Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2249 2022-06-04 17:02:00 |
2 Format correction Meta information modification 2249 2022-06-06 04:07:50 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Mora, J. Autologous Stem-Cell Transplantation for High-Risk Neuroblastoma. Encyclopedia. Available online: https://encyclopedia.pub/entry/23726 (accessed on 27 July 2024).
Mora J. Autologous Stem-Cell Transplantation for High-Risk Neuroblastoma. Encyclopedia. Available at: https://encyclopedia.pub/entry/23726. Accessed July 27, 2024.
Mora, Jaume. "Autologous Stem-Cell Transplantation for High-Risk Neuroblastoma" Encyclopedia, https://encyclopedia.pub/entry/23726 (accessed July 27, 2024).
Mora, J. (2022, June 04). Autologous Stem-Cell Transplantation for High-Risk Neuroblastoma. In Encyclopedia. https://encyclopedia.pub/entry/23726
Mora, Jaume. "Autologous Stem-Cell Transplantation for High-Risk Neuroblastoma." Encyclopedia. Web. 04 June, 2022.
Autologous Stem-Cell Transplantation for High-Risk Neuroblastoma
Edit

Curing high-risk neuroblastoma (HR-NB) is a challenging endeavor, which involves the optimal application of several therapeutic modalities.  High-dose therapy transformed clinical practice, legislation, and public health policy, and it drove a two-decade period of entrepreneurial oncology. However, no ASCT strategies remain for any solid tumor indication in adults. As with most solid malignancies, higher dosing of cytotoxic agents has not resulted in a clear benefit in survival for HR-NB patients, whereas the long-term toxicity has been well defined. Fortunately, novel approaches such as anti-GD2 immunotherapy have demonstrated a significant survival benefit with a much less adverse impact on the patient’s wellbeing. 

neuroblastoma autologous stem-cell transplant myeloablative therapy hematopoietic stem cells

1. Hematopoietic Stem-Cell Transplant: Historical Review

The era of hematopoietic stem-cell transplantation began after the first atomic bomb explosion, with the pioneering observations by Leon Jacobson et al. in 1949. Mice were protected from the lethal effects of ionizing radiation on bone marrow (BM) by protecting their spleens with lead [1]. Subsequently, Lorenz and colleagues showed that surviving lethal radiation could also be achieved by infusions of BM [2]. The discovery showing that all hematopoietic stem cells arise from pluripotent transplantable stem cells led to the use of BM transplantation as a treatment for hematological malignancies. In the years 1959–1965, three landmark papers were published that described the first experiences with transplantation of BM into leukemia patients [3][4][5]. In the first study, two patients with acute lymphoblastic leukemia received high-dose total body irradiation (TBI), followed by BM grafts from twin siblings [3]. This was the first example of patients given supralethal irradiation who showed hematological recovery.
The second half of the 1960s provided a significant evolution of high-dose myeloablative conditioning regimens. These consisted of maximally tolerated doses of TBI and/or chemotherapeutic agents such as cyclophosphamide, which not only served to kill cancer cells, but also suppressed the immune system of the host so that BM grafts would not be rejected [6]. The 1980s and 1990s showed how successful stem-cell transplantation could be safely applied with refined supportive care and broader application using alternative graft donors and sources. T-cell depletion and immunosuppressive agents were shown to minimize graft-versus-host disease. Drugs such as busulfan were identified as alternatives to TBI [7]. In 1990, E. Donnall Thomas won a Nobel Prize for his pioneering research on stem-cell transplantation for the treatment of hematopoietic diseases, laying the foundations of modern stem-cell replacement therapy for human diseases including cancer. In the year 2000 alone, approximately 18,000 patients worldwide were treated with myeloablative approaches, a strategy accepted as the standard of care for a range of specific indications.

2. The Basic Principle of Transplantation Expanded to Solid Tumors: More Is Not Always Better

With the improvement of supportive therapy and sophistication in transplant-related techniques, treatment intensity for cancer became highly regarded in the 1990s partly because of the lessons from Hodgkin’s lymphoma. Higher cumulative dosage or higher dose intensity (DI) could be applied since myelotoxicity was no longer limiting in the face of stem-cell rescue. Investigative trials assumed that tumor response correlated positively with the dosage or intensity of drug(s) administered, and that this response would translate into improved overall survival (OS). Among the many preclinical studies, a steep log–linear dose–response curve was consistently documented for some cytotoxic agents (Figure 1) [8]. Increased chemotherapy drug dosage correlated with a log-fold increase in cell kill. It was postulated that, if myelotoxicity (the major dose-limiting toxicity of chemotherapeutic agents) could be reversed by stem-cell rescue, cure might be possible by increasing the dose or dose intensity of treatment. The principle supported autologous stem-cell transplant (ASCT) strategies. This linear–log relationship between chemotherapy agent dose and tumor cell killing suggested that, if the drug dose is increased without increasing toxicity to the patient, then a multiple log increase in tumor cell death would be observed [9][10]. Few prior clinical studies were available but suggested a positive relationship [9][10]. In addition to the limitation of extrapolating in vitro studies to human beings, there are underlying assumptions of this “cure by dosing” hypothesis, i.e., that the dose–response curve in patients is indeed linear, that drug resistance is overcome by increasing treatment intensity, that myelotoxicity is the only dose-limiting concern, and that response will translate into improved survival. Decades later, this hypothesis has been tested in multiple cancer types, and its success has been revealed to be disease-specific and qualified.
Figure 1. Dose–response curve showing log-fold increase in cell killing (Y-axis = survivor cells) with increase in dosage (X-axis) as the basis for high-dose therapy strategies. Adapted from original in Frei E III, Canellos GP: Dose: A critical factor in cancer chemotherapy. Am J Med 69:585–594, 1980. [9].
Chemoresistance, be it intrinsic or acquired, is a key factor impacting response and the ability to achieve a cure. In chemotherapy-sensitive solid cancers, a high initial response rate to chemotherapy is commonly followed by subsequent relapse and resistance to further treatment. Tumors fitting this model usually show a dose–response behavior to drugs given in the conventional dose range. In principle, the drugs causing this effect should be appropriate for further dose escalation, with dose-limiting toxicity (DLT) primarily affecting the hematopoietic organ rescuable with hematopoietic stem cells. Ideally, their nonhematologic effects should be limited, and morbidity and mortality should be minimal. Inherent to this approach is the assumption that resistance can be overcome with the increase in dose, which can be achieved with stem-cell support [11]. Alkylating agents are the most appropriate agents for dose escalation [10]. There was evidence from cell lines and animal models of a steep dose–response curve for these compounds [10], and resistance to alkylating agents had been overcome by a 5–10-fold increase in dose. Their DLT was primarily hematologic, and nonhematologic toxicities at high doses were non-overlapping. In contrast, anthracyclines are not suitable for dose escalation not only because of their cardiac and mucosal toxicities, but also because their cytotoxicity in preclinical studies leveled off. Antimetabolites and vinca alkaloids are also not suitable agents for high-dose therapy (HDT) not only because of their non-marrow toxicity limiting significant escalation, but also because their dose–response curves in vitro and in vivo are not linear [10]. Dose–response curves for most chemotherapy agents were determined using in vitro and in animal models, with tumor cell killing being directly proportional to both dose and duration of exposure (Figure 1, red line). Importantly, the steepness of the dose–response curve for most drugs showed a distinct plateau above which more drug does not appear to kill more tumor cells (Figure 1, green line) [7]. Furthermore, in humans, it is yet mostly unknown whether a plateau (Figure 1, green line) is reached when drugs are administered at the maximal tolerated dose (MTD).
The concept of HDT refers to pulsed chemotherapy, which achieves high peak drug concentrations that theoretically increase the overall exposure of tumor cells to supratherapeutic levels of cytotoxic drugs [12]. The concept of “dose intensity” (DI), however, is not the same as HDT. DI is defined as the amount of drug administered per unit of time (e.g., mg/m2/week). A dose-intensive regimen may or may not yield high peak concentrations (HDT). Continuous administration of cyclophosphamide might be quite dose-intensive, but it could be associated with lower peak concentrations and less acute toxicity than a similarly dose-intensive, pulsed, high-dose regimen. Different trials have shown the importance of dose intensity in the treatment of small round cell tumors including sarcomas [13][14] and neuroblastomas [15].
Myelosuppression associated with cytotoxic therapy often limited the amount of drug that could be administered. This limitation was overcome to some degree with the use of hematopoietic cytokines such as granulocyte colony-stimulating factor (G-CSF) and techniques to support hematopoiesis such as autologous peripheral blood stem cells and/or BM. Prospective studies in adults with solid tumors showed that dose escalation with cytokine support did not increase cure rates in patients with chemotherapy-responsive solid tumors (testis cancer, small-cell lung cancer (SCLC), advanced ovarian cancer, non-Hodgkin’s lymphoma, or bladder cancer [16]). The many reasons explaining this lack of benefit are as follows: (1) cytotoxic agents produced toxicity to nonhematopoietic tissues that limited the capacity to escalate doses further; (2) patients received extensive prior treatments that resulted in impairment of the BM function sufficient to blunt the response to cytokines; (3) the use of CSFs did not allow dose escalation beyond twofold higher than conventional doses.
Many clinical trials involving a variety of adult solid tumors were performed in the 1990s testing myeloablative therapy with BM transplant [11]. For the most common tumors (breast, ovary, and SCLC) compelling evidence to support the use of myeloablative chemotherapy in standard patient management did not emerge. Randomized data comparing standard therapy to HDT was found unconvincing. In breast cancer, eight randomized trials were reported, and the data did not support the use of HDT in breast cancer. In SCLC, insufficient evidence supported the change in practice. Selection bias was found to account for favorable results in uncontrolled studies in these tumors, as many authors pointed out by reviewing outcomes of HDT candidates who received only standard therapy [11]. HDT was ultimately determined to be ineffective and to have significantly greater toxicity. From its birth in the 1980s to its abandonment in the late 1990s, HDT transformed clinical practice, legislation, and public health policy, as well as drove a long period of entrepreneurial oncology. It also gave rise to one of the most serious cases of research misconduct of the 20th century [17]. Today, no ASCT strategies remain for any solid tumor indication in adults. For pediatric tumors, except for specific subgroups of lymphomas and germ-cell tumors, no other extracranial solid tumor indication of ASCT has been established as standard management for high-risk cases. ASCT strategies have failed to show benefit for Ewing sarcomas [18], Wilms tumors [19], non-rhabdomyosarcoma soft-tissue sarcomas [20], rhabdomyosarcomas [21], osteosarcomas [22], hepatoblastomas [23], and rhabdoid tumors [24].

3. Anti-GD2 Immunotherapy and ASCT

Fortunately, chemo-resistant NB is highly responsive to treatments far less toxic than myeloablative therapy, namely, anti-GD2 antibody + granulocyte–macrophage colony-stimulating factor (GM-CSF) [25] ± low-dose chemotherapy [26]. It is noteworthy that two anti-GD2 mAbs—dinutuximab and naxitamab—have received approval from the Food and Drug Administration in the USA.
Immunotherapy targeting GD2 emerged with the aim to eliminate chemotherapy-refractory disease, similar to HDT strategies, in the 1990s and early 2000s. The murine monoclonal antibody (mAb) 3F8 underwent extensive preclinical testing and, when administered with GM-CSF, led to significant responses in patients with refractory NB [27]. Clinical trials of anti-GD2 mAb therapy demonstrated significantly improved EFS and, for the first time, OS when used after major responses to standard induction therapy and ASCT, leading to regulatory approval in the United States and Europe [28][29]. The problem is that the two strategies (ASCT and anti-GD2 immunotherapy) targeting minimal residual disease (MRD) have not been formally evaluated independently.
The only randomized controlled trial involved HR-NB patients who received either anti-GD2 mAb dinutuximab-containing immunotherapy or cis-RA after ASCT (ANBL0032) [28]. The primary objective was an intention-to-treat comparison. At 2 years, the estimated EFS was 66% in the dinutuximab-containing immunotherapy arm and 46% in the standard therapy group. A subsequent report provided longer follow-up data on survival [30]. The survival difference between the treatment groups remained statistically significant with a 5 year EFS of 56.6% for patients randomized to triple (dinutuximab, IL-2, and GM-CSF) immunotherapy versus 46.1% for those randomized to cis-RA only (p = 0.042). The 5 year OS was 73.2% versus 56.6% for triple immunotherapy versus cis-RA only patients, respectively (p = 0.045). The role of anti-GD2 immunotherapy after HDT and ASCT was clearly shown to be relevant for the survival of patients with HR-NB, and anti-GD2 immunotherapy was then adopted worldwide as the standard of care for all HR-NB patients in remission post ASCT. These results, however, question even further the role of HDT and ASCT in the era of anti-GD2 immunotherapy given that the landmark CCG-3891 study, as described earlier [31], did not show a benefit in OS when tested without anti-GD2 mAb. More recently, a detailed analysis by the SIOPEN of risk factors and the relevance for each of the sequential therapies within the HR-NB multimodal treatment program (chemotherapy, ASCT, and immunotherapy), revealed that the impact of immunotherapy on EFS is significantly influenced by stage and pattern of metastases, but only borderline related to ASCT [32].
Initial evidence of the superiority of anti-GD2 immunotherapy over HDT and ASCT for the management of patients with HR-NB came from the MSKCC group. This group had a great interest in and experience with transplant during the HDT era [33][34][35][36][37][38] but discontinued it in 2003 when their analyses of sequential studies with or without ASCT and/or mAb 3F8 showed no survival benefit [39]. In 2016, they first reported similar outcomes for HR-NB patients whose consolidative therapy of first CR/VGPR included 3F8/GM-CSF + isotretinoin with or without prior HDT and ASCT [40][41]. They were able to accrue a large cohort of more than 100 patients treated without ASCT, a remarkable group of patients given that ASCT has remained in all major studies since the year 2000 and continues today. Independently, another group of researchers were able to recruit a cohort of non-ASCT patients (n = 54) adding to that from MSKCC, increasing the population of HR-NB patients managed in the current era without ASCT and showing no difference in survival [42]. Recent advances in anti-GD2 immunotherapy, such as more potent mAbs [43], the lesser use of toxic IL-2 [29], and the systematic use of GM-CSF, may account for the current lack of survival advantage of ASCT. With all this evidence (or lack of it), discontinuing ASCT for HR-NB would be the logical step forward and consistent with the general consensus among (pediatric and adult) oncologists that this highly toxic treatment from the 20th century should no longer be recommended as long as anti-GD2 immunotherapy is broadly available to all children affected by HR-NB.

References

  1. Jacobson, L.O.; Marks, E.K.; Robson, M.J.; Gaston, E.O.; Zirkle, R.E. Effect of spleen protection on mortality following X-irradiation. J. Lab. Clin. Med. 1949, 34, 1538–1543.
  2. Lorenz, E.; Uphoff, D.; Reid, T.R.; Shelton, E. Modification of irradiation injury in mice and guinea pigs by bone marrow injections. J. Natl. Cancer Inst. 1951, 12, 197–201.
  3. Thomas, E.D.; Lochte, H.L., Jr.; Cannon, J.H.; Sahler, O.D.; Ferrebee, J.W. Supralethal whole body irradiation and isologous marrow transplantation in man. J. Clin. Investig. 1959, 38, 1709–1716.
  4. Mathé, G.; Amiel, J.L.; Schwarzenberg, L.; Catton, A.; Schneider, M. Adoptive immunotherapy of acute leukemia: Experimental and clinical results. Cancer Res. 1965, 25, 1525–1531.
  5. McGovern, J.J., Jr.; Russel, P.S.; Atkins, L.; Webster, E.W. Treatment of terminal leukemic relapse by total-body irradiation and intravenous infusion of stored autologous bone marrow obtained during remission. N. Engl. J. Med. 1959, 260, 675–683.
  6. Santos, G.W.; Sensenbrenner, L.L.; Burke, P.J.; Colvin, M.; Owens Jr, A.H.; Bias, W.B.; Slavin, R.E. Marrow transplantation in man following cyclophosphamide. Transplant. Proc. 1971, 3, 400–404.
  7. Santos, G.W. Busulfan (Bu) and cyclophosphamide (Cy) or marrow transplantation. Bone Marrow Transplant. 1989, 4, 236–239.
  8. Frei, E., III; Teicher, B.A.; Holden, S.A.; Cathcart, K.N.; Wang, Y.Y. Preclinical studies and clinical correlation of the effect of alkylating dose. Cancer Res. 1988, 48, 6417–6423.
  9. Frei, E.J.; Canellos, G.P. Dose: A critical factor in cancer chemotherapy. Am. J. Med. 1980, 69, 585–594.
  10. Frei, E., III; Antman, K.; Teicher, B.; Eder, P.; Schnipper, L. Bone marrow autotransplantation for solid tumors prospects. J. Clin. Oncol. 1989, 7, 515–526.
  11. MacNeil, M.; Eisenhauer, E.A. Adults: High-dose chemotherapy: Is it standard management for any common solid tumor? Ann. Oncol. 1999, 10, 1145–1161.
  12. Livingston, R.B. Dose intensity and high dose therapy. Cancer 1994, 74, 1177–1182.
  13. Marina, N.M.; Pappo, A.S.; Parham, D.M.; Cain, A.M.; Rao, B.N.; Poquette, C.A.; Pratt, C.B.; Greenwald, C.; Meyer, W.H. Chemotherapy dose-intensification for pediatric patients with Ewing’s family of tumors and desmoplastic small round-cell tumors: A feasibility study at St. Jude Children’s Research Hospital. J. Clin. Oncol. 1999, 17, 180–190.
  14. Whelan, J.; Khan, A.; Sharma, A.; Rothermundt, C.; Dileo, P.; Michelagnoli, M.; Seddon, B.; Strausss, S. Interval compressed vincristine, doxorubicin, cyclophosphamide alternating with ifosfamide, etoposide in patients with advanced Ewing’s and other Small Round Cell Sarcomas. Clin. Sarcoma Res. 2012, 21, 12.
  15. Cheung, N.-K.V.; Heller, G. Chemotherapy Dose Intensity Correlates Strongly With Response, Median Survival, and Median Progression-Free Survival in Metastatic Neuroblastoma. J. Clin. Oncol. 1991, 9, 1050–1058.
  16. Savarese, D.M.F.; Hsieh, C.-C.; Stewart, F.M. Clinical Impact of Chemotherapy Dose Escalation in Patients With Hematologic Malignancies and Solid Tumors. J. Clin. Oncol. 1997, 15, 2981–2995.
  17. Rettig, R.A.; Jacobson, P.D.; Farquhar, C.M.; Aubry, W.M. False Hope: Bone Marrow Transplantation for Breast Cancer; Oxford University Press: London, UK, 2007; ISBN 978-0195187762.
  18. Haveman, L.M.; van Ewijk, R.; van Dalen, E.C.; Breunis, W.B.; Kremer, L.C.; van den Berg, H.; Dirksen, U.; Merks, J.H. High-dose chemotherapy followed by autologous haematopoietic cell transplantation for children, adolescents, and young adults with primary metastatic Ewing sarcoma. Cochrane Database Syst. Rev. 2021, 9, CD011405.
  19. Delafoy, M.; Verschuur, A.; Scheleirmacher, G.; Tabone, M.D.; Sudour-Bonnange, H.; Thébaud, E.; Freycon, C.; Notz-Carrère, A.; Boulanger, C.; Pellier, I.; et al. High-dose chemotherapy followed by autologous stem cell rescue in Wilms tumors: French report on toxicity and efficacy. Pediatric Blood Cancer 2022, 69, e29431.
  20. Peinemann, F.; Enk, H.; Smith, L.A. Autologous hematopoietic stem cell transplantation following high-dose chemotherapy for nonrhabdomyosarcoma soft tissue sarcomas. Cochrane Database Syst. Rev. 2017, 4, CD008216.
  21. Peinemann, F.; Kröger, N.; Bartel, C.; Grouven, U.; Pittler, M.; Erttmann, R.; Kulig, M. High-dose chemotherapy followed by autologous stem cell transplantation for metastatic rhabdomyosarcoma--a systematic review. PLoS ONE 2011, 6, e17127.
  22. Sauerbrey, A.; Bielack, S.; Kempf-Bielack, B.; Zoubek, A.; Paulussen, M.; Zintl, F. High-dose chemotherapy (HDC) and autologous hematopoietic stem cell transplantation (ASCT) as salvage therapy for relapsed osteosarcoma. Bone Marrow Transplant. 2001, 27, 933–937.
  23. Häberle, B.; Maxwell, R.; von Schweinitz, D.; Schmid, I. High Dose Chemotherapy with Autologous Stem Cell Transplantation in Hepatoblastoma does not Improve Outcome. Results of the GPOH Study HB99. Klin. Pädiatrie 2019, 231, 283–290.
  24. Furtwängler, R.; Kager, L.; Melchior, P.; Rübe, C.; Ebinger, M.; Nourkami-Tutdibi, N.; Niggli, F.; Warmann, S.; Hubertus, J.; Amman, G.; et al. High-dose treatment for malignant rhabdoid tumor of the kidney: No evidence for improved survival-The Gesellschaft für Pädiatrische Onkologie und Hämatologie (GPOH) experience. Pediatric Blood Cancer 2018, 65.
  25. Kushner, B.H.; Cheung, I.Y.; Modak, S.; Basu, E.M.; Roberts, S.S.; Cheung, N.-K. Humanized 3F8 Anti-GD2 Monoclonal Antibody Dosing with Granulocyte-Macrophage Colony-Stimulating Factor in Pa-tients With Resistant Neuroblastoma: A Phase 1 Clinical Trial. JAMA Oncol. 2018, 4, 1729–1735.
  26. Mody, R.; Naranjo, A.; Van Ryn, C.; Yu, A.L.; London, W.B.; Shulkin, B.L.; Parisi, M.T.; Servaes, S.-E.-N.; Diccianni, M.B.; Sondel, P.M.; et al. Irinotecan-temozolomide with temsirolimus or dinutuximab in children with refractory or relapsed neuroblastoma (COG ANBL1221): An open-label, randomised, phase 2 trial. Lancet Oncol. 2017, 18, 946–957.
  27. Kushner, B.H.; Kramer, K.; Cheung, N.-K.V. Phase II trial of the anti-G(D2) monoclonal antibody 3F8 and granulocyte macrophage colony-stimulating factor for neuro-blastoma. J. Clin. Oncol. 2001, 19, 4189–4194.
  28. Yu, A.L.; Gilman, A.L.; Ozkaynak, M.F.; London, W.B.; Kreissman, S.G.; Chen, H.X.; Smith, M.; Anderson, B.; Villablanca, J.G.; Matthay, K.K.; et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N. Engl. J. Med. 2010, 363, 1324–1334.
  29. Ladenstein, R.; Pötschger, U.; Valteau-Couanet, D.; Luksch, R.; Castel, V.; Yaniv, I.; Laureys, G.; Brock, P.; Michon, J.M.; Owens, C.; et al. Interleukin 2 with anti-GD2 antibody ch14.18/CHO (dinutuximab beta) in patients with high-risk neuroblastoma (HR-NBL1/SIOPEN): A multicentre, randomised, phase 3 trial. Lancet Oncol. 2018, 19, 1617–1629.
  30. Yu, A.L.; Gilman, A.L.; Ozkaynak, M.F.; Naranjo, A.; Diccianni, M.B.; Gan, J.; Hank, J.A.; Batova, A.; London, W.B.; Tenney, S.C.; et al. Long-Term Follow-up of a Phase III Study of ch14.18 (Dinutuximab) þ Cytokine Immunotherapy in Children with High-Risk Neuroblastoma: COG Study ANBL0032. Clin. Cancer Res. 2021, 27, 2179–2189.
  31. Matthay, K.K.; Villablanca, J.G.; Seeger, R.C.; Stram, D.O.; Harris, R.E.; Ramsay, N.K.; Swift, P.; Shimada, H.; Black, C.T.; Brodeur, G.M.; et al. Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. Children’s Cancer Group. N. Engl. J. Med. 1999, 341, 1165–1173.
  32. Ladenstein, R.; Pötschger, U.; Valteau-Couanet, D.; Luksch, R.; Castel, V.; Ash, S.; Laureys, G.; Brock, P.; Michon, J.M.; Owens, C.; et al. Investigation of the Role of Dinutuximab Beta-Based Immunotherapy in the SIOPEN High-Risk Neuroblastoma 1 Trial (HR-NBL1). Cancers 2020, 12, 309.
  33. Kushner, B.H.; O’Reilly, R.J.; Mandell, L.R.; Gulati, S.C.; LaQuaglia, M.; Cheung, N.K. Myeloablative combination chemotherapy without total body irradiation for neuroblastoma. J. Clin. Oncol. 1991, 9, 274–279.
  34. Kushner, B.H.; Gulati, S.C.; O’Reilly, R.J.; Heller, G.; Cheung, N.-K.V. Autografting with bone marrow exposed to multiple courses of very high dose cyclophosphamide in vivo and to 4-hydroperoxy-cyclophosphamide in vitro. Med. Pediatric Oncol. 1990, 18, 454–458.
  35. Kushner, B.H.; Hadju, S.I.; Gulati, S.C.; Erlandson, R.A.; Exelby, P.R.; Lieberman, P.H. Extracranial primitive neuroectodermal tumors: The Memorial Sloan-Kettering Cancer Center experience. Cancer 1991, 67, 1825–1829.
  36. Kushner, B.H.; Gulati, S.C.; Kwon, J.H.; O’Reilly, R.J.; Exelby, P.R.; Cheung, N.-K.V. High-dose melphalan with 6-hydroxydopamine-purged autologous bone marrow transplantation for poor-risk neuroblas-toma. Cancer 1991, 68, 242–247.
  37. Kushner, B.H.; Cheung, N.-K.V.; Kramer, K.; Dunkel, I.J.; Calleja, E.; Boulad, F. Topotecan combined with myeloablative doses of thiotepa and carboplatin for neuroblastoma, brain tumors, and other poor-risk solid tumors in children and young adults. Bone Marrow Transplant. 2001, 28, 551–556.
  38. Kushner, B.H.; Kramer, K.; Modak, S.; Kernan, N.A.; Reich, L.M.; Danis, K.; Cheung, N.-K.V. Topotecan, thiotepa, and carboplatin for neuroblastoma: Failure to prevent relapse in the central nervous system. Bone Marrow Transplant. 2006, 37, 271–276.
  39. Cheung, N.-K.V.; Cheung, I.Y.; Kushner, B.H.; Ostrovnaya, I.; Kramer, K.; Modak, S. Murine anti-GD2 monoclonal antibody 3F8 combined with granulocyte-macrophage colony stimulating factor and 13-cis-retinoic acid in high-risk patients with stage 4 neuroblastoma in first remission. J. Clin. Oncol. 2012, 30, 3264–3270.
  40. Kushner, B.H.; Ostrovnaya, I.; Cheung, I.Y.; Kuk, D.; Modak, S.; Kramer, K.; Roberts, S.S.; Basu, E.M.; Yataghene, K.; Cheung, N.-K.V. Lack of survival advantage with autologous stem-cell transplantation in high-risk neuroblastoma consolidated by anti-GD2 immunotherapy and isotretinoin. Oncotarget 2016, 7, 4155–4166.
  41. Kushner, B.H.; LaQuaglia, M.P.; Modak, S.; Wolden, S.L.; Basu, E.M.; Roberts, S.S.; Kramer, K.; Yataghene, K.; Cheung, I.Y.; Cheung, N.-K.V. MYCN-amplified stage 2/3 neuroblastoma: Excellent survival in the era of anti-GD2 immunotherapy. Oncotarget 2017, 8, 95293–95302.
  42. Mora, J.; Castañeda, A.; Flores, M.A.; Santa-María, V.; Garraus, M.; Gorostegui, M.; Simao, M.; Perez-Jaume, S.; Mañe, S. The Role of Autologous Stem-Cell Transplantation in High-Risk Neuroblastoma Consolidated by anti-GD2 Immunothera-py. Results of Two Consecutive Studies. Front. Pharmacol. 2020, 11, 1699.
  43. Mora, J.; Castañeda, A.; Gorostegui, M.; Santa-María, V.; Garraus, M.; Muñoz, J.P.; Varo, A.; Perez-Jaume, S.; Mañe, S. Naxitamab combined with granulocyte-macrophage colony-stimulating factor as consolidation for high-risk neuroblastoma patients in complete remission. Pediatric Blood Cancer 2021, 68, e29121.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 394
Revisions: 2 times (View History)
Update Date: 06 Jun 2022
1000/1000
Video Production Service