1
|
Shang Q, Wang Y, Lu A, Jia Y, Zuo Y, Zeng H, Zhang L. Impact of pre-infusion disease burden on outcomes in pediatric relapsed/refractory B-cell lymphoblastic leukemia following anti-CD19 CAR T-cell therapy. Leuk Lymphoma 2024:1-10. [PMID: 39378242 DOI: 10.1080/10428194.2024.2406958] [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: 04/21/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024]
Abstract
Anti-CD19 chimeric antigen receptor (CAR) T-cell therapies have demonstrated high efficacy in pediatric patients with relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). Despite this success, the challenge of post-infusion relapse persists. In our study, we evaluate 116 children with R/R B-ALL who received anti-CD19 CAR T-cell therapy at our center. All patients were included in the response analysis and assessed for survival and toxicity. The CR rate was 98.3%, with 90.5% achieving minimal residual disease negative (MRD)- CR by day 28 (d28). The overall survival (OS) and event-free survival (EFS) were 69.3%±4.5% and 59.0%±4.6%, respectively, with a median follow-up duration of 47.9 months. The patients with pre-infusion MRD ≥ 1% was associated with lower 4-year OS (p = 0.006) and EFS (p = 0.027) comparing to those with MRD < 1%. The incidences of grade ≥ 3 cytokine release syndrome (CRS) and neurotoxicity were21.6 and 5.0%, respectively. Therefore, pre-infusion disease burden is a predictor of long-term outcome following anti-CD19 CAR T-cell therapy for pediatric R/R B-ALL.
Collapse
Affiliation(s)
- Qianwen Shang
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Yu Wang
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Aidong Lu
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Yueping Jia
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Yingxi Zuo
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Huimin Zeng
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Leping Zhang
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| |
Collapse
|
2
|
Choe M, Summers C. The Road More or Less Traveled- Examining the Role of Consolidative Allogeneic Hematopoietic Stem Cell Transplantation After Chimeric Antigen Receptor T Cell Therapy in B-cell ALL. Semin Hematol 2024; 61:314-320. [PMID: 39370355 DOI: 10.1053/j.seminhematol.2024.08.004] [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: 06/04/2024] [Revised: 07/25/2024] [Accepted: 08/19/2024] [Indexed: 10/08/2024]
Abstract
Treatment with CD19-targeted chimeric antigen receptor T cell therapy (CD19-CART) has improved salvage rates in children and adults with relapsed and/or refractory B-cell acute lymphoblastic leukemia (ALL). However, not all patients treated with CD19-CAR T cells achieve long-term remission. The role of allogeneic hematopoietic stem cell transplantation as consolidative therapy remains undefined. We aim to review the current literature published to date regarding prognostic markers indicating durable ALL response to CD19-CART and risk factors for relapse after CD19-CART to identify patient cohorts who may benefit from consolidative hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Michelle Choe
- Clinical Research Division, Fred Hutch Cancer Center, Seattle, Washington, USA; Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Corinne Summers
- Department of Pediatrics, University of Washington, Seattle, Washington, USA; Seattle Children's Therapeutics, Seattle Children's Research Institute, Seattle, Washington, USA.
| |
Collapse
|
3
|
Huang Y, Yu W. Advances in Immune Checkpoint Therapy in Hepatocellular Carcinoma. Br J Hosp Med (Lond) 2024; 85:1-21. [PMID: 39347660 DOI: 10.12968/hmed.2024.0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The incidence and lethality of hepatocellular carcinoma (HCC) are increasing annually, and traditional treatments have been proven to be ineffective for patients with advanced stages of the disease. In recent years, immune checkpoint therapy has rapidly evolved, demonstrating promising results across a wide range of cancers and offering new hope for cancer treatment. However, the efficacy of immune checkpoint therapy in HCC varies greatly among individuals, with only a small proportion of HCC patients responding positively. A major cause of immune resistance and poor efficacy in HCC patients is immune evasion, which is often due to insufficient infiltration of immune cells. Understanding the mechanisms underlying immune evasion is crucial for enhancing the efficacy of immune therapies. In this review, we aim to summarize the mechanisms of immune evasion observed during immune checkpoint therapy and discuss future directions for this therapeutic approach. Our goal is to provide insights that could help overcome immune evasion, thereby improving the efficacy of immune therapies and extending patient survival time.
Collapse
Affiliation(s)
- Yamei Huang
- Department of Pathology and Pathophysiology, Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Weiping Yu
- Department of Pathology and Pathophysiology, Medical School of Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Dreyzin A, Rankin AW, Luciani K, Gavrilova T, Shah NN. Overcoming the challenges of primary resistance and relapse after CAR-T cell therapy. Expert Rev Clin Immunol 2024; 20:745-763. [PMID: 38739466 PMCID: PMC11180598 DOI: 10.1080/1744666x.2024.2349738] [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: 12/17/2023] [Accepted: 04/26/2024] [Indexed: 05/16/2024]
Abstract
INTRODUCTION While CAR T-cell therapy has led to remarkable responses in relapsed B-cell hematologic malignancies, only 50% of patients ultimately have a complete, sustained response. Understanding the mechanisms of resistance and relapse after CAR T-cell therapy is crucial to future development and improving outcomes. AREAS COVERED We review reasons for both primary resistance and relapse after CAR T-cell therapies. Reasons for primary failure include CAR T-cell manufacturing problems, suboptimal fitness of autologous T-cells themselves, and intrinsic features of the underlying cancer and tumor microenvironment. Relapse after initial response to CAR T-cell therapy may be antigen-positive, due to CAR T-cell exhaustion or limited persistence, or antigen-negative, due to antigen-modulation on the target cells. Finally, we discuss ongoing efforts to overcome resistance to CAR T-cell therapy with enhanced CAR constructs, manufacturing methods, alternate cell types, combinatorial strategies, and optimization of both pre-infusion conditioning regimens and post-infusion consolidative strategies. EXPERT OPINION There is a continued need for novel approaches to CAR T-cell therapy for both hematologic and solid malignancies to obtain sustained remissions. Opportunities for improvement include development of new targets, optimally combining existing CAR T-cell therapies, and defining the role for adjunctive immune modulators and stem cell transplant in enhancing long-term survival.
Collapse
Affiliation(s)
- Alexandra Dreyzin
- Pediatric Oncology Branch, Center of Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Division of Pediatric Oncology, Children's National Hospital, Washington DC, USA
| | - Alexander W Rankin
- Pediatric Oncology Branch, Center of Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Katia Luciani
- School of Medicine, University of Limerick, Limerick, Ireland
| | | | - Nirali N Shah
- Pediatric Oncology Branch, Center of Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Shang Q, Xue L, Lu A, Jia Y, Zuo Y, Zeng H, Zhang L. Efficacy and Safety of Children With Relapsed/Refractory B-Cell Acute Lymphoblastic Leukemia After Anti-CD19 CAR T-Cell Therapy Without Bridging Transplantation. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:392-399.e5. [PMID: 38429221 DOI: 10.1016/j.clml.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Anti-CD19 chimeric antigen receptor (CAR) T-cell therapies have demonstrated significant efficacy in achieving complete remission (CR) in pediatric patients with relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). However, a considerable number of patients experience relapse within 1 year after CAR T-cell therapy, leading to an extremely poor prognosis, particularly in patients without bridging transplantation. MATERIALS AND METHODS In our study, we investigated 42 children with R/R B-ALL who underwent anti-CD19 CAR T-cell therapy without bridging transplantation at our center. All patients were included in the response analysis and evaluated for survival and toxicity. RESULTS The cohort that received the CAR T-cell infusion exhibited a 100% CR rate by day 28 (d28). The overall survival (OS) at 4 years was 61.3% ± 8.5%, and the event-free survival (EFS) was 55.9% ± 7.9%, with a median follow-up duration of 50.1 months. Minimal residual disease (MRD) ≥1% was associated with inferior outcomes, resulting in lower 4-year OS (P = .033) and EFS (P = .014) compared to MRD<1%. The incidences of grade ≥3 cytokine release syndrome (CRS) and neurotoxicity were 26.8% and 23.8%, respectively. Furthermore, MRD≥1% was identified as an independent factor associated with increased severity of CRS and occurrence of neurotoxicity. CONCLUSION These findings suggest that reducing the pre-infusion MRD could serve as an effective treatment strategy to enhance the outcomes of CAR T-cell therapy.
Collapse
Affiliation(s)
- Qianwen Shang
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Lian Xue
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Aidong Lu
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Yueping Jia
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - YingXi Zuo
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Huimin Zeng
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China
| | - Leping Zhang
- Department of Pediatrics, Peking University People's Hospital, Peking University, Beijing, China.
| |
Collapse
|
6
|
Hadiloo K, Taremi S, Safa SH, Amidifar S, Esmaeilzadeh A. The new era of immunological treatment, last updated and future consideration of CAR T cell-based drugs. Pharmacol Res 2024; 203:107158. [PMID: 38599467 DOI: 10.1016/j.phrs.2024.107158] [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: 12/11/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/12/2024]
Abstract
Cancer treatment is one of the fundamental challenges in clinical setting, especially in relapsed/refractory malignancies. The novel immunotherapy-based treatments bring new hope in cancer therapy and achieve various treatment successes. One of the distinguished ways of cancer immunotherapy is adoptive cell therapy, which utilizes genetically modified immune cells against cancer cells. Between different methods in ACT, the chimeric antigen receptor T cells have more investigation and introduced a promising way to treat cancer patients. This technology progressed until it introduced six US Food and Drug Administration-approved CAR T cell-based drugs. These drugs act against hematological malignancies appropriately and achieve exciting results, so they have been utilized widely in cell therapy clinics. In this review, we introduce all CAR T cells-approved drugs based on their last data and investigate them from all aspects of pharmacology, side effects, and compressional. Also, the efficacy of drugs, pre- and post-treatment steps, and expected side effects are introduced, and the challenges and new solutions in CAR T cell therapy are in the last speech.
Collapse
Affiliation(s)
- Kaveh Hadiloo
- Department of immunology, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran; School of Medicine, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran
| | - Siavash Taremi
- Department of immunology, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran; School of Medicine, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran
| | - Salar Hozhabri Safa
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran
| | - Sima Amidifar
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran; Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran.
| |
Collapse
|
7
|
Oporto Espuelas M, Burridge S, Kirkwood AA, Bonney D, Watts K, Shenton G, Jalowiec KA, O'Reilly MA, Roddie C, Castleton A, Clesham K, Nicholson E, Alajangi R, Prabhu S, George L, Uttenthal B, Gabelli M, Neill L, Besley C, Chaganti S, Wynn RF, Bartram J, Chiesa R, Lucchini G, Pavasovic V, Rao A, Rao K, Silva J, Samarasinghe S, Vora A, Clark P, Cummins M, Marks DI, Amrolia P, Hough R, Ghorashian S. Intention-to-treat outcomes utilising a stringent event definition in children and young people treated with tisagenlecleucel for r/r ALL through a national access scheme. Blood Cancer J 2024; 14:66. [PMID: 38622139 PMCID: PMC11018620 DOI: 10.1038/s41408-024-01038-2] [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: 12/01/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 04/17/2024] Open
Abstract
CAR T-cell therapy has transformed relapsed/refractory (r/r) B-cell precursor acute lymphoblastic leukaemia (B-ALL) management and outcomes, but following CAR T infusion, interventions are often needed. In a UK multicentre study, we retrospectively evaluated tisagenlecleucel outcomes in all eligible patients, analysing overall survival (OS) and event-free survival (EFS) with standard and stringent definitions, the latter including measurable residual disease (MRD) emergence and further anti-leukaemic therapy. Both intention-to-treat and infused cohorts were considered. We collected data on feasibility of delivery, manufacture, toxicity, cause of therapy failure and followed patients until death from any cause. Of 142 eligible patients, 125 received tisagenlecleucel, 115/125 (92%) achieved complete remission (CR/CRi). Severe cytokine release syndrome and neurotoxicity occurred in 16/123 (13%) and 10/123 (8.1%), procedural mortality was 3/126 (2.4%). The 2-year intent to treat OS and EFS were 65.2% (95%CI 57.2-74.2%) and 46.5% (95%CI 37.6-57.6%), 2-year intent to treat stringent EFS was 35.6% (95%CI 28.1-44.9%). Median OS was not reached. Sixty-two responding patients experienced CAR T failure by the stringent event definition. Post failure, 1-year OS and standard EFS were 61.2% (95%CI 49.3-75.8) and 55.3% (95%CI 43.6-70.2). Investigation of CAR T-cell therapy for B-ALL delivered on a country-wide basis, including following patients beyond therapy failure, provides clinicians with robust outcome measures. Previously, outcomes post CAR T-cell therapy failure were under-reported. Our data show that patients can be successfully salvaged in this context with good short-term survival.
Collapse
Affiliation(s)
- Macarena Oporto Espuelas
- Infection, Immunity and Inflammation, UCL Great Ormond Ormond Street Institute of Child Health, London, UK.
| | - Saskia Burridge
- Department of Haematology, Great Ormond Street Hospital, London, UK
| | - Amy A Kirkwood
- Cancer Research UK & Cancer Trials Centre, UCL, London, UK
| | - Denise Bonney
- Department of Blood and Bone Marrow Transplant, Royal Manchester Children's Hospital, Manchester, UK
| | - Kelly Watts
- Department of Blood and Bone Marrow Transplant, Royal Manchester Children's Hospital, Manchester, UK
| | - Geoff Shenton
- Great North Children's Hospital, Newcastle upon Tyne, UK
| | - Katarzyna A Jalowiec
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Maeve A O'Reilly
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Claire Roddie
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Anna Castleton
- Department of Haematology, The Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Katherine Clesham
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Emma Nicholson
- Department of Haematology/Bone Marrow Transplantation, The Royal Marsden NHS Foundation Trust, London, UK
- Institute of Cancer Research, London, UK
| | - Rajesh Alajangi
- Department of Haematology/Bone Marrow Transplant, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Shilpa Prabhu
- Department of Haematology/Bone Marrow Transplant, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Lindsay George
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
| | - Ben Uttenthal
- Cambridge University Hospital NHS Foundation Trust, Cambridge, UK
| | - Maria Gabelli
- Department of Bone Marrow Transplant, Great Ormond Street Hospital, London, UK
- Pediatric Onco-hematology and Hematopoietic Stem Cell Transplantation, Woman and Child Health Department, University of Padova, Padua, Italy
| | - Lorna Neill
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Caroline Besley
- Department of Haematology/Bone Marrow Transplant, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Sridhar Chaganti
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
| | - Robert F Wynn
- Department of Blood and Bone Marrow Transplant, Royal Manchester Children's Hospital, Manchester, UK
| | - Jack Bartram
- Department of Haematology, Great Ormond Street Hospital, London, UK
| | - Robert Chiesa
- Department of Bone Marrow Transplant, Great Ormond Street Hospital, London, UK
| | - Giovanna Lucchini
- Department of Bone Marrow Transplant, Great Ormond Street Hospital, London, UK
| | - Vesna Pavasovic
- Department of Haematology, Great Ormond Street Hospital, London, UK
| | - Anupama Rao
- Department of Haematology, Great Ormond Street Hospital, London, UK
| | - Kanchan Rao
- Department of Bone Marrow Transplant, Great Ormond Street Hospital, London, UK
| | - Juliana Silva
- Department of Bone Marrow Transplant, Great Ormond Street Hospital, London, UK
| | | | - Ajay Vora
- Department of Haematology, Great Ormond Street Hospital, London, UK
| | | | | | - David I Marks
- Department of Haematology, University Hospitals Bristol, Bristol, UK
| | - Persis Amrolia
- Infection, Immunity and Inflammation, UCL Great Ormond Ormond Street Institute of Child Health, London, UK
- Department of Bone Marrow Transplant, Great Ormond Street Hospital, London, UK
| | - Rachael Hough
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Sara Ghorashian
- Department of Haematology, Great Ormond Street Hospital, London, UK
- Developmental Biology and Cancer, UCL Great Ormond Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
8
|
Gye A, Lourenco RDA, Goodall S. Discrete Event Simulation to Incorporate Infusion Wait-Time When Assessing Cost-Effectiveness of a Chimeric-Antigen Receptor T Cell Therapy. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2024; 27:415-424. [PMID: 38301961 DOI: 10.1016/j.jval.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 12/21/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024]
Abstract
OBJECTIVES The main objective was to use discrete event simulation to model the impact of wait-time, defined as the time between leukapheresis and chimeric antigen receptor (CAR-T) infusion, when assessing the cost-effectiveness of tisagenlecleucel in young patients with relapsed/refractory acute lymphoblastic leukemia. METHODS The movement of patients through the model was determined by parametric time-to-event distributions, with the competing risk of an event determining the costs and quality-adjusted life-years (QALYs) assigned. Cost-effectiveness was expressed using the incremental cost-effectiveness ratio (ICER) for tisagenlecleucel compared with chemotherapy over the lifetime. RESULTS The base case generated a total of 5.79 QALYs and $622 872 for tisagenlecleucel and 1.19 QALYs and $181 219 for blinatumomab, resulting in an ICER of $96 074 per QALY. An increase in mean CAR-T wait-time to 6.20 months reduced the benefit and costs of tisagenlecleucel to 2.78 QALYs and $294 478 because of fewer patients proceeding to infusion, reducing the ICER to $71 112 per QALY. Alternatively, when the cost of tisagenlecleucel was assigned pre-infusion in sensitivity analysis, the ICER increased with increasing wait-time. CONCLUSIONS Under a payment arrangement where CAR-T cost is incurred post-infusion, the loss of benefit to patients is not reflected in the ICER. This may be misguiding to decision makers, where cost-effectiveness ratios are used to guide resource allocation. discrete event simulation is an important tool for economic modeling of CAR-T as it is amenable to capturing the impact of wait-time, facilitating better understanding of factors affecting service delivery and consequently informed decision making to deliver faster access to CAR-T for patients.
Collapse
Affiliation(s)
- Amy Gye
- Centre for Health Economics Research and Evaluation, University of Technology Sydney, Sydney, NSW, Australia.
| | - Richard De Abreu Lourenco
- Centre for Health Economics Research and Evaluation, University of Technology Sydney, Sydney, NSW, Australia
| | - Stephen Goodall
- Centre for Health Economics Research and Evaluation, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
9
|
Molinos-Quintana Á, Alonso-Saladrigues A, Herrero B, Caballero-Velázquez T, Galán-Gómez V, Panesso M, Torrebadell M, Delgado-Serrano J, Pérez de Soto C, Faura A, González-Martínez B, Castillo-Robleda A, Diaz-de-Heredia C, Pérez-Martínez A, Pérez-Hurtado JM, Rives S, Pérez-Simón JA. Impact of disease burden and late loss of B cell aplasia on the risk of relapse after CD19 chimeric antigen receptor T Cell (Tisagenlecleucel) infusion in pediatric and young adult patients with relapse/refractory acute lymphoblastic leukemia: role of B-cell monitoring. Front Immunol 2024; 14:1280580. [PMID: 38292483 PMCID: PMC10825008 DOI: 10.3389/fimmu.2023.1280580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction Loss of B-cell aplasia (BCA) is a well-known marker of functional loss of CD19 CAR-T. Most relapses and loss of BCA occur in the first months after CD19 CAR-T infusion. In addition, high tumor burden (HTB) has shown to have a strong impact on relapse, especially in CD19-negative. However, little is known about the impact of late loss of BCA or the relationship between BCA and pre-infusion tumor burden in patients infused with tisagenlecleucel for relapsed/refractory B-cell acute lymphoblastic leukemia. Therefore, the optimal management of patients with loss of BCA is yet to be defined. Methods We conducted a Spanish, multicentre, retrospective study in patients infused with tisagenlecleucel after marketing authorization. A total of 73 consecutively treated patients were evaluated. Results Prior to infusion, 39 patients had HTB (≥ 5% bone marrow blasts) whereas 34 had a low tumor burden (LTB) (<5% blasts). Complete remission was achieved in 90.4% of patients, of whom 59% relapsed. HTB was associated with inferior outcomes, with a 12-month EFS of 19.3% compared to 67.2% in patients with LTB (p<0.001) with a median follow-up of 13.5 months (95% CI 12.4 - 16.2). In the HTB subgroup relapses were mainly CD19-negative (72%) whereas in the LTB subgroup they were mainly CD19-positive (71%) (p=0.017). In the LTB group, all CD19-positive relapses were preceded by loss of BCA whereas only 57% (4/7) of HTB patients experienced CD19-positive relapse. We found a positive correlation between loss of BCA and CD19-positive relapse (R-squared: 74) which persisted beyond six months post-infusion. We also explored B-cell recovery over time using two different definitions of loss of BCA and found a few discrepancies. Interestingly, transient immature B-cell recovery followed by BCA was observed in two pediatric patients. In conclusion, HTB has an unfavorable impact on EFS and allo-SCT might be considered in all patients with HTB, regardless of BCA. In patients with LTB, loss of BCA preceded all CD19-positive relapses. CD19-positive relapse was also frequent in patients who lost BCA beyond six months post-infusion. Therefore, these patients are still at significant risk for relapse and close MRD monitoring and/or therapeutic interventions should be considered.
Collapse
Affiliation(s)
- Águeda Molinos-Quintana
- Pediatric Unit, Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS)/CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Anna Alonso-Saladrigues
- CAR T-cell Unit. Leukemia and Lymphoma Department. Pediatric Cancer Center Barcelona (PCCB). Hospital Sant Joan de Déu de Barcelona, Barcelona, Spain
| | - Blanca Herrero
- Pediatric Hemato-Oncology Department, Peditric University Hospital del Niño Jesús, Madrid, Spain
| | - Teresa Caballero-Velázquez
- Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS)/CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Víctor Galán-Gómez
- Pediatric Hemato-Oncology Department, University Hospital La Paz, Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - Melissa Panesso
- Division of Pediatric Hematology and Oncology, Hospital Universitari Vall d’Hebron, Vall d’Hebron Research Institute (VHIR), Barcelona, Spain
| | - Montserrat Torrebadell
- CAR T-cell Unit. Leukemia and Lymphoma Department. Pediatric Cancer Center Barcelona (PCCB). Hospital Sant Joan de Déu de Barcelona, Barcelona, Spain
| | - Javier Delgado-Serrano
- Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS)/CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Concepción Pérez de Soto
- Pediatric Unit, Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS)/CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Anna Faura
- CAR T-cell Unit. Leukemia and Lymphoma Department. Pediatric Cancer Center Barcelona (PCCB). Hospital Sant Joan de Déu de Barcelona, Barcelona, Spain
| | - Berta González-Martínez
- Pediatric Hemato-Oncology Department, University Hospital La Paz, Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - Ana Castillo-Robleda
- Pediatric Hemato-Oncology Department, Peditric University Hospital del Niño Jesús, Madrid, Spain
| | - Cristina Diaz-de-Heredia
- Division of Pediatric Hematology and Oncology, Hospital Universitari Vall d’Hebron, Vall d’Hebron Research Institute (VHIR), Barcelona, Spain
| | - Antonio Pérez-Martínez
- Pediatric Hemato-Oncology Department, University Hospital La Paz, Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - José María Pérez-Hurtado
- Pediatric Unit, Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS)/CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Susana Rives
- Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Sant Joan de Déu, Leukemia and Pediatric Hematology Disorders, Developmental Tumors Biology Group, Barcelona, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red De Enfermedades Raras (CIBERER), Madrid, Spain
| | - José Antonio Pérez-Simón
- Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS)/CSIC, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
10
|
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
|
11
|
Talleur AC, Naik S, Gottschalk S. Preventing relapse after CD19 CAR T-cell therapy for pediatric ALL: the role of transplant and enhanced CAR T cells. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:91-96. [PMID: 38066941 PMCID: PMC10727085 DOI: 10.1182/hematology.2023000424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
CD19-specific chimeric antigen receptor (CAR) T-cell therapy has become an integral part of our treatment armamentarium for pediatric patients with relapsed or refractory B-cell acute lymphoblastic leukemia (B-ALL). However, despite initial remission rates of greater than 80%, durable remission occurs in only 40% to 50% of patients. In this review we summarize our current knowledge of the role of consolidative hematopoietic cell transplantation in the management of pediatric patients who achieved a minimal residual disease-negative complete response post CD19 CAR T-cell therapy. In addition, we review approaches to enhance effector function CD19 CAR T cells, focusing on how to improve persistence and prevent the emergence of CD19- B-ALL blasts.
Collapse
Affiliation(s)
- Aimee C Talleur
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN
| | - Swati Naik
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
12
|
McClory SE, Maude SL. The Current State of Chimeric Antigen Receptor T Cell Therapy for B Lymphoblastic Leukemia. Hematol Oncol Clin North Am 2023; 37:1041-1052. [PMID: 37500380 DOI: 10.1016/j.hoc.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Over the past decade, CAR T cell therapy has transformed the treatment of relapsed or refractory B-ALL in children and adults. CD19-directed CAR T cells can induce complete remissions in a large majority of patients with B-ALL, and up to half of these patients will go on to maintain durable remissions. However, significant challenges remain for patients who relapse or do not respond. This review will discuss the history of CAR T cell therapy for B-ALL, the treatment considerations for CAR T cell recipients, and current clinical trials and future directions for CAR T cell therapy in B-ALL.
Collapse
Affiliation(s)
- Susan E McClory
- Division of Oncology, Cancer Immunotherapy Program, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon L Maude
- Division of Oncology, Cancer Immunotherapy Program, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Epperly R, Shulkin BL, Bag AK, Cheng C, Inaba H, Lucas JT, Naik S, Triplett BM, Gottschalk S, Talleur AC. CD19 CAR T-cell therapy demonstrates activity against extramedullary disease in pediatric patients with B-ALL. Blood Adv 2023; 7:6320-6324. [PMID: 37595052 PMCID: PMC10589782 DOI: 10.1182/bloodadvances.2023010461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/18/2023] [Accepted: 08/10/2023] [Indexed: 08/20/2023] Open
Affiliation(s)
- Rebecca Epperly
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Barry L. Shulkin
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN
| | - Asim K. Bag
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - John T. Lucas
- Department of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Swati Naik
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Brandon M. Triplett
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Aimee C. Talleur
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
14
|
Myers RM, Jacoby E, Pulsipher MA, Pasquini MC, Grupp SA, Shah NN, Laetsch TW, Curran KJ, Schultz LM. INSPIRED Symposium Part 1: Clinical Variables Associated with Improved Outcomes for Children and Young Adults treated with Chimeric Antigen Receptor T cells for B cell Acute Lymphoblastic Leukemia. Transplant Cell Ther 2023; 29:598-607. [PMID: 37481241 PMCID: PMC11031134 DOI: 10.1016/j.jtct.2023.07.016] [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: 07/02/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy (CAR-T) targeting the CD19 antigen on B cell acute lymphoblastic leukemia (B-ALL) has transitioned from a highly investigational therapy with limited access to a commercial therapy with established toxicities, response and survival rates, and access in numerous countries. With more than a decade of clinical study and 5 years of commercial access, data showing associations with success and failure have emerged. To address functional limitations of CAR-T and overcome constrained sample sizes when studying single-trial or single-center data, collaborative groups, including the Pediatric Real World CAR Consortium, the CAR-Multicenter Analysis, the Center for International Blood and Marrow Transplant Research, and the International BFM Study Group, among others, have been retrospectively interrogating the amassed clinical experience. The high patient numbers and varied clinical experiences compiled by these groups have defined clinical variables impacting CAR-T outcomes. Here we review published CAR-T trials and consortium/collaborative outcomes to establish variables associated with optimal response to CAR-T in children and young adults with B-ALL. We focus on findings with clinical relevance that have emerged, including data implicating pretreatment disease burden, presence of extramedullary disease, nonresponse to prior CD19 antigen targeting (blinatumomab therapy), CAR T cell dose, and fludarabine pharmacokinetics as factors impacting post-CAR-T survival. Additionally, we address the role of collaborative efforts going forward in guiding clinical practice evolution and further optimizing post-CAR-T outcomes.
Collapse
Affiliation(s)
- Regina M Myers
- Division of Oncology, Center for Childhood Cancer Research and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Elad Jacoby
- Division of Pediatric Hematology, Oncology and BMT, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Michael A Pulsipher
- Intermountain Primary Children's Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, Utah
| | - Marcelo C Pasquini
- Medical College of Wisconsin/Center for International Blood and Marrow Transplant Research, Milwaukee, Wisconsin
| | - Stephan A Grupp
- Division of Oncology, Center for Childhood Cancer Research and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Theodore W Laetsch
- Division of Oncology, Center for Childhood Cancer Research and Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Kevin J Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Liora M Schultz
- Department of Pediatrics, Division of Hematology and Oncology, Stanford University School of Medicine, Palo Alto, California, USA.
| |
Collapse
|
15
|
McNerney KO, Si Lim SJ, Ishikawa K, Dreyzin A, Vatsayan A, Chen JJ, Baggott C, Prabhu S, Pacenta HL, Philips C, Rossoff J, Stefanski HE, Talano JA, Moskop A, Verneris M, Myers D, Karras NA, Brown P, Bonifant CL, Qayed M, Hermiston M, Satwani P, Krupski C, Keating AK, Baumeister SHC, Fabrizio VA, Chinnabhandar V, Egeler E, Mavroukakis S, Curran KJ, Mackall CL, Laetsch TW, Schultz LM. HLH-like toxicities predict poor survival after the use of tisagenlecleucel in children and young adults with B-ALL. Blood Adv 2023; 7:2758-2771. [PMID: 36857419 PMCID: PMC10275701 DOI: 10.1182/bloodadvances.2022008893] [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: 09/07/2022] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 03/03/2023] Open
Abstract
Chimeric antigen receptor-associated hemophagocytic lymphohistiocytosis (HLH)-like toxicities (LTs) involving hyperferritinemia, multiorgan dysfunction, coagulopathy, and/or hemophagocytosis are described as occurring in a subset of patients with cytokine release syndrome (CRS). Case series report poor outcomes for those with B-cell acute lymphoblastic leukemia (B-ALL) who develop HLH-LTs, although larger outcomes analyses of children and young adults (CAYAs) with B-ALL who develop these toxicities after the administration of commercially available tisagenlecleucel are not described. Using a multi-institutional database of 185 CAYAs with B-ALL, we conducted a retrospective cohort study including groups that developed HLH-LTs, high-grade (HG) CRS without HLH-LTs, or no to low-grade (NLG) CRS without HLH-LTs. Primary objectives included characterizing the incidence, outcomes, and preinfusion factors associated with HLH-LTs. Among 185 CAYAs infused with tisagenlecleucel, 26 (14.1%) met the criteria for HLH-LTs. One-year overall survival and relapse-free survival were 25.7% and 4.7%, respectively, in those with HLH-LTs compared with 80.1% and 57.6%, respectively, in those without. In multivariable analysis for death, meeting criteria for HLH-LTs carried a hazard ratio of 4.61 (95% confidence interval, 2.41-8.83), controlling for disease burden, age, and sex. Patients who developed HLH-LTs had higher pretisagenlecleucel disease burden, ferritin, and C-reactive protein levels and lower platelet and absolute neutrophil counts than patients with HG- or NLG-CRS without HLH-LTs. Overall, CAYAs with B-ALL who developed HLH-LTs after tisagenlecleucel experienced high rates of relapse and nonrelapse mortality, indicating the urgent need for further investigations into prevention and optimal management of patients who develop HLH-LTs after tisagenlecleucel.
Collapse
Affiliation(s)
- Kevin O. McNerney
- Cancer and Blood Disorders Institute, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Stephanie J. Si Lim
- Division of Oncology, Department of Pediatrics, John A. Burns School of Medicine, University of Hawai’i at Manoa, Honolulu, HI
| | - Kyle Ishikawa
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawai’i at Manoa, Honolulu, HI
| | - Alexandra Dreyzin
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC
| | - Anant Vatsayan
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC
| | - John J. Chen
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawai’i at Manoa, Honolulu, HI
| | - Christina Baggott
- Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA
| | - Snehit Prabhu
- Center for Cancer Cell Therapy, Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, CA
| | - Holly L. Pacenta
- Department of Pediatrics, University of Texas Southwestern Medical Center/Children’s Health, Dallas, TX
- Division of Hematology and Oncology, Cook Children’s Medical Center, Fort Worth, TX
| | - Christine Philips
- Division of Pediatrics, University of Cincinnati, Cincinnati, OH
- Cincinnati Children’s Hospital Medical Center, Cancer and Blood Disease Institute, Cincinnati, OH
| | - Jenna Rossoff
- Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL
| | | | - Julie-An Talano
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Department of Pediatrics, Medical College of Wisconsin and Children’s Wisconsin, Milwaukee, WI
| | - Amy Moskop
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Department of Pediatrics, Medical College of Wisconsin and Children’s Wisconsin, Milwaukee, WI
| | - Michael Verneris
- University of Colorado School of Medicine, Children’s Hospital of Colorado, Aurora, CO
| | - Doug Myers
- Department of Hematology, Oncology and Blood and Marrow Transplantation, Children’s Mercy Hospital, University of Missouri Kansas City, Kansas City, MO
| | - Nicole A. Karras
- Department of Pediatrics, City of Hope National Medical Center, Duarte, CA
| | - Patrick Brown
- Department of Oncology, Sidney Kimmel Cancer Center, John Hopkins University School of Medicine, Baltimore, MD
| | - Challice L. Bonifant
- Department of Oncology, Sidney Kimmel Cancer Center, John Hopkins University School of Medicine, Baltimore, MD
| | - Muna Qayed
- Division of Pediatric Hematology/Oncology and Bone Marrow Transplantation, Aflac Cancer and Blood Disorders Center, Emory University and Children’s Healthcare of Atlanta, Atlanta, GA
| | - Michelle Hermiston
- University of California San Francisco Benioff Children's Hospital, San Francisco, CA
| | - Prakash Satwani
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Medical Center, New York, NY
| | - Christa Krupski
- Division of Pediatrics, University of Cincinnati, Cincinnati, OH
- Cincinnati Children’s Hospital Medical Center, Cancer and Blood Disease Institute, Cincinnati, OH
| | - Amy K. Keating
- University of Colorado School of Medicine, Children’s Hospital of Colorado, Aurora, CO
| | - Susanne H. C. Baumeister
- Division of Pediatric Hematology-Oncology, Boston Children’s Hospital, Boston, MA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Vanessa A. Fabrizio
- University of Colorado School of Medicine, Children’s Hospital of Colorado, Aurora, CO
| | - Vasant Chinnabhandar
- Division of Pediatric Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Emily Egeler
- Center for Cancer Cell Therapy, Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, CA
| | - Sharon Mavroukakis
- Center for Cancer Cell Therapy, Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, CA
| | - Kevin J. Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, Cornell University, New York, NY
| | - Crystal L. Mackall
- Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, CA
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA
| | - Theodore W. Laetsch
- Department of Pediatrics and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Liora M. Schultz
- Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA
| |
Collapse
|
16
|
Talleur AC, Pui CH, Karol SE. What is Next in Pediatric B-cell Precursor Acute Lymphoblastic Leukemia. LYMPHATICS 2023; 1:34-44. [PMID: 38269058 PMCID: PMC10804398 DOI: 10.3390/lymphatics1010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Cure rates now exceed 90% in many contemporary trials for children with B-cell acute lymphoblastic leukemia (ALL). However, treatment remains suboptimal and therapy is toxic for all patients. New treatment options potentially offer the chance to reduce both treatment resistance and toxicity. Here, we review recent advances in ALL diagnostics, chemotherapy, and immunotherapy. In addition to describing recently published results, we also attempt to project the impact of these new developments into the future to imagine what B-ALL therapy may look like in the next few years.
Collapse
Affiliation(s)
- Aimee C Talleur
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Seth E Karol
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
17
|
Elsallab M, Ellithi M, Hempel S, Abdel-Azim H, Abou-El-Enein M. Long-term response to autologous anti-CD19 chimeric antigen receptor T cells in relapsed or refractory B cell acute lymphoblastic leukemia: a systematic review and meta-analysis. Cancer Gene Ther 2023; 30:845-854. [PMID: 36750666 PMCID: PMC10281866 DOI: 10.1038/s41417-023-00593-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/14/2022] [Accepted: 01/19/2023] [Indexed: 02/09/2023]
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy is an effective treatment approach for patients with relapsed or refractory acute lymphoblastic leukemia (R/R B-ALL). However, identifying the factors that influence long-term response to this therapy is necessary to optimize patient selection and treatment allocation. We conducted a literature review and meta-analysis to investigate the use of autologous anti-CD19 CAR T cell therapy in both pediatric and adult patients with R/R B-ALL, using several databases including MEDLINE, Cochrane Central, ScienceDirect, Web of Science, Journals@Ovid, Embase, and clinicaltrial.gov. A total of 38 reports were analyzed, which enrolled 2134 patients. Time-to-event endpoints were estimated using reconstructed patient survival data. The study explored key modulators of response, including costimulatory domains, disease status, age, and lymphodepletion. The median overall survival and event-free survival were 36.2 months [95% CI 28.9, NR] and 13.3 months [95% CI 12.2, 17], respectively. The overall response rate was 76% [95% CI 71, 81]. The use of 4-1BB costimulatory domain in the CAR construct, administration of low-dose cyclophosphamide lymphodepletion, and pretreatment morphologic remission were associated with better overall survival, with hazard ratios of 0.72, 0.56, and 0.66, respectively. Morphologic remission and 4-1BB domain were associated with better event-free survival, with hazard ratios of 0.66 and 0.72, respectively. These findings suggest that CAR T cell therapy may offer long-term benefits to patients with R/R B-ALL. However, further research is needed to optimize patient selection and better understand the impact of various factors on the outcome of CAR T cell therapy.
Collapse
Affiliation(s)
- Magdi Elsallab
- Harvard-MIT Center for Regulatory Science, Harvard Medical School, Boston, MA, USA.
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA.
- USC/CHLA Cell Therapy Program, University of Southern California, and Children's Hospital Los Angeles, Los Angeles, CA, USA.
| | - Moataz Ellithi
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Susanne Hempel
- Southern California Evidence Review Center, University of Southern California, Los Angeles, CA, USA
| | - Hisham Abdel-Azim
- Loma Linda University School of Medicine, Cancer Center, Children Hospital and Medical Center, Loma Linda, CA, USA
| | - Mohamed Abou-El-Enein
- USC/CHLA Cell Therapy Program, University of Southern California, and Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Myers RM, Shah NN, Pulsipher MA. How I use risk factors for success or failure of CD19 CAR T cells to guide management of children and AYA with B-cell ALL. Blood 2023; 141:1251-1264. [PMID: 36416729 PMCID: PMC10082355 DOI: 10.1182/blood.2022016937] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/17/2022] [Accepted: 11/20/2022] [Indexed: 11/24/2022] Open
Abstract
By overcoming chemotherapeutic resistance, chimeric antigen receptor (CAR) T cells facilitate deep, complete remissions and offer the potential for long-term cure in a substantial fraction of patients with chemotherapy refractory disease. However, that success is tempered with 10% to 30% of patients not achieving remission and over half of patients treated eventually experiencing relapse. With over a decade of experience using CAR T cells in children, adolescents, and young adults (AYA) to treat relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL) and 5 years since the first US Food and Drug Administration approval, data defining the nuances of patient-specific risk factors are emerging. With the commercial availability of 2 unique CD19 CAR T-cell constructs for B-ALL, in this article, we review the current literature, outline our approach to patients, and discuss how individual factors inform strategies to optimize outcomes in children and AYA receiving CD19 CAR T cells. We include data from both prospective and recent large retrospective studies that offer insight into understanding when the risks of CAR T-cell therapy failure are high and offer perspectives suggesting when consolidative hematopoietic cell transplantation or experimental CAR T-cell and/or alternative immunotherapy should be considered. We also propose areas where prospective trials addressing the optimal use of CAR T-cell therapy are needed.
Collapse
Affiliation(s)
- Regina M. Myers
- Division of Oncology, Cell Therapy and Transplant Section, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Nirali N. Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Michael A. Pulsipher
- Division of Hematology and Oncology, Intermountain Primary Children’s Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, UT
| |
Collapse
|
19
|
McNerney KO, Richards RM, Aguayo-Hiraldo P, Calkoen FG, Talano JA, Moskop A, Balduzzi A, Krajewski J, Dave H, Vatsayan A, Callahan C, Liu H, Li Y, Davis KL, Maude SL. SARS-CoV-2 infections in pediatric and young adult recipients of chimeric antigen receptor T-cell therapy: an international registry report. J Immunother Cancer 2023; 11:jitc-2022-005957. [PMID: 36707090 PMCID: PMC9884906 DOI: 10.1136/jitc-2022-005957] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2023] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Immunocompromised patients are at increased risk of SARS-CoV-2 infections. Patients undergoing chimeric antigen receptor (CAR) T-cell therapy for relapsed/refractory B-cell malignancies are uniquely immunosuppressed due to CAR T-mediated B-cell aplasia (BCA). While SARS-CoV-2 mortality rates of 33%-40% are reported in adult CAR T-cell recipients, outcomes in pediatric and young adult CAR T-cell recipients are limited. METHODS We created an international retrospective registry of CAR T recipients aged 0-30 years infected with SARS-CoV-2 within 2 months prior to or any time after CAR T infusion. SARS-CoV-2-associated illness was graded as asymptomatic, mild, moderate, or severe COVID-19, or multisystem inflammatory syndrome in children (MIS-C). To assess for risk factors associated with significant SARS-CoV-2 infections (infections requiring hospital admission for respiratory distress or supplemental oxygen), univariate and multivariable regression analyses were performed. RESULTS Nine centers contributed 78 infections in 75 patients. Of 70 SARS-CoV-2 infections occurring after CAR T infusion, 13 (18.6%) were classified as asymptomatic, 37 (52.9%) mild, 11 (15.7%) moderate, and 6 (8.6%) severe COVID-19. Three (4.3%) were classified as MIS-C. BCA was not significantly associated with infection severity. Prior to the emergence of the Omicron variant, of 47 infections, 19 (40.4%) resulted in hospital admission and 7 (14.9%) required intensive care, while after the emergence of the Omicron variant, of 23 infections, only 1 (4.3%) required admission and the remaining 22 (95.7%) had asymptomatic or mild COVID-19. Death occurred in 3 of 70 (4.3%); each death involved coinfection or life-threatening condition. In a multivariable model, factors associated with significant SARS-CoV-2 infection included having two or more comorbidities (OR 7.73, CI 1.05 to 74.8, p=0.048) and age ≥18 years (OR 9.51, CI 1.90 to 82.2, p=0.014). In the eight patients infected with SARS-CoV-2 before CAR T, half of these patients had their CAR T infusion delayed by 15-30 days. CONCLUSIONS In a large international cohort of pediatric and young adult CAR-T recipients, SARS-CoV-2 infections resulted in frequent hospital and intensive care unit admissions and were associated with mortality in 4.3%. Patients with two or more comorbidities or aged ≥18 years were more likely to experience significant illness. Suspected Omicron infections were associated with milder disease.
Collapse
Affiliation(s)
- Kevin Owen McNerney
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA,Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rebecca M Richards
- Hematology, Oncology, and Bone Marrow Transplant, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Paibel Aguayo-Hiraldo
- Cancer and Blood Disease Institute, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Friso G Calkoen
- Division of Pediatric Oncology, Princess Maxima Center, Utrecht, The Netherlands
| | - Julie-An Talano
- Division of Hematology/Oncology/Blood and Marrow TransplantationDepartment of Pediatrics, Medical College of Wisconsin and Children’s Wisconsin, Milwaukee, Wisconsin, USA
| | - Amy Moskop
- Division of Hematology/Oncology/Blood and Marrow TransplantationDepartment of Pediatrics, Medical College of Wisconsin and Children’s Wisconsin, Milwaukee, Wisconsin, USA
| | - Adriana Balduzzi
- Clinica Pediatrica Università degli Studi di Milano Bicocca, Fondazione IRCCS San Gerardo dai Tintori, Milan, Italy
| | - Jennifer Krajewski
- Pediatric Blood and Marrow Transplantation, Hackensack Meridian School of Medicine, Hackensack, New Jersey, USA
| | - Hema Dave
- Cancer Immunology and Microbial Oncology Research Program, Children's National Hospital, Washington, District of Columbia, USA
| | - Anant Vatsayan
- Center for Cancer and Blood Disorders, Children's National Hospital, Washington, District of Columbia, USA
| | - Colleen Callahan
- Division of Oncology and Cancer Immunotherapy Program, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hongyan Liu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yimei Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kara Lynn Davis
- Pediatrics, Stanford University School of Medicine, Stanford, California, USA,Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, California, USA
| | - Shannon L Maude
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,Division of Oncology, Center for Childhood Cancer Research and Cancer Immunotherapy Program, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
20
|
Kyriakidis I, Mantadakis E, Stiakaki E, Groll AH, Tragiannidis A. Infectious Complications of Targeted Therapies in Children with Leukemias and Lymphomas. Cancers (Basel) 2022; 14:cancers14205022. [PMID: 36291806 PMCID: PMC9599435 DOI: 10.3390/cancers14205022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/09/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Targeted therapies in children with hematological malignancies moderate the effects of cytotoxic therapy, thus improving survival rates. They have emerged over the last decade and are used in combination with or after the failure of conventional chemotherapy and as bridging therapy prior to hematopoietic stem cell transplantation (HSCT). Nowadays, there is a growing interest in their efficacy and safety in pediatric patients with refractory or relapsed disease. The compromised immune system, even prior to therapy, requires prompt monitoring and treatment. In children with hematological malignancies, targeted therapies are associated with a comparable incidence of infectious complications to adults. The exact impact of these agents that have different mechanisms of action and are used after conventional chemotherapy or HSCT is difficult to ascertain. Clinicians should be cautious of severe infections after the use of targeted therapies, especially when used in combination with chemotherapy. Abstract The aim of this review is to highlight mechanisms of immunosuppression for each agent, along with pooled analyses of infectious complications from the available medical literature. Rituximab confers no increase in grade ≥3 infectious risks, except in the case of patients with advanced-stage non-Hodgkin lymphoma. Gemtuzumab ozogamicin links with high rates of grade ≥3 infections which, however, are comparable with historical cohorts. Pembrolizumab exhibits a favorable safety profile in terms of severe infections. Despite high rates of hypogammaglobulinemia (HGG) with blinatumomab, low-grade ≥3 infection rates were observed, especially in the post-reinduction therapy of relapsed B-acute lymphoblastic leukemia. Imatinib and nilotinib are generally devoid of severe infectious complications, but dasatinib may slightly increase the risk of opportunistic infections. Data on crizotinib and pan-Trk inhibitors entrectinib and larotrectinib are limited. CAR T-cell therapy with tisagenlecleucel is associated with grade ≥3 infections in children and is linked with HGG and the emergence of immune-related adverse events. Off-label therapies inotuzumab ozogamicin, brentuximab vedotin, and venetoclax demonstrate low rates of treatment-related grade ≥3 infections, while the addition of bortezomib to standard chemotherapy in T-cell malignancies seems to decrease the infection risk during induction. Prophylaxis, immune reconstitution, and vaccinations for each targeted agent are discussed, along with comparisons to adult studies.
Collapse
Affiliation(s)
- Ioannis Kyriakidis
- Department of Pediatric Hematology-Oncology & Autologous Hematopoietic Stem Cell Transplantation Unit, University Hospital of Heraklion & Laboratory of Blood Diseases and Childhood Cancer Biology, School of Medicine, University of Crete, 70013 Heraklion, Greece
| | - Elpis Mantadakis
- Department of Paediatrics, Paediatric Hematology/Oncology Unit, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Eftichia Stiakaki
- Department of Pediatric Hematology-Oncology & Autologous Hematopoietic Stem Cell Transplantation Unit, University Hospital of Heraklion & Laboratory of Blood Diseases and Childhood Cancer Biology, School of Medicine, University of Crete, 70013 Heraklion, Greece
| | - Andreas H. Groll
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Pediatric Hematology and Oncology, University Children’s Hospital Münster, D-48149 Münster, Germany
| | - Athanasios Tragiannidis
- Pediatric and Adolescent Hematology-Oncology Unit, 2nd Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, 54636 Thessaloniki, Greece
- Correspondence: ; Fax: +30-2310-994803
| |
Collapse
|
21
|
Talleur AC, Myers R, Annesley C, Shalabi H. Chimeric Antigen Receptor T-cell Therapy: Current Status and Clinical Outcomes in Pediatric Hematologic Malignancies. Hematol Oncol Clin North Am 2022; 36:701-727. [PMID: 35780062 DOI: 10.1016/j.hoc.2022.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Chimeric antigen receptor T-cell (CART) therapy has transformed the treatment paradigm for pediatric patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL), with complete remission rates in key pivotal CD19-CART trials ranging from 65% to 90%. Alongside this new therapy, new toxicity profiles and treatment limitations have emerged, necessitating toxicity consensus grading systems, cooperative group trials, and novel management approaches. This review highlights the results of key clinical trials of CART for pediatric hematologic malignancies, discusses the most common toxicities seen to date, and elucidates challenges, opportunities, and areas of active research to optimize this therapy.
Collapse
Affiliation(s)
- Aimee C Talleur
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS1130, Memphis, TN 38105, USA
| | - Regina Myers
- Division of Oncology, Children's Hospital of Philadelphia, Office 2568A, 3500 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Colleen Annesley
- Seattle Children's Research Institute, 4800 Sand Point Way NE, M/S MB8.501, Seattle, WA 98145-5005, USA
| | - Haneen Shalabi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room 1W-5750, 9000 Rockville Pike, Bethesda, MD 20892-1104, USA.
| |
Collapse
|
22
|
Maron GM, Hijano DR, Epperly R, Su Y, Tang L, Hayden RT, Naik S, Karol SE, Gottschalk S, Triplett BM, Talleur AC. Infectious Complications in Pediatric, Adolescent and Young Adult Patients Undergoing CD19-CAR T Cell Therapy. Front Oncol 2022; 12:845540. [PMID: 35356197 PMCID: PMC8959860 DOI: 10.3389/fonc.2022.845540] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/04/2022] [Indexed: 12/20/2022] Open
Abstract
CD19-specific chimeric antigen receptor (CAR) T cell therapy has changed the treatment paradigm for pediatric, adolescent and young adult (AYA) patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). However, data on the associated infectious disease challenges in this patient population are scarce. Knowledge of infections presenting during treatment, and associated risk factors, is critical for pediatric cellular therapy and infectious disease specialists as we seek to formulate effective anti-infective prophylaxis, infection monitoring schemas, and empiric therapy regimens. In this work we describe our institutional experience in a cohort of 38 pediatric and AYA patients with CD19-positive malignancy treated with lymphodepleting chemotherapy (fludarabine/cyclophosphamide) followed by a single infusion of CD19-CAR T cells (total infusions, n=39), including tisagenlecleucel (n=19; CD19/4-1BB) or on an institutional clinical trial (n=20; CD19/4-1BB; NCT03573700). We demonstrate that infections were common in the 90 days post CAR T cells, with 19 (50%) patients experiencing a total of 35 infections. Most of these (73.7%) occurred early post infusion (day 0 to 28; infection density of 2.36 per 100 patient days-at-risk) compared to late post infusion (day 29 to 90; infection density 0.98 per 100 patient days-at-risk), respectively. Bacterial infections were more frequent early after CAR T cell therapy, with a predominance of bacterial blood stream infections. Viral infections occurred throughout the post infusion period and included primarily systemic reactivations and gastrointestinal pathogens. Fungal infections were rare. Pre-infusion disease burden, intensity of bridging chemotherapy, lymphopenia post lymphodepleting chemotherapy/CAR T cell infusion and development of CAR-associated hemophagocytic lymphohistiocytosis (carHLH) were all significantly associated with either infection density or time to first infection post CAR T cell infusion. A subset of patients (n=6) had subsequent CAR T cell reinfusion and did not appear to have increased risk of infectious complications. Our experience highlights the risk of infections after CD19-CAR T cell therapy, and the need for continued investigation of infectious outcomes as we seek to improve surveillance, prophylaxis and treatment algorithms.
Collapse
Affiliation(s)
- Gabriela M. Maron
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Diego R. Hijano
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Rebecca Epperly
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Yin Su
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Li Tang
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Randall T. Hayden
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Swati Naik
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Seth E. Karol
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Brandon M. Triplett
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Aimee C. Talleur
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
23
|
Jacoby E, Bielorai B, Hutt D, Itzhaki O, Adam E, Bar D, Besser MJ, Toren A. Parameters of long‐term response with
CD28
‐based
CD19 chimaeric antigen receptor‐modified
T cells in children and young adults with
B‐acute lymphoblastic leukaemia. Br J Haematol 2022; 197:475-481. [DOI: 10.1111/bjh.18105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Elad Jacoby
- Division of Pediatric Hematology and Oncology The Edmond and Lily Safra Children's Hospital, Sheba Medical Center Tel Hashomer Israel
- Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
| | - Bella Bielorai
- Division of Pediatric Hematology and Oncology The Edmond and Lily Safra Children's Hospital, Sheba Medical Center Tel Hashomer Israel
- Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
| | - Daphna Hutt
- Division of Pediatric Hematology and Oncology The Edmond and Lily Safra Children's Hospital, Sheba Medical Center Tel Hashomer Israel
| | - Orit Itzhaki
- Ella Institute of Immuno‐Oncology Sheba Medical Center Tel Hashomer Israel
| | - Etai Adam
- Division of Pediatric Hematology and Oncology The Edmond and Lily Safra Children's Hospital, Sheba Medical Center Tel Hashomer Israel
- Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
| | - Diana Bar
- Division of Pediatric Hematology and Oncology The Edmond and Lily Safra Children's Hospital, Sheba Medical Center Tel Hashomer Israel
| | - Michal J. Besser
- Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
- Ella Institute of Immuno‐Oncology Sheba Medical Center Tel Hashomer Israel
| | - Amos Toren
- Division of Pediatric Hematology and Oncology The Edmond and Lily Safra Children's Hospital, Sheba Medical Center Tel Hashomer Israel
- Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
| |
Collapse
|