1
|
McCall D, Alqahtani S, Budak M, Sheikh I, Fan AE, Ramakrishnan R, Nunez C, Roth M, Garcia MB, Gibson A, Daver N, Garces S, Short NJ, Issa GC, Ravandi F, DiNardo CD, Montalban Bravo G, Garcia-Manero G, Cuglievan B, Kadia T. Cladribine-Based Therapy for Acute Myeloid Leukemia in Child, Adolescent, and Early Young Adult Patients: The MD Anderson Cancer Center Experience. Cancers (Basel) 2024; 16:3886. [PMID: 39594839 PMCID: PMC11592422 DOI: 10.3390/cancers16223886] [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: 10/11/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Cladribine-based combination chemotherapy has demonstrated promising efficacy in patients with relapsed/refractory adult acute myeloid leukemia (AML), prompting its increased utilization in the frontline; in pediatrics, it has been typically reserved for relapsed or refractory cases. While fludarabine has been used more commonly as a purine analog in intensive regimens, cladribine may be an important alternative. Methods: We performed a retrospective study at MD Anderson Cancer Center from January 2015 to July 2023, which included patients aged 1-21 years with refractory or relapsed AML who received cladribine outside of a transplant conditioning. Results: A total of 30 patients were included, with a median age of 20 years (range, 2-21), and 55% being male. Similar to adults, cladribine exhibited good tolerability in pediatric and adolescent patients, with the most common adverse events being febrile neutropenia and myelosuppression. The most common grade 3 or 4 adverse events included febrile neutropenia (55%) and sepsis (26%), and there were no treatment discontinuations due to adverse events. Among patients with a median number of 2 (0-7) prior treatments, the overall response rate (CR/CRi) was 45%, and median event-free and overall survival were 6 and 12 months, respectively. Disease progression resulted in 4 deaths within 30 days of treatment. Conclusions: Cladribine was tolerated in pediatrics. No new safety signals were seen with cladribine regimens in this cohort. Response assessment is limited due to the heavily pretreated cohort. Further prospective studies are warranted on the safety and efficacy of cladribine and establish its role in pediatric, adolescent, and early young adult patients with AML.
Collapse
Affiliation(s)
- David McCall
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shaikha Alqahtani
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Moriah Budak
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Irtiza Sheikh
- Department of Pediatric Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Aaron E. Fan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ramya Ramakrishnan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cesar Nunez
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael Roth
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Miriam B. Garcia
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amber Gibson
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sofia Garces
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicholas J. Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ghayas C. Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Courtney D. DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Branko Cuglievan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
2
|
Sisinni L, Monserrate GXA, Hurtado JMP, Panesso M, Molina B, Fuentes C, Fuster JL, Verdu-Amoros J, Regueiro A, Palomo P, Beléndez C, Pascual A, Badell I, Mozo Y, Bueno D, Pérez-Martínez A, Fernández JM, Vicent MG, de Heredia CD. Haploidentical versus Cord Blood Transplantation in Pediatric AML. A Retrospective Outcome Analysis on Behalf of the Pediatric Subcommittee of GETH (Grupo Español de Trasplante Hematopoyético). Transplant Cell Ther 2024; 30:1015.e1-1015.e13. [PMID: 39067788 DOI: 10.1016/j.jtct.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Haploidentical stem cell transplantation (Haplo-SCT) and cord blood transplantation (CBT) are both effective alternative treatments in patients suffering from acute myeloid leukemia (AML) and lacking a matched HLA donor. In the last years, many centers have abandoned CBT procedures mostly due to concern about poorer immune recovery compared with Haplo-SCT. We conducted a retrospective multicenter study comparing the outcomes using both alternative approaches in AML. A total of 122 transplants (86 Haplo-SCTs and 36 CBTs) from 12 Spanish centers were collected from 2007 to 2021. Median age at hematopoietic stem cell transplantation (HSCT) was 7 years (0.4-20). Thirty-nine patients (31.9%) showed positive minimal residual disease (MRD) at HSCT and a previous HSCT was performed in 37 patients (30.3%). The median infused cellularity was 14.4 × 106/kg CD34+ cells (6.0-22.07) for Haplo-SCT and 4.74 × 105/kg CD34+ cells (0.8-9.4) for CBT. Median time to neutrophil engraftment was 14 days (7-44) for Haplo-SCT and 17 days (8-29) for CBT (P = .03). The median time to platelet engraftment was 14 days (6-70) for Haplo-SCT and 43 days (10-151) for CBT (P < .001). Graft rejection was observed in 13 Haplo-SCTs (15%) and in 6 CBTs (16%). The cumulative incidence of acute graft versus host disease (GvHD) grades II-IV was 54% and 51% for Haplo-SCT and CBT, respectively (P = .50). The cumulative incidence of severe acute GvHD (grades III-IV) was 22% for Haplo-SCT and 25% for CBT (P = .90). There was a tendency to a higher risk of chronic GvHD in the Haplo-SCT group being the cumulative incidence of 30% for Haplo-SCT and 12% for CBT (P = .09). The cumulative incidence of relapse was 28% and 20% for Haplo-SCT and CBT, respectively (P = .60). We did not observe statistically significant differences in outcome measures between Haplo-SCT and CBT procedures: 5-year overall survival (OS) was 64% versus 57% (P = .50), 5-year disease-free survival (DFS) 58% versus 57% (P = .80), GvHD-free and relapse-free survival (GFRFS) 41% versus 54% (P = .30), and cumulative incidence of transplant-related mortality (TRM) 14% versus 15% (P = .80), respectively. In the multivariate analysis, MRD positivity and a disease status >CR1 at the time of HSCT were significantly associated with poorer outcomes (P < .05). In conclusion, our study supports that both haploidentical and cord blood transplantation show comparable outcomes in pediatric AML patients. We obtained comparable survival rates, although CBT showed a trend to lower rates of chronic GvHD and higher GFRFS, demonstrating that it should still be considered a valuable option, particularly for pediatric patients.
Collapse
Affiliation(s)
- Luisa Sisinni
- Hematología y Oncología Pediátrica, Hospital Universitario La Paz, Madrid.
| | | | | | - Melissa Panesso
- Servicio de Oncología y Hematología Pediátrica, Unidad HSCT. Hospital Universitari Vall d'Hebron, Barcelona
| | - Blanca Molina
- Hematología-Oncología Pediátrica, Hospital Niño Jesús, Madrid
| | | | - José Luís Fuster
- Sección de Oncohematología Pediátrica, Hospital Clínico Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia
| | - Jaime Verdu-Amoros
- Hematología Pediátrica, Hospital Clínico Universitario; INCLIVA-Biomedical Research Institute, Valencia
| | | | - Pilar Palomo
- Hematología Pediátrica, Hospital Universitario Central de Asturia, Oviedo
| | | | | | - Isabel Badell
- Hematología-Oncología Pediátrica, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Yasmina Mozo
- Hematología y Oncología Pediátrica, Hospital Universitario La Paz, Madrid
| | - David Bueno
- Hematología y Oncología Pediátrica, Hospital Universitario La Paz, Madrid
| | | | | | | | - Cristina Díaz de Heredia
- Servicio de Oncología y Hematología Pediátrica, Unidad HSCT. Hospital Universitari Vall d'Hebron, Barcelona
| |
Collapse
|
3
|
Verma A, Chi YY, Malvar J, Lamble A, Chaudhury S, Agarwal A, Li HT, Liang G, Leong R, Brown PA, Kaplan J, Schafer ES, Slone T, Pauly M, Chang BH, Stieglitz E, Wayne AS, Hijiya N, Bhojwani D. Nivolumab Plus 5-Azacitidine in Pediatric Relapsed/Refractory Acute Myeloid Leukemia (AML): Phase I/II Trial Results from the Therapeutic Advances in Childhood Leukemia and Lymphoma (TACL) Consortium. Cancers (Basel) 2024; 16:496. [PMID: 38339248 PMCID: PMC10854518 DOI: 10.3390/cancers16030496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/11/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Improvements in survival have been made over the past two decades for childhood acute myeloid leukemia (AML), but the approximately 40% of patients who relapse continue to have poor outcomes. A combination of checkpoint-inhibitor nivolumab and azacitidine has demonstrated improvements in median survival in adults with AML. This phase I/II study with nivolumab and azacitidine in children with relapsed/refractory AML (NCT03825367) was conducted through the Therapeutic Advances in Childhood Leukemia & Lymphoma consortium. Thirteen patients, median age 13.7 years, were enrolled. Patients had refractory disease with multiple reinduction attempts. Twelve evaluable patients were treated at the recommended phase II dose (established at dose level 1, 3 mg/kg/dose). Four patients (33%) maintained stable disease. This combination was well tolerated, with no dose-limiting toxicities observed. Grade 3-4 adverse events (AEs) were primarily hematological. Febrile neutropenia was the most common AE ≥ grade 3. A trend to improved quality of life was noted. Increases in CD8+ T cells and reductions in CD4+/CD8+ T cells and demethylation were observed. The combination was well tolerated and had an acceptable safety profile in pediatric patients with relapsed/refractory AML. Future studies might explore this combination for the maintenance of remission in children with AML at high risk of relapse.
Collapse
Affiliation(s)
- Anupam Verma
- Center for Cancer and Blood Disorders, Pediatric Hematology Oncology Branch, Children’s National Hospital, Washington, DC 20010, USA
- Division of Pediatric Hematology Oncology, Primary Children’s Hospital, University of Utah, Salt Lake City, UT 84113, USA
| | - Yueh-Yun Chi
- Division of Hematology-Oncology, Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA; (Y.-Y.C.); (A.S.W.); (D.B.)
| | - Jemily Malvar
- Division of Hematology-Oncology, Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (J.M.)
| | - Adam Lamble
- Department of Pediatric Hematology Oncology, Seattle Children’s Hospital, Seattle, WA 98105, USA;
| | - Sonali Chaudhury
- Department of Pediatric Hematology Oncology, Ann and Robert Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA;
| | - Archana Agarwal
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT 84108, USA;
| | - Hong-Tao Li
- Department of Urology, University of Southern California, Los Angeles, CA 90033, USA; (H.-T.L.); (G.L.)
| | - Gangning Liang
- Department of Urology, University of Southern California, Los Angeles, CA 90033, USA; (H.-T.L.); (G.L.)
| | - Roy Leong
- Division of Hematology-Oncology, Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (J.M.)
| | | | - Joel Kaplan
- Department of Pediatric Hematology Oncology, Atrium Health Levine Children’s Hospital, Wake Forrest University, Charlotte, NC 28203, USA;
| | - Eric S. Schafer
- Division of Pediatric Hematology/Oncology, Baylor College of Medicine, Texas Children’s Cancer and Hematology Center, Houston, TX 77030, USA;
| | - Tamra Slone
- Department of Pediatric Hematology Oncology, UT Southwestern, Dallas, TX 75235, USA;
| | - Melinda Pauly
- Department of Pediatric Hematology Oncology, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA;
| | - Bill H. Chang
- Division of Pediatric Hematology Oncology, Oregon Health and Science University, Portland, OR 97239, USA;
| | - Elliot Stieglitz
- Department of Pediatric Oncology, University of California, San Francisco Benioff Children’s Hospitals, San Francisco, CA 94158, USA;
| | - Alan S. Wayne
- Division of Hematology-Oncology, Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA; (Y.-Y.C.); (A.S.W.); (D.B.)
| | - Nobuko Hijiya
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Columbia University Medical Center, New York, NY 10032, USA;
| | - Deepa Bhojwani
- Division of Hematology-Oncology, Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA; (Y.-Y.C.); (A.S.W.); (D.B.)
| |
Collapse
|
4
|
Lee JH, Ju HY, Hyun JK, Kim SJ, Cho HW, Lee JK, Lee JW, Sung KW, Yoo KH. Treatment outcome and prognostic factors in relapsed pediatric acute myeloid leukemia. Blood Res 2023; 58:181-186. [PMID: 37926557 PMCID: PMC10758629 DOI: 10.5045/br.2023.2023152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Background Despite improved outcomes for pediatric patients with acute myeloid leukemia (AML), the prognosis for relapse remains poor. This study aimed to examine the clinical factors associated with prognosis in relapsed pediatric AML. Methods We conducted a chart review of pediatric patients with AML who experienced their first relapse and received treatment at our institution between 2008 and 2019. Risk stratification at diagnosis was performed according to the definition suggested by the ongoing AML 2012 study in Korea, and the clinical factors associated with prognosis were analyzed. Results A total of 27 pediatric patients with relapsed AML were identified. The 5-year overall survival (OS) and event-free survival (EFS) rates were 32.9% and 32.9%, respectively. A duration ≥12 months from diagnosis to relapse had a favorable impact on survival outcomes (5-yr OS, 64.0% vs. 15.7%; P=0.007). Patients who achieved complete remission (CR) after 1 course of chemotherapy following relapse (N=15) had a 5-year OS rate of 59.3%, while none of the other patients survived (P<0.0001). Additionally, the 5-year OS differed significantly based on the risk group at initial diagnosis (62.3% [favorable and intermediate prognosis groups, N=11] vs. 13.3% [poor prognosis group, N=15]; P=0.014). Conclusion Patients with a longer duration of CR before relapse, who achieved CR following 1 course of reinduction chemotherapy, and were in the favorable or intermediate prognosis group at diagnosis demonstrated better outcomes. These findings emphasize the importance of tailoring treatment strategies based on the expected prognosis at relapse in pediatric patients with AML.
Collapse
Affiliation(s)
- Jung Hwan Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Young Ju
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ju Kyung Hyun
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - So Jin Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae Kyung Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Science and Technology, SAIHST, Sungkyunkwan University School of Medicine, Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| |
Collapse
|
5
|
Kelvin JM, Jain J, Thapa A, Qui M, Birnbaum LA, Moore SG, Zecca H, Summers RJ, Switchenko JM, Costanza E, Uricoli B, Wang X, Jui NT, Fu H, Du Y, DeRyckere D, Graham DK, Dreaden EC. Constitutively Synergistic Multiagent Drug Formulations Targeting MERTK, FLT3, and BCL-2 for Treatment of AML. Pharm Res 2023; 40:2133-2146. [PMID: 37704893 DOI: 10.1007/s11095-023-03596-9] [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: 03/15/2023] [Accepted: 08/26/2023] [Indexed: 09/15/2023]
Abstract
PURPOSE Although high-dose, multiagent chemotherapy has improved leukemia survival rates, treatment outcomes remain poor in high-risk subsets, including acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) in infants. The development of new, more effective therapies for these patients is therefore an urgent, unmet clinical need. METHODS The dual MERTK/FLT3 inhibitor MRX-2843 and BCL-2 family protein inhibitors were screened in high-throughput against a panel of AML and MLL-rearranged precursor B-cell ALL (infant ALL) cell lines. A neural network model was built to correlate ratiometric drug synergy and target gene expression. Drugs were loaded into liposomal nanocarriers to assess primary AML cell responses. RESULTS MRX-2843 synergized with venetoclax to reduce AML cell density in vitro. A neural network classifier based on drug exposure and target gene expression predicted drug synergy and growth inhibition in AML with high accuracy. Combination monovalent liposomal drug formulations delivered defined drug ratios intracellularly and recapitulated synergistic drug activity. The magnitude and frequency of synergistic responses were both maintained and improved following drug formulation in a genotypically diverse set of primary AML bone marrow specimens. CONCLUSIONS We developed a nanoscale combination drug formulation that exploits ectopic expression of MERTK tyrosine kinase and dependency on BCL-2 family proteins for leukemia cell survival in pediatric AML and infant ALL cells. We demonstrate ratiometric drug delivery and synergistic cell killing in AML, a result achieved by a systematic, generalizable approach of combination drug screening and nanoscale formulation that may be extended to other drug pairs or diseases in the future.
Collapse
Affiliation(s)
- James M Kelvin
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Juhi Jain
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
- Department of Pediatrics, University of Arizona College of Medicine, and Banner University Medical Center Tucson, Tucson, AZ, 85724, USA
| | - Aashis Thapa
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Min Qui
- Department of Pharmacology and Chemical Biology, Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Lacey A Birnbaum
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Samuel G Moore
- Systems Mass Spectrometry Core Facility, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Henry Zecca
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Ryan J Summers
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Jeffrey M Switchenko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Emma Costanza
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Biaggio Uricoli
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Xiaodong Wang
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Nathan T Jui
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Deborah DeRyckere
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Douglas K Graham
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA.
| | - Erik C Dreaden
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA.
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA.
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
6
|
Horgan C, Mullanfiroze K, Rauthan A, Patrick K, Butt NA, Mirci-Danicar O, O’Connor O, Furness C, Deshpande A, Lawson S, Broderick V, Evans P, Gibson B, Roberts W, Ali S, Galani S, Kirkwood AA, Jovanovic J, Dillon R, Virgo P, James B, Rao K, Amrolia PJ, Wynn RF. T-cell replete cord transplants give superior outcomes in high-risk and relapsed/refractory pediatric myeloid malignancy. Blood Adv 2023; 7:2155-2165. [PMID: 36649566 PMCID: PMC10206437 DOI: 10.1182/bloodadvances.2022009253] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/22/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023] Open
Abstract
Stem cell transplant (SCT) outcomes in high-risk and relapsed/refractory (R/R) pediatric acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) have been historically poor. Cord blood (CB) allows T-cell replete CB transplant (TRCB), enabling enhanced graft-versus-leukemia. We consecutively collected data from 367 patients undergoing TRCB (112 patients) or other cell source (255 patients) SCT for pediatric AML/MDS in the United Kingdom and Ireland between January 2014 and December 2021. Data were collected about the patient's demographics, disease, and its treatment; including previous transplant, measurable residual disease (MRD) status at transplant, human leukocyte antigen-match, relapse, death, graft versus host disease (GvHD), and transplant-related mortality (TRM). Univariable and multivariable analyses were undertaken. There was a higher incidence of poor prognosis features in the TRCB cohort: 51.4% patients were MRD positive at transplant, 46.4% had refractory disease, and 21.4% had relapsed after a previous SCT, compared with 26.1%, 8.6%, and 5.1%, respectively, in the comparator group. Event free survival was 64.1% within the TRCB cohort, 50% in MRD-positive patients, and 79% in MRD-negative patients. To allow for the imbalance in baseline characteristics, a multivariable analysis was performed where the TRCB cohort had significantly improved event free survival, time to relapse, and reduced chronic GvHD, with some evidence of improved overall survival. The effect appeared similar regardless of the MRD status. CB transplant without serotherapy may be the optimal transplant option for children with myeloid malignancy.
Collapse
Affiliation(s)
- Claire Horgan
- Royal Manchester Children’s Hospital, Manchester, United Kingdom
| | | | - Archana Rauthan
- Great Ormond Street Hospital for Children, London, United Kingdom
| | - Katharine Patrick
- Sheffield Children’s NHS Foundation Trust, Sheffield, United Kingdom
| | | | | | - Olya O’Connor
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Caroline Furness
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
- The Institute of Cancer Research, Sutton, United Kingdom
| | | | - Sarah Lawson
- Birmingham Children’s Hospital, Birmingham, United Kingdom
| | | | - Pamela Evans
- Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Brenda Gibson
- Royal Hospital for Children, Glasgow, United Kingdom
| | - Wing Roberts
- Great North Children’s Hospital, Victoria Wing, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Salah Ali
- Leeds Children’s Hospital, Clarendon Wing, Leeds General Infirmary, Leeds, United Kingdom
| | - Sevasti Galani
- CR UK & UCL Cancer Trials Centre, UCL Cancer Institute, University College London, London, United Kingdom
| | - Amy A. Kirkwood
- CR UK & UCL Cancer Trials Centre, UCL Cancer Institute, University College London, London, United Kingdom
| | - Jelena Jovanovic
- Department of Medical and Molecular Genetics, King’s College London, Strand, London, United Kingdom
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King’s College London, Strand, London, United Kingdom
| | - Paul Virgo
- Department of Immunology and Immunogenetics, North Bristol NHS Trust, Bristol, United Kingdom
| | - Beki James
- Leeds Children’s Hospital, Clarendon Wing, Leeds General Infirmary, Leeds, United Kingdom
| | - Kanchan Rao
- Great Ormond Street Hospital for Children, London, United Kingdom
| | | | - Robert F. Wynn
- Royal Manchester Children’s Hospital, Manchester, United Kingdom
| |
Collapse
|
7
|
Trabal A, Gibson A, He J, McCall D, Roth M, Nuñez C, Garcia M, Buzbee M, Toepfer L, Bidikian A, Daver N, Kadia T, Short NJ, Issa GC, Ravandi F, DiNardo CD, Montalban Bravo G, Garces S, Marcogliese A, Paek H, Dreyer Z, Brackett J, Redell M, Yi J, Garcia-Manero G, Konopleva M, Stevens A, Cuglievan B. Venetoclax for Acute Myeloid Leukemia in Pediatric Patients: A Texas Medical Center Experience. Cancers (Basel) 2023; 15:cancers15071983. [PMID: 37046645 PMCID: PMC10093646 DOI: 10.3390/cancers15071983] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/07/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
The BCL-2 inhibitor venetoclax improves survival for adult patients with acute myeloid leukemia (AML) in combination with lower-intensity therapies, but its benefit in pediatric patients with AML remains unclear. We retrospectively reviewed two Texas Medical Center institutions’ experience with venetoclax in 43 pediatric patients with AML; median age 17 years (range, 0.6–21). This population was highly refractory; 44% of patients (n = 19) had ≥3 prior lines of therapy, 37% (n = 16) had received a prior bone marrow transplant, and 81% (n = 35) had unfavorable genetics KMT2A (n = 17), WT1 (n = 13), FLT3-ITD (n = 10), monosomy 7 (n = 5), TP53 (n = 3), Inv(3) (n = 3), IDH1/2 (n = 2), monosomy 5 (n = 1), NUP98 (n = 1) and ASXL1 (n = 1). The majority (86%) received venetoclax with a hypomethylating agent. Grade 3 or 4 adverse events included febrile neutropenia in 37% (n = 16), non-febrile neutropenia in 12% (n = 5), anemia in 14% (n = 6), and thrombocytopenia in 14% (n = 6). Of 40 patients evaluable for response, 10 patients (25%) achieved complete response (CR), 6 patients (15%) achieved CR with incomplete blood count recovery (CRi), and 2 patients (5%) had a partial response, (CR/CRi composite = 40%; ORR = 45%). Eleven (25%) patients received a hematopoietic stem cell transplant following venetoclax combination therapy, and six remain alive (median follow-up time 33.6 months). Median event-free survival and overall survival duration was 3.7 months and 8.7 months, respectively. Our findings suggest that in pediatric patients with AML, venetoclax is well-tolerated, with a safety profile similar to that in adults. More studies are needed to establish an optimal venetoclax-based regimen for the pediatric population.
Collapse
|
8
|
Wang B, Wen X, Zhang R, Zhu G, Wu Y, Zhang Y, Lin W, Yu J, Fan J, Li J, Yang J, Qin M, Zheng H. Homoharringtonine-Based Induction Therapy Reduces the Recurrence Rate of Pediatric Acute Myeloid Leukemia After Allogeneic Hematopoietic Stem Cell Transplantation. Cell Transplant 2023; 32:9636897231183559. [PMID: 37470325 PMCID: PMC10363872 DOI: 10.1177/09636897231183559] [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: 03/17/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 07/21/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is an effective treatment for acute myeloid leukemia (AML). Pediatric patients with AML who relapse after HSCT have an extremely poor prognosis. We performed a retrospective study of pediatric patients diagnosed with AML from August 2015 to October 2019 who were treated with HSCT. Kaplan-Meier analyses were used to evaluate overall survival (OS), event-free survival (EFS), and cumulative recurrence rate (CRR). Cox regression analysis was used to determine the association between the baseline characteristics and relapse. A total of 37 pediatric patients met the inclusion criteria. Twenty-eight (75.7%) patients survived, and 9 (24.3%) patients died. The OS rates of AML patients treated with HSCT were 89.2% ± 5.1%, 75.7% ± 7.1%, and 75.7% ± 7.1% at 1, 3, and 5 years, respectively, and the CRRs were 11.4% ± 5.4%, 24.7% ± 7.7%, and 33.1% ± 10.4% at 1, 3, and 5 years after HSCT, respectively; four of nine children who relapsed after transplantation died. Induction with etoposide rather than homoharringtonine and fungal infections could be high-risk factors for recurrence after transplantation. The association between homoharringtonine-based induction therapy and a low recurrence rate persisted after adjusting for age, sex, risk stratification, fusion genes, and fungal infections. This study clarifies the clinical features and poor prognosis of post-transplant relapse in pediatric AML and indicates the urgent need for effective therapy for patients who relapse after HSCT.
Collapse
Affiliation(s)
- Bin Wang
- Transplantation Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Xiaojia Wen
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Ruidong Zhang
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Guanghua Zhu
- Transplantation Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Ying Wu
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Yuanyuan Zhang
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Wei Lin
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jiaole Yu
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jia Fan
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jing Li
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jun Yang
- Transplantation Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Maoquan Qin
- Transplantation Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Huyong Zheng
- Leukemia Department, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Clinical Discipline of Pediatric Hematology, National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| |
Collapse
|
9
|
Luo Y, Xu Y, Li X, Shi X, Huang P, Chen Y, He Z. A Prognostic Model of Seven Immune Genes to Predict Overall Survival in Childhood Acute Myeloid Leukemia. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7724220. [PMID: 36518627 PMCID: PMC9744619 DOI: 10.1155/2022/7724220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/04/2022] [Accepted: 11/05/2022] [Indexed: 12/05/2024]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is one of the most common hematological malignancies and accounts for about 20% of childhood leukemias. Currently, immunotherapy is one of the recommended treatment schemes for recurrent AML patients to improve their survival rates. Nonetheless, low remission and high mortality rates are observed in recurrent AML and challenge the prognosis of AML patients. To address this problem, we aimed to establish and verify a reliable prognostic risk model using immune-related genes to improve the prognostic evaluation and recommendation for personalized treatment of AML. METHODS Transcriptome data and clinical data were acquired from the TARGET database while immune genes were sourced from InnateDB and ImmPort Shared databases. The mRNA expression profile matrix of immune genes from 62 normal samples and 1408 AML cases was extracted from the transcriptome data and subjected to differential expression (DE) analysis. The entire cohort of DE immune genes was randomly divided into the test group and training group. The prognostic model associated with immune genes was constructed using the training group. The test group and entire cohort were employed for model validation. Lastly, we analyzed the potential clinical application of the model and its association with immune cell infiltration. RESULTS In total, 751 DE immune genes were differentially regulated, including 552 upregulated and 199 downregulated. Based on these DE genes, we developed and validated a prognostic risk model composed of seven immune genes, GDF1, TPM2, IL1R1, PSMD4, IL5RA, DHCR24, and IL12RB2. This model is able to predict the 5-year survival rate more accurately compared with age, gender, and risk stratification. Further analysis showed that CD8+ T-cell contents and neutrophil infiltration decreased but macrophage infiltration increased as the risk score increased. CONCLUSIONS A seven-immune gene model of AML was developed and validated. We propose this model as an independent prognostic variable able to estimate the 5-year survival rate. In addition, the model can also reflect the immune microenvironment of AML patients.
Collapse
Affiliation(s)
- Yan Luo
- Suzhou Medical College of Soochow University, Suzhou 215325, China
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi 563000, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, 563003 Guizhou, China
| | - Yanpeng Xu
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi 563000, China
| | - Xue Li
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Xiaoqi Shi
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Pei Huang
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi 563000, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, 563003 Guizhou, China
| | - Yan Chen
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi 563000, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, 563003 Guizhou, China
| | - Zhixu He
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi 563000, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, 563003 Guizhou, China
| |
Collapse
|
10
|
Hematopoietic Cell Transplantation in the Treatment of Pediatric Acute Myelogenous Leukemia and Myelodysplastic Syndromes: Guidelines from the American Society of Transplantation and Cellular Therapy. Transplant Cell Ther 2022; 28:530-545. [DOI: 10.1016/j.jtct.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/20/2022]
|
11
|
Relapsed acute myeloid leukemia in children and adolescents: current treatment options and future strategies. Leukemia 2022; 36:1951-1960. [PMID: 35668109 DOI: 10.1038/s41375-022-01619-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/18/2022] [Accepted: 05/26/2022] [Indexed: 11/08/2022]
Abstract
Pediatric acute myeloid leukemia (AML) develops from clonal expansion of hematopoietic precursor cells and is characterized by morphologic and cytomolecular heterogeneity. Although the past 40 years have seen significant improvements in overall survival, the prevailing treatment challenges in pediatric AML are the prevention of relapse and the management of relapsed disease. Approximately 25% of children and adolescents with AML suffer disease relapse and face a poor prognosis. Our greater understanding of the genomic, epigenomic, metabolomic, and immunologic pathophysiology of relapsed AML allows for better therapeutic strategies that are being developed for pediatric clinical trials. The development of biologically rational agents is critical as conventional chemotherapeutic salvage regimens are not effective for all patients and pose risk of organ toxicity in heavily pretreated patients. Another major barrier to improvement in outcomes for relapsed pediatric AML is the historic lack of availability and participation in clinical trials. There are ongoing efforts to launch multinational clinical trials of emerging therapies. The purpose of this review is to summarize currently available and newly developed therapies for relapsed pediatric AML.
Collapse
|
12
|
Pommert L, Schafer ES, Malvar J, Gossai N, Florendo E, Pulakanti K, Heimbruch K, Stelloh C, Chi YY, Sposto R, Rao S, Van Huynh T, Brown P, Chang BH, Colace SI, Hermiston ML, Heym K, Hutchinson RJ, Kaplan JA, Mody R, O’Brien TA, Place AE, Shaw PH, Ziegler DS, Wayne A, Bhojwani D, Burke MJ. Decitabine and vorinostat with FLAG chemotherapy in pediatric relapsed/refractory AML: Report from the therapeutic advances in childhood leukemia and lymphoma (TACL) consortium. Am J Hematol 2022; 97:613-622. [PMID: 35180323 PMCID: PMC8986610 DOI: 10.1002/ajh.26510] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 12/17/2022]
Abstract
Survival outcomes for relapsed/refractory pediatric acute myeloid leukemia (R/R AML) remain dismal. Epigenetic changes can result in gene expression alterations which are thought to contribute to both leukemogenesis and chemotherapy resistance. We report results from a phase I trial with a dose expansion cohort investigating decitabine and vorinostat in combination with fludarabine, cytarabine, and G-CSF (FLAG) in pediatric patients with R/R AML [NCT02412475]. Thirty-seven patients enrolled with a median age at enrollment of 8.4 (range, 1-20) years. There were no dose limiting toxicities among the enrolled patients, including two patients with Down syndrome. The recommended phase 2 dose of decitabine in combination with vorinostat and FLAG was 10 mg/m2 . The expanded cohort design allowed for an efficacy evaluation and the overall response rate among 35 evaluable patients was 54% (16 complete response (CR) and 3 complete response with incomplete hematologic recovery (CRi)). Ninety percent of responders achieved minimal residual disease (MRD) negativity (<0.1%) by centralized flow cytometry and 84% (n = 16) successfully proceeded to hematopoietic stem cell transplant. Two-year overall survival was 75.6% [95%CI: 47.3%, 90.1%] for MRD-negative patients vs. 17.9% [95%CI: 4.4%, 38.8%] for those with residual disease (p < .001). Twelve subjects (34%) had known epigenetic alterations with 8 (67%) achieving a CR, 7 (88%) of whom were MRD negative. Correlative pharmacodynamics demonstrated the biologic activity of decitabine and vorinostat and identified specific gene enrichment signatures in nonresponding patients. Overall, this therapy was well-tolerated, biologically active, and effective in pediatric patients with R/R AML, particularly those with epigenetic alterations.
Collapse
Affiliation(s)
- Lauren Pommert
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Eric S. Schafer
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Jemily Malvar
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Nathan Gossai
- Department of Pediatrics, Center for Cancer and Blood Disorders, Children’s Minnesota, Minneapolis, MN
| | - Ellynore Florendo
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA
| | | | - Katelyn Heimbruch
- Blood Research Institute, Versiti, Milwaukee, WI
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Cary Stelloh
- Blood Research Institute, Versiti, Milwaukee, WI
| | - Yueh-Yun Chi
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, C
| | - Richard Sposto
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, C
| | - Sridhar Rao
- Blood Research Institute, Versiti, Milwaukee, WI
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Division of Pediatric Hematology-Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Thu Van Huynh
- Department of Pediatrics, Children’s Hospital of Orange County, Orange, CA
| | - Patrick Brown
- Division of Pediatric Oncology, Johns Hopkins University, Baltimore, MD
| | - Bill H. Chang
- Department of Pediatrics, Oregon Health and Science University, Portland, OR
| | - Susan I. Colace
- Department of Pediatrics, Hematology and Oncology, Nationwide Children’s Hospital, Columbus, OH
| | - Michelle L. Hermiston
- Division of Hematology/Oncology, University of California, San Francisco Benioff Children’s Hospital, San Francisco, CA
| | - Kenneth Heym
- Department of Pediatrics, Cook Children’s Medical Center, Fort Worth, TX
| | - Raymond J. Hutchinson
- Department of Pediatric and Communicable Diseases Division of Pediatric Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI
| | - Joel A. Kaplan
- Department of Pediatrics, Carolinas Medical Center/Levine Cancer Institute, Charlotte, NC
| | - Rajen Mody
- Department of Pediatric and Communicable Diseases Division of Pediatric Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI
| | - Tracey A. O’Brien
- Cord & Marrow Transplant Program, Centre for Children’s Cancer & Blood Disorders, Sydney Children’s Hospital, Sydney, Australia
| | - Andrew E. Place
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Peter H. Shaw
- Cancer and Blood Disorders Institute, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - David S. Ziegler
- Kids Cancer Centre, Sydney Children’s Hospital, Randwick, Australia
- School of Women’s and Children’s Health, University of New South Wales, Sydney, Australia
| | - Alan Wayne
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, C
| | - Deepa Bhojwani
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, C
| | - Michael J. Burke
- Division of Pediatric Hematology-Oncology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
13
|
Bednarski JJ, Zimmerman C, Berrien-Elliott MM, Foltz JA, Becker-Hapak M, Neal CC, Foster M, Schappe T, McClain E, Pence PP, Desai S, Kersting-Schadek S, Wong P, Russler-Germain DA, Fisk B, Lie WR, Eisele J, Hyde S, Bhatt ST, Griffith OL, Griffith M, Petti AA, Cashen AF, Fehniger TA. Donor memory-like NK cells persist and induce remissions in pediatric patients with relapsed AML after transplant. Blood 2022; 139:1670-1683. [PMID: 34871371 PMCID: PMC8931511 DOI: 10.1182/blood.2021013972] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/18/2021] [Indexed: 11/20/2022] Open
Abstract
Pediatric and young adult (YA) patients with acute myeloid leukemia (AML) who relapse after allogeneic hematopoietic cell transplantation (HCT) have an extremely poor prognosis. Standard salvage chemotherapy and donor lymphocyte infusions (DLIs) have little curative potential. Previous studies showed that natural killer (NK) cells can be stimulated ex vivo with interleukin-12 (IL-12), -15, and -18 to generate memory-like (ML) NK cells with enhanced antileukemia responses. We treated 9 pediatric/YA patients with post-HCT relapsed AML with donor ML NK cells in a phase 1 trial. Patients received fludarabine, cytarabine, and filgrastim followed 2 weeks later by infusion of donor lymphocytes and ML NK cells from the original HCT donor. ML NK cells were successfully generated from haploidentical and matched-related and -unrelated donors. After infusion, donor-derived ML NK cells expanded and maintained an ML multidimensional mass cytometry phenotype for >3 months. Furthermore, ML NK cells exhibited persistent functional responses as evidenced by leukemia-triggered interferon-γ production. After DLI and ML NK cell adoptive transfer, 4 of 8 evaluable patients achieved complete remission at day 28. Two patients maintained a durable remission for >3 months, with 1 patient in remission for >2 years. No significant toxicity was experienced. This study demonstrates that, in a compatible post-HCT immune environment, donor ML NK cells robustly expand and persist with potent antileukemic activity in the absence of exogenous cytokines. ML NK cells in combination with DLI present a novel immunotherapy platform for AML that has relapsed after allogeneic HCT. This trial was registered at https://clinicaltrials.gov as #NCT03068819.
Collapse
Affiliation(s)
| | - Clare Zimmerman
- Division of Hematology and Oncology, Department of Pediatrics, and
| | - Melissa M Berrien-Elliott
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Jennifer A Foltz
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Michelle Becker-Hapak
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Carly C Neal
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Mark Foster
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Timothy Schappe
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Ethan McClain
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Patrick P Pence
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Sweta Desai
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Samantha Kersting-Schadek
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Pamela Wong
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - David A Russler-Germain
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Bryan Fisk
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | | | - Jeremy Eisele
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Stephanie Hyde
- Division of Hematology and Oncology, Department of Pediatrics, and
| | - Sima T Bhatt
- Division of Hematology and Oncology, Department of Pediatrics, and
| | - Obi L Griffith
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Malachi Griffith
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Allegra A Petti
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO
| | - Amanda F Cashen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
14
|
White T, Kaspers G, Abrahamsson J, Arad-Cohen N, Cianci D, Fernandez J, Ha SY, Hasle H, De Moerloose B, Zwaan CM, Goemans BF. Clinical outcomes of second relapsed and refractory first relapsed paediatric AML: A retrospective study within the NOPHO-DB SHIP consortium. Br J Haematol 2022; 197:755-765. [PMID: 35118649 PMCID: PMC9303517 DOI: 10.1111/bjh.18039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/24/2021] [Accepted: 12/30/2021] [Indexed: 01/27/2023]
Abstract
As treatments for second relapsed and refractory first relapsed paediatric AML transition from purely palliative to more commonly curative in nature, comparative data is necessary for evaluating the effectiveness of emerging treatment options. Furthermore, little is known about predictors of prognosis following third-line therapy. From 2004 until 2019, 277 of the 869 patients enrolled in NOPHO-DB SHIP consortium trials experienced a first relapse and, of these patients, 98 experienced refractory first relapse and 59 a second relapse. Data on patient and disease characteristics within this cohort of 157 patients was analysed to determine probability of overall survival (pOS) and to identify factors influencing survival. Data on early treatment response and complete remission were not available. One and 5-year pOS were 22 ± 3% and 14 ± 3%, respectively. There was no statistically significant difference in survival between refractory first relapsed and second relapsed AML. Factors influencing prognosis included: late relapse, type of third-line treatment, FLT3 mutational status, and original treatment protocol. These data provide a baseline for evaluating the effectiveness of emerging therapies for the treatment of children with refractory first relapsed and second relapsed paediatric AML and evidence that select patients receiving third-line therapy can be cured.
Collapse
Affiliation(s)
- Tara White
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Gertjan Kaspers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Pediatric Oncology, Amsterdam UMC, Emma Children's Hospital, Vrije Universiteit, Amsterdam, The Netherlands.,Dutch Childhood Oncology Group (DCOG), Utrecht, The Netherlands
| | - Jonas Abrahamsson
- Department of Paediatrics, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Nira Arad-Cohen
- Pediatric Hemato-Oncology Department, Ruth Rappaport Children's Hospital, Haifa, Israel
| | - Daniela Cianci
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jose Fernandez
- Pediatric Oncology and Haematology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Shau-Yin Ha
- Department of Paediatrics & Adolescent Medicine, Hong Kong Children 's Hospital, Ngau Tau Kok, Hong Kong
| | - Henrik Hasle
- Department of Pediatrics, Aarhus University Hospital, Aarhus, Denmark
| | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Bianca F Goemans
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
15
|
Christodoulou I, Ho WJ, Marple A, Ravich JW, Tam A, Rahnama R, Fearnow A, Rietberg C, Yanik S, Solomou EE, Varadhan R, Koldobskiy MA, Bonifant CL. Engineering CAR-NK cells to secrete IL-15 sustains their anti-AML functionality but is associated with systemic toxicities. J Immunother Cancer 2021; 9:jitc-2021-003894. [PMID: 34896980 PMCID: PMC8655609 DOI: 10.1136/jitc-2021-003894] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2021] [Indexed: 01/15/2023] Open
Abstract
Background The prognosis of patients with recurrent/refractory acute myelogenous leukemia (AML) remains poor and cell-based immunotherapies hold promise to improve outcomes. Natural Killer (NK) cells can elicit an antileukemic response via a repertoire of activating receptors that bind AML surface ligands. NK-cell adoptive transfer is safe but thus far has shown limited anti-AML efficacy. Here, we aimed to overcome this limitation by engineering NK cells to express chimeric antigen receptors (CARs) to boost their anti-AML activity and interleukin (IL)-15 to enhance their persistence. Methods We characterized in detail NK-cell populations expressing a panel of AML (CD123)-specific CARs and/or IL-15 in vitro and in AML xenograft models. Results CARs with 2B4.ζ or 4-1BB.ζ signaling domains demonstrated greater cell surface expression and endowed NK cells with improved anti-AML activity in vitro. Initial in vivo testing revealed that only 2B4.ζ Chimeric Antigen Receptor (CAR)-NK cells had improved anti-AML activity in comparison to untransduced (UTD) and 4-1BB.ζ CAR-NK cells. However, the benefit was transient due to limited CAR-NK-cell persistence. Transgenic expression of secretory interleukin (sIL)-15 in 2B4.ζ CAR and UTD NK cells improved their effector function in the setting of chronic antigen simulation in vitro. Multiparameter flow analysis after chronic antigen exposure identified the expansion of unique NK-cell subsets. 2B4.ζ/sIL-15 CAR and sIL-15 NK cells maintained an overall activated NK-cell phenotype. This was confirmed by transcriptomic analysis, which revealed a highly proliferative and activated signature in these NK-cell groups. In vivo, 2B4.ζ/sIL-15 CAR-NK cells had potent anti-AML activity in one model, while 2B4.ζ/sIL-15 CAR and sIL-15 NK cells induced lethal toxicity in a second model. Conclusion Transgenic expression of CD123-CARs and sIL-15 enabled NK cells to function in the setting of chronic antigen exposure but was associated with systemic toxicities. Thus, our study provides the impetus to explore inducible and controllable expression systems to provide cytokine signals to AML-specific CAR-NK cells before embarking on early-phase clinical testing.
Collapse
Affiliation(s)
- Ilias Christodoulou
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Internal Medicine, University of Patras School of Health Sciences, Patras, Western Greece, Greece
| | - Won Jin Ho
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Marple
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jonas W Ravich
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ada Tam
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ruyan Rahnama
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Adam Fearnow
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Cambrynne Rietberg
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Sean Yanik
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Elena E Solomou
- Department of Internal Medicine, University of Patras School of Health Sciences, Patras, Western Greece, Greece
| | - Ravi Varadhan
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael A Koldobskiy
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Challice L Bonifant
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA .,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Kobayashi S, Sano H, Mochizuki K, Ohara Y, Takahashi N, Kudo S, Ikeda K, Ohto H, Kikuta A. Effects of second transplantation with T-cell-replete haploidentical graft using low-dose anti-thymocyte globulin on long-term overall survival in pediatric patients with relapse of leukemia after first allogeneic transplantation. Int J Hematol 2021; 115:414-423. [PMID: 34822127 DOI: 10.1007/s12185-021-03266-9] [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: 09/06/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 10/19/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the preferred treatment for children with high-risk hematologic malignancies, but post-allo-HSCT relapse has a poor prognosis and limited treatment options. We evaluated the feasibility, outcome, and risk factors influencing survival after T-cell-replete haploidentical HSCT with low-dose anti-thymocyte globulin (ATG) in 30 patients with post-allo-HSCT relapse of acute lymphoblastic leukemia and acute myeloid leukemia. Overall, 50% of the patients had complete remission (CR) before the second transplant and the overall survival (OS) rate was 52%. In surviving patients (median follow-up 614 days), Kaplan-Meier analysis revealed estimated 2-year leukemia-free survival and OS rates of 48.1% and 61.1%, respectively. Cumulative incidences of 2-year non-relapse mortality and relapse were 24.7% and 36.3%, respectively. Achieving CR before the second allo-HSCT was a predominant independent prognostic factor identified in the multivariate analysis, with a significantly improved 2-year OS rate of 86.7%. T-cell-replete haplo-HSCT with low-dose ATG for second allo-HSCT may benefit a selected patient population.
Collapse
Affiliation(s)
- Shogo Kobayashi
- Department of Pediatric Oncology, Fukushima Medical University School of Medicine, 1 Hikariga-oka, Fukushima City, Fukushima, 960-1295, Japan.
| | - Hideki Sano
- Department of Pediatric Oncology, Fukushima Medical University School of Medicine, 1 Hikariga-oka, Fukushima City, Fukushima, 960-1295, Japan
| | - Kazuhiro Mochizuki
- Department of Pediatric Oncology, Fukushima Medical University School of Medicine, 1 Hikariga-oka, Fukushima City, Fukushima, 960-1295, Japan
| | - Yoshihiro Ohara
- Department of Pediatric Oncology, Fukushima Medical University School of Medicine, 1 Hikariga-oka, Fukushima City, Fukushima, 960-1295, Japan
| | - Nobuhisa Takahashi
- Department of Pediatric Oncology, Fukushima Medical University School of Medicine, 1 Hikariga-oka, Fukushima City, Fukushima, 960-1295, Japan
| | - Shingo Kudo
- Department of Pediatric Oncology, Fukushima Medical University School of Medicine, 1 Hikariga-oka, Fukushima City, Fukushima, 960-1295, Japan
| | - Kazuhiko Ikeda
- Department of Blood Transfusion and Transplantation Immunology, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Hitoshi Ohto
- Department of Blood Transfusion and Transplantation Immunology, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, Japan
| | - Atsushi Kikuta
- Department of Pediatric Oncology, Fukushima Medical University School of Medicine, 1 Hikariga-oka, Fukushima City, Fukushima, 960-1295, Japan
| |
Collapse
|
17
|
Kada A, Kikuta A, Saito AM, Kato K, Iguchi A, Yabe H, Ishida H, Hyakuna N, Takahashi Y, Nagasawa M, Hashii Y, Umeda K, Matsumoto K, Fujisaki H, Yano M, Nakazawa Y, Sano H. Single-Arm Non-Blinded Multicenter Clinical Trial on T-Cell-Replete Haploidentical Stem Cell Transplantation Using Low-Dose Antithymocyte Globulin for Relapsed and Refractory Pediatric Acute Leukemia. Kurume Med J 2021; 66:161-168. [PMID: 34421094 DOI: 10.2739/kurumemedj.ms663004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Although approximately 70% of pediatric hematological malignancies are curable, approximately 30% remain fatal. No standard treatment is available in patients showing relapse and those with refractory disease. Although different methods are adopted in different hospitals, its efficacy is extremely limited. In recent years, haploidentical stem cell transplantation, involving high-dose cyclophosphamide administration post-transplanta tion, has been used, mainly in adults; however, its application is limited to removal of alloreactive T cells. Multicenter single-arm clinical trials of T-cell replete haploidentical stem cell transplantation (TCR-haplo-SCT) will be conducted in children with relapsed and refractory acute leukemia. After myeloablative conditioning using total body irradiation or busulfan, intensive graft versus host disease prophylaxis is administered, consisting of low-dose rabbit anti-human thymocyte globulin, tacrolimus, methotrexate, and prednisolone. An external control group is set up for the study. The treatment period is around 3 months, and the follow-up period is 2 years from transplantation completion.The aim of this study is to verify the efficacy and safety of TCR-haplo-SCT and present it as a new immune cell therapy for improving survival rate in children with relapsed and refractory acute leukemia.
Collapse
Affiliation(s)
- Akiko Kada
- Clinical Research Center, National Hospital Organization Nagoya Medical Center
| | - Atsushi Kikuta
- Department of Pediatric Oncology, Fukushima Medical University Hospital
| | - Akiko M Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center
| | - Koji Kato
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital
| | | | - Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine
| | | | | | - Yoshiyuki Takahashi
- Department of Pediatrics/Developmental Pediatrics, Nagoya University Graduate School of Medicine
| | | | - Yoshiko Hashii
- Department of Pediatrics, Osaka University Graduate School of Medicine
| | | | - Kimikazu Matsumoto
- Children's Cancer Center, National Center for Child Health and Development
| | - Hiroyuki Fujisaki
- Department of Pediatrics Hematology/Oncology, Osaka City General Hospital
| | | | - Yozo Nakazawa
- Department of Pediatrics, Shinshu University School of Medicine
| | - Hideki Sano
- Department of Pediatric Oncology, Fukushima Medical University Hospital
| |
Collapse
|
18
|
Argenziano M, Tortora C, Paola AD, Pota E, Martino MD, Pinto DD, Leva CD, Rossi F. Eltrombopag and its iron chelating properties in pediatric acute myeloid leukemia. Oncotarget 2021; 12:1377-1387. [PMID: 34262648 PMCID: PMC8274721 DOI: 10.18632/oncotarget.28000] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/11/2021] [Indexed: 01/08/2023] Open
Abstract
Pediatric acute myeloid leukemia (AML) represents 20% of total childhood leukemia diagnoses and is characterized by poor prognosis with a long-term survival rate around the 50%, when patients are properly treated. The standard treatment for pediatric AML currently consists in a combination of cytarabine (Ara-C) and antracycline. Iron plays an important role in cancer development and progression. Targeting iron and its metabolism mediators could be a novel therapeutic strategy in cancer.Deferasirox (DFX) inhibits cancer cell proliferation and its use as an antiblastic drug could be suggested. Eltrombopag (ELT), a thrombopoietin receptor agonist used in immunethrombocytopenia, shows anticancer properties related to its emerging iron chelating properties. We compare the anticancer effect of classically used cytarabine with DFX and ELT effects in a pediatric AML cell line, THP-1, in order to identify innovative and more effective therapeutic strategies. ELT and DFX reduce intracellular iron concentration by inhibiting its uptake and by promoting its release. In particular, even though further investigations are needed to better understand the extact underlying action mechanisms, we demonstrated that ELT improves cytarabine antineoplastic activity in pediatric AML cell line.
Collapse
Affiliation(s)
- Maura Argenziano
- Department of Woman, Child and General and Specialist Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Chiara Tortora
- Department of Woman, Child and General and Specialist Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Alessandra Di Paola
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Elvira Pota
- Department of Woman, Child and General and Specialist Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Martina Di Martino
- Department of Woman, Child and General and Specialist Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Daniela Di Pinto
- Department of Woman, Child and General and Specialist Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Caterina Di Leva
- Department of Woman, Child and General and Specialist Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Francesca Rossi
- Department of Woman, Child and General and Specialist Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| |
Collapse
|
19
|
Arora S, Pushpam D, Tiwari A, Choudhary P, Chopra A, Gupta R, Kumar R, Bakhshi S. Allogeneic hematopoietic stem cell transplant in pediatric acute myeloid leukemia: Lessons learnt from a tertiary care center in India. Pediatr Transplant 2021; 25:e13918. [PMID: 33142026 DOI: 10.1111/petr.13918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/09/2020] [Accepted: 10/18/2020] [Indexed: 01/10/2023]
Abstract
There is paucity of data on outcomes of MSD-HSCT in children with relapsed or high-risk AML from developing countries, which have unique challenges including adverse host factors and resource constraints. We retrospectively reviewed records of children (age ≤ 18 years) who underwent MSD-HSCT for AML at our center from 2009 to 2019 to evaluate clinical outcome and its predictors using Cox proportional hazards model. There were 46 children (36 boys and 10 girls) with mean age 10.7 ± 4.8 years. Indication for HSCT was relapsed AML in CR2 (n = 37), primary refractory (n = 3), or relapsed refractory disease (n = 3); high-risk (n = 1) or secondary (n = 2) AML in CR1. Five-year EFS and OS were 33.3 ± 7.2% and 36.3 ± 7.6%, respectively. On multivariate analysis, CR1 duration less than 12 months, presence of active disease at transplant, and use of bone marrow stem cell graft were associated with poorer EFS and OS. There was one (2.2%) TRM, while disease relapse occurred in 20/40 patients who underwent HSCT in remission. Though the 5-year EFS and OS were inferior to results reported from high-income countries, relapse (and not TRM) was the major cause of treatment failure. A well-sustained CR1, achievement of disease remission, and use of peripheral blood allograft seem imperative to a successful transplant. Targeted therapy along with HSCT may be the option for those with early relapse.
Collapse
Affiliation(s)
- Shalabh Arora
- Department of Medical Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Deepam Pushpam
- Department of Medical Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Akash Tiwari
- Department of Medical Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | | | - Anita Chopra
- Unit of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Ritu Gupta
- Unit of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Rajive Kumar
- Unit of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| |
Collapse
|
20
|
Salaverria C, Plenert E, Vasquez R, Fuentes-Alabi S, Tomlinson GA, Sung L. Paediatric relapsed acute leukaemia: curative intent chemotherapy improves quality of life. BMJ Support Palliat Care 2021:bmjspcare-2020-002722. [PMID: 33455915 DOI: 10.1136/bmjspcare-2020-002722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/18/2020] [Accepted: 01/03/2021] [Indexed: 01/25/2023]
Abstract
OBJECTIVES Paediatric patients with leukaemia with relapse or induction failure have poor prognosis. Anticipated quality of life (QoL) is important in treatment decision making. The objective was to determine if curative intent at relapse or induction failure, when compared with palliative intent, was associated with child's physical health, pain or general fatigue and parents' QoL over time among patients with paediatric leukaemia in El Salvador. METHODS This was a prospective observational cohort study. Children 2-18 years with acute leukaemia at first relapse or induction failure were eligible. Assessments occurred every 2 months for up to 2 years using validated proxy report and self-report scales, where guardians were the primary respondents. Initial curative or palliative intent was categorised at enrolment by physicians. The impact of initial intent on QoL was assessed using linear mixed effects models and interaction between QoL and time. RESULTS Of the 60 families enrolled, initial treatment intent was curative in 31 (51.7%) and palliative in 29 (48.3%). During the 2-year observation period, 44 children died. Initial curative intent significantly improved child's physical health (estimate=8.4, 95% CI 5.1 to 11.6), pain (estimate=5.4, 95% CI 1.5 to 9.2) and fatigue (estimate=6.6, 95% CI 3.2 to 9.9) compared with palliative intent, but not parents' QoL (estimate=1.0, 95% CI -0.8 to 2.8). CONCLUSIONS Among paediatric patients with acute leukaemia at relapse or induction failure, initial curative intent treatment plan was associated with better physical health, pain and fatigue when compared with palliative intent. A curative approach may be a reasonable option for patients with acute leukaemia even when prognosis is poor.
Collapse
Affiliation(s)
- Carmen Salaverria
- Division of Hematology and Oncology, Hospital Nacional De Ninos Benjamin Bloom, San Salvador, El Salvador
| | - Erin Plenert
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Roberto Vasquez
- Division of Hematology and Oncology, Hospital Nacional De Ninos Benjamin Bloom, San Salvador, El Salvador
| | - Soad Fuentes-Alabi
- Division of Hematology and Oncology, Hospital Nacional De Ninos Benjamin Bloom, San Salvador, El Salvador
| | - George A Tomlinson
- Department of Medicine, Toronto General Hospital, Toronto, Ontario, Canada
| | - Lillian Sung
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Moritake H, Tanaka S, Miyamura T, Nakayama H, Shiba N, Shimada A, Terui K, Yuza Y, Koh K, Goto H, Kakuda H, Saito A, Hasegawa D, Iwamoto S, Taga T, Adachi S, Tomizawa D. The outcomes of relapsed acute myeloid leukemia in children: Results from the Japanese Pediatric Leukemia/Lymphoma Study Group AML-05R study. Pediatr Blood Cancer 2021; 68:e28736. [PMID: 32991072 DOI: 10.1002/pbc.28736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/04/2020] [Accepted: 09/06/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND The prognosis of children with acute myeloid leukemia (AML) has improved with the efficacy of hematopoietic cell transplantation (HCT) as a second-line therapy and improvements in supportive care following anthracycline- and cytarabine-based chemotherapy; however, the outcomes of children with relapsed AML still remain unsatisfactory. PROCEDURE In order to identify prognostic factors and improve their prognosis, we analyzed 111 patients who relapsed after treatment with the Japanese Pediatric Leukemia/Lymphoma Study Group (JPLSG) AML-05 protocol and who were registered in the retrospective JPLSG AML-05R study. RESULTS The 5-year overall survival rate was 36.1%. The major determinant of survival was duration from the diagnosis to relapse. The mean duration in the nonsurviving group (10.1 ± 4.1 months) was shorter than that in the surviving group (16.3 ± 8.3 months) (P < .01). Moreover, achieving a second complete remission (CR2) prior to HCT was associated with a good prognosis (P < .01). Etoposide, cytarabine, and mitoxantrone (ECM)- or fludarabine, cytarabine, and granulocyte colony-stimulating factor (FLAG)-based regimens were therefore recommended for reinduction therapy (P < .01). A genetic analysis also revealed the prognostic significance of FMS-like tyrosine kinase 3 (FLT3)-internal tandem duplication as a poor prognostic marker (P = .04) and core binding factor-AML, t(8;21), and inv(16) as good prognostic markers (P < .01). CONCLUSIONS Achieving a CR2 prior to HCT is important in order to improve the prognosis of relapsed pediatric AML. Recent molecular targeted therapies, such as FLT3 inhibitors, may contribute to overcome their prognoses. Larger prospective investigations are necessary to establish individualized treatment strategies for patients with relapsed childhood AML.
Collapse
Affiliation(s)
- Hiroshi Moritake
- Division of Pediatrics, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Shiro Tanaka
- Department of Clinical Biostatistics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takako Miyamura
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hideki Nakayama
- Department of Pediatrics, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Norio Shiba
- Department of Pediatrics, Yokohama City University Hospital, Yokohama, Japan
| | - Akira Shimada
- Department of Pediatrics, Okayama University, Okayama, Japan
| | - Kiminori Terui
- Department of Pediatrics, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yuki Yuza
- Department of Hematology/Oncology, Tokyo Metropolitan Children's Medical Center, Fuchu, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Hiroaki Goto
- Division of Hemato-oncology/Regenerative Medicine, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Harumi Kakuda
- Department of Hematology/Oncology, Chiba Children's Hospital, Chiba, Japan
| | - Akiko Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Daisuke Hasegawa
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan
| | - Shotaro Iwamoto
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Japan
| | - Takashi Taga
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Japan
| | - Souichi Adachi
- Department of Human Health Sciences, Kyoto University, Kyoto, Japan
| | - Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
22
|
Lamble AJ, Gardner R. CAR T cells for other pediatric non-B-cell hematologic malignancies. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2020; 2020:494-500. [PMID: 33275678 PMCID: PMC7727568 DOI: 10.1182/hematology.2020000134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
As CAR T-cell therapy has advanced in B-cell acute lymphoblastic leukemia, research is now underway to develop similar therapies for other lymphoid and myeloid malignancies for pediatric patients. Barriers, including antigen selection and on-target/off-tumor toxicity, have prevented the rapid development of immune-based therapies for T-lineage and myeloid malignancies. More recently, unique strategies have been developed to overcome these barriers, with several products advancing to clinical trials. For T-lineage diseases, targets have focused on CD5, CD7, and CD38, whereas myeloid disease targets have predominately focused on CD123, CD33, and, more recently, CLL-1. This review provides a comprehensive overview of these targets and approaches to overcoming safety concerns in the development of CAR T-cell therapies for pediatric patients with T-lineage and myeloid malignancies.
Collapse
Affiliation(s)
- Adam J. Lamble
- Division of Hematology-Oncology, Seattle Children’s Hospital, Seattle, WA; and
- University of Washington School of Medicine, Seattle, WA
| | - Rebecca Gardner
- Division of Hematology-Oncology, Seattle Children’s Hospital, Seattle, WA; and
- University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
23
|
Expanded activated autologous lymphocyte infusions improve outcomes of low- and intermediate-risk childhood acute myeloid leukemia with low level of minimal residual disease. Cancer Lett 2020; 493:128-132. [PMID: 32829005 DOI: 10.1016/j.canlet.2020.08.003] [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: 02/25/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 11/22/2022]
Abstract
The presence of minimal residual disease (MRD) is a risk factor for relapse among children with acute myeloid leukemia (AML), and eliminating MRD can usually improve survival rates. To investigate the effect of expanded activated autologous lymphocytes (EAALs) combined with chemotherapy on eliminating MRD and improving survival rates of children with AML, we retrospectively analyzed the results of 115 children with low- or intermediate-risk AML with MRD treated at the Pediatric Hematological Center, Peking University People's Hospital, between January 2010 and January 2016. The patients were assigned to the chemotherapy plus EAAL (combined therapy) group (n = 61) and chemotherapy group (n = 54). The MRD-negativity rates were 95.1% (58/61) in the combined therapy group and 63.0% (34/54) in the chemotherapy group (P < 0.0001) during consolidation treatment. The 5-year event-free survival rate was higher in the combined therapy group than in the chemotherapy group (86.3 ± 4.6% vs. 72.1 ± 6.1%, P = 0.025). No severe adverse event was observed after EAAL infusion. The present study showed that EAAL combined with chemotherapy could improve the MRD-negativity rate and event-free survival rate among children with AML with low level MRD-positive status.
Collapse
|
24
|
Wang Y, Su H, Yan M, Zhang L, Tang J, Li Q, Gu X, Gong Q. Interleukin-33 Promotes Cell Survival via p38 MAPK-Mediated Interleukin-6 Gene Expression and Release in Pediatric AML. Front Immunol 2020; 11:595053. [PMID: 33324412 PMCID: PMC7726021 DOI: 10.3389/fimmu.2020.595053] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 10/28/2020] [Indexed: 11/30/2022] Open
Abstract
Acute myeloid leukemia (AML) is a fatal disease characterized by the accumulation of immature myeloid blasts in the bone marrow (BM). Cytokine provide signals for leukemia cells to improve their survival in the BM microenvironment. Previously, we identified interleukin-33 (IL-33) as a promoter of cell survival in a human AML cell line and primary mouse leukemia cells. In this study, we report that the cell surface expression of IL-33–specific receptor, Interleukin 1 Receptor Like 1 (IL1RL1), is elevated in BM cells from AML patients at diagnosis, and the serum level of IL-33 in AML patients is higher than that of healthy donor controls. Moreover, IL-33 levels are found to be positively associated with IL-6 levels in pediatric patients with AML. In vitro, IL-33 treatment increased IL-6 mRNA expression and protein level in BM and peripheral blood (PB) cells from AML patients. Evidence was also provided that IL-33 inhibits cell apoptosis by activating p38 mitogen-activated protein kinase (MAPK) pathway using human AML cell line and AML patient samples. Finally, we confirmed that IL-33 activated IL-6 expression in a manner that required p38 MAPK pathway using clinical AML samples. Taken together, we identified a potential mechanism of IL-33–mediated survival involving p38 MAPK in pediatric AML patients that would facilitate future drug development.
Collapse
Affiliation(s)
- Yiqian Wang
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Haibo Su
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Muxia Yan
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Li Zhang
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jiancheng Tang
- Department of Anesthesiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Quanxin Li
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xiaoqiong Gu
- Department of Blood Transfusion, Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qing Gong
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
25
|
Hoffman AE, Schoonmade LJ, Kaspers GJ. Pediatric relapsed acute myeloid leukemia: a systematic review. Expert Rev Anticancer Ther 2020; 21:45-52. [PMID: 33111585 DOI: 10.1080/14737140.2021.1841640] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Pediatric relapsed acute myeloid leukemia (AML) remains lethal in the majority of cases, despite intensive therapy. Randomized trials are largely lacking, and the main issues of optimal therapy and prognostic factors remain unclear. Area covered: This systematic review includes all literature evaluating treatment outcome after first relapse. We searched databases PubMed and Embase.com. Twelve out of six thousand articles were ultimately included, based on age of the population (<21 years), relapsed AML, and information on clinical outcome (second complete remission (CR2), disease-free survival (DFS), event-free survival (EFS) and overall survival (OS)). There was only one randomized clinical trial reported. This review shows that there is no standard treatment for relapsed AML in children, and that outcome varies for CR2 and (2- to 10-year) OS rates, mean 64% (range, 50-75%), and 31% (16-43%), respectively. Children treated with chemotherapy only in first complete remission (CR1) tend to have better outcome after relapse than children receiving allo-SCT in CR1. Allo-SCT seems to be the most effective consolidation therapy in children achieving CR2, after relapse. Duration of CR1 was the most frequently reported statistically significant prognostic factor. Through randomized clinical trials, better knowledge of prognostic factors enabling risk-stratified treatment, and of more effective and less toxic therapies, should contribute to better clinical outcome for children with relapsed AML. Expert opinion: Outcome of pediatric relapsed AML has improved to OS rates up to 40%. However, there is a lack of knowledge on (independent) prognostic factors, optimal reinduction chemotherapy, timing of allo-SCT, and late effects. International collaboration should enable large, randomized clinical trials addressing these issues.
Collapse
Affiliation(s)
- Anne E Hoffman
- Emma Children's Hospital, Amsterdam UMC, Pediatric Oncology, Vrije Universiteit Amsterdam , Amsterdam
| | - Linda J Schoonmade
- Medical Library, Vrije Universiteit Amsterdam , Amsterdam, The Netherlands
| | - Gertjan Jl Kaspers
- Emma Children's Hospital, Amsterdam UMC, Pediatric Oncology, Vrije Universiteit Amsterdam , Amsterdam.,Princess Máxima Center For Pediatric Oncolocy , The Netherlands, Utrecht
| |
Collapse
|
26
|
Xue YJ, Cheng YF, Lu AD, Wang Y, Zuo YX, Yan CH, Suo P, Zhang LP, Huang XJ. Efficacy of Haploidentical Hematopoietic Stem Cell Transplantation Compared With Chemotherapy as Postremission Treatment of Children With Intermediate-risk Acute Myeloid Leukemia in First Complete Remission. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 21:e126-e136. [PMID: 33060049 DOI: 10.1016/j.clml.2020.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND The role of haploidentical hematopoietic stem cell transplantation (haplo-HSCT) for children with intermediate-risk acute myeloid leukemia (IR-AML) in first complete remission has been controversial. The present study compared the effect of chemotherapy with unmanipulated haplo-HSCT as treatment of patients with IR-AML in first complete remission (CR1). PATIENTS AND METHODS We retrospectively analyzed the outcomes of 80 children with IR-AML and compared the effects of chemotherapy (n = 47) with those of haplo-HSCT (n = 33) as treatment in CR1. RESULTS The 3-year overall survival, event-free survival (EFS), and cumulative incidence of relapse (CIR) was 85.4% ± 4.1%, 73.2% ± 5.0%, and 25.4% ± 4.5%, respectively. Compared with the chemotherapy group, the patients in the haplo-HSCT group had a lower CIR (P = .059) and better EFS (P = .108), but roughly equivalent overall survival (P = .841). Multivariate analysis revealed chemotherapy and minimal residual disease (MRD) of ≥ 10-3 after induction therapy as independent risk factors affecting CIR and EFS. EFS (P = .045) and CIR (P = .045) differed significantly between the 2 treatment groups in patients with MRD of ≥ 10-3 after induction therapy. CONCLUSION Haplo-HSCT might be a feasible option for children with IR-AML in CR1, especially for patients with MRD of ≥ 10-3 after induction therapy.
Collapse
Affiliation(s)
- Yu-Juan Xue
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Yi-Fei Cheng
- Department of Hematology, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Peking-Tsinghua Center for Life Sciences and Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| | - Ai-Dong Lu
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Yu Wang
- Department of Hematology, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Peking-Tsinghua Center for Life Sciences and Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying-Xi Zuo
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Chen-Hua Yan
- Department of Hematology, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Peking-Tsinghua Center for Life Sciences and Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| | - Pan Suo
- Department of Hematology, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Peking-Tsinghua Center for Life Sciences and Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| | - Le-Ping Zhang
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China.
| | - Xiao-Jun Huang
- Department of Hematology, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Peking-Tsinghua Center for Life Sciences and Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
27
|
Garg A, Ganguly S, Vishnubhatla S, Chopra A, Bakhshi S. Outpatient ADE (cytarabine, daunorubicin, and etoposide) is feasible and effective for the first relapse of pediatric acute myeloid leukemia: A prospective, phase II study. Pediatr Blood Cancer 2020; 67:e28404. [PMID: 32672904 DOI: 10.1002/pbc.28404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/26/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Approximately 40% children with acute myeloid leukemia (AML) invariably relapse, after attaining the first complete remission (CR), with dismal long-term outcome. There is little consensus regarding choice of optimal induction chemotherapy regimen for relapsed pediatric AML. PROCEDURE A prospective single arm phase II study (CTRI/2017/02/007757) was carried out at our center to evaluate the safety and efficacy of outpatient cytarabine, daunorubicin, and etoposide (ADE) regimen in pediatric AML (≤18 years) at the first relapse. Response evaluation was done by bone marrow aspiration morphology along with minimal residual disease (MRD) assessment. All adverse events including need and duration of hospitalization, transfusion support, and antimicrobial use were recorded. RESULTS Total 45 patients were included with median age of 12 years. The CR rate of the cohort was 66% and 54% of patients were MRD negative. The estimated 2-year event-free survival (EFS) and overall survival (OS) were 29% (±7%) and 34% (±7%), respectively. The presence of fever at relapse was associated with inferior CR rate (P = .001), positive MRD (P = .01), and inferior EFS (P = .02), while not achieving nadir absolute neutrophil count of zero during induction was associated with inferior CR rate (P = .03) and inferior OS (P = .04). Approximately all patients developed ≥Grade 3 cytopenia and febrile neutropenia. Twenty-six (59%) patients required hospitalization for management of toxicity and there were four (9%) deaths attributed to infection. CONCLUSION ADE is an effective induction regimen for pediatric AML patients at the first relapse with reasonable toxicity profile. Outpatient administration of the regimen is feasible in the presence of proper support structure and rigorous follow up.
Collapse
Affiliation(s)
- Arun Garg
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Shuvadeep Ganguly
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | | | - Anita Chopra
- Department of Laboratory Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
28
|
Successful Treatment of Relapsed Pediatric Acute Myeloid Leukemia Presenting as Central Nervous System Myeloid Sarcoma: A Single-Institution Case Series. J Pediatr Hematol Oncol 2020; 42:319-321. [PMID: 30543582 DOI: 10.1097/mph.0000000000001383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Relapsed acute myeloid leukemia presenting as an isolated central nervous system myeloid sarcoma (CNS MS) is very rare and generally entails poor outcomes. CNS MS treatment is not well defined and can include systemic chemotherapy, intrathecal chemotherapy, radiation therapy, or hematopoietic stem cell transplant. Thiotepa, vinorelbine, topotecan, and clofarabine (TVTC) has been successful for reinduction therapy in relapsed/refractory leukemia to induce remission before hematopoietic stem cell transplant. There is no published evidence of TVTC being utilized for CNS MS. In this series, we report 2 symptomatic patients with isolated CNS MS at relapse who demonstrated near complete resolution after reinduction with TVTC and additional intrathecal chemotherapy.
Collapse
|
29
|
Venetoclax for paediatric acute myeloid leukaemia: a step forward. Lancet Oncol 2020; 21:476-478. [DOI: 10.1016/s1470-2045(20)30136-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/16/2020] [Accepted: 02/17/2020] [Indexed: 11/20/2022]
|
30
|
Hazar V, Karasu GT, Uygun V, Özbek N, Karakükçü M, Öztürk G, Daloğlu H, Kılıç SÇ, Aksu T, Ünal E, Koçak Ü, Yeşilipek A, Akçay A, Gürsel O, Küpesiz A, Okur FV, İleri T, Kansoy S, Bayram İ, Karagün BŞ, Gökçe M, Kaya Z, Ok Bozkaya İ, Patıroğlu T, Aksoylar S. Role of a second transplantation for children with acute leukemia following posttransplantation relapse: a study by the Turkish Bone Marrow Transplantation Study Group. Leuk Lymphoma 2020; 61:1465-1474. [PMID: 32037917 DOI: 10.1080/10428194.2020.1716220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
We examined outcomes of 51 pediatric patients with relapsed acute leukemia (AL) who underwent a second allogeneic hematopoietic stem cell transplantation (alloHSCT). After a median follow-up of 941 days (range, 69-2842 days), leukemia-free survival (LFS) and overall survival (OS) at 3 years were 26.6% and 25.6%, respectively. The nonrelapse mortality rate (NMR) and cumulative incidence of relapse (CIR) were 36.4% and 42.4%, respectively. The Cox regression analysis demonstrated that the risk factors at second transplantation for predicting limited LFS were active disease (hazard ratio (HR) = 5.1), reduced intensity conditioning (RIC) (HR = 5.0), matched unrelated donor (MUD) (HR = 3.4) and performance score <80 (HR = 3.2). Pediatric patients with AL who relapsed after their first alloHSCT may survive with a second alloHSCT. Disease status, conditioning intensity, donor type, and performance score at the second transplantation are the relevant risk factors. A score based on these factors may predict the results of the second transplantation.
Collapse
Affiliation(s)
- Volkan Hazar
- Pediatric BMT Unit, Medical Park Göztepe Hospital, Istanbul, Turkey
| | | | - Vedat Uygun
- Pediatric BMT Unit, Medical Park Antalya Hospital, Antalya, Turkey
| | - Namık Özbek
- Pediatric BMT Unit, Ankara Dışkapı Child Health and Diseases Hematology Oncology Training and Research Hospital, Ankara, Turkey
| | - Musa Karakükçü
- Pediatric BMT Unit, Erciyes University Faculty of Medicine, Kayseri, Turkey
| | - Gülyüz Öztürk
- Pediatric BMT Unit, Acıbadem University Faculty of Medicine, Acıbadem Atakent Hospital, Istanbul, Turkey
| | - Hayriye Daloğlu
- Pediatric BMT Unit, Medical Park Antalya Hospital, Antalya, Turkey
| | - Suar Çakı Kılıç
- Pediatric BMT Unit, Medical Park Göztepe Hospital, Istanbul, Turkey
| | - Tekin Aksu
- Pediatric BMT Unit, Ankara Dışkapı Child Health and Diseases Hematology Oncology Training and Research Hospital, Ankara, Turkey
| | - Ekrem Ünal
- Pediatric BMT Unit, Erciyes University Faculty of Medicine, Kayseri, Turkey
| | - Ülker Koçak
- Pediatric BMT Unit, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Akif Yeşilipek
- Pediatric BMT Unit, Medical Park Antalya Hospital, Antalya, Turkey
| | - Arzu Akçay
- Pediatric BMT Unit, Acıbadem University Faculty of Medicine, Acıbadem Atakent Hospital, Istanbul, Turkey
| | - Orhan Gürsel
- Pediatric BMT Unit, University of Health Sciences Faculty of Medicine, Ankara GATA Hospital, Ankara, Turkey
| | - Alphan Küpesiz
- Pediatric BMT Unit, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Fatma Visal Okur
- Pediatric BMT Unit, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Talia İleri
- Pediatric BMT Unit, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Savaş Kansoy
- Pediatric BMT Unit, Ege University Faculty of Medicine, Izmir, Turkey
| | - İbrahim Bayram
- Pediatric BMT Unit, Çukurova University Faculty of Medicine, Adana, Turkey
| | | | - Müge Gökçe
- Pediatric BMT Unit, Yüzüncü Yıl University Faculty of Medicine, GOP Hospital, Istanbul, Turkey
| | - Zühre Kaya
- Pediatric BMT Unit, Gazi University Faculty of Medicine, Ankara, Turkey
| | - İkbal Ok Bozkaya
- Pediatric BMT Unit, Ankara Dışkapı Child Health and Diseases Hematology Oncology Training and Research Hospital, Ankara, Turkey
| | - Türkan Patıroğlu
- Pediatric BMT Unit, Erciyes University Faculty of Medicine, Kayseri, Turkey
| | - Serap Aksoylar
- Pediatric BMT Unit, Ege University Faculty of Medicine, Izmir, Turkey
| |
Collapse
|
31
|
Smith AM, Zhang CRC, Cristino AS, Grady JP, Fink JL, Moore AS. PTEN deletion drives acute myeloid leukemia resistance to MEK inhibitors. Oncotarget 2019; 10:5755-5767. [PMID: 31645898 PMCID: PMC6791388 DOI: 10.18632/oncotarget.27206] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 08/12/2019] [Indexed: 12/31/2022] Open
Abstract
Kinases such as MEK are attractive targets for novel therapy in cancer, including acute myeloid leukaemia (AML). Acquired and inherent resistance to kinase inhibitors, however, is becoming an increasingly important challenge for the clinical success of such therapeutics, and often arises from mutations in the drug-binding domain of the target kinase. To identify possible causes of resistance to MEK inhibition, we generated a model of resistance by long-term treatment of AML cells with AZD6244 (selumetinib). Remarkably, resistance to MEK inhibition was due to acquired PTEN haploinsufficiency, rather than mutation of MEK. Resistance via this mechanism was confirmed using CRISPR/Cas9 technology targeting exon 5 of PTEN. While PTEN loss has been previously implicated in resistance to a number of other therapeutic agents, this is the first time that it has been shown directly and in AML.
Collapse
Affiliation(s)
- Amanda M Smith
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Australia.,Current address: Washington University in Saint Louis, Saint Louis, Missouri, United States of America
| | - Christine R C Zhang
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Australia.,Current address: Washington University in Saint Louis, Saint Louis, Missouri, United States of America
| | - Alexandre S Cristino
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Australia.,Current address: Griffith Institute for Drug Discovery, Brisbane Innovation Park, Nathan, Australia
| | - John P Grady
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Australia.,Current address: Garvan Institute of Medical Research, Darlinghurst, Australia
| | - J Lynn Fink
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Australia
| | - Andrew S Moore
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Australia.,Oncology Services Group, Queensland Children's Hospital, South Brisbane, Australia.,Child Health Research Centre, The University of Queensland, South Brisbane, Australia.,Current address: Washington University in Saint Louis, Saint Louis, Missouri, United States of America
| |
Collapse
|
32
|
Zhang N, Chen Y, Lou S, Shen Y, Deng J. A six-gene-based prognostic model predicts complete remission and overall survival in childhood acute myeloid leukemia. Onco Targets Ther 2019; 12:6591-6604. [PMID: 31496748 PMCID: PMC6701647 DOI: 10.2147/ott.s218928] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/26/2019] [Indexed: 01/07/2023] Open
Abstract
Objective Acute myeloid leukemia (AML) is a malignant clonal disorder. Despite enormous progress in its diagnosis and treatment, the mortality rate of AML remains high. The aim of this study was to identify prognostic biomarkers by using the gene expression profile dataset from public database, and to improve the risk-stratification criteria of survival for patients with AML. Materials and methods The gene expression data and clinical parameter were acquired from the Therapeutically Applicable Research to Generate Effective Treatment (TARGET) database. A total of 856 differentially expressed genes (DEGs) were obtained from the childhood AML patients classified into first complete remission (CR1) group (n=791) and not CR group (n=249). We performed a series of bioinformatics analysis to screen key genes and pathways, further comprehending these DEGs through Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Results Six genes (SLC17A7, MSX2, CDC26, MSLN, CTSZ and DEFA3) identified by univariate, Kaplan-Meier survival and multivariate Cox regression analyses were used to develop the prognostic model. Further analysis showed that the survival estimations in the high-risk group had an increased risk of death compared with the low-risk group based on the model. The area under the curve of the receiver operator characteristic curve in the prognostic model for predicting the overall survival was 0.729, confirming good prognostic model. We also performed a nomogram to provide an individual patient with the overall probability, and internal validation in the TARGET cohort. Conclusion We identified a six-gene prognostic signature for risk-stratifying in patients with childhood AML. The risk classification model can be used to predict CR markers and may assist clinicians in providing realize the individualized treatment in this patient population.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Ying Chen
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Shifeng Lou
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Yan Shen
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Jianchuan Deng
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, People's Republic of China
| |
Collapse
|
33
|
High-dose etoposide and cyclophosphamide in adults and children with primary refractory and multiply relapsed acute leukaemias: The Royal Marsden experience. Leuk Res 2019; 85:106217. [PMID: 31493701 DOI: 10.1016/j.leukres.2019.106217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/11/2019] [Accepted: 08/12/2019] [Indexed: 11/23/2022]
Abstract
INTRODUCTION For patients with primary refractory and relapsed acute leukaemias allogeneic stem cell transplantation is the only hope for cure, but morphological remission is not always achieved after standard salvage regimens. Here we review the experience with high-dose etoposide and cyclophosphamide (HD-Et/Cy) in relapsed/refractory acute leukaemias at the Royal Marsden Hospital. PATIENTS AND METHODS Twenty-three patients (15 adults, 8 children) with refractory/relapsed acute myeloblastic (n = 18; 78%), lymphoblastic (n = 4; 17%) or biphenotypic (n = 1; 4%) leukaemia who had failed to respond to at least one previous line of chemotherapy received HD-Et/Cy at our institution between 2006 and 2015. RESULTS Overall response rate was 21.7% (95%CI 4.0-40.0). Median overall survival was 14.8 months (95%CI 9.1-49.1). Eight (35%) patients (7 AML, 1 biphenotypic leukaemia) proceeded to allogeneic transplant after one cycle of HD-Et/Cy: four of them (50%; 3 adults, 1 child) in complete remission and another four children (50%) with aplastic bone marrow with scattered blasts. Among the transplant recipients, three with AML (38%), ie. one adult (responder) and two children with aplastic bone marrow with scattered blasts, became long-term survivors 9.8, 4.4 and 2.5 years post-HD-Et/Cy, respectively. Toxicity profile was comparable to similar regimens with no treatment-related deaths. The most common grade 3-4 toxicity was febrile neutropenia (96%). CONCLUSIONS HD-Et/Cy can salvage patients with refractory/relapsed AML who remain candidates for allogeneic stem cell transplantation after failure of standard salvage regimens and do not have access to clinical trials.
Collapse
|
34
|
Selim A, Alvaro F, Cole CH, Fraser CJ, Mechinaud F, O'Brien TA, Shaw PJ, Tapp H, Teague L, Nivison-Smith I, Moore AS. Hematopoietic stem cell transplantation for children with acute myeloid leukemia in second remission: A report from the Australasian Bone Marrow Transplant Recipient Registry and the Australian and New Zealand Children's Haematology Oncology Group. Pediatr Blood Cancer 2019; 66:e27812. [PMID: 31111633 DOI: 10.1002/pbc.27812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/10/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Approximately one-third of children with acute myeloid leukemia (AML) relapse, requiring re-treatment and allogeneic hematopoietic stem cell transplantation (HSCT). Although achieving second complete remission (CR2) prior to HSCT is desirable, once CR2 is attained, it is unclear if there is any benefit from further chemotherapy prior to HSCT. Moreover, although pre-HSCT minimal residual disease (MRD) has prognostic value in acute lymphoblastic leukemia, the benefit of MRD reduction after achieving CR prior to HSCT is less clear for AML. PROCEDURE To address these questions, we analyzed data from pediatric transplant centers in Australia and New Zealand concerning relapsed childhood AML cases occurring between 1998 and 2013. Given the retrospective nature of our analysis and assay data available, we analyzed patients on the basis of measurable residual disease (MeRD) by any methodology, rather than MRD in the conventional sense. RESULTS We observed improved overall survival (OS) in children receiving two chemotherapy cycles, compared to one cycle or three or more cycles pre-HSCT. Improved OS with two cycles remained significant for patients without MeRD after cycle 1. CONCLUSIONS These data suggest that a second chemotherapy cycle pre-HSCT may improve survival by lowering disease burden. Prospective trials assessing strategies to reduce pre-HSCT MRD in relapsed childhood AML are warranted.
Collapse
Affiliation(s)
- Adrian Selim
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | | | - Catherine H Cole
- Department of Haematology/Oncology, Princess Margaret Hospital for Children, Perth, Australia
| | - Chris J Fraser
- Oncology Services Group, Queensland Children's Hospital, Brisbane, Australia
| | | | - Tracey A O'Brien
- Kids' Cancer Centre, Sydney Children's Hospital, Sydney, Australia
| | - Peter J Shaw
- Oncology Unit, The Children's Hospital at Westmead, Sydney, Australia
| | - Heather Tapp
- Department of Clinical Haematology/Oncology, Women's and Children's Hospital, Adelaide, Australia
| | | | - Ian Nivison-Smith
- Australasian Bone Marrow Transplant Recipient Registry, Sydney, Australia
| | - Andrew S Moore
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia.,Oncology Services Group, Queensland Children's Hospital, Brisbane, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, Australia
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Incorporation of minimal residual disease (MRD) testing in acute lymphoblastic leukemia (ALL) and acute myeloblastic leukemia (AML) has transformed the landscape of hematopoietic cell transplantation (HCT). Pre-HCT MRD has allowed prognostication of HCT outcomes for high-risk leukemia patients, whereas the detection of post-HCT MRD has allowed for interventions to decrease relapse. RECENT FINDINGS In this review, we emphasize studies from the past two decades that highlight the critical role of MRD in HCT in pediatric ALL and AML. Advances in MRD detection methodology, using next-generation sequencing, have improved the sensitivity of MRD testing allowing for more accurate predictions of HCT outcomes for patients with relapsed and refractory ALL and AML. In addition, novel pre-HCT therapies, especially immunotherapy in ALL, have dramatically increased the number of patients who achieve MRD-negative remissions pre-HCT, resulting in improved HCT outcomes. Post-HCT MRD remains a challenge and new therapeutic interventions are needed to reduce post-HCT relapse. SUMMARY As immunotherapy increases pre-HCT MRD-negative remissions, and next-generation sequencing-MRD is incorporated to improve the sensitivity of MRD detection, future clinical studies will investigate less toxic HCT approaches to reduce long-term sequelae and to identify which patients may benefit most from early post-HCT intervention to reduce relapse.
Collapse
Affiliation(s)
- Agne Taraseviciute
- Division of Pediatric Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | | |
Collapse
|
36
|
Broglie L, Gershan J, Burke MJ. Checkpoint inhibition of PD-L1 and CTLA-4 in a child with refractory acute leukemia. Int J Hematol Oncol 2019; 8:IJH10. [PMID: 30863527 PMCID: PMC6410023 DOI: 10.2217/ijh-2018-0009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/11/2018] [Indexed: 01/22/2023] Open
Abstract
Children with multiple relapsed or refractory leukemia have dismal survival. Research has identified engagement of immune checkpoint receptors (e.g., PD-1, PD-L1 and CTLA-4) as a mechanism for treatment resistance. For adult cancer, inhibitors of PD-1 (nivolumab) and CTLA-4 (ipilimumab) have shown promise with response rates ranging from 7 to 40%. In vitro studies using acute myeloid leukemia cell lines have shown that acute myeloid leukemia blasts may similarly utilize the PD-1/PD-L1 axis to evade an anticancer immune response. We report the first case of a pediatric patient with multiple relapsed/refractory leukemia treated with nivolumab, ipilimumab and 5-azacytidine who tolerated therapy with brief improvement of symptoms.
Collapse
Affiliation(s)
- Larisa Broglie
- Department of Pediatrics, Division of Hematology/Oncology & Blood & Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI, 53226, USA,Department of Pediatric, Division of Hematology/Oncology & Blood & Marrow Transplantation, Columbia University Medical Center, New York, NY, 10027, USA
| | - Jill Gershan
- Department of Pediatrics, Division of Hematology/Oncology & Blood & Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Michael J Burke
- Department of Pediatrics, Division of Hematology/Oncology & Blood & Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI, 53226, USA,*Author for correspondence: Tel.: +1 414 955 4170; Fax: +1 414 955 6543;
| |
Collapse
|
37
|
Fedorova NE, Chernoryzh YY, Vinogradskaya GR, Emelianova SS, Zavalyshina LE, Yurlov KI, Zakirova NF, Verbenko VN, Kochetkov SN, Kushch AA, Ivanov AV. Inhibitor of polyamine catabolism MDL72.527 restores the sensitivity to doxorubicin of monocytic leukemia Thp-1 cells infected with human cytomegalovirus. Biochimie 2018; 158:82-89. [PMID: 30578923 DOI: 10.1016/j.biochi.2018.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/17/2018] [Indexed: 12/17/2022]
Abstract
Leukemic cells from different patients exhibit different sensitivity to anticancer drugs including doxorubicin (DOX). Resistance to chemotherapy decreases efficacy of the treatment and promotes cancer recurrence and metastases. One of the approaches to overcome drug resistance includes E2F1-mediated regulation of the р73 protein that belongs to the р53 family. Its ΔNp73 isoform exhibits pro-oncogenic effects, and TAp73 - anti-oncogenic effects. Human cytomegalovirus (HCMV), often found in tumors, suppresses pro-apoptotic pathways and E2F1/p73 in particular. The activity of E2F1 and p73 transcription factors is linked to metabolism of biogenic polyamines. Therefore, it could be suggested that compounds that target polyamine-metabolizing enzymes can sensitize HCMV-infected hematological malignancies to doxorubicin. Here we report that HCMV infection of ТНР-1 monocytic leukemic cells considerably elevates E2F1 levels and shifts the balance between the р73 isoforms towards ΔNp73 leading to survival of DOX-treated leukemic cells. In contrast, MDL72.527, an inhibitor of polyamine catabolism, decreases ΔNp73/ТАр73 ratio and thus restores sensitivity of the cells to DOX. Our findings indicate the combination of doxorubicin and MDL72.527 may present a novel strategy for therapy of leukemia in patients with and without HCMV infection.
Collapse
Affiliation(s)
- Natalia E Fedorova
- Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Yana Yu Chernoryzh
- Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Galina R Vinogradskaya
- Konstantinov Petersburg Nuclear Physics Institute, National Research Center "Kurchatov Institute", Gatchina, Leningrad Region, Russia
| | - Svetlana S Emelianova
- Konstantinov Petersburg Nuclear Physics Institute, National Research Center "Kurchatov Institute", Gatchina, Leningrad Region, Russia
| | - Larisa E Zavalyshina
- Educational Institution of Further Professional Education «Russian Medical Academy of Continuous Professional Education» of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Kirill I Yurlov
- Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Natalia F Zakirova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Valery N Verbenko
- Konstantinov Petersburg Nuclear Physics Institute, National Research Center "Kurchatov Institute", Gatchina, Leningrad Region, Russia
| | - Sergey N Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alla A Kushch
- Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Alexander V Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
38
|
Mustafa O, Abdalla K, AlAzmi AA, Elimam N, Abrar MB, Jastaniah W. FLAG/FLAG-IDA regimen for children with relapsed/refractory acute leukemia in the era of targeted novel therapies. J Oncol Pharm Pract 2018; 25:1831-1838. [PMID: 30518307 DOI: 10.1177/1078155218817816] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Outcomes of relapsed/refractory childhood acute leukemia remain poor. We analyzed the safety/efficacy of fludarabine, cytarabine, and granulocyte colony stimulating factor, with/without idarubicin (FLAG ± IDA) as salvage therapy compared with recent published results of novel therapies. METHODS This retrospective study included children aged 1 to 15 years with relapsed/refractory acute leukemia who received FLAG ± IDA salvage therapy from January 2000 to December 2014. Patients with infant leukemia, mixed lineage leukemia, Philadelphia-positive acute leukemia, or secondary leukemia were excluded. RESULT Fifty patients were identified: 25 with acute lymphoblastic leukemia and 25 with acute myeloid leukemia. The median age at initiation of FLAG ± IDA was seven years. Site of relapse was the bone marrow in 29, isolated central nervous system in 11, and combined in 10 patients. FLAG ± IDA was used after first relapse in 68% and after multiple relapses in 32%. Complete remission was achieved in 34 (68%) patients. No variables predictive of complete remission were identified. Grade 3 or greater toxicity was observed in 96% and 6% died from toxicity. Toxicities included hematologic toxicity (96%), infection (52%), and enterocolitis (28%). Twenty-four of 50 (48%) patients achieved a sustained complete remission and survived to bone marrow transplantation. The five-year overall survival was 23.9% ± 6.9%. Patients achieving second complete remission and patients proceeding to bone marrow transplantation following second complete remission demonstrated significantly improved overall survival (p = 0.001). CONCLUSION Despite a 68% complete remission rate using FLAG ± IDA, only 48% of patients survived to bone marrow transplantation. The regimen was associated with 96% toxicity and only one in four patients was alive at five years. This underscores the need to find more effective lower toxicity salvage regimens.
Collapse
Affiliation(s)
- Omima Mustafa
- Princess Noorah Oncology Center, King Abdulaziz Medical City, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Khalid Abdalla
- Princess Noorah Oncology Center, King Abdulaziz Medical City, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Aeshah A AlAzmi
- Department of Pharmaceutical Care, Clinical Pharmacy, King Abdulaziz Medical City, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Naglla Elimam
- Princess Noorah Oncology Center, King Abdulaziz Medical City, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Mohammed Burhan Abrar
- Princess Noorah Oncology Center, King Abdulaziz Medical City, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Wasil Jastaniah
- Princess Noorah Oncology Center, King Abdulaziz Medical City, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia.,Department of Pediatrics, Faculty of Medicine, Umm AlQura University, Makkah, Saudi Arabia
| |
Collapse
|
39
|
Identifying Prognostic Factors That Influence Outcome of Childhood Acute Myeloid Leukemia in First Relapse in Saudi Arabia: Results of the Multicenter SAPHOS Study. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:773-780. [DOI: 10.1016/j.clml.2018.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/28/2018] [Accepted: 09/04/2018] [Indexed: 01/17/2023]
|
40
|
Tai J, Wang S, Zhang J, Ge W, Liu Y, Li X, Liu Y, Deng Z, He L, Wang G, Ni X. Up-regulated lipocalin-2 in pediatric thyroid cancer correlated with poor clinical characteristics. Eur Arch Otorhinolaryngol 2018; 275:2823-2828. [PMID: 30182199 DOI: 10.1007/s00405-018-5118-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/31/2018] [Indexed: 11/25/2022]
Abstract
BACKGROUND The incidence of thyroid cancer is fast increasing in both adults and children. The pediatric thyroid cancer had often already progressed to a more advanced stage of the disease at diagnosis. Early detection of pediatric thyroid cancer has been a problem for many years. Lipocalin-2 (Lcn2) has been reported to be over-expressed in cancers of diverse histological origin and it facilitates tumorigenesis by promoting survival, growth, and metastasis. METHODS The plasma Lcn2 concentration of 28 Chinese papillary thyroid cancer (PTC) children and 24 healthy controls was measured. Immunostaining for Ki-67 of tumor tissue from PTC children was performed. The expression levels of Lcn2 and NFκB in PTC tissue and peri-carcinoma tissue of PTC children were measured through Western blot. RESULTS The plasma concentration of Lcn2 was significantly elevated in pediatric PTC patients compared with healthy controls. Besides, the plasma Lcn2 concentration significantly correlated with clinical characteristics, NFκB level, and Ki-67 positive rate of nucleus in tissue of PTC. CONCLUSION This is the first study to evaluate the plasma Lcn2 in pediatric PTC patients. It is possible that the plasma Lcn2 may be a new biomarker of pediatric thyroid cancer. Further studies are needed to explore the definite role and mechanism of Lcn2 in thyroid cancer, which will help to explore novel diagnostic or therapeutic strategies.
Collapse
Affiliation(s)
- Jun Tai
- Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China.,Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China
| | - Shengcai Wang
- Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China.,Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China
| | - Jie Zhang
- Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China.,Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China
| | - Wentong Ge
- Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China.,Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China
| | - Yuanhu Liu
- Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China.,Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China
| | - Xiaodan Li
- Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China.,Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China
| | - Yuwei Liu
- Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China.,Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China
| | - Zhijuan Deng
- Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China.,Department of Pathology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Lejian He
- Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China.,Department of Pathology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Guoliang Wang
- Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China.
| | - Xin Ni
- Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China. .,Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China.
| |
Collapse
|
41
|
Zhang X, Wu H, Fan H, Su B, Zhang G, Dong L. Clinical characteristics and prognosis of pediatric patients with B cell acute lymphoblastic leukemia relapse. Oncol Lett 2018; 16:2929-2934. [PMID: 30127881 PMCID: PMC6096220 DOI: 10.3892/ol.2018.8974] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 05/16/2018] [Indexed: 12/15/2022] Open
Abstract
The aim of the present study was to investigate the clinical characteristics and prognosis of pediatric patients with B cell acute lymphoblastic leukemia (B-ALL) relapse. A total of 390 pediatric patients diagnosed as B-ALL and receiving regular chemotherapy in Jining First People's Hospital from August 2010 to May 2016 were selected. The clinical characteristics, therapeutic response and prognosis were compared between the two groups. There were significant differences in the comparisons of age, leukocyte count in the initial diagnosis and glucocorticoid sensitive test between B-cell ALL (B-ALL) relapse group and non-relapse group; the minimal residual disease (MRD) levels of pediatric patients in the two groups at 33 days and 12 weeks were significantly different. The 3-year event-free survival (EFS) rates of pediatric patients with early, medium and late B-ALL relapse were 12.5±7.8%, 33.1±9.8% and 63.6±6.1%, respectively, and the prognosis of late relapse was significantly better than that of early relapse (P<0.001). The 3-year EFS rates of pediatric patients with bone marrow relapse in standard risk group, intermediate risk group and high risk group were 29.1±6.9, 31.3±6.5 and 28.3±6.3%, respectively; there were no statistically significant differences (P=0.387, P>0.05). Pediatric patients with B-ALL relapse are characterized by higher onset age (≥10 years old), high leukocyte count and hormone insensitivity. Dynamic monitoring of MRD level in B-ALL pediatric patients can predict the relapse.
Collapse
Affiliation(s)
- Xiaohong Zhang
- Department of Pediatrics, Jining First People's Hospital, Jining, Shandong 272000, P.R. China
| | - Haixia Wu
- Department of Pediatrics, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Hong Fan
- Department of Nursing, People's Hospital of Zhangqiu District, Jinan, Shandong 250000, P.R. China
| | - Baifang Su
- Department of Science and Education, People's Hospital of Zhangqiu District, Jinan, Shandong 250000, P.R. China
| | - Ge Zhang
- Department of Obstetrics, People's Hospital of Zhangqiu District, Jinan, Shandong 250000, P.R. China
| | - Lin Dong
- Department of Hematology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250000, P.R. China
| |
Collapse
|
42
|
Niktoreh N, Lerius B, Zimmermann M, Gruhn B, Escherich G, Bourquin JP, Dworzak M, Sramkova L, Rossig C, Creutzig U, Reinhardt D, Rasche M. Gemtuzumab ozogamicin in children with relapsed or refractory acute myeloid leukemia: a report by Berlin-Frankfurt-Münster study group. Haematologica 2018; 104:120-127. [PMID: 30093401 PMCID: PMC6312035 DOI: 10.3324/haematol.2018.191841] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022] Open
Abstract
Despite intensified salvage treatments, children with relapsed/refractory acute myeloid leukemia (AML) have poor survival. We evaluated gemtuzumab ozogamicin (CD33-targeted drug) used on a compassionate basis in patients diagnosed from 1995 until 2014 within Acute Myeloid Leukemia Berlin-Frankfurt-Münster studies, and identified 76 patients (<18 years) with highly-advanced and pre-treated AML [refractory de novo acute myeloid leukemia (n=10), de novo AML refractory to relapse (1st early: n=41; 1st late: n=10; 2nd or more: n=10), and secondary AML (n=5)]. At doses of 2.5–10 mg/m2, gemtuzumab ozogamicin was administered in 1-4 cycles as single agent (47%), combined with cytarabine (47%), or others (6%). Most common grade 3/4 adverse events were infections or febrile neutropenia (78% of severe adverse events), infusion-related immunological reactions (6%), and gastrointestinal symptoms (5%). Three patients experienced veno-occlusive disease (one fatal due to exacerbation of a pre-existing cardiomyopathy). Sixty-four percent received subsequent hematopoietic stem cell transplantation. Probability of 4-year overall survival was 18±5% in all, 27±7% in patients with and 0% in patients without hematopoietic stem cell transplantation (P<0.0001). Administration of gemtuzumab ozogamicin on a patient-specific, compassionate use basis was frequently considered in our study group and proved to be effective for bridging children with very advanced AML to hematopoietic stem cell transplantation. Uniform prospective studies for these patients are urgently needed.
Collapse
Affiliation(s)
- Naghmeh Niktoreh
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Germany
| | - Beate Lerius
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Germany
| | - Martin Zimmermann
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Germany
| | - Bernd Gruhn
- Department of Pediatrics, Jena University Hospital, Germany
| | - Gabriele Escherich
- Department of Pediatric Hematology and Oncology, Eppendorf University Hospital, Hamburg, Germany
| | - Jean-Pierre Bourquin
- Division of Pediatric Hematology/Oncology, University Children's Hospital Zurich, Switzerland
| | - Michael Dworzak
- St. Anna Children's Hospital and Children's Cancer Research Institute, Department of Pediatrics, Medical University of Vienna, Austria
| | - Lucie Sramkova
- Department of Pediatric Hematology and Oncology, 2 Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Claudia Rossig
- University Children's Hospital Münster, Pediatric Hematology and Oncology, Germany
| | - Ursula Creutzig
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Germany
| | - Dirk Reinhardt
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Germany
| | - Mareike Rasche
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Germany
| |
Collapse
|
43
|
van Eijkelenburg NKA, Rasche M, Ghazaly E, Dworzak MN, Klingebiel T, Rossig C, Leverger G, Stary J, De Bont ESJM, Chitu DA, Bertrand Y, Brethon B, Strahm B, van der Sluis IM, Kaspers GJL, Reinhardt D, Zwaan CM. Clofarabine, high-dose cytarabine and liposomal daunorubicin in pediatric relapsed/refractory acute myeloid leukemia: a phase IB study. Haematologica 2018; 103:1484-1492. [PMID: 29773602 PMCID: PMC6119144 DOI: 10.3324/haematol.2017.187153] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 05/16/2018] [Indexed: 12/29/2022] Open
Abstract
Survival in children with relapsed/refractory acute myeloid leukemia is unsatisfactory. Treatment consists of one course of fludarabine, cytarabine and liposomal daunorubicin, followed by fludarabine and cytarabine and stem-cell transplantation. Study ITCC 020/I-BFM 2009-02 aimed to identify the recommended phase II dose of clofarabine replacing fludarabine in the abovementioned combination regimen (3+3 design). Escalating dose levels of clofarabine (20-40 mg/m2/day × 5 days) and liposomal daunorubicin (40-80 mg/m2/day) were administered with cytarabine (2 g/m2/day × 5 days). Liposomal DNR was given on day 1, 3 and 5 only. The cohort at the recommended phase II dose was expanded to make a preliminary assessment of anti-leukemic activity. Thirty-four children were enrolled: refractory 1st (n=11), early 1st (n=15), ≥2nd relapse (n=8). Dose level 3 (30 mg/m2clofarabine; 60 mg/m2liposomal daunorubicin) appeared to be safe only in patients without subclinical fungal infections. Infectious complications were dose-limiting. The recommended phase II dose was 40 mg/m2 clofarabine with 60 mg/m2 liposomal daunorubicin. Side-effects mainly consisted of infections. The overall response rate was 68% in 31 response evaluable patients, and 80% at the recommended phase II dose (n=10); 22 patients proceeded to stem cell transplantation. The 2-year probability of event-free survival (pEFS) was 26.5±7.6 and probability of survival (pOS) 32.4±8.0%. In the 21 responding patients, the 2-year pEFS was 42.9±10.8 and pOS 47.6±10.9%. Clofarabine exposure in plasma was not significantly different from that in single-agent studies. In conclusion, clofarabine was well tolerated and showed high response rates in relapsed/refractory pediatric acute myeloid leukemia. Patients with (sub) clinical fungal infections should be treated with caution. Clofarabine has been taken forward in the Berlin-Frankfurt-Münster study for newly diagnosed acute myeloid leukemia. The Study ITCC-020 was registered as EUDRA-CT 2009-009457-13; Dutch Trial Registry number 1880.
Collapse
Affiliation(s)
- Natasha K A van Eijkelenburg
- Department of Pediatric Oncology/Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,European Consortium for Innovative Therapies for Children with Cancer (ITCC), Villejuif, France
| | - Mareike Rasche
- Department of Pediatric Oncology, University Children's Hospital, Essen, Germany
| | - Essam Ghazaly
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, UK
| | - Michael N Dworzak
- Children's Cancer Research Institute and St. Anna Children's Hospital, Department of Pediatrics, Medical University of Vienna, Austria
| | - Thomas Klingebiel
- Pediatric Hematology/Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Claudia Rossig
- Pediatric Hematology and Oncology, University Children's Hospital, Münster, Germany
| | - Guy Leverger
- Department of Pediatric Hematology and Oncology, AP-HP, GH HUEP, Trousseau Hospital, Paris, France
| | - Jan Stary
- Department of Pediatric Hematology and Oncology, 2Faculty of Medicine, Charles University Prague, University Hospital Motol, Czech Republic
| | - Eveline S J M De Bont
- Department of Pediatric Oncology, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Dana A Chitu
- Clinical Trial Center, Department of Hematology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Yves Bertrand
- Pediatric Hematology Department, IHOP and Claude Bernard University, Lyon, France
| | - Benoit Brethon
- Department of Pediatric Hematology, Robert Debré Hospital, Paris, France
| | - Brigitte Strahm
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University of Freiburg, Germany
| | - Inge M van der Sluis
- Department of Pediatric Oncology/Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,European Consortium for Innovative Therapies for Children with Cancer (ITCC), Villejuif, France
| | - Gertjan J L Kaspers
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Pediatric Oncology, VU University Medical Center, Amsterdam, the Netherlands.,I-BFM-AML committee, Kiel, Germany
| | - Dirk Reinhardt
- European Consortium for Innovative Therapies for Children with Cancer (ITCC), Villejuif, France.,I-BFM-AML committee, Kiel, Germany
| | - C Michel Zwaan
- Department of Pediatric Oncology/Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands .,Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,European Consortium for Innovative Therapies for Children with Cancer (ITCC), Villejuif, France
| |
Collapse
|
44
|
Yaniv I, Krauss AC, Beohou E, Dalissier A, Corbacioglu S, Zecca M, Afanasyev BV, Berger M, Diaz MA, Kalwak K, Sedlacek P, Varotto S, Peters C, Bader P. Second Hematopoietic Stem Cell Transplantation for Post-Transplantation Relapsed Acute Leukemia in Children: A Retrospective EBMT-PDWP Study. Biol Blood Marrow Transplant 2018; 24:1629-1642. [PMID: 29548831 DOI: 10.1016/j.bbmt.2018.03.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/06/2018] [Indexed: 02/05/2023]
Abstract
Outcome data were collected from the European Society for Blood and Marrow Transplantation registry on 373 children from 120 centers with relapsed leukemia (214 with acute lymphoblastic leukemia [ALL] and 159 with acute myelogenous leukemia [AML]) who underwent second allogeneic hematopoietic stem cell transplantation (HSCT) between 2004 and 2013. Overall survival (OS) was 38% at 2 years and 29% at 5 years, and leukemia-free survival (LFS) was 30% at 2 years and 25% at 5 years. Median follow-up after second HSCT was 36.4 months in the ALL group and 50.2 months in the AML group. In the ALL group, OS was 43% at 2 years and 33% at 5 years, and LFS was 34% at 2 years and 31% at 5 years. In the AML group, OS was 32% at 2 years and 24% at 5 years, and LFS was 24% at 2 years and 17% at 5 years. The 2-year nonrelapse mortality (NRM) rate was 22% in the ALL group and 18% in the AML group. Favorable prognostic factors (P < .05) for OS and LFS included >12 months between transplantations and chronic graft-versus-host disease after the first HSCT (in both groups), complete response before the second HSCT (ALL group only), and age >12 years (AML group only). Findings were more consistent over time in the ALL group, with no significant differences between 2-year and 5-year rates of relapse, NRM, and LFS. Children with relapsed acute leukemias have a substantial likelihood of long-term survival following second HSCT. Given the many novel targeted and immunomodulation therapies currently under development, it is important to identify specific patient subpopulations that may benefit from a second HSCT compared with those better suited to new approaches.
Collapse
Affiliation(s)
- Isaac Yaniv
- Rina Zaizov Hematology-Oncology Division, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Aviva C Krauss
- Rina Zaizov Hematology-Oncology Division, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Eric Beohou
- EBMT Paris study office/CEREST-TC, Department of Haematology, Saint Antoine Hospital, INSERM UMR 938, Paris, France
| | - Arnaud Dalissier
- EBMT Paris study office/CEREST-TC, Department of Haematology, Saint Antoine Hospital, INSERM UMR 938, Paris, France
| | - Selim Corbacioglu
- Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Regensburg, Germany
| | - Marco Zecca
- Pediatric Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Boris V Afanasyev
- Hematology and Transplantation Department, Pavlov First Saint Petersburg State Medical University, Saint Petersburg, Russian Federation
| | - Massimo Berger
- Pediatric Onco-Hematology, Regina Margherita Children Hospital, Torino, Italy
| | - Miguel Angel Diaz
- Division of Hematopoietic Stem Cell Transplantation, "Nino Jesus" Children Hospital, Madrid, Spain
| | - Krzysztof Kalwak
- Department of Pediatric Hematology/Oncology and Bone Marrow Transplantation, Cape of Hope Wroclaw Medical University, Wroclaw, Poland
| | - Petr Sedlacek
- Pediatric Hematology-Oncology, University Hospital Motol, Prague, Czech Republic
| | - Stefania Varotto
- Oncoematologia Pediatrica, Azienda Ospedaliera-Universita, Padova, Italy
| | - Christina Peters
- AustriaStem Cell Transplantation Unit, St. Anna Children's Hospital, Vienna, Austria
| | - Peter Bader
- Department for Children and Adolescents; Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|
45
|
Masetti R, Castelli I, Astolfi A, Bertuccio SN, Indio V, Togni M, Belotti T, Serravalle S, Tarantino G, Zecca M, Pigazzi M, Basso G, Pession A, Locatelli F. Genomic complexity and dynamics of clonal evolution in childhood acute myeloid leukemia studied with whole-exome sequencing. Oncotarget 2018; 7:56746-56757. [PMID: 27462774 PMCID: PMC5302950 DOI: 10.18632/oncotarget.10778] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/10/2016] [Indexed: 11/25/2022] Open
Abstract
Despite significant improvement in treatment of childhood acute myeloid leukemia (AML), 30% of patients experience disease recurrence, which is still the major cause of treatment failure and death in these patients. To investigate molecular mechanisms underlying relapse, we performed whole-exome sequencing of diagnosis-relapse pairs and matched remission samples from 4 pediatric AML patients without recurrent cytogenetic alterations. Candidate driver mutations were selected for targeted deep sequencing at high coverage, suitable to detect small subclones (0.12%). BiCEBPα mutation was found to be stable and highly penetrant, representing a separate biological and clinical entity, unlike WT1 mutations, which were extremely unstable. Among the mutational patterns underlying relapse, we detected the acquisition of proliferative advantage by signaling activation (PTPN11 and FLT3-TKD mutations) and the increased resistance to apoptosis (hyperactivation of TYK2). We also found a previously undescribed feature of AML, consisting of a hypermutator phenotype caused by SETD2 inactivation. The consequent accumulation of new mutations promotes the adaptability of the leukemia, contributing to clonal selection. We report a novel ASXL3 mutation characterizing a very small subclone (<1%) present at diagnosis and undergoing expansion (60%) at relapse. Taken together, these findings provide molecular clues for designing optimal therapeutic strategies, in terms of target selection, adequate schedule design and reliable response-monitoring techniques.
Collapse
Affiliation(s)
- Riccardo Masetti
- Department of Pediatrics "Lalla Seràgnoli", Hematology-Oncology Unit, University of Bologna, Bologna, Italy
| | - Ilaria Castelli
- Department of Pediatrics "Lalla Seràgnoli", Hematology-Oncology Unit, University of Bologna, Bologna, Italy
| | - Annalisa Astolfi
- Interdepartmental Centre of Cancer Research "G. Prodi", University of Bologna, Bologna, Italy
| | - Salvatore Nicola Bertuccio
- Department of Pediatrics "Lalla Seràgnoli", Hematology-Oncology Unit, University of Bologna, Bologna, Italy
| | - Valentina Indio
- Interdepartmental Centre of Cancer Research "G. Prodi", University of Bologna, Bologna, Italy
| | - Marco Togni
- Department of Pediatrics "Lalla Seràgnoli", Hematology-Oncology Unit, University of Bologna, Bologna, Italy.,Current address: Stem Cell Group, University College London Cancer Institute, University College London, London, United Kingdom
| | - Tamara Belotti
- Department of Pediatrics "Lalla Seràgnoli", Hematology-Oncology Unit, University of Bologna, Bologna, Italy
| | - Salvatore Serravalle
- Department of Pediatrics "Lalla Seràgnoli", Hematology-Oncology Unit, University of Bologna, Bologna, Italy
| | - Giuseppe Tarantino
- Interdepartmental Centre of Cancer Research "G. Prodi", University of Bologna, Bologna, Italy
| | - Marco Zecca
- Department of Pediatric Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Martina Pigazzi
- Department of Woman and Child Health, Laboratory of Hematology-Oncology, University of Padova, Padova, Italy
| | - Giuseppe Basso
- Department of Woman and Child Health, Laboratory of Hematology-Oncology, University of Padova, Padova, Italy
| | - Andrea Pession
- Department of Pediatrics "Lalla Seràgnoli", Hematology-Oncology Unit, University of Bologna, Bologna, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology-Oncology, IRCCS Ospedale Bambino Gesù, Rome, Italy.,University of Pavia, Pavia, Italy
| |
Collapse
|
46
|
Wang J, Yuan L, Cheng H, Fei X, Yin Y, Gu J, Xue S, He J, Yang F, Wang X, Yang Y, Zhang W. Salvaged allogeneic hematopoietic stem cell transplantation for pediatric chemotherapy refractory acute leukemia. Oncotarget 2018; 9:3143-3159. [PMID: 29423036 PMCID: PMC5790453 DOI: 10.18632/oncotarget.22809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/27/2017] [Indexed: 01/31/2023] Open
Abstract
There is an ongoing debate concerning the performance of salvaged allogeneic hematopoietic stem cell transplantation (allo-HSCT) in pediatric patients with acute refractory leukemia, in whom the prognosis is quite dismal. Few studies have ever been conducted on this subject. This may be partly due to missed opportunities by majority of the patients in such situations. To investigate the feasibility, evaluate the efficiency, and identify the prognostic factors of allo-HSCT in this sub-setting, the authors performed a single institution-based retrospective analysis. A total of 44 patients, of whom 28 had acute myeloid leukemia (AML), 13 had acute lymphocytic leukemia (ALL), and 3 had mixed phenotype leukemia (MPL), were enrolled in this study. With a median follow-up of 19 months, the estimated 2-year overall survival (OS) and progression free survival (PFS) were 34.3% (95% CI, 17.9–51.4%) and 33.6% (95% CI, 18.0–50.1%), respectively. The estimated 2-year incidence rates of relapse and non-relapse mortality (NRM) were 43.8% (95% CI 26.4–60.0%) and 19.6% (95% CI 9.1–32.9%), respectively. The estimated 100-day cumulative incidence of acute graft versus host disease (aGvHD) was 43.6% (95% CI 28.7–57.5%), and the 1-year cumulative incidence of chronic GvHD (cGvHD) was 45.5% (95% CI 30.5–59.3%). Compared with the previous studies, the multivariate analysis in this study additionally identified that female donors and cGvHD were associated with lower relapse and better PFS and OS. Male recipients, age younger than 10 years, a diagnosis of ALL, and the intermediate-adverse cytogenetic risk group were associated with increased relapse. On the contrary, extramedullary disease (EMD) and aGvHD were only linked to worse PFS. These data suggested that although only one-third of the patients would obtain PFS over 2 years, salvaged allo-HSCT is still the most reliable and best therapeutic strategy for refractory pediatric acute leukemia. If probable, choosing a female donor, better management of aGvHD, and induction of cGvHD promotes patient survival.
Collapse
Affiliation(s)
- Jingbo Wang
- Department of Hematology, China Aerospace Central Hospital, Beijing, China
| | - Lei Yuan
- Department of Hematology, China Aerospace Central Hospital, Beijing, China
| | - Haoyu Cheng
- Department of Hematology, China Aerospace Central Hospital, Beijing, China
| | - Xinhong Fei
- Department of Hematology, China Aerospace Central Hospital, Beijing, China
| | - Yumin Yin
- Department of Hematology, China Aerospace Central Hospital, Beijing, China
| | - Jiangying Gu
- Department of Hematology, China Aerospace Central Hospital, Beijing, China
| | - Song Xue
- Department of Hematology, China Aerospace Central Hospital, Beijing, China
| | - Junbao He
- Department of Hematology, China Aerospace Central Hospital, Beijing, China
| | - Fan Yang
- Department of Hematology, China Aerospace Central Hospital, Beijing, China
| | - Xiaocan Wang
- Department of Hematology, China Aerospace Central Hospital, Beijing, China
| | - Yixin Yang
- Department of Hematology, China Aerospace Central Hospital, Beijing, China
| | - Weijie Zhang
- Department of Hematology, China Aerospace Central Hospital, Beijing, China
| |
Collapse
|
47
|
Alexander TB, Bhakta N, Kolb EA, Rubnitz JE. Opportunities for expanding clinical trial enrollment for relapsed and refractory pediatric acute myeloid leukemia in the United States and Canada. Pediatr Blood Cancer 2017; 64. [PMID: 28509405 DOI: 10.1002/pbc.26632] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/14/2017] [Accepted: 04/08/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Thomas B Alexander
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina
| | - Nickhill Bhakta
- Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - E Anders Kolb
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Jeffrey E Rubnitz
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
48
|
Nakayama H, Tomizawa D, Tanaka S, Iwamoto S, Shimada A, Saito AM, Yamashita Y, Moritake H, Terui K, Taga T, Matsuo H, Kosaka Y, Koh K, Hosoi H, Kurosawa H, Isoyama K, Horibe K, Mizutani S, Adachi S. Fludarabine, cytarabine, granulocyte colony-stimulating factor and idarubicin for relapsed childhood acute myeloid leukemia. Pediatr Int 2017; 59:1046-1052. [PMID: 28771903 DOI: 10.1111/ped.13378] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 06/21/2017] [Accepted: 07/05/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND The combination of fludarabine (Flu), high-dose cytarabine (Ara-C) and granulocyte colony-stimulating factor (G-CSF; FLAG), with anthracyclines has become standard chemotherapy for refractory acute myeloid leukemia (AML) in European children and adults. To clarify the efficacy and the safety of FLAG-idarubicin (IDA) for children prospectively, we planned a multicenter phase II study (AML-R11) by the Japanese Pediatric Leukemia/Lymphoma Study Group. METHODS Patients with AML aged between 2 and 20 years old, who had the first bone marrow (BM) relapse or induction failure, were enrolled. The FLAG-IDA regimen consisted of Flu 30 mg/m2 for 5 days, Ara-C 2 g/m2 for 5 days, G-CSF (lenograstim) 5 μg/kg for 6 days and IDA 10 mg/m2 for 3 days. The primary endpoint was remission rate after therapy. RESULTS Due to drug supply issues, the trial was suspended after the inclusion of seven eligible patients. There were six cases of early relapse within 1 year of the first remission. All seven patients completed the therapy and no early death was observed. Hematological toxicity was common, and one patient developed grade 4 non-hematological toxicity of bacterial meningitis. Although only one patient with late relapse achieved complete remission, minimal residual disease was positive on both flow cytometry and Wilms' tumor 1 mRNA. Two patients were alive in remission following hematopoietic stem cell transplantation, whereas the other five patients died of either the disease or treatment-related causes. CONCLUSION FLAG-IDA might be tolerable for children with refractory AML although the efficacy should be further investigated.
Collapse
Affiliation(s)
- Hideki Nakayama
- Department of Pediatrics, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan.,Department of Pediatrics, National Hospital Organization Fukuoka-Higashi Medical Center, Fukuoka, Japan.,Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Shiro Tanaka
- Department of Clinical Biostatistics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shotaro Iwamoto
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Akira Shimada
- Department of Pediatrics, Okayama University, Okayama, Japan
| | - Akiko M Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Yuka Yamashita
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Hiroshi Moritake
- Department of Pediatrics, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Kiminori Terui
- Department of Pediatrics, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Takashi Taga
- Department of Pediatrics, Shiga University of Medical Science, Ohtsu, Japan
| | - Hidemasa Matsuo
- School of Human Health Science, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshiyuki Kosaka
- Department of Pediatric Hematology/Oncology, Hyougo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Hajime Hosoi
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Keiichi Isoyama
- Department of Pediatrics, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Keizo Horibe
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Shuki Mizutani
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Souichi Adachi
- School of Human Health Science, Faculty of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
49
|
Abstract
Immune surveillance comprising of adaptive and innate immune systems is naturally designed to eliminate cancer development; overexpression of inhibitory receptors and their ligands prevent this check and lead to evasion and hence cancer progression and metastasis. The use of tumor-specific monoclonal antibodies (MAbs) targeting these checkpoint regulators is promising and has led to this novel field of cancer immunotherapy. The first antibody directed against cytotoxic T-lymphocyte associated protein 4 (CTLA-4), ipilimumab, showed promising results in clinical trials and was approved by the US Food and Drug Administration (FDA) for the treatment of metastatic melanoma in 2011. Since then, various other immune checkpoint inhibitors are being studied in preclinical and clinical trial phases, targeting programmed-death-1 (PD-1) and its ligand programmed death ligand 1 (PD-L1), T cell lymphocyte activation gene-3 (LAG-3), and others. Results from clinical trials are promising, and currently this approach has proven effective and safe in patients with solid tumors and some hematological malignancies in adults. In general, CTLA-4 and PD-1 inhibitors are well tolerated; however, the augmented immune response enabled by this class of agents is associated with a unique group of side effects called immune-related adverse events (irAEs). Experience in pediatrics using immune checkpoint inhibitors for hematological malignancies is limited to Hodgkin's disease and non-Hodgkin's lymphoma as in the ongoing Children's Oncology Group (COG) protocol ADVL1412. Therapeutic advances in childhood leukemia and lymphoma (TACL) consortium will initiate an early phase clinical trial with PD-1 inhibitor nivolumab in relapsed/refractory acute myeloid leukemia (AML) in the next few months.
Collapse
Affiliation(s)
- Kara L Davis
- a Bass Center for Childhood Cancer and Blood Disorders, Department of Pediatrics , Stanford University School of Medicine , Palo Alto , California
| | - Archana M Agarwal
- b Department of Pathology , University of Utah and ARUP Laboratories , Salt Lake City, Utah
| | - Anupam R Verma
- c Primary Children's Hospital, Pediatric Hematology Oncology, Department of Pediatrics , University of Utah , Salt Lake City, Utah
| |
Collapse
|
50
|
Integrating Biological and Mathematical Models to Explain and Overcome Drug Resistance in Cancer. Part 1: Biological Facts and Studies in Drug Resistance. CURRENT STEM CELL REPORTS 2017. [DOI: 10.1007/s40778-017-0097-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|