data, was utilized to assemble a high-risk neuroblastoma cohort (n = 952) from the Pediatric Health Information System (PHIS) Database and validated at a single institution [sensitivity 89.1%; positive predictive value (PPV) 96.1%]. This cohort provides a data source for future comparative effectiveness and clinical epidemiology studies in high-risk neuroblastoma patients.
Collapse
Affiliation(s)
- Ami V. Desai
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA,Center for Pharmacoepidemiology Research and Training, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Marko Kavcic
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yuan-Shung Huang
- Center for Pediatric Clinical Effectiveness, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Nicole Herbst
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Brian T. Fisher
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA,Center for Pediatric Clinical Effectiveness, The Children's Hospital of Philadelphia, Philadelphia, PA,Division of Infectious Diseases, The Children's Hospital of Philadelphia, Philadelphia, PA,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Alix E Seif
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Yimei Li
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Sean Hennessy
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA,Center for Pharmacoepidemiology Research and Training, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Richard Aplenc
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA,Center for Pharmacoepidemiology Research and Training, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA,Center for Pediatric Clinical Effectiveness, The Children's Hospital of Philadelphia, Philadelphia, PA,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Rochelle Bagatell
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
37
|
Treatment of high-risk neuroblastoma in children: recent clinic trial results. ACTA ACUST UNITED AC 2013. [DOI: 10.4155/cli.13.90] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
38
|
Kreissman SG, Seeger RC, Matthay KK, London WB, Sposto R, Grupp SA, Haas-Kogan DA, Laquaglia MP, Yu AL, Diller L, Buxton A, Park JR, Cohn SL, Maris JM, Reynolds CP, Villablanca JG. Purged versus non-purged peripheral blood stem-cell transplantation for high-risk neuroblastoma (COG A3973): a randomised phase 3 trial. Lancet Oncol 2013; 14:999-1008. [PMID: 23890779 DOI: 10.1016/s1470-2045(13)70309-7] [Citation(s) in RCA: 215] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Myeloablative chemoradiotherapy and immunomagnetically purged autologous bone marrow transplantation has been shown to improve outcome for patients with high-risk neuroblastoma. Currently, peripheral blood stem cells (PBSC) are infused after myeloablative therapy, but the effect of purging is unknown. We did a randomised study of tumour-selective PBSC purging in stem-cell transplantation for patients with high-risk neuroblastoma. METHODS Between March 16, 2001, and Feb 24, 2006, children and young adults (<30 years) with high-risk neuroblastoma were randomly assigned at diagnosis by a web-based system (in a 1:1 ratio) to receive either non-purged or immunomagnetically purged PBSC. Randomisation was done in blocks stratified by International Neuroblastoma Staging System stage, age, MYCN status, and International Neuroblastoma Pathology classification. Patients and treating physicians were not masked to treatment assignment. All patients were treated with six cycles of induction chemotherapy, myeloablative consolidation, and radiation therapy to the primary tumour site plus meta-iodobenzylguanidine avid metastases present before myeloablative therapy, followed by oral isotretinoin. PBSC collection was done after two induction cycles. For purging, PBSC were mixed with carbonyl iron and phagocytic cells removed with samarium cobalt magnets. Remaining cells were mixed with immunomagnetic beads prepared with five monoclonal antibodies targeting neuroblastoma cell surface antigens and attached cells were removed using samarium cobalt magnets. Patients underwent autologous stem-cell transplantation with PBSC as randomly assigned after six cycles of induction therapy. The primary endpoint was event-free survival and was analysed by intention-to-treat. The trial is registered with ClinicalTrials.gov, number NCT00004188. FINDINGS 495 patients were enrolled, of whom 486 were randomly assigned to treatment: 243 patients to receive non-purged PBSC and 243 to received purged PBSC. PBSC were collected from 229 patients from the purged group and 236 patients from the non-purged group, and 180 patients from the purged group and 192 from the non-purged group received transplant. 5-year event-free survival was 40% (95% CI 33-46) in the purged group versus 36% (30-42) in the non-purged group (p=0·77); 5-year overall survival was 50% (95% CI 43-56) in the purged group compared with 51% (44-57) in the non-purged group (p=0·81). Toxic deaths occurred in 15 patients during induction (eight in the purged group and seven in the non-purged group) and 12 during consolidation (eight in the purged group and four in the non-purged group). The most common adverse event reported was grade 3 or worse stomatitis during both induction (87 of 242 patients in the purged group and 93 of 243 patients in the non-purged group) and consolidation (131 of 177 in the purged group vs 145 of 191 in the non-purged group). Serious adverse events during induction were grade 3 or higher decreased cardiac function (four of 242 in the purged group and five of 243 in the non-purged group) and elevated creatinine (five of 242 in the purged group and six of 243 non-purged group) and during consolidation were sinusoidal obstructive syndrome (12 of 177 in the purged group and 17 of 191 in the non-purged group), acute vascular leak (11 of 177 in the purged group and nine of 191 in the non-purged group), and decreased cardiac function (one of 177 in the purged group and four of 191 in the non-purged group). INTERPRETATION Immunomagnetic purging of PBSC for autologous stem-cell transplantation did not improve outcome, perhaps because of incomplete purging or residual tumour in patients. Non-purged PBSC are acceptable for support of myeloablative therapy of high-risk neuroblastoma.
Collapse
|