1
|
Kalwak K, Moser LM, Pötschger U, Bader P, Kleinschmidt K, Meisel R, Dalle JH, Yesilipek A, Balduzzi A, Krivan G, Goussetis E, Staciuk R, Sedlacek P, Pichler H, Svec P, Gabriel M, Güngör T, Bilic E, Buechner J, Renard M, Vettenranta K, Ifversen M, Diaz-de-Heredia C, Stein J, Toporski J, Bierings M, Peters C, Ansari M, Locatelli F. Comparable outcomes after busulfan- or treosulfan-based conditioning for allo-HSCT in children with ALL: results of FORUM. Blood Adv 2025; 9:741-751. [PMID: 39602342 PMCID: PMC11869852 DOI: 10.1182/bloodadvances.2024014548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/09/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
ABSTRACT The superiority of total body irradiation (TBI)-based vs chemotherapy conditioning for allogeneic hematopoietic stem cell transplantation (allo-HSCT) in children with acute lymphoblastic leukemia (ALL) has been established in the international, prospective phase-3 FORUM study, randomizing 417 patients aged 4-18 years in complete remission (CR), who received allo-HSCT from HLA-matched sibling or unrelated donors. Because of the unavailability of TBI in some regions and to accommodate individual contraindications, this study reports the prespecified comparison of outcomes of patients receiving busulfan (BU)- or treosulfan (TREO)-based regimens from 2013 to 2018. Overall, 180 and 128 patients received BU/thiotepa (THIO)/fludarabine (FLU) or TREO/THIO/FLU, respectively. Data were analyzed as of February 2023, with a median follow-up of 4.2 years (range, 0.3-9.1). 3-year overall survival was 0.71 (BU, 95% confidence interval [0.64-0.77]) and 0.72 (TREO, [0.63-0.79]) and 3-year event-free survival was 0.60 (BU, [0.53-0.67]) and 0.55 (TREO, [0.46-0.63]). The 3-year cumulative incidence of relapse (BU, 0.31 [0.25-0.38]; TREO, 0.36 [0.27-0.44]); and nonrelapse mortality (BU, 0.08 [0.05-0.13]; TREO, 0.09 [0.05-0.15]) were comparable. One case of fatal veno-occlusive disease occurred in each group. No significant differences in acute and chronic graft-versus-host disease (GVHD) or 3-year GVHD-free and relapse-free survival (BU, 0.48 [0.41-0.55]; TREO, 0.45 [0.37-0.54]) were recorded. Outcomes for patients in first and second CR were similar irrespective of the regimen. In conclusion, BU/THIO/FLU or TREO/THIO/FLU regimens can be an alternative to TBI for patients with ALL aged >4 years with contraindications or lack of access to TBI. This trial was registered at www.ClinicalTrials.gov as #NCT01949129.
Collapse
Affiliation(s)
- Krzysztof Kalwak
- Department of Pediatric Hematology, Oncology, and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Laura M. Moser
- Division for Stem Cell Transplantation and Immunology, Department of Pediatrics, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | | | - Peter Bader
- Division for Stem Cell Transplantation and Immunology, Department of Pediatrics, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Katharina Kleinschmidt
- Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation, University Children’s Hospital Regensburg, Regensburg, Germany
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Jean-Hugues Dalle
- Pediatric Hematology and Immunology Department, Robert Debré Hospital, Groupe Hospitalo-Universitaire Assistance Publique Hôpitaux de Paris Nord, Université Paris Cité, Paris, France
| | | | - Adriana Balduzzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Pediatric Hematopoietic Stem Cell Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Gergely Krivan
- Pediatric Hematology and Stem Cell Transplantation Department, National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Evgenios Goussetis
- Stem Cell Transplant Unit, Agia Sofia Children's Hospital, Athens, Greece
| | - Raquel Staciuk
- Hospital de Pediatría “Prof. Dr Juan P. Garrahan,” Buenos Aires, Argentina
| | - Petr Sedlacek
- Department of Pediatric Hematology and Oncology, University Hospital Motol, Prague, Czech Republic
| | - Herbert Pichler
- St. Anna Children’s Cancer Research Institute, Vienna, Austria
- Department of Pediatric Hematology and Oncology, St. Anna Children’s Hospital, Medical University of Vienna, Vienna, Austria
| | - Peter Svec
- Department of Pediatric Hematology and Oncology, National Institute of Children’s Diseases, Comenius University, Bratislava, Slovakia
| | | | - Tayfun Güngör
- Division of Hematology/Oncology/Immunology, Gene Therapy, and Stem Cell Transplantation, University Children's Hospital Zurich, Eleonore Foundation and Children’s Research Center, Zürich, Switzerland
| | - Ernest Bilic
- Division for Hematology and Oncology, Department of Pediatrics Zagreb, University Hospital Center, Zagreb, Croatia
| | - Jochen Buechner
- Department of Pediatric Hematology and Oncology, Oslo University Hospital, Oslo, Norway
| | - Marleen Renard
- Department of Paediatric Oncology, University Hospital Leuven, Leuven, Belgium
| | - Kim Vettenranta
- University of Helsinki and the Hospital for Children and Adolescents, University of Helsinki, Helsinki, Finland
| | - Marianne Ifversen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Cristina Diaz-de-Heredia
- Division of Pediatric Hematology and Oncology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Jerry Stein
- Schneider Children's Medical Center of Israel and Sackler Faculty of Medicine Tel Aviv University, Petah Tikva, Israel
| | - Jacek Toporski
- Department Cell Therapy and Allogeneic Stem Cell Transplant, Karolinska University Hospital, Stockholm, Sweden
| | - Marc Bierings
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Christina Peters
- St. Anna Children’s Cancer Research Institute, Vienna, Austria
- Department of Pediatric Hematology and Oncology, St. Anna Children’s Hospital, Medical University of Vienna, Vienna, Austria
| | - Marc Ansari
- CANSEARCH Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Geneva Hospitals, Geneva, Switzerland
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
2
|
van Kalsbeek RJ, Feijen EAM, Bresters D, Kremer LCM, Pluijm SMF, Asogwa OA, Dulmen-den Broeder EV, van den Heuvel-Eibrink MM, Janssens GO, Tissing WJ, Loonen JJ, Neggers SJCMM, van der Pal HJH, Ronckers CM, Teepen JC, de Vries ACH, Louwerens M, van der Heiden-van der Loo M, Prevaes SMPJ, Versluys AB. Cyclophosphamide is not associated with clinically relevant late pulmonary dysfunction in Dutch survivors of childhood cancer - The DCCSS-LATER 2 PULM sub-study. Respir Med 2025; 237:107948. [PMID: 39793859 DOI: 10.1016/j.rmed.2025.107948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND Treatment for childhood cancer may increase the risk of long-term pulmonary complications and dysfunction. Pulmonary surveillance is recommended after established pulmonary toxic exposures, including bleomycin, busulfan, carmustine (BCNU), lomustine (CCNU), radiotherapy to a field exposing the lungs, and pulmonary surgery. However, the role of cyclophosphamide as a pulmonary toxic agent is debated. AIM To establish whether cyclophosphamide is associated with late pulmonary dysfunction among survivors of childhood cancer. METHODS In this multicenter Dutch Childhood Cancer Survivor Study (DCCSS)-LATER 2 PULM sub-study, we included 828 survivors with a median follow-up of 26.6 years, treated with cyclophosphamide and/or established pulmonary toxic treatment, or neither. Pulmonary function tests were used to measure the primary outcomes of diffusion impairment (diffusing capacity for carbon monoxide (DLCO) z-score), restriction (total lung capacity (TLC) z-score), and obstruction (forced expiratory volume in the first second/forced vital capacity (FEV1/FVC) z-score). Secondary outcomes comprised chronic cough, recurrent respiratory tract infections, shortness of breath, and supplemental oxygen need. RESULTS Diffusion and restrictive abnormalities were highly prevalent among those treated with established pulmonary toxic treatment, with cyclophosphamide (41.0 % and 50.4 %, respectively) and without (34.3 % and 41.9 %, respectively), and occurred less frequently in survivors treated with cyclophosphamide only (12.9 % and 7.3 %, respectively) or in survivor controls (9.9 % and 12.4 %, respectively). In multivariable analyses, cyclophosphamide did not have a clinically relevant effect on the primary or secondary outcomes. CONCLUSIONS This study suggests that cyclophosphamide is not associated with clinically relevant pulmonary dysfunction in long-term childhood cancer survivors.
Collapse
Affiliation(s)
- R J van Kalsbeek
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - E A M Feijen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - D Bresters
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Leiden University Medical Center/Willem-Alexander Children's Hospital, Leiden, the Netherlands
| | - L C M Kremer
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, the Netherlands; Utrecht University and Utrecht Medical Center, Utrecht, the Netherlands
| | - S M F Pluijm
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - O A Asogwa
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Leiden University Medical Center, Leiden, the Netherlands
| | | | - M M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Utrecht University and Utrecht Medical Center, Utrecht, the Netherlands
| | - G O Janssens
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - W J Tissing
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - J J Loonen
- Department of Hematology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - S J C M M Neggers
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Internal Medicine, Section Endocrinology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - H J H van der Pal
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - C M Ronckers
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Division of Childhood Cancer Epidemiology, Institute of Medical Biometrics, Epidemiology and Computer Science, University Medicine, Johannes Gutenberg University, Mainz, Germany
| | - J C Teepen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - A C H de Vries
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - M Louwerens
- Leiden University Medical Center, Leiden, the Netherlands
| | | | - S M P J Prevaes
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | - A B Versluys
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Wilhelmina Children's Hospital, Utrecht, the Netherlands.
| |
Collapse
|
3
|
Sharma A. How I treat sickle cell disease with gene therapy. Blood 2024; 144:2693-2705. [PMID: 39356871 PMCID: PMC11830977 DOI: 10.1182/blood.2024024519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 10/04/2024] Open
Abstract
ABSTRACT In 2023, 2 different gene therapies were approved for individuals with severe sickle cell disease (SCD). The small number of patients treated on the pivotal clinical trials that led to these approvals have experienced dramatic short-term reductions in the occurrence of painful vaso-occlusive crises, but the long-term safety and efficacy of these genetic therapies are yet to be ascertained. Several challenges and treatment-related concerns have emerged in regard to administering these therapies in clinical practice. This article discusses the selection and preparation of individuals with SCD who wish to receive autologous gene therapy, as well as the salient features of the care needed to support them through a long and arduous treatment process. I specifically focus on postinfusion care, as it relates to immune monitoring and infection prevention. Compared with allogeneic hematopoietic cell transplantation, delivering autologous gene therapy to an individual with SCD has distinct nuances that require awareness and special interventions. Using clinical vignettes derived from real-life patients, I provide perspectives on the complex decision-making process for gene therapy for SCD based on currently available data and make recommendations for evaluating and supporting these patients.
Collapse
Affiliation(s)
- Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
4
|
Mao J, Ge J, Ding S, Sun Z, Nan F, Yu H, Ding J, Wang X, Liu Z, Zhang M, Fu X. TBI/Cy followed by auto-HSCT is a good choice next to allo-HSCT for patients with T-LBL/ALL. Sci Rep 2024; 14:22356. [PMID: 39333693 PMCID: PMC11437004 DOI: 10.1038/s41598-024-72897-9] [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: 04/10/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024] Open
Abstract
The aim of this retrospective study was to evaluate the efficiency and safety of total body irradiation plus cyclophosphamide (TBI/Cy) followed by autogenetic hematopoietic stem cell transplantation (auto-HSCT) in T-LBL/ALL patients that cannot receive allogeneic hematopoietic stem cell transplant (allo-HSCT). Between 2013 and 2023, 24 patients received auto-HSCT following by TBI/Cy, 26 patients underwent allo-HSCT, all patients achieved completed hematopoietic reconstitution after HSCT. The progression free survival (PFS) and overall survival (OS) had no statistically significant differences between the two groups (P = 0.791, HR 1.127, 95%CI 0.456-2.785; P = 0.456, HR 0.685, 95%CI 0.256-1.828). Although the cumulative incidence of relapse was lower for patients who received allo-HSCT than auto-HSCT (P = 0.033, HR 3.707, 95%CI 1.188-11.570, 2-year relapse 11.5% vs. 33.3%), the incidence of non-relapse mortality (NRM) was higher than that in the auto-HSCT group (P = 0.014, HR 0.000, 95%CI -1.000 - -1.000, 2-year NRM, 23.1% vs. 0%). Trough Landmark analysis, the two groups showed a statistically significant difference in 3-year PFS and 4-year OS curves (Figure S2A&B, P = 0.039, HR 0.426, 95%CI 0.163-1.117; P = 0.014, HR 0.317, 95%CI 0.113-0.887). By COX analysis, poor baseline performance status (ECOG-PS ≥ 2) and CNS involvement were risk factors for PFS and OS. In conclusion, TBI/Cy followed by auto-HSCT is a good choice next to allo-HSCT for patients with T-LBL/ALL.
Collapse
Affiliation(s)
- Jinghua Mao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jurui Ge
- Department of Oncology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shiqi Ding
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenchang Sun
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feifei Nan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Yu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiayin Ding
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinyi Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhuling Liu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Xiaorui Fu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
5
|
Uhlving HH, Specht L, Masmas TN, Bernsdorf M, Ifversen M. Late effects following HSCT for childhood ALL: A national single-center study using three different modalities of delivery of total body irradiation. Pediatr Blood Cancer 2024; 71:e31163. [PMID: 38943233 DOI: 10.1002/pbc.31163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND Total body irradiation (TBI) is a pivotal part of conditioning prior to hematopoietic stem cell transplantation (HSCT) for childhood acute lymphoblastic leukemia (ALL), yet evidence is sparse regarding the effect of TBI delivery techniques on acute and late toxicities. DESIGN In a national cohort of pediatric HSCT-recipients, we compared three TBI schedules; 12 Gray (Gy) delivered as (i) 4 Gy daily fractions from 2008 to 2011 (n = 12); (ii) 2 Gy fractions twice daily with two-dimensional (2D) planning technology from 2012 to 2015 (n = 16); and (iii) 2 Gy twice daily with three-dimensional (3D) planning intensity-modulated radiotherapy (IMRT) from 2016 to 2020 (n = 14). RESULTS The 5-year event-free survival was 75.0%, 81.3%, and 81.3% in Cohorts 1, 2, and 3, respectively. Acute toxicity assessed as maximum ferritin and C-reactive protein during the first 3 months post HSCT did not differ between cohorts, nor did the time to first hospital discharge (median 28, 32, and 31 days, p = .25). The incidences of acute graft-versus-host disease (GvHD) (66%, 56%, 71%) and chronic GvHD (25%, 31%, 14%) were comparable. Pulmonary function assessed by spirometry did not differ significantly. The 5-year cataract-free survival was 33.3%, 79%, and 100% in Cohorts 1, 2, and 3, respectively. We found a nonsignificant tendency toward more endocrinopathies in Cohort 1 compared to Cohorts 2 and 3. CONCLUSION The change of modality did not result in more relapses. More fractionation led to improvement with a lower incidence of cataract and a tendency toward fewer endocrinopathies. The effect of 3D-planning-IMRT technology requires further evaluation in larger studies.
Collapse
Affiliation(s)
- Hilde Hylland Uhlving
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Lena Specht
- Department of Radiation Oncology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Tania Nicole Masmas
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Mogens Bernsdorf
- Department of Radiation Oncology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Marianne Ifversen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
6
|
Naik S, Li Y, Talleur AC, Selukar S, Ashcraft E, Cheng C, Madden RM, Mamcarz E, Qudeimat A, Sharma A, Srinivasan A, Suliman AY, Epperly R, Obeng EA, Velasquez MP, Langfitt D, Schell S, Métais JY, Arnold PY, Hijano DR, Maron G, Merchant TE, Akel S, Leung W, Gottschalk S, Triplett BM. Memory T-cell enriched haploidentical transplantation with NK cell addback results in promising long-term outcomes: a phase II trial. J Hematol Oncol 2024; 17:50. [PMID: 38937803 PMCID: PMC11212178 DOI: 10.1186/s13045-024-01567-0] [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: 05/06/2024] [Accepted: 06/13/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Relapse remains a challenge after transplantation in pediatric patients with hematological malignancies. Myeloablative regimens used for disease control are associated with acute and long-term adverse effects. We used a CD45RA-depleted haploidentical graft for adoptive transfer of memory T cells combined with NK-cell addback and hypothesized that maximizing the graft-versus-leukemia (GVL) effect might allow for reduction in intensity of conditioning regimen. METHODS In this phase II clinical trial (NCT01807611), 72 patients with hematological malignancies (complete remission (CR)1: 25, ≥ CR2: 28, refractory disease: 19) received haploidentical CD34 + enriched and CD45RA-depleted hematopoietic progenitor cell grafts followed by NK-cell infusion. Conditioning included fludarabine, thiotepa, melphalan, cyclophosphamide, total lymphoid irradiation, and graft-versus-host disease (GVHD) prophylaxis consisted of a short-course sirolimus or mycophenolate mofetil without serotherapy. RESULTS The 3-year overall survival (OS) and event-free-survival (EFS) for patients in CR1 were 92% (95% CI:72-98) and 88% (95% CI: 67-96); ≥ CR2 were 81% (95% CI: 61-92) and 68% (95% CI: 47-82) and refractory disease were 32% (95% CI: 11-54) and 20% (95% CI: 6-40). The 3-year EFS for all patients in morphological CR was 77% (95% CI: 64-87) with no difference amongst recipients with or without minimal residual disease (P = 0.2992). Immune reconstitution was rapid, with mean CD3 and CD4 T-cell counts of 410/μL and 140/μL at day + 30. Cumulative incidence of acute GVHD and chronic GVHD was 36% and 26% but most patients with acute GVHD recovered rapidly with therapy. Lower rates of grade III-IV acute GVHD were observed with NK-cell alloreactive donors (P = 0.004), and higher rates of moderate/severe chronic GVHD occurred with maternal donors (P = 0.035). CONCLUSION The combination of a CD45RA-depleted graft and NK-cell addback led to robust immune reconstitution maximizing the GVL effect and allowed for use of a submyeloablative, TBI-free conditioning regimen that was associated with excellent EFS resulting in promising long-term outcomes in this high-risk population. The trial is registered at ClinicalTrials.gov (NCT01807611).
Collapse
Affiliation(s)
- Swati Naik
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA.
| | - Ying Li
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Aimee C Talleur
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Subodh Selukar
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Emily Ashcraft
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Cheng Cheng
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Renee M Madden
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ewelina Mamcarz
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Amr Qudeimat
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Akshay Sharma
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ashok Srinivasan
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ali Y Suliman
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Rebecca Epperly
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Esther A Obeng
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - M Paulina Velasquez
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Deanna Langfitt
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Sarah Schell
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Jean-Yves Métais
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Paula Y Arnold
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Diego R Hijano
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Gabriela Maron
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Thomas E Merchant
- Department of Radiation Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Salem Akel
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Wing Leung
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Brandon M Triplett
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
7
|
Otth M, Kasteler R, Mulder RL, Agrusa J, Armenian SH, Barnea D, Bergeron A, Bhatt NS, Bourke SJ, Constine LS, Goutaki M, Green DM, Hennewig U, Houdouin V, Hudson MM, Kremer L, Latzin P, Ng A, Oeffinger KC, Schindera C, Skinner R, Sommer G, Srinivasan S, Stokes DC, Versluys B, Waespe N, Weiner DJ, Dietz AC, Kuehni CE. Recommendations for surveillance of pulmonary dysfunction among childhood, adolescent, and young adult cancer survivors: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group. EClinicalMedicine 2024; 69:102487. [PMID: 38420219 PMCID: PMC10900250 DOI: 10.1016/j.eclinm.2024.102487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
Childhood, adolescent, and young adult (CAYA) cancer survivors are at risk of pulmonary dysfunction. Current follow-up care guidelines are discordant. Therefore, the International Late Effects of Childhood Cancer Guideline Harmonization Group established and convened a panel of 33 experts to develop evidence-based surveillance guidelines. We critically reviewed available evidence regarding risk factors for pulmonary dysfunction, types of pulmonary function testing, and timings of surveillance, then we formulated our recommendations. We recommend that CAYA cancer survivors and healthcare providers are aware of reduced pulmonary function risks and pay vigilant attention to potential symptoms of pulmonary dysfunction, especially among survivors treated with allogeneic haematopoietic stem cell transplantation, thoracic radiotherapy, and thoracic surgery. Based on existing limited evidence and current lack of interventions, our panel recommends pulmonary function testing only for symptomatic survivors. Since scarce existing evidence informs our recommendation, we highlight the need for prospective collaborative studies to address pulmonary function knowledge gaps among CAYA cancer survivors.
Collapse
Affiliation(s)
- Maria Otth
- Department of Oncology, Haematology, Immunology, Stem Cell Transplantation and Somatic Gene Therapy, University Children's Hospital Zurich, Zurich, Switzerland
- Pediatric Hematology-Oncology Center, Children's Hospital of Eastern Switzerland, St Gallen, Switzerland
- Department of Health Sciences and Medicine, University of Lucerne, Lucerne, Switzerland
| | - Rahel Kasteler
- Department of Oncology, Haematology, Immunology, Stem Cell Transplantation and Somatic Gene Therapy, University Children's Hospital Zurich, Zurich, Switzerland
- Pediatric Hematology-Oncology Center, Children's Hospital of Eastern Switzerland, St Gallen, Switzerland
| | - Renée L. Mulder
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jennifer Agrusa
- Department of Pediatric Hematology Oncology, C.S. Mott Children's Hospital, University of Michigan, Michigan, USA
| | | | - Dana Barnea
- Department of Hematology and Department of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Anne Bergeron
- Hôpitaux Universitaires de Genève, Université de Genève, Genève, Switzerland
| | - Neel S. Bhatt
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Stephen J. Bourke
- Department of Respiratory Medicine, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Louis S. Constine
- Departments of Radiation Oncology and Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Myrofora Goutaki
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Daniel M. Green
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ulrike Hennewig
- University Hospital of Giessen and Marburg, Pediatric Hematology and Oncology, Giessen, Germany
| | | | - Melissa M. Hudson
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Leontien Kremer
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Philipp Latzin
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Antony Ng
- Department of Paediatric Oncology, Royal Hospital for Children, Bristol, UK
| | - Kevin C. Oeffinger
- Department of Community and Family Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Christina Schindera
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Division of Pediatric Oncology/Haematology, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Roderick Skinner
- Department of Paediatric and Adolescent Haematology/Oncology, Great North Children's Hospital and Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Grit Sommer
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Saumini Srinivasan
- Division of Pulmonology, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Dennis C. Stokes
- Division of Pulmonology, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Birgitta Versluys
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Nicolas Waespe
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Pediatric Hematology and Oncology, University Children's Hospital Bern, University of Bern, Bern, Switzerland
- CANSEARCH Research Platform for Paediatric Oncology and Haematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Daniel J. Weiner
- Division of Pediatric Pulmonology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Claudia E. Kuehni
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Pediatric Hematology and Oncology, University Children's Hospital Bern, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Ferrer C, Huertas C, Ocanto A, García D, Plaza R, Mínguez C, de la Monja P, Escribano A, Pérez A, Sáez M. Evolution of dosimetric treatment planning for pediatric total lymphoid irradiation (TLI): a single-institution experience. Rep Pract Oncol Radiother 2024; 28:772-783. [PMID: 38515822 PMCID: PMC10954272 DOI: 10.5603/rpor.98734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 12/20/2023] [Indexed: 03/23/2024] Open
Abstract
Background Total lymphoid irradiation (TLI) is a conditioning regimen in allogeneic hematopoietic stem cell transplantation (allo-HSCT) which may reduce long-term toxicities attributed to other techniques, such as total body irradiation (TBI). At our institution, TLI treatments were first planned with the three-dimensional conformal radiation therapy (3D-CRT) technique and later with volumetric modulated arc therapy (VMAT). With the recent availability of a basic helical tomotherapy (HT), the possible dosimetric gain of the latter for TLI is studied. Materials and methods 22 pediatric patients were planned for VMAT and HT, prescribed to 8 Gy in 4 fractions. VMAT was planned with template based on a single cost function, using the Monaco treatment planning system (TPS). HT plans were planned using Accuray Precision TPS for a basic HT without the dynamic jaws feature or VOLO-Ultra algorithm. Plan quality was analyzed based on four quality indices, mean and maximum doses to planning target volume (PTV) and organs at risk (OARs), dose gradient and integral doses. Differences were analyzed with Wilcoxon signed-rank test. Results HT plans resulted in improved conformity (CI) and homogeneity indices (HI) (p < 0.05) but less steep dose gradient (p = 0.181). VMAT plans created larger areas with high doses within the PTV, while comparable doses to OARs, except mainly for the spinal marrow, for which a reduction of 37.7% in D2% was obtained (p < 0.05). Integral dose for non-tumor tissue was 11.3% lower with the VMAT template (p < 0.05). Conclusion HT achieves better conformity and homogeneity even without its more advanced features. Nevertheless, the VMAT template achieves dosimetric results close to those of HT, both with similar clinical outcome.
Collapse
Affiliation(s)
- Carlos Ferrer
- Medical Physics and Radiation Protection Department, H.U. La Paz, Madrid, Spain
| | - Concepción Huertas
- Medical Physics and Radiation Protection Department, H.U. La Paz, Madrid, Spain
| | | | - David García
- Medical Physics and Radiation Protection Department, H.U. La Paz, Madrid, Spain
| | - Rodrigo Plaza
- Medical Physics and Radiation Protection Department, H.U. La Paz, Madrid, Spain
| | - Cristina Mínguez
- Medical Physics and Radiation Protection Department, H.U. La Paz, Madrid, Spain
| | | | - Anne Escribano
- Radiation Oncology Department, H.U. La Paz, Madrid, Spain
| | - Antonio Pérez
- Pediatric Hemato-Oncology Department, H.U. La Paz, Madrid, Spain
| | - Moisés Sáez
- Medical Physics and Radiation Protection Department, H.U. La Paz, Madrid, Spain
| |
Collapse
|
9
|
Ollivier L, Debbi K, To NH, Cailleteau A, Supiot S, Mervoyer A, Guimas V, Belkacémi Y. Is oligometastatic disease an applicable and useful concept in haematologic malignancies? A narrative review of radiation therapy standards, modern techniques, and innovations. Cancer Radiother 2024; 28:119-130. [PMID: 38143233 DOI: 10.1016/j.canrad.2023.08.008] [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: 07/24/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 12/26/2023]
Abstract
PURPOSE Haematologic malignancies are particular in that they can generally be cured, even when distant metastases are present at diagnosis, unlike solid malignancies. Systemic treatments, including chemotherapy, targeted therapies, and immunotherapy, are the standard of care with excellent results. The considerable progress made in the management of these diseases in the last 20years has redefined the role of radiation therapy as minor in many clinical situations. We propose a literature review of data, showing that radiation therapy still has a role in curative, salvage, and palliative therapy situations. MATERIAL AND METHODS A document and literature search was carried out in the following databases: Medline and ClinicalTrial.gov, for the terms "radiotherapy", "haematologic malignancies", "Hodgkin lymphoma", "non-Hodgkin lymphoma", "CAR T cells", "multiple myeloma", "solitary plasmocytoma", "intensity-modulated radiotherapy", "extracranial stereotactic body radiation therapy" and "proton therapy references". RESULTS Haemopathological malignancies include a wide range of diseases and radiation therapy indications have been assessed over the past 20years. Currently, radiation therapy is indicated for localized disease (solitary plasmocytoma), as an adjuvant (Hodgkin lymphoma), in palliative settings, or after systemic treatment in relapsed patients (chimeric antigen receptor [CAR] T-cells) with a low recurrence burden, which can therefore be considered "oligorecurrence". Radiation therapy, through total body irradiation, has important indications, thanks to its immunomodulatory and/or myeloablative effects. Moreover, recent technological developments have made possible significant improvement in safety, contributing to radiation therapy being positioned in the treatment strategy of several indications. CONCLUSIONS Given the effectiveness of systemic treatments in hematologic malignancies, the oligometastasis stage is of little importance. A curative intent after local radiation therapy, even advanced stage, is possible, both with residual disease for advanced Hodgkin lymphoma, aggressive non-Hodgkin lymphoma, or solitary plasmocytoma, and even without evidence of disease after chemotherapy for Hodgkin or non-Hodgkin lymphoma. The role of new treatments, such as CAR T cells, allows us to consider radiation therapy after systemic treatment of relapsed diseases with low volume recurrence, which can be considered oligorecurrence.
Collapse
Affiliation(s)
- L Ollivier
- Service d'oncologie radiothérapie, Institut de cancérologie de l'Ouest, centre René-Gauducheau, Saint-Herblain, France.
| | - K Debbi
- Department of Radiation Oncology, centre Sein Henri-Mondor, CHU Henri-Mondor, AP-HP, university Paris Est Créteil (Upec), Créteil, France
| | - N-H To
- Department of Radiation Oncology, centre Sein Henri-Mondor, CHU Henri-Mondor, AP-HP, university Paris Est Créteil (Upec), Créteil, France; Institut Mondor de recherche biomédicale (IMRB), Inserm U955, i-Biot, Créteil, France
| | - A Cailleteau
- Service d'oncologie radiothérapie, Institut de cancérologie de l'Ouest, centre René-Gauducheau, Saint-Herblain, France
| | - S Supiot
- Service d'oncologie radiothérapie, Institut de cancérologie de l'Ouest, centre René-Gauducheau, Saint-Herblain, France
| | - A Mervoyer
- Service d'oncologie radiothérapie, Institut de cancérologie de l'Ouest, centre René-Gauducheau, Saint-Herblain, France
| | - V Guimas
- Service d'oncologie radiothérapie, Institut de cancérologie de l'Ouest, centre René-Gauducheau, Saint-Herblain, France
| | - Y Belkacémi
- Department of Radiation Oncology, centre Sein Henri-Mondor, CHU Henri-Mondor, AP-HP, university Paris Est Créteil (Upec), Créteil, France; Institut Mondor de recherche biomédicale (IMRB), Inserm U955, i-Biot, Créteil, France
| |
Collapse
|
10
|
Zhang C, Hou Y, Yang Y, Zhang J, Zheng X, Yan J. Second haploidentical bone marrow transplantation with antithymocyte antibody-containing conditioning regimen for graft failure in eight patients with severe aplastic anemia. Sci Rep 2024; 14:2293. [PMID: 38280947 PMCID: PMC10821899 DOI: 10.1038/s41598-024-52917-4] [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: 04/07/2023] [Accepted: 01/25/2024] [Indexed: 01/29/2024] Open
Abstract
The effects of a second haploidentical bone marrow transplantation with an antithymocyte antibody-containing conditioning regimen after graft failure in patients with severe aplastic anemia remain unclear. Eight severe aplastic anemia patients with graft failure with a median age of 12.5 (range, 3-22) years were retrospectively reviewed. At the second transplantation, they received a median mononuclear cell number of 15.7 (range, 11.2-20.9) × 108/kg or a median CD34+ cell number of 6.2 (range, 2.5-17.5) × 106/kg. They were all successfully engrafted, with a median time of 12.5 (range, 11-16) days for neutrophils and 24 (range, 14-50) days for platelets. Three patients developed skin acute graft-versus-host disease Grades I-II, and another 3 developed limited chronic graft-versus-host disease. All patients successfully recovered after treatment with methylprednisolone (0.5-1 mg/kg/day) and tacrolimus. One patient each died of respiratory failure caused by multidrug-resistant Klebsiella pneumoniae at 8 months and invasive fungal disease at 23 months after transplantation. Six patients survived with a 5-year estimated overall survival of 75% and a median follow-up time of 61 (range, 8-129) months. A second haploidentical bone marrow transplantation with an antithymocyte antibody-containing conditioning regimen was feasible for saving severe aplastic anemia patients with graft failure.
Collapse
Affiliation(s)
- Chengtao Zhang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, ShaHeKou District, Dalian, 116027, China
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Blood Stem Cell Transplantation Institute, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, The Second Hospital of Dalian Medical University, Dalian, 116027, China
| | - Yutong Hou
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, ShaHeKou District, Dalian, 116027, China
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Blood Stem Cell Transplantation Institute, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, The Second Hospital of Dalian Medical University, Dalian, 116027, China
| | - Yan Yang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, ShaHeKou District, Dalian, 116027, China
| | - Jingjing Zhang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, ShaHeKou District, Dalian, 116027, China.
- Department of Pediatric, Pediatric Oncology and Hematology Center, The Second Hospital of Dalian Medical University, Dalian, 116027, China.
| | - Xiaoli Zheng
- Department of Hematology, Air Force Medical Center, PLA, No. 3 Fuchen Road, Haidian District, Beijing, 100142, China.
| | - Jinsong Yan
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, ShaHeKou District, Dalian, 116027, China.
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Blood Stem Cell Transplantation Institute, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, The Second Hospital of Dalian Medical University, Dalian, 116027, China.
- Department of Pediatric, Pediatric Oncology and Hematology Center, The Second Hospital of Dalian Medical University, Dalian, 116027, China.
| |
Collapse
|
11
|
McNerney KO, Moskop A, Winestone LE, Baggott C, Talano JA, Schiff D, Rossoff J, Modi A, Verneris MR, Laetsch TW, Schultz L. Practice Preferences for Consolidative Hematopoietic Stem Cell Transplantation Following Tisagenlecleucel in Children and Young Adults with B Cell Acute Lymphoblastic Leukemia. Transplant Cell Ther 2024; 30:75.e1-75.e11. [PMID: 37816472 DOI: 10.1016/j.jtct.2023.10.004] [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: 07/18/2023] [Revised: 08/26/2023] [Accepted: 10/04/2023] [Indexed: 10/12/2023]
Abstract
Treatment with tisagenlecleucel (tisa-cel) achieves excellent complete remission rates in children and young adults with relapsed or refractory B cell acute lymphoblastic leukemia (B-ALL), but approximately 50% maintain long-term remission. Consolidative hematopoietic stem cell transplantation (cHSCT) is a potential strategy to reduce relapse risk, but it carries substantial short- and long-term toxicities. Additionally, several strategies for management of B cell recovery (BCR) and next-generation sequencing (NGS) positivity post-tisa-cel exist, without an accepted standard. We hypothesized that practice preferences surrounding cHSCT, as well as management of BCR and NGS positivity, varies across tisa-cel-prescribing physicians and sought to characterize current practice preferences. A survey focusing on preferences regarding the use of cHSCT, management of BCR, and NGS positivity was distributed to physicians who prescribe tisa-cel for children and young adults with B-ALL. Responses were collected from August 2022 to April 2023. Fifty-nine unique responses were collected across 43 institutions. All respondents prescribed tisa-cel for children and young adults. The clinical focus of respondents was HSCT in 71%, followed by leukemia/lymphoma in 24%. For HSCT-naive patients receiving tisa-cel, 57% of respondents indicated they made individualized decisions for cHSCT based on patient factors, whereas 22% indicated they would avoid cHSCT and 21% indicated they would pursue cHSCT when feasible. Certain factors influenced >50% of respondents towards recommending cHSCT (either an increased likelihood of recommending or always recommending), including preinfusion disease burden >25%, primary refractory B-ALL, M3 bone marrow following reinduction for relapse, KMT2A-rearranged B-ALL, history of blinatumomab nonresponse, and HSCT-naive status. Most respondents indicated they would pursue HSCT for HSCT-naive, total body irradiation (TBI) recipients with BCR before 6 months post-tisa-cel or with NGS positivity at 1 or 3 months post-tisa-cel, although there was variability in responses regarding whether to proceed to HSCT directly or provide intervening therapy prior to HSCT. Fewer respondents recommended HSCT for BCR or NGS positivity in patients with a history of HSCT, in noncandidates for TBI, and in patients with trisomy 21. The results of this survey indicate there exists significant practice variability regarding the use of cHSCT, as well as interventions for post-tisa-cel BCR or NGS positivity. These results highlight areas in which ongoing clinical trials could inform more standardized practice.
Collapse
Affiliation(s)
- Kevin O McNerney
- Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.
| | - Amy Moskop
- Division of HematologyOncologyBlood and Marrow Transplantation, Department of Pediatrics, Medical College of Wisconsin and Children's Wisconsin, Milwaukee, Wisconsin
| | - Lena E Winestone
- Division of Allergy, Immunology, and BMT, Department of Pediatrics, University of California San Francisco Benioff Children's Hospitals, San Francisco, California; UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Christina Baggott
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Julie-An Talano
- Division of HematologyOncologyBlood and Marrow Transplantation, Department of Pediatrics, Medical College of Wisconsin and Children's Wisconsin, Milwaukee, Wisconsin
| | - Deborah Schiff
- Department of Pediatric Hematology and Oncology, Rady Children's Hospital, San Diego, California
| | - Jenna Rossoff
- Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Arunkumar Modi
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Michael R Verneris
- University of Colorado School of Medicine, Children's Hospital of Colorado, Aurora, Colorado
| | - Theodore W Laetsch
- Department of Pediatrics and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Liora Schultz
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
12
|
Ansari F, Behfar M, Jafari L, Mohseni R, Naji P, Karamlou Y, Amirzade-Iranaq MH, Hamidieh AA. A comprehensive comparison between TBI vs non-TBI-based conditioning regimen in pediatric patients with acute lymphoblastic leukemia: A systematic review and meta-analysis. Leuk Res 2023; 135:107416. [PMID: 37918224 DOI: 10.1016/j.leukres.2023.107416] [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: 03/11/2023] [Revised: 09/11/2023] [Accepted: 10/25/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION We aimed to evaluate the efficacy, safety, and latent toxicity of total body irradiation (TBI)-based conditioning regimens compared to non-TBI regimens for pediatric patients (under 18 years old) with acute lymphoblastic leukemia (ALL) undergoing allogeneic hematopoietic stem cell transplantation (HSCT). METHODS A systematic search was performed on MEDLINE, Scopus, WOS, and PMC. Also, a search for grey literature was performed on Google Scholar and relevant articles' references were included. Relevant articles which met the inclusion criteria were retrieved up to October 31th, 2022. CMA version 2 was used for the quantitative synthesis of the data. RESULTS Eight studies on efficacy and safety of TBI and non-TBI as a conditioning regimen were analyzed and six comparative studies on late toxicity were investigated. The meta-analysis revealed a hazard ratio (HR) of 1.508 (95% CI 0.96-2.35) for overall survival (OS) in instances of non-TBI conditioning. Also, an HR of 1.503 (95% CI 1.006-2.25) for disease-free- survival (DFS) favoring TBI-based conditioning. Late complications were reported to be significantly higher in the TBI conditioning regimen group than in the non-TBI group. CONCLUSION It appears that non-TBI regimens are as effective as TBI regimens in pediatrics with ALL regarding OS. Occurrence of latent toxicity is higher with TBI conditioning regimen. Conversely, TBI-based regimens are superior to non-TBI conditioning regimens regarding DFS. Considering all aspects, non-TBI conditioning regimens can be an alternative treatment option for pediatric ALL undergoing HSCT.
Collapse
Affiliation(s)
- Faezeh Ansari
- Pediatric Cell and Gene Therapy Research Centre, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Behfar
- Pediatric Cell and Gene Therapy Research Centre, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Jafari
- Pediatric Cell and Gene Therapy Research Centre, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Rashin Mohseni
- Pediatric Cell and Gene Therapy Research Centre, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Naji
- Pediatric Cell and Gene Therapy Research Centre, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Yalda Karamlou
- Pediatric Cell and Gene Therapy Research Centre, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Amirzade-Iranaq
- Universal Network of Interdisciplinary Research in Oral and Maxillofacial Surgery (UNIROMS), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Centre, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Hao C, Ladbury C, Wong J, Dandapani S. Modern Radiation for Hematologic Stem Cell Transplantation: Total Marrow and Lymphoid Irradiation or Intensity-Modulated Radiation Therapy Total Body Irradiation. Surg Oncol Clin N Am 2023; 32:475-495. [PMID: 37182988 DOI: 10.1016/j.soc.2023.03.001] [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: 05/16/2023]
Abstract
The development of large-field intensity-modulated radiation therapy (IMRT) has enabled the implementation of total marrow irradiation (TMI), total marrow and lymphoid irradiation (TMLI), and IMRT total body irradiation (TBI). IMRT TBI limits doses to organs at risk, primarily the lungs and in some cases the kidneys and lenses, which may mitigate complications. TMI/TMLI allows for dose escalation above TBI radiation therapy doses to malignant sites while still sparing organs at risk. Although still sparingly used, these techniques have established feasibility and demonstrated promise in reducing the adverse effects of TBI while maintaining and potentially improving survival outcomes.
Collapse
Affiliation(s)
- Claire Hao
- Department of Radiation Oncology, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Colton Ladbury
- Department of Radiation Oncology, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Jeffrey Wong
- Department of Radiation Oncology, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Savita Dandapani
- Department of Radiation Oncology, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA.
| |
Collapse
|
14
|
Ozono S, Sakashita K, Yoshida N, Kakuda H, Watanabe K, Maeda M, Ishida Y, Manabe A, Taga T, Muramatsu H. A nationwide survey of late effects in survivors of juvenile myelomonocytic leukemia in Japan. Pediatr Blood Cancer 2023; 70:e30126. [PMID: 36495260 DOI: 10.1002/pbc.30126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/21/2022] [Accepted: 11/09/2022] [Indexed: 12/14/2022]
Abstract
We conducted a cross-sectional study using a questionnaire to explore the late effects in survivors of allogenic hematopoietic stem cell transplantation (HSCT) for juvenile myelomonocytic leukemia (JMML). The attending pediatric hematologists/oncologists completed the questionnaires. Of the 30 survivors, approximately 83% showed more than one late effect. The identified late effects included endocrine, dental, skin, ophthalmologic, musculoskeletal, pulmonary, neurocognitive, and cardiovascular dysfunction. The prevalence of short stature, pulmonary, cardiovascular, and nephrological complications was significantly elevated among survivors who were 12 years or more lapsed after HSCT. Therefore, a multidisciplinary follow-up system for survivors of JMML is crucial.
Collapse
Affiliation(s)
- Shuichi Ozono
- Department of Pediatrics and Child Health, Kurume University School of Medicine, Kurume, Japan
- Japan Children's Cancer Group (JCCG), Tokyo, Japan
| | - Kazuo Sakashita
- Japan Children's Cancer Group (JCCG), Tokyo, Japan
- Department of Hemato-Oncology, Nagano Prefectural Children's Hospital, Nagano, Japan
| | - Nao Yoshida
- Japan Children's Cancer Group (JCCG), Tokyo, Japan
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Harumi Kakuda
- Japan Children's Cancer Group (JCCG), Tokyo, Japan
- Department of Hemato-Oncology, Chiba Prefectural Children's Hospital, Chiba, Japan
| | - Kenichiro Watanabe
- Japan Children's Cancer Group (JCCG), Tokyo, Japan
- Department of Hemato-Oncology, Shizuoka Prefectural Children's Hospital, Shizuoka, Japan
| | - Miho Maeda
- Japan Children's Cancer Group (JCCG), Tokyo, Japan
- Department of Pediatrics, Nippon Medical School, Tokyo, Japan
| | - Yasushi Ishida
- Japan Children's Cancer Group (JCCG), Tokyo, Japan
- Department of Pediatrics, Ehime Prefectural Central Hospital, Ehime, Japan
| | - Atsushi Manabe
- Japan Children's Cancer Group (JCCG), Tokyo, Japan
- Department of Pediatrics, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Takashi Taga
- Japan Children's Cancer Group (JCCG), Tokyo, Japan
- Department of Pediatrics, Shiga Medical College, School of Medicine, Shiga, Japan
| | - Hideki Muramatsu
- Japan Children's Cancer Group (JCCG), Tokyo, Japan
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
15
|
Moreno C, Ramos-Elbal E, Velasco P, Aguilar Y, Gonzáález Martínez B, Fuentes C, Molinos Á, Guerra-García P, Palomo P, Verdu J, Adán Pedroso RM, Vagace JM, López-Duarte M, Regueiro A, Tasso M, Dapena JL, Salinas JA, Navarro S, Bautista F, Lassaletta Á, Lendínez F, Rives S, Pascual A, Rodríguez A, Pérez-Hurtado JM, Fernández JM, Pérez-Martínez A, González-Vicent M, Díaz de Heredia C, Fuster JL. Haploidentical vs. HLA-matched donor hematopoietic stem-cell transplantation for pediatric patients with acute lymphoblastic leukemia in second remission: A collaborative retrospective study of the Spanish Group for Bone Marrow Transplantation in Children (GETMON/GETH) and the Spanish Childhood Relapsed ALL Board (ReALLNet). Front Pediatr 2023; 11:1140637. [PMID: 37020654 PMCID: PMC10067875 DOI: 10.3389/fped.2023.1140637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/23/2023] [Indexed: 04/07/2023] Open
Abstract
Introduction Studies addressing the role of haploidentical as alternative to HLA-matched donors for stem cell transplantation (SCT) often include patients with diverse hematological malignancies in different remission statuses. Methods We compared outcomes of children with acute lymphoblastic leukemia (ALL) undergoing SCT in second complete remission (CR2) from haploidentical (n = 25) versus HLA-matched donor (n = 51). Results Patients were equally distributed across both groups according to age, immunophenotype, time to and site of relapse, relapse risk-group allocation, and minimal residual disease (MRD) before SCT. Incidence of graft failure, acute graft versus host disease (GVHD), and other early complications did not differ between both groups. We found no differences in overall survival (58.7% versus 59.5%; p = .8), leukemia free survival (LFS) (48% versus 36.4%; p = .5), event free survival (40% versus 34.4%; p = .69), cumulative incidence (CI) of subsequent relapse (28% versus 40.9%; p = .69), treatment related mortality (24% versus 23.6%; p = .83), CI of cGVHD (4.5% versus 18.7%; p = .2), and chronic GVHD-free and leukemia-free survival (44% versus 26.3%; p = .3) after haploidentical donor SCT. Chronic GVHD (HR = 0.09; p=.02) had protective impact, and MRD ≥ 0.01% before SCT (HR = 2.59; p=.01) had unfavorable impact on LFS. Discussion These results support the role of haploidentical donor SCT in children with ALL in CR2.
Collapse
Affiliation(s)
- Celia Moreno
- Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | | | - Pablo Velasco
- Hospital Universitario Vall d’Hebron, Vall d’Hebron Institut de Recerca, Barcelona, Spain
| | | | - Berta Gonzáález Martínez
- Hospital Universitario La Paz, IdiPAZ, Instituto de Investigación, Hospital Universitario La Paz, Madrid, Spain
| | | | | | - Pilar Guerra-García
- Hospital Universitario La Paz, IdiPAZ, Instituto de Investigación, Hospital Universitario La Paz, Madrid, Spain
- Hospital Universitario 12 de octubre, Madrid, Spain
| | - Pilar Palomo
- Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Jaime Verdu
- Hospital Universitario de Valencia, Valencia, Spain
| | | | | | - Mónica López-Duarte
- Hospital de Valdecilla, Instituto de Investigación Sanitaria Valdecilla, IDIVAL, Santander, Spain
| | - Alexandra Regueiro
- Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | - María Tasso
- Hospital General Universitario Doctor Balmis, Alicante, Spain
| | - José Luis Dapena
- Pediatric Cancer Center, Hospital Sant Joan de Déu, Barcelona, Spain
- Institut de Recerca San Joan de Déu,Barcelona, Spain
| | | | - Samuel Navarro
- Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | | | | | | | - Susana Rives
- Pediatric Cancer Center, Hospital Sant Joan de Déu, Barcelona, Spain
- Institut de Recerca San Joan de Déu,Barcelona, Spain
| | | | | | | | | | - Antonio Pérez-Martínez
- Hospital Universitario La Paz, IdiPAZ, Instituto de Investigación, Hospital Universitario La Paz, Madrid, Spain
| | | | | | - José Luis Fuster
- Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
- Correspondence: José Luis Fuster
| |
Collapse
|
16
|
Liu KX, Poux N, Shin KY, Moore N, Chen YH, Margossian S, Whangbo JS, Duncan CN, Lehmann LE, Marcus KJ. Comparison of Pulmonary Toxicity after Total Body Irradiation- and Busulfan-Based Myeloablative Conditioning for Allogeneic Hematopoietic Stem Cell Transplantation in Pediatric Patients. Transplant Cell Ther 2022; 28:502.e1-502.e12. [PMID: 35623615 PMCID: PMC11075968 DOI: 10.1016/j.jtct.2022.05.028] [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: 11/09/2021] [Revised: 05/14/2022] [Accepted: 05/16/2022] [Indexed: 11/22/2022]
Abstract
Pulmonary toxicity after allogeneic hematopoietic stem cell transplantation (allo-HSCT) for childhood leukemia and myelodysplastic syndrome (MDS), along with the impact of different myeloablative conditioning regimens, remain incompletely described. Here we compared the acute and long-term incidence of pulmonary toxicity (PT) after total body irradiation (TBI)- and busulfan-based myeloablative conditioning. We conducted this retrospective cohort study of 311 consecutive pediatric patients with leukemia or MDS who underwent allo-HSCT at Dana-Farber Cancer Institute/Boston Children's Hospital between 2008 and 2018. PT was graded using Common Terminology Criteria for Adverse Events version 5.0. The primary objective was to compare the cumulative incidence of grade ≥3 and grade 5 PT after TBI-based and busulfan-based myeloablative conditioning using Gray's test. Secondary objectives were to determine factors associated with PT and overall survival (OS) using competing risk analysis and Cox regression analyses, respectively. There was no significant difference between the TBI-conditioned group (n = 227) and the busulfan-conditioned group (n = 84) in the incidence of grade ≥3 PT (29.2% versus 34.7% at 2 years; P = .26) or grade 5 pulmonary toxicity (6.2% versus 6.1% at 2 years; P = .47). Age (hazard ratio [HR], 1.70, 95% confidence interval [CI], 1.11 to 2.59; P = .01), grade ≥2 PT prior to allo-HSCT or preexisting pulmonary conditions (HR, 1.84, 95% CI, 1.24 to 2.72; P < .01), acute graft-versus-host disease (GVHD) (HR, 2.50; 95% CI, 1.51 to 4.14; P < .01), and chronic GVHD (HR, 2.61; 95% CI, 1.26 to 5.42; P = .01) were associated with grade ≥3 PT on multivariable analysis. Grade ≥3 PT was associated with worse OS (81.1% versus 61.5% at 2 years; P < .01). In pediatric allo-HSCT recipients, rates of PT were similar in recipients of TBI-based and recipients of busulfan-based myeloablative conditioning regimens. Age, the presence of PT or preexisting pulmonary conditions prior to transplantation, and the development of either acute or chronic GVHD were associated with grade ≥3 PT post-transplantation. Furthermore, the occurrence of grade 3-4 PT post-transplantation was associated with inferior OS.
Collapse
Affiliation(s)
- Kevin X Liu
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Kee-Young Shin
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Yu-Hui Chen
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Steven Margossian
- Pediatric Stem Cell Transplant, Division of Pediatric Oncology, Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jennifer S Whangbo
- Division of Hematology/Oncology, Stem Cell Transplant Program, Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Christine N Duncan
- Pediatric Stem Cell Transplant, Division of Pediatric Oncology, Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Leslie E Lehmann
- Pediatric Stem Cell Transplant, Division of Pediatric Oncology, Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Karen J Marcus
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
17
|
Rossini L, Durante C, Bresolin S, Opocher E, Marzollo A, Biffi A. Diagnostic Strategies and Algorithms for Investigating Cancer Predisposition Syndromes in Children Presenting with Malignancy. Cancers (Basel) 2022; 14:cancers14153741. [PMID: 35954404 PMCID: PMC9367486 DOI: 10.3390/cancers14153741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Here we provide an overview of several genetically determined conditions that predispose to the development of solid and hematologic malignancies in children. Diagnosing these conditions, whose prevalence is estimated around 10% in children with cancer, is useful to warrant personalized oncologic treatment and follow-up, as well as psychological and genetic counseling to these children and their families. We reviewed the most recent studies focusing on the prevalence of cancer predisposition syndromes in cancer-bearing children and the most-used clinical screening tools. Our work highlighted the value of clinical screening tools in the management of young cancer patients, especially in settings where genetic testing is not promptly accessible. Abstract In the past recent years, the expanding use of next-generation sequencing has led to the discovery of new cancer predisposition syndromes (CPSs), which are now known to be responsible for up to 10% of childhood cancers. As knowledge in the field is in constant evolution, except for a few “classic” CPSs, there is no consensus about when and how to perform germline genetic diagnostic studies in cancer-bearing children. Several clinical screening tools have been proposed to help identify the patients who carry higher risk, with heterogeneous strategies and results. After introducing the main clinical and molecular features of several CPSs predisposing to solid and hematological malignancies, we compare the available clinical evidence on CPS prevalence in pediatric cancer patients and on the most used decision-support tools in identifying the patients who could benefit from genetic counseling and/or direct genetic testing. This analysis highlighted that a personalized stepwise approach employing clinical screening tools followed by sequencing in high-risk patients might be a reasonable and cost-effective strategy in the care of children with cancer.
Collapse
Affiliation(s)
- Linda Rossini
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Via Giustiniani 3, 35128 Padua, Italy; (L.R.); (C.D.); (S.B.); (E.O.)
| | - Caterina Durante
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Via Giustiniani 3, 35128 Padua, Italy; (L.R.); (C.D.); (S.B.); (E.O.)
| | - Silvia Bresolin
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Via Giustiniani 3, 35128 Padua, Italy; (L.R.); (C.D.); (S.B.); (E.O.)
- Maternal and Child Health Department, Padua University, Via Giustiniani, 3, 35128 Padua, Italy
| | - Enrico Opocher
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Via Giustiniani 3, 35128 Padua, Italy; (L.R.); (C.D.); (S.B.); (E.O.)
| | - Antonio Marzollo
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Via Giustiniani 3, 35128 Padua, Italy; (L.R.); (C.D.); (S.B.); (E.O.)
- Correspondence: (A.M.); (A.B.)
| | - Alessandra Biffi
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Via Giustiniani 3, 35128 Padua, Italy; (L.R.); (C.D.); (S.B.); (E.O.)
- Maternal and Child Health Department, Padua University, Via Giustiniani, 3, 35128 Padua, Italy
- Correspondence: (A.M.); (A.B.)
| |
Collapse
|
18
|
Long-Term Health Effects of Curative Therapies on Heart, Lungs, and Kidneys for Individuals with Sickle Cell Disease Compared to Those with Hematologic Malignancies. J Clin Med 2022; 11:jcm11113118. [PMID: 35683502 PMCID: PMC9181610 DOI: 10.3390/jcm11113118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 12/30/2022] Open
Abstract
The goal of curing children and adults with sickle cell disease (SCD) is to maximize benefits and minimize intermediate and long-term adverse outcomes so that individuals can live an average life span with a high quality of life. While greater than 2000 individuals with SCD have been treated with curative therapy, systematic studies have not been performed to evaluate the long-term health effects of hematopoietic stem cell transplant (HSCT) in this population. Individuals with SCD suffer progressive heart, lung, and kidney disease prior to curative therapy. In adults, these sequalae are associated with earlier death. In comparison, individuals who undergo HSCT for cancer are heavily pretreated with chemotherapy, resulting in potential acute and chronic heart, lung, and kidney disease. The long-term health effects on the heart, lung, and kidney for children and adults undergoing HSCT for cancer have been extensively investigated. These studies provide the best available data to extrapolate the possible late health effects after curative therapy for SCD. Future research is needed to evaluate whether HSCT abates, stabilizes, or exacerbates heart, lung, kidney, and other diseases in children and adults with SCD receiving myeloablative and non-myeloablative conditioning regimens for curative therapy.
Collapse
|
19
|
Poyer F, Füreder A, Holter W, Peters C, Boztug H, Dworzak M, Engstler G, Friesenbichler W, Köhrer S, Lüftinger R, Ronceray L, Witt V, Pichler H, Attarbaschi A. Relapsed acute lymphoblastic leukaemia after allogeneic stem cell transplantation: a therapeutic dilemma challenging the armamentarium of immunotherapies currently available (case reports). Ther Adv Hematol 2022; 13:20406207221099468. [PMID: 35646299 PMCID: PMC9134426 DOI: 10.1177/20406207221099468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 04/19/2022] [Indexed: 12/05/2022] Open
Abstract
While survival rates in paediatric acute lymphoblastic leukaemia (ALL) nowadays
exceed 90%, systemic ALL relapse, especially after haemopoietic stem cell
transplantation (HSCT), is associated with a poor outcome. As there is currently
no standardized treatment for this situation, individualized treatment is often
pursued. Exemplified by two clinical scenarios, the aim of this article is to
highlight the challenge for treating physicians to find a customized treatment
strategy integrating the role of conventional chemotherapy, immunotherapeutic
approaches and second allogeneic HSCT. Case 1 describes a 2-year-old girl with
an early isolated bone marrow relapse of an infant
KMT2A-rearranged B-cell precursor ALL after allogeneic HSCT.
After bridging chemotherapy and lymphodepleting chemotherapy, chimeric antigen
receptor (CAR) T-cells (tisagenlecleucel) were administered for remission
induction, followed by a second HSCT from the 9/10 human leukocyte antigen
(HLA)-matched mother. Case 2 describes a 16-year-old girl with a late, isolated
bone marrow relapse of B-cell precursor ALL after allogeneic HSCT who
experienced severe treatment toxicities including stage IV renal insufficiency.
After dose-reduced bridging chemotherapy, CAR T-cells (tisagenlecleucel) were
administered for remission induction despite a CD19- clone without
prior lymphodepletion due to enhanced persisting toxicity. This was followed by
a second allogeneic HSCT from the haploidentical mother. While patient 2
relapsed around Day + 180 after the second HSCT, patient 1 is still in complete
remission >360 days after the second HSCT. Both cases demonstrate the
challenges associated with systemic ALL relapse after first allogeneic HSCT,
including chemotherapy-resistant disease and persisting organ damage inflicted
by previous therapy. Immunotherapeutic approaches, such as CAR T-cells, can
induce remission and enable a second allogeneic HSCT. However, optimal therapy
for systemic ALL relapse after first HSCT remains to be defined.
Collapse
Affiliation(s)
- Fiona Poyer
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Anna Füreder
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Holter
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Christina Peters
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Heidrun Boztug
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Michael Dworzak
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Gernot Engstler
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Waltraud Friesenbichler
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Stefan Köhrer
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Roswitha Lüftinger
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Leila Ronceray
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Volker Witt
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Herbert Pichler
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Andishe Attarbaschi
- Department of Pediatric Haematology and Oncology, St. Anna Children’s Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
20
|
Otth M, Yammine S, Usemann J, Latzin P, Mader L, Spycher B, Güngör T, Scheinemann K, Kuehni CE. Longitudinal lung function in childhood cancer survivors after hematopoietic stem cell transplantation. Bone Marrow Transplant 2022; 57:207-214. [PMID: 34750563 PMCID: PMC8821009 DOI: 10.1038/s41409-021-01509-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 11/08/2022]
Abstract
Longitudinal data on pulmonary function after pediatric allogeneic or autologous hematopoietic stem cell transplantation (HSCT) are rare. We examined pulmonary function and associated risk factors in 5-year childhood cancer survivors (CCSs) longitudinally. We included 74 CCSs diagnosed between 1976 and 2010, treated with HSCT, and with at least two pulmonary function tests performed during follow-up. Median follow-up was 9 years (range 6-13). We described pulmonary function as z-scores for lung volumes (forced vital capacity [FVC], residual volume [RV], total lung capacity [TLC]), flows (forced expiratory volume in 1 s [FEV1], maximal mid-expiratory flow [MMEF]), and diffusion capacity for carbon monoxide (DLCO) and assessed associations with potential risk factors using multivariable regression analysis. The median z-scores for FEV1, FVC, and TLC were below the expected throughout the follow-up period. This was not the case for RV, MMEF and DLCO. Female gender, radiotherapy to the chest, and relapse were associated with lower z-scores of FEV1, FVC, MMEF, RV or DLCO. Childhood cancer survivors after HSCT are at risk of pulmonary dysfunction. The complex and multifactorial etiology of pulmonary dysfunction emphasizes the need for longitudinal prospective studies to better characterize the course and causes of pulmonary function impairment in CCSs.
Collapse
Affiliation(s)
- Maria Otth
- Childhood Cancer Research Group, Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
- Division of Oncology- Hematology, Department of Pediatrics, Kantonsspital Aarau, Aarau, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Department of Oncology, Hematology, Immunology, Stem Cell Transplantation and Somatic Gene Therapy, University Children's Hospital Zurich - Eleonore Foundation, Zurich, Switzerland
| | - Sophie Yammine
- Division of Respiratory Medicine, Department of Pediatrics, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Jakob Usemann
- Division of Respiratory Medicine, Department of Pediatrics, University Children's Hospital Zurich-Eleonore Foundation, Zurich, Switzerland
- University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Philipp Latzin
- Division of Respiratory Medicine, Department of Pediatrics, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Luzius Mader
- Childhood Cancer Research Group, Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
| | - Ben Spycher
- Childhood Cancer Research Group, Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
| | - Tayfun Güngör
- Department of Oncology, Hematology, Immunology, Stem Cell Transplantation and Somatic Gene Therapy, University Children's Hospital Zurich - Eleonore Foundation, Zurich, Switzerland
| | - Katrin Scheinemann
- Division of Oncology- Hematology, Department of Pediatrics, Kantonsspital Aarau, Aarau, Switzerland
- University of Basel, Basel, Switzerland
- Department of Pediatrics, McMaster University Hamilton, Hamilton, ON, Canada
| | - Claudia E Kuehni
- Childhood Cancer Research Group, Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland.
- Division of Hematology/Oncology, Department of Pediatrics, Inselspital, University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
21
|
Buechner J, Caruana I, Künkele A, Rives S, Vettenranta K, Bader P, Peters C, Baruchel A, Calkoen FG. Chimeric Antigen Receptor T-Cell Therapy in Paediatric B-Cell Precursor Acute Lymphoblastic Leukaemia: Curative Treatment Option or Bridge to Transplant? Front Pediatr 2022; 9:784024. [PMID: 35145941 PMCID: PMC8823293 DOI: 10.3389/fped.2021.784024] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/02/2021] [Indexed: 01/02/2023] Open
Abstract
Chimeric antigen receptor T-cell therapy (CAR-T) targeting CD19 has been associated with remarkable responses in paediatric patients and adolescents and young adults (AYA) with relapsed/refractory (R/R) B-cell precursor acute lymphoblastic leukaemia (BCP-ALL). Tisagenlecleucel, the first approved CD19 CAR-T, has become a viable treatment option for paediatric patients and AYAs with BCP-ALL relapsing repeatedly or after haematopoietic stem cell transplantation (HSCT). Based on the chimeric antigen receptor molecular design and the presence of a 4-1BB costimulatory domain, tisagenlecleucel can persist for a long time and thereby provide sustained leukaemia control. "Real-world" experience with tisagenlecleucel confirms the safety and efficacy profile observed in the pivotal registration trial. Recent guidelines for the recognition, management and prevention of the two most common adverse events related to CAR-T - cytokine release syndrome and immune-cell-associated neurotoxicity syndrome - have helped to further decrease treatment toxicity. Consequently, the questions of how and for whom CD19 CAR-T could substitute HSCT in BCP-ALL are inevitable. Currently, 40-50% of R/R BCP-ALL patients relapse post CD19 CAR-T with either CD19- or CD19+ disease, and consolidative HSCT has been proposed to avoid disease recurrence. Contrarily, CD19 CAR-T is currently being investigated in the upfront treatment of high-risk BCP-ALL with an aim to avoid allogeneic HSCT and associated treatment-related morbidity, mortality and late effects. To improve survival and decrease long-term side effects in children with BCP-ALL, it is important to define parameters predicting the success or failure of CAR-T, allowing the careful selection of candidates in need of HSCT consolidation. In this review, we describe the current clinical evidence on CAR-T in BCP-ALL and discuss factors associated with response to or failure of this therapy: product specifications, patient- and disease-related factors and the impact of additional therapies given before (e.g., blinatumomab and inotuzumab ozogamicin) or after infusion (e.g., CAR-T re-infusion and/or checkpoint inhibition). We discuss where to position CAR-T in the treatment of BCP-ALL and present considerations for the design of supportive trials for the different phases of disease. Finally, we elaborate on clinical settings in which CAR-T might indeed replace HSCT.
Collapse
Affiliation(s)
- Jochen Buechner
- Department of Pediatric Hematology and Oncology, Oslo University Hospital, Oslo, Norway
| | - Ignazio Caruana
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, Würzburg, Germany
| | - Annette Künkele
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Susana Rives
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu de Barcelona, Institut per la Recerca Sant Joan de Déu, Barcelona, Spain
| | - Kim Vettenranta
- University of Helsinki and Children's Hospital, University of Helsinki, Helsinki, Finland
| | - Peter Bader
- Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Department for Children and Adolescents, University Hospital, Goethe University, Frankfurt, Germany
| | - Christina Peters
- St. Anna Children's Hospital, Medical University Vienna, Vienna, Austria
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - André Baruchel
- Université de Paris et Institut de Recherche Saint-Louis (EA 35-18) and Hôpital Universitaire Robert Debré (APHP), Paris, France
| | - Friso G. Calkoen
- Department of Stem Cell Transplantation and Cellular Therapy, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| |
Collapse
|
22
|
Andolina JR, Fries C, Boulware R, Vargas A, Fraint E, Barth M, Ambrusko S, Comito M, Monteleone P. Successful Bone Marrow Transplantation With Intensive Post-transplant Intrathecal Chemotherapy for CNS Relapsed AML in 2 Infants. J Pediatr Hematol Oncol 2022; 44:e264-e267. [PMID: 33843815 DOI: 10.1097/mph.0000000000002151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 02/15/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Infant acute myeloid leukemia is a rare but aggressive form of leukemia. OBSERVATION We report 2 children who presented with hyperleukocytosis, subsequently diagnosed with infant acute myeloid leukemia, and both developed isolated central nervous system relapse while on chemotherapy. Both infants underwent successful bone marrow transplantation with myeloablative conditioning (thiotepa, busulfan, and cyclophosphamide) without radiation, followed by 12 empiric post-transplant lumbar punctures with intrathecal cytarabine. Both patients tolerated these therapies well, and are without infections, chronic graft-versus-host disease, or any post-transplant sequelae. CONCLUSION Nonradiation-based conditioning followed by empiric central nervous system-directed intrathecal chemotherapy may be considered for high-risk infants with leukemia.
Collapse
Affiliation(s)
- Jeffrey R Andolina
- Department of Pediatrics, Golisano Children's Hospital, University of Rochester Medical Center, Rochester
| | - Carol Fries
- Department of Pediatrics, Golisano Children's Hospital, University of Rochester Medical Center, Rochester
| | - Renee Boulware
- Department of Pediatrics, Golisano Children's Hospital, University of Rochester Medical Center, Rochester
| | - Alyssa Vargas
- Department of Pediatrics, Golisano Children's Hospital, University of Rochester Medical Center, Rochester
| | - Ellen Fraint
- Department of Pediatrics, Children's Hospital at Montefiore, New York
| | - Matthew Barth
- Department of Pediatrics, Oshei Children's Hospital, Roswell Park Cancer Institute, Buffalo
| | - Steven Ambrusko
- Department of Pediatrics, Oshei Children's Hospital, Roswell Park Cancer Institute, Buffalo
| | - Melanie Comito
- Department of Pediatrics, Upstate Golisano Children's Hospital, Syracuse, NY
| | - Philip Monteleone
- Department of Pediatrics, Upstate Golisano Children's Hospital, Syracuse, NY
| |
Collapse
|
23
|
Incidence of subsequent malignancies after total body irradiation-based allogeneic HSCT in children with ALL - long-term follow-up from the prospective ALL-SCT 2003 trial. Leukemia 2022; 36:2567-2576. [PMID: 36097283 PMCID: PMC9613465 DOI: 10.1038/s41375-022-01693-z] [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: 07/20/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/24/2022]
Abstract
Total body irradiation (TBI)-based conditioning is associated with superior leukemia-free survival in children with ALL undergoing HSCT. However, the risk for subsequent malignant neoplasms (SMN) remains a significant concern. We analyzed 705 pediatric patients enrolled in the prospective ALL-SCT-BFM-2003 trial and its subsequent registry. Patients >2 years received conditioning with TBI 12 Gy/etoposide (n = 558) and children ≤2 years of age or with contraindications for TBI received busulfan/cyclophosphamide/etoposide (n = 110). The 5- and 10-year cumulative incidence of SMN was 0.02 ± 0.01 and 0.13 ± 0.03, respectively. In total, 39 SMN (34 solid tumors, 5 MDS/AML) were diagnosed in 33 patients at a median of 5.8 years (1.7-13.4), exclusively in the TBI group. Of 33 affected patients, 21 (64%) are alive at a median follow-up of 5.1 years (0-9.9) after diagnosis of their first SMN. In univariate analysis, neither age at HSCT, donor type, acute GVHD, chronic GVHD, nor CMV constituted a significant risk factor for SMN. The only significant risk factor was TBI versus non-TBI based conditioning. This analysis confirms and quantifies the increased risk of SMN in children with ALL after conditioning with TBI. Future strategies to avoid TBI will need careful tailoring within prospective, controlled studies to prevent unfavorable outcomes.
Collapse
|
24
|
Hoeben BAW, Wong JYC, Fog LS, Losert C, Filippi AR, Bentzen SM, Balduzzi A, Specht L. Total Body Irradiation in Haematopoietic Stem Cell Transplantation for Paediatric Acute Lymphoblastic Leukaemia: Review of the Literature and Future Directions. Front Pediatr 2021; 9:774348. [PMID: 34926349 PMCID: PMC8678472 DOI: 10.3389/fped.2021.774348] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/03/2021] [Indexed: 12/13/2022] Open
Abstract
Total body irradiation (TBI) has been a pivotal component of the conditioning regimen for allogeneic myeloablative haematopoietic stem cell transplantation (HSCT) in very-high-risk acute lymphoblastic leukaemia (ALL) for decades, especially in children and young adults. The myeloablative conditioning regimen has two aims: (1) to eradicate leukaemic cells, and (2) to prevent rejection of the graft through suppression of the recipient's immune system. Radiotherapy has the advantage of achieving an adequate dose effect in sanctuary sites and in areas with poor blood supply. However, radiotherapy is subject to radiobiological trade-offs between ALL cell destruction, immune and haematopoietic stem cell survival, and various adverse effects in normal tissue. To diminish toxicity, a shift from single-fraction to fractionated TBI has taken place. However, HSCT and TBI are still associated with multiple late sequelae, leaving room for improvement. This review discusses the past developments of TBI and considerations for dose, fractionation and dose-rate, as well as issues regarding TBI setup performance, limitations and possibilities for improvement. TBI is typically delivered using conventional irradiation techniques and centres have locally developed heterogeneous treatment methods and ways to achieve reduced doses in several organs. There are, however, limitations in options to shield organs at risk without compromising the anti-leukaemic and immunosuppressive effects of conventional TBI. Technological improvements in radiotherapy planning and delivery with highly conformal TBI or total marrow irradiation (TMI), and total marrow and lymphoid irradiation (TMLI) have opened the way to investigate the potential reduction of radiotherapy-related toxicities without jeopardising efficacy. The demonstration of the superiority of TBI compared with chemotherapy-only conditioning regimens for event-free and overall survival in the randomised For Omitting Radiation Under Majority age (FORUM) trial in children with high-risk ALL makes exploration of the optimal use of TBI delivery mandatory. Standardisation and comprehensive reporting of conventional TBI techniques as well as cooperation between radiotherapy centres may help to increase the ratio between treatment outcomes and toxicity, and future studies must determine potential added benefit of innovative conformal techniques to ultimately improve quality of life for paediatric ALL patients receiving TBI-conditioned HSCT.
Collapse
Affiliation(s)
- Bianca A. W. Hoeben
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Jeffrey Y. C. Wong
- Department of Radiation Oncology, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, United States
| | - Lotte S. Fog
- Alfred Health Radiation Oncology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Christoph Losert
- Department of Radiation Oncology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Andrea R. Filippi
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Søren M. Bentzen
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Adriana Balduzzi
- Stem Cell Transplantation Unit, Clinica Paediatrica Università degli Studi di Milano Bicocca, Monza, Italy
| | - Lena Specht
- Department of Oncology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Vogel J, Hui S, Hua CH, Dusenbery K, Rassiah P, Kalapurakal J, Constine L, Esiashvili N. Pulmonary Toxicity After Total Body Irradiation - Critical Review of the Literature and Recommendations for Toxicity Reporting. Front Oncol 2021; 11:708906. [PMID: 34513689 PMCID: PMC8428368 DOI: 10.3389/fonc.2021.708906] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Total body irradiation is an effective conditioning regimen for allogeneic stem cell transplantation in pediatric and adult patients with high risk or relapsed/refractory leukemia. The most common adverse effect is pulmonary toxicity including idiopathic pneumonia syndrome (IPS). As centers adopt more advanced treatment planning techniques for TBI, total marrow irradiation (TMI), or total marrow and lymphoid irradiation (TMLI) there is a greater need to understand treatment-related risks for IPS for patients treated with conventional TBI. However, definitions of IPS as well as risk factors for IPS remain poorly characterized. In this study, we perform a critical review to further evaluate the literature describing pulmonary outcomes after TBI. MATERIALS AND METHODS A search of publications from 1960-2020 was undertaken in PubMed, Embase, and Cochrane Library. Search terms included "total body irradiation", "whole body radiation", "radiation pneumonias", "interstitial pneumonia", and "bone marrow transplantation". Demographic and treatment-related data was abstracted and evidence quality supporting risk factors for pulmonary toxicity was evaluated. RESULTS Of an initial 119,686 publications, 118 met inclusion criteria. Forty-six (39%) studies included a definition for pulmonary toxicity. A grading scale was provided in 20 studies (17%). In 42% of studies the lungs were shielded to a set mean dose of 800cGy. Fourteen (12%) reported toxicity outcomes by patient age. Reported pulmonary toxicity ranged from 0-71% of patients treated with TBI, and IPS ranged from 1-60%. The most common risk factors for IPS were receipt of a TBI containing regimen, increasing dose rate, and lack of pulmonary shielding. Four studies found an increasing risk of pulmonary toxicity with increasing age. CONCLUSIONS Definitions of IPS as well as demographic and treatment-related risk factors remain poorly characterized in the literature. We recommend routine adoption of the diagnostic workup and the definition of IPS proposed by the American Thoracic Society. Additional study is required to determine differences in clinical and treatment-related risk between pediatric and adult patients. Further study using 3D treatment planning is warranted to enhance dosimetric precision and correlation of dose volume histograms with toxicities.
Collapse
Affiliation(s)
- Jennifer Vogel
- Department of Radiation Oncology, Bon Secours Merch Health St. Francis Cancer Center, Greenville, SC, United States
| | - Susanta Hui
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - Chia-Ho Hua
- Department of Radiation Oncology, St Jude Children’s Research Hospital, Memphis, TN, United States
| | - Kathryn Dusenbery
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Premavarthy Rassiah
- Department of Radiation Oncology, University of Utah Huntsman Cancer Hospital, Salt Lake City, UT, United States
| | - John Kalapurakal
- Department of Radiation Oncology, Northwestern University School of Medicine, Chicago, IL, United States
| | - Louis Constine
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY, United States
| | - Natia Esiashvili
- Department of Radiation Oncology, Emory School of Medicine, Atlanta, GA, United States
| |
Collapse
|
26
|
The Role of Allogeneic Hematopoietic Stem Cell Transplantation in Pediatric Leukemia. J Clin Med 2021; 10:jcm10173790. [PMID: 34501237 PMCID: PMC8432223 DOI: 10.3390/jcm10173790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/08/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) offers potentially curative treatment for many children with high-risk or relapsed acute leukemia (AL), thanks to the combination of intense preparative radio/chemotherapy and the graft-versus-leukemia (GvL) effect. Over the years, progress in high-resolution donor typing, choice of conditioning regimen, graft-versus-host disease (GvHD) prophylaxis and supportive care measures have continuously improved overall transplant outcome, and recent successes using alternative donors have extended the potential application of allotransplantation to most patients. In addition, the importance of minimal residual disease (MRD) before and after transplantation is being increasingly clarified and MRD-directed interventions may be employed to further ameliorate leukemia-free survival after allogeneic HSCT. These advances have occurred in parallel with continuous refinements in chemotherapy protocols and the development of targeted therapies, which may redefine the indications for HSCT in the coming years. This review discusses the role of HSCT in childhood AL by analysing transplant indications in both acute lymphoblastic and acute myeloid leukemia, together with current and most promising strategies to further improve transplant outcome, including optimization of conditioning regimen and MRD-directed interventions.
Collapse
|
27
|
Prednisolone poor response is not an indication for HSCT in pediatric B-cell precursor acute lymphoblastic leukemia in first remission: results from JACLS ALL-02 study. Int J Hematol 2021; 113:893-902. [PMID: 33641058 DOI: 10.1007/s12185-021-03110-0] [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: 12/11/2020] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 10/22/2022]
Abstract
Approximately 90% of pediatric acute lymphoblastic leukemia (ALL) cases are curable with intensified chemotherapy, but very high-risk patients may require hematopoietic stem cell transplantation (HSCT). A suitable indication for HSCT in the first complete remission (CR1) should be defined to protect patients from long-term complications. We report the outcomes of HSCT in CR1 from the Japan Association of Childhood Leukemia Study (JACLS) ALL-02 study and reassess indications for HSCT. Of 1114 patients, 71 (6.4%) received HSCT in CR1. Indications included high-risk cytogenetic abnormalities and non-CR on day 33. Patients with B-cell precursor (BCP) ALL and a prednisolone poor response (PPR) received HSCT when leukocyte antigen-matched siblings were available. The 4-year overall survival (OS) of transplanted patients was 78.8% (confidence interval 67.3-86.6). Multivariate analysis revealed that cord blood transplantation was associated with poor OS. For BCP-ALL patients with PPR who achieved CR1 after induction therapy, HSCT in CR1 showed excellent outcomes (4-year OS 90.9%) but demonstrated no survival advantage as the outcome with chemotherapy was also excellent (4-year OS 97.0%). This study suggests that in BCP-ALL patients PPR is not an indication for HSCT in CR1. Precise evaluation of treatment responses would increase sophistication of indications for HSCT in CR1.
Collapse
|
28
|
Gabriel M, Hoeben BAW, Uhlving HH, Zajac-Spychala O, Lawitschka A, Bresters D, Ifversen M. A Review of Acute and Long-Term Neurological Complications Following Haematopoietic Stem Cell Transplant for Paediatric Acute Lymphoblastic Leukaemia. Front Pediatr 2021; 9:774853. [PMID: 35004543 PMCID: PMC8734594 DOI: 10.3389/fped.2021.774853] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/02/2021] [Indexed: 12/02/2022] Open
Abstract
Despite advances in haematopoietic stem cell transplant (HSCT) techniques, the risk of serious side effects and complications still exists. Neurological complications, both acute and long term, are common following HSCT and contribute to significant morbidity and mortality. The aetiology of neurotoxicity includes infections and a wide variety of non-infectious causes such as drug toxicities, metabolic abnormalities, irradiation, vascular and immunologic events and the leukaemia itself. The majority of the literature on this subject is focussed on adults. The impact of the combination of neurotoxic drugs given before and during HSCT, radiotherapy and neurological complications on the developing and vulnerable paediatric and adolescent brain remains unclear. Moreover, the age-related sensitivity of the nervous system to toxic insults is still being investigated. In this article, we review current evidence regarding neurotoxicity following HSCT for acute lymphoblastic leukaemia in childhood. We focus on acute and long-term impacts. Understanding the aetiology and long-term sequelae of neurological complications in children is particularly important in the current era of immunotherapy for acute lymphoblastic leukaemia (such as chimeric antigen receptor T cells and bi-specific T-cell engager antibodies), which have well-known and common neurological side effects and may represent a future treatment modality for at least a fraction of HSCT-recipients.
Collapse
Affiliation(s)
- Melissa Gabriel
- Cancer Centre for Children, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Bianca A W Hoeben
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Hilde Hylland Uhlving
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Olga Zajac-Spychala
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Poznań, Poland
| | - Anita Lawitschka
- Haematopoietic Stem Cell Transplant Unit, St. Anna Children's Hospital, Medical University Vienna, Vienna, Austria
| | - Dorine Bresters
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Marianne Ifversen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
29
|
Diesch-Furlanetto T, Gabriel M, Zajac-Spychala O, Cattoni A, Hoeben BAW, Balduzzi A. Late Effects After Haematopoietic Stem Cell Transplantation in ALL, Long-Term Follow-Up and Transition: A Step Into Adult Life. Front Pediatr 2021; 9:773895. [PMID: 34900873 PMCID: PMC8652149 DOI: 10.3389/fped.2021.773895] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
Haematopoietic stem cell transplant (HSCT) can be a curative treatment for children and adolescents with very-high-risk acute lymphoblastic leukaemia (ALL). Improvements in supportive care and transplant techniques have led to increasing numbers of long-term survivors worldwide. However, conditioning regimens as well as transplant-related complications are associated with severe sequelae, impacting patients' quality of life. It is widely recognised that paediatric HSCT survivors must have timely access to life-long care and surveillance in order to prevent, ameliorate and manage all possible adverse late effects of HSCT. This is fundamentally important because it can both prevent ill health and optimise the quality and experience of survival following HSCT. Furthermore, it reduces the impact of preventable chronic illness on already under-resourced health services. In addition to late effects, survivors of paediatric ALL also have to deal with unique challenges associated with transition to adult services. In this review, we: (1) provide an overview of the potential late effects following HSCT for ALL in childhood and adolescence; (2) focus on the unique challenges of transition from paediatric care to adult services; and (3) provide a framework for long-term surveillance and medical care for survivors of paediatric ALL who have undergone HSCT.
Collapse
Affiliation(s)
- Tamara Diesch-Furlanetto
- Division of Pediatric Oncology/Hematology, University Children's Hospital Basel (UKB), University of Basel, Basel, Switzerland
| | - Melissa Gabriel
- Cancer Centre for Children, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Olga Zajac-Spychala
- Department of Pediatric Oncology, Hematology and Transplantology, University of Medical Sciences, Poznań, Poland
| | - Alessandro Cattoni
- Clinica Pediatrica, University degli Studi di Milano-Bicocca, Fondazione Monza e Brianza per il Bambino e la sua Mamma (MBBM), San Gerardo Hospital, Monza, Italy
| | - Bianca A W Hoeben
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Adriana Balduzzi
- Clinica Pediatrica, University degli Studi di Milano-Bicocca, Fondazione Monza e Brianza per il Bambino e la sua Mamma (MBBM), San Gerardo Hospital, Monza, Italy
| |
Collapse
|
30
|
Peters C, Dalle JH, Locatelli F, Poetschger U, Sedlacek P, Buechner J, Shaw PJ, Staciuk R, Ifversen M, Pichler H, Vettenranta K, Svec P, Aleinikova O, Stein J, Güngör T, Toporski J, Truong TH, Diaz-de-Heredia C, Bierings M, Ariffin H, Essa M, Burkhardt B, Schultz K, Meisel R, Lankester A, Ansari M, Schrappe M, von Stackelberg A, Balduzzi A, Corbacioglu S, Bader P. Total Body Irradiation or Chemotherapy Conditioning in Childhood ALL: A Multinational, Randomized, Noninferiority Phase III Study. J Clin Oncol 2020; 39:295-307. [PMID: 33332189 PMCID: PMC8078415 DOI: 10.1200/jco.20.02529] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Total body irradiation (TBI) before allogeneic hematopoietic stem cell transplantation (HSCT) in pediatric patients with acute lymphoblastic leukemia (ALL) is efficacious, but long-term side effects are concerning. We investigated whether preparative combination chemotherapy could replace TBI in such patients.
Collapse
Affiliation(s)
- Christina Peters
- St. Anna Children's Hospital, Children's Cancer Research Institute, University Vienna, Vienna, Austria
| | - Jean-Hugues Dalle
- Hôpital Robert Debré, GH APHP-Nord Université de Paris, Paris, France
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza University of Rome, Rome, Italy
| | | | - Petr Sedlacek
- Department of Pediatric Hematology and Oncology, Motol University Hospital, Prague, Czech Republic
| | - Jochen Buechner
- Department of Pediatric Hematology and Oncology, Oslo University Hospital, Oslo, Norway
| | - Peter J Shaw
- The Children`s Hospital at Westmead, Sydney, Australia
| | | | | | - Herbert Pichler
- St. Anna Children's Hospital, Children's Cancer Research Institute, University Vienna, Vienna, Austria
| | - Kim Vettenranta
- Children's Hospital, University of Helsinki, Helsinki, Finland
| | - Peter Svec
- National Institute of Children's Diseases, Bratislava, Slovakia
| | - Olga Aleinikova
- Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Borovlyani, Belarus
| | - Jerry Stein
- Schneider Children's Medical Center of Israel, Sackler Faculty of Medicine, Tel Aviv University, Petach-Tikva, Israel
| | | | | | - Tony H Truong
- Alberta Children's Hospital Calgary, Calgary, Alberta, Canada
| | | | - Marc Bierings
- Princess Máxima Center for Pediatric Oncology, Bilthoven, the Netherlands
| | | | - Mohammed Essa
- King Abdullah Specialist Children's Hospital, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | | | - Kirk Schultz
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Arjan Lankester
- Willem-Alexander Children's Hospital, Leiden, the Netherlands
| | - Marc Ansari
- Geneva University Hospital, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | - Peter Bader
- Goethe University, University Hospital Frankfurt, Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Frankfurt am Main, Germany
| |
Collapse
|
31
|
Towards new long-term composite 'Quality of Survival' endpoints. Bone Marrow Transplant 2020; 55:1898-1899. [PMID: 32533090 DOI: 10.1038/s41409-020-0883-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/04/2020] [Accepted: 03/19/2020] [Indexed: 11/08/2022]
|
32
|
Summers C, Sheth VS, Bleakley M. Minor Histocompatibility Antigen-Specific T Cells. Front Pediatr 2020; 8:284. [PMID: 32582592 PMCID: PMC7283489 DOI: 10.3389/fped.2020.00284] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/06/2020] [Indexed: 01/05/2023] Open
Abstract
Minor Histocompatibility (H) antigens are major histocompatibility complex (MHC)/Human Leukocyte Antigen (HLA)-bound peptides that differ between allogeneic hematopoietic stem cell transplantation (HCT) recipients and their donors as a result of genetic polymorphisms. Some minor H antigens can be used as therapeutic T cell targets to augment the graft-vs.-leukemia (GVL) effect in order to prevent or manage leukemia relapse after HCT. Graft engineering and post-HCT immunotherapies are being developed to optimize delivery of T cells specific for selected minor H antigens. These strategies have the potential to reduce relapse risk and thereby permit implementation of HCT approaches that are associated with less toxicity and fewer late effects, which is particularly important in the growing and developing pediatric patient. Most minor H antigens are expressed ubiquitously, including on epithelial tissues, and can be recognized by donor T cells following HCT, leading to graft-vs.-host disease (GVHD) as well as GVL. However, those minor H antigens that are expressed predominantly on hematopoietic cells can be targeted for selective GVL. Once full donor hematopoietic chimerism is achieved after HCT, hematopoietic-restricted minor H antigens are present only on residual recipient malignant hematopoietic cells, and these minor H antigens serve as tumor-specific antigens for donor T cells. Minor H antigen-specific T cells that are delivered as part of the donor hematopoietic stem cell graft at the time of HCT contribute to relapse prevention. However, in some cases the minor H antigen-specific T cells delivered with the graft may be quantitatively insufficient or become functionally impaired over time, leading to leukemia relapse. Following HCT, adoptive T cell immunotherapy can be used to treat or prevent relapse by delivering large numbers of donor T cells targeting hematopoietic-restricted minor H antigens. In this review, we discuss minor H antigens as T cell targets for augmenting the GVL effect in engineered HCT grafts and for post-HCT immunotherapy. We will highlight the importance of these developments for pediatric HCT.
Collapse
Affiliation(s)
- Corinne Summers
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Vipul S Sheth
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Marie Bleakley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States
| |
Collapse
|