Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 141(6): 609-619, 2023 02 09.
Article in English | MEDLINE | ID: mdl-36351239

ABSTRACT

Children living in poverty experience excessive relapse and death from newly diagnosed acute lymphoblastic leukemia (ALL). The influence of household poverty and neighborhood social determinants on outcomes from chimeric antigen receptor (CAR) T-cell therapy for relapsed/refractory (r/r) leukemia is poorly described. We identified patients with r/r CD19+ ALL/lymphoblastic lymphoma treated on CD19-directed CAR T-cell clinical trials or with commercial tisagenlecleucel from 2012 to 2020. Socioeconomic status (SES) was proxied at the household level, with poverty exposure defined as Medicaid-only insurance. Low-neighborhood opportunity was defined by the Childhood Opportunity Index. Among 206 patients aged 1 to 29, 35.9% were exposed to household poverty, and 24.9% had low-neighborhood opportunity. Patients unexposed to household poverty or low-opportunity neighborhoods were more likely to receive CAR T-cell therapy with a high disease burden (>25%), a disease characteristic associated with inferior outcomes, as compared with less advantaged patients (38% vs 30%; 37% vs 26%). Complete remission (CR) rate was 93%, with no significant differences by household poverty (P = .334) or neighborhood opportunity (P = .504). In multivariate analysis, patients from low-opportunity neighborhoods experienced an increased hazard of relapse as compared with others (P = .006; adjusted hazard ratio [HR], 2.3; 95% confidence interval [CI], 1.3-4.1). There was no difference in hazard of death (P = .545; adjusted HR, 1.2; 95% CI, 0.6-2.4). Among children who successfully receive CAR T-cell therapy, CR and overall survival are equitable regardless of proxied SES and neighborhood opportunity. Children from more advantaged households and neighborhoods receive CAR T-cell therapy with a higher disease burden. Investigation of multicenter outcomes and access disparities outside of clinical trial settings is warranted.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Receptors, Chimeric Antigen , Humans , Child , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Recurrence , Antigens, CD19 , Poverty
2.
Blood ; 139(14): 2173-2185, 2022 04 07.
Article in English | MEDLINE | ID: mdl-34871373

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy can induce durable remissions of relapsed/refractory B-acute lymphoblastic leukemia (ALL). However, case reports suggested differential outcomes mediated by leukemia cytogenetics. We identified children and young adults with relapsed/refractory CD19+ ALL/lymphoblastic lymphoma treated on 5 CD19-directed CAR T-cell (CTL019 or humanized CART19) clinical trials or with commercial tisagenlecleucel from April 2012 to April 2019. Patients were hierarchically categorized according to leukemia cytogenetics: High-risk lesions were defined as KMT2A (MLL) rearrangements, Philadelphia chromosome (Ph+), Ph-like, hypodiploidy, or TCF3/HLF; favorable as hyperdiploidy or ETV6/RUNX1; and intermediate as iAMP21, IKZF1 deletion, or TCF3/PBX1. Of 231 patients aged 1 to 29, 74 (32%) were categorized as high risk, 28 (12%) as intermediate, 43 (19%) as favorable, and 86 (37%) as uninformative. Overall complete remission rate was 94%, with no difference between strata. There was no difference in relapse-free survival (RFS; P = .8112), with 2-year RFS for the high-risk group of 63% (95% confidence interval [CI], 52-77). There was similarly no difference seen in overall survival (OS) (P = .5488), with 2-year OS for the high-risk group of 70% (95% CI, 60-82). For patients with KMT2A-rearranged infant ALL (n = 13), 2-year RFS was 67% (95% CI, 45-99), and OS was 62% (95% CI, 40-95), with multivariable analysis demonstrating no increased risk of relapse (hazard ratio, 0.70; 95% CI, 0.21-2.90; P = .7040) but a higher proportion of relapses associated with myeloid lineage switch and a 3.6-fold increased risk of all-cause death (95% CI, 1.04-12.75; P = .0434). CTL019/huCART19/tisagenlecleucel are effective at achieving durable remissions across cytogenetic categories. Relapsed/refractory patients with high-risk cytogenetics, including KMT2A-rearranged infant ALL, demonstrated high RFS and OS probabilities at 2 years.


Subject(s)
Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Antigens, CD19 , Child , Cytogenetic Analysis , Humans , Infant , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/therapeutic use , Recurrence , Young Adult
3.
Ann Surg ; 278(3): e589-e597, 2023 09 01.
Article in English | MEDLINE | ID: mdl-36538614

ABSTRACT

OBJECTIVE: Develop a predictive model to identify patients with 1 pathologic lymph node (pLN) versus >1 pLN using machine learning applied to gene expression profiles and clinical data as input variables. BACKGROUND: Standard management for clinically detected melanoma lymph node metastases is complete therapeutic LN dissection (TLND). However, >40% of patients with a clinically detected melanoma lymph node will only have 1 pLN on final review. Recent data suggest that targeted excision of just the single enlarged LN may provide excellent regional control, with less morbidity than TLND. The selection of patients for less morbid surgery requires accurate identification of those with only 1 pLN. METHODS: The Cancer Genome Atlas database was used to identify patients who underwent TLND for melanoma. Pathology reports in The Cancer Genome Atlas were reviewed to identify the number of pLNs. Patients were included for machine learning analyses if RNA sequencing data were available from a pLN. After feature selection, the top 20 gene expression and clinical input features were used to train a ridge logistic regression model to predict patients with 1 pLN versus >1 pLN using 10-fold cross-validation on 80% of samples. The model was then tested on the remaining holdout samples. RESULTS: A total of 153 patients met inclusion criteria: 64 with one pLN (42%) and 89 with >1 pLNs (58%). Feature selection identified 1 clinical (extranodal extension) and 19 gene expression variables used to predict patients with 1 pLN versus >1 pLN. The ridge logistic regression model identified patient groups with an accuracy of 90% and an area under the receiver operating characteristic curve of 0.97. CONCLUSIONS: Gene expression profiles together with clinical variables can distinguish melanoma metastasis patients with 1 pLN versus >1 pLN. Future models trained using positron emission tomography/computed tomography imaging, gene expression, and relevant clinical variables may further improve accuracy and may predict patients who can be managed with a targeted LN excision rather than a complete TLND.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Lymphatic Metastasis/pathology , Skin Neoplasms/genetics , Skin Neoplasms/surgery , Skin Neoplasms/pathology , Melanoma/genetics , Melanoma/surgery , Melanoma/pathology , Lymph Nodes/pathology , Decision Making , Lymph Node Excision , Retrospective Studies
4.
Pediatr Blood Cancer ; 67(3): e28092, 2020 03.
Article in English | MEDLINE | ID: mdl-31793170

ABSTRACT

Twenty-eight patients were maintained on subcutaneous immunoglobulin replacement for persistent B-cell aplasia and agammaglobulinemia following CD19-targeted chimeric antigen receptor T-cell therapy for B-cell lymphoblastic leukemia. Patients were transitioned from intravenous to subcutaneous immunoglobulin replacement at a median of 11.5 months (range, 4-20). Increasing serum IgG level was significantly associated with a lower rate of sinopulmonary infection (P = 0.0072). The median serum IgG level during infection-free periods was 1000 mg/dL (range, 720-1430), which was significantly higher than IgG levels in patients with sinopulmonary infections. As such, we recommend maintaining a goal IgG level > 1000 mg/dL to provide optimal protection.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Immunoglobulins/administration & dosage , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/transplantation , Adolescent , Adult , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Injections, Subcutaneous , Male , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Retrospective Studies , T-Lymphocytes/immunology , Young Adult
5.
JAMA ; 322(17): 1673-1681, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31688884

ABSTRACT

Importance: Children, adolescents, and young adults with acute myeloid leukemia are at high risk of life-threatening invasive fungal disease with both yeasts and molds. Objective: To compare the efficacy of caspofungin vs fluconazole prophylaxis against proven or probable invasive fungal disease and invasive aspergillosis during neutropenia following acute myeloid leukemia chemotherapy. Design, Setting, and Participants: This multicenter, randomized, open-label, clinical trial enrolled patients aged 3 months to 30 years with newly diagnosed de novo, relapsed, or secondary acute myeloid leukemia being treated at 115 US and Canadian institutions (April 2011-November 2016; last follow-up June 30, 2018). Interventions: Participants were randomly assigned during the first chemotherapy cycle to prophylaxis with caspofungin (n = 257) or fluconazole (n = 260). Prophylaxis was administered during the neutropenic period following each chemotherapy cycle. Main Outcomes and Measures: The primary outcome was proven or probable invasive fungal disease as adjudicated by blinded central review. Secondary outcomes were invasive aspergillosis, empirical antifungal therapy, and overall survival. Results: The second interim efficacy analysis and an unplanned futility analysis based on 394 patients appeared to have suggested futility, so the study was closed to accrual. Among the 517 participants who were randomized (median age, 9 years [range, 0-26 years]; 44% female), 508 (98%) completed the trial. The 23 proven or probable invasive fungal disease events (6 caspofungin vs 17 fluconazole) included 14 molds, 7 yeasts, and 2 fungi not further categorized. The 5-month cumulative incidence of proven or probable invasive fungal disease was 3.1% (95% CI, 1.3%-7.0%) in the caspofungin group vs 7.2% (95% CI, 4.4%-11.8%) in the fluconazole group (overall P = .03 by log-rank test) and for cumulative incidence of proven or probable invasive aspergillosis was 0.5% (95% CI, 0.1%-3.5%) with caspofungin vs 3.1% (95% CI, 1.4%-6.9%) with fluconazole (overall P = .046 by log-rank test). No statistically significant differences in empirical antifungal therapy (71.9% caspofungin vs 69.5% fluconazole, overall P = .78 by log-rank test) or 2-year overall survival (68.8% caspofungin vs 70.8% fluconazole, overall P = .66 by log-rank test) were observed. The most common toxicities were hypokalemia (22 caspofungin vs 13 fluconazole), respiratory failure (6 caspofungin vs 9 fluconazole), and elevated alanine transaminase (4 caspofungin vs 8 fluconazole). Conclusions and Relevance: Among children, adolescents, and young adults with acute myeloid leukemia, prophylaxis with caspofungin compared with fluconazole resulted in significantly lower incidence of invasive fungal disease. The findings suggest that caspofungin may be considered for prophylaxis against invasive fungal disease, although study interpretation is limited by early termination due to an unplanned interim analysis that appeared to have suggested futility. Trial Registration: ClinicalTrials.gov Identifier: NCT01307579.


Subject(s)
Antifungal Agents/therapeutic use , Caspofungin/therapeutic use , Fluconazole/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Mycoses/prevention & control , Adolescent , Adult , Antifungal Agents/adverse effects , Aspergillosis/epidemiology , Aspergillosis/prevention & control , Caspofungin/adverse effects , Child , Child, Preschool , Early Termination of Clinical Trials , Female , Fluconazole/adverse effects , Humans , Infant , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/complications , Male , Neutropenia/complications , Young Adult
6.
Blood ; 128(3): 360-70, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27166358

ABSTRACT

The mechanisms underlying the maintenance of long-lasting humoral immunity are not well understood. Studies in mice indicate that plasma cells (PCs) can survive up to a lifetime, even in the absence of regeneration by B cells, implying the presence of long-lived PCs as a mechanism for long-lasting immunity. Evidence from humans treated with anti-CD20, which depletes circulating B cells, also suggests B-cell-independent long-term survival of some PCs. On the other hand, antibody responses may be sustained solely by short-lived PCs with repopulation from clonally related memory B cells. To explore PC longevity and humoral immunity in humans, we investigated the fate of PCs and their antibodies in adult and pediatric patients who received chimeric antigen receptor-based adoptive T-cell immunotherapy targeting CD19 to treat B-cell lineage malignancies (CTL019). Treatment with CTL019 is frequently associated with B-cell aplasia that can persist for years. Serum antibody titers to vaccine-related antigens were measured, and quantitative assessment of B cells and PCs in blood and bone marrow was performed at various time points before and after CTL019 therapy. While total serum immunoglobulin concentrations decline following CTL019-induced B-cell aplasia, several vaccine/pathogen-specific serum immunoglobulin G and A (IgG and IgA) titers remain relatively stable for at least 6 and 12 months posttreatment, respectively. Analysis of bone marrow biopsies after CTL019 revealed 8 patients with persistence of antibody-secreting PCs at least 25 months post-CTL019 infusion despite absence of CD19(+)CD20(+) B cells. These results provide strong evidence for the existence of memory B-cell-independent, long-lived PCs in humans that contribute to long-lasting humoral immunity.


Subject(s)
Adoptive Transfer , Antigens, CD19 , Lymphoma, B-Cell , Plasma Cells , T-Lymphocytes , Adolescent , Adult , Antigens, CD19/blood , Antigens, CD19/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Child , Female , Humans , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lymphoma, B-Cell/blood , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/therapy , Male , Middle Aged , Plasma Cells/immunology , Plasma Cells/metabolism , Plasma Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , T-Lymphocytes/transplantation
7.
Blood Adv ; 8(9): 2182-2192, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38386999

ABSTRACT

ABSTRACT: Relapse after CD19-directed chimeric antigen receptor (CAR)-modified T cells remains a substantial challenge. Short CAR T-cell persistence contributes to relapse risk, necessitating novel approaches to prolong durability. CAR T-cell reinfusion (CARTr) represents a potential strategy to reduce the risk of or treat relapsed disease after initial CAR T-cell infusion (CARTi). We conducted a retrospective review of reinfusion of murine (CTL019) or humanized (huCART19) anti-CD19/4-1BB CAR T cells across 3 clinical trials or commercial tisagenlecleucel for relapse prevention (peripheral B-cell recovery [BCR] or marrow hematogones ≤6 months after CARTi), minimal residual disease (MRD) or relapse, or nonresponse to CARTi. The primary endpoint was complete response (CR) at day 28 after CARTr, defined as complete remission with B-cell aplasia. Of 262 primary treatments, 81 were followed by ≥1 reinfusion (investigational CTL019, n = 44; huCART19, n = 26; tisagenlecleucel, n = 11), representing 79 patients. Of 63 reinfusions for relapse prevention, 52% achieved CR (BCR, 15/40 [38%]; hematogones, 18/23 [78%]). Lymphodepletion was associated with response to CARTr for BCR (odds ratio [OR], 33.57; P = .015) but not hematogones (OR, 0.30; P = .291). The cumulative incidence of relapse was 29% at 24 months for CR vs 61% for nonresponse to CARTr (P = .259). For MRD/relapse, CR rate to CARTr was 50% (5/10), but 0/8 for nonresponse to CARTi. Toxicity was generally mild, with the only grade ≥3 cytokine release syndrome (n = 6) or neurotoxicity (n = 1) observed in MRD/relapse treatment. Reinfusion of CTL019/tisagenlecleucel or huCART19 is safe, may reduce relapse risk in a subset of patients, and can reinduce remission in CD19+ relapse.


Subject(s)
Antigens, CD19 , Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Child , Antigens, CD19/immunology , Antigens, CD19/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Child, Preschool , Female , Male , Receptors, Chimeric Antigen/therapeutic use , Adolescent , Recurrence , Retrospective Studies , Infant , Receptors, Antigen, T-Cell/therapeutic use , Treatment Outcome , T-Lymphocytes/immunology
8.
Asia Pac J Oncol Nurs ; 10(11): 100281, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38023730

ABSTRACT

The treatment landscape for pediatric cancers over the last 11 years has undergone a dramatic change, especially with relapsed and refractory B-cell acute lymphoblastic leukemia (ALL), due to the introduction of chimeric antigen receptor-T (CAR-T) cell therapy. Because of the success of CAR-T cell therapy in patients with relapsed and refractory B-cell ALL, this promising therapy is undergoing trials in multiple other pediatric malignancies. This article will focus on the introduction of CAR-T cell therapy in pediatric B-cell ALL and discuss past and current trials. We will also discuss trials for CAR-T cell therapy in other pediatric malignancies. This information was gathered through a comprehensive literature review along with using first hand institutional experience. Due to the potential severe toxicities related to CAR-T cell therapy, safe practices and monitoring are key. These authors demonstrate that nurses have a profound responsibility in preparing and caring for patients and families, monitoring and managing side effects in these patients, ensuring that study guidelines are followed, and providing continuity for patients, families, and referring providers. Education of nurses is crucial for improved patient outcomes.

9.
J Immunother Cancer ; 11(1)2023 01.
Article in English | MEDLINE | ID: mdl-36707090

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.


Subject(s)
COVID-19 , Coronavirus Infections , Pneumonia, Viral , Receptors, Chimeric Antigen , Humans , Child , Young Adult , Adolescent , Adult , COVID-19/complications , SARS-CoV-2 , Retrospective Studies , Pneumonia, Viral/complications , Coronavirus Infections/complications , Betacoronavirus , Neoplasm Recurrence, Local , Registries , Cell- and Tissue-Based Therapy
10.
Lancet Haematol ; 8(10): e711-e722, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34560014

ABSTRACT

BACKGROUND: CNS relapse of acute lymphocytic leukaemia is difficult to treat. Durable remissions of relapsed or refractory B-cell acute lymphocytic leukaemia have been observed following treatment with CD19-directed chimeric antigen receptor (CAR) T cells; however, most trials have excluded patients with active CNS disease. We aimed to assess the safety and activity of CAR T-cell therapy in patients with a history of CNS relapsed or refractory B-cell acute lymphocytic leukaemia. METHODS: In this post-hoc analysis, we included 195 patients (aged 1-29 years; 110 [56%] male and 85 [44%] female) with relapsed or refractory CD19-positive acute lymphocytic leukaemia or lymphocytic lymphoma from five clinical trials (Pedi CART19, 13BT022, ENSIGN, ELIANA, and 16CT022) done at the Children's Hospital of Philadelphia (Philadelphia, PA, USA), in which participants received CD19-directed CAR T-cell therapy between April 17, 2012, and April 16, 2019. The trials required control of CNS disease at enrolment and infusion and excluded treatment in the setting of acute neurological toxic effects (>grade 1 in severity) or parenchymal lesions deemed to increase the risk of neurotoxicity. 154 patients from Pedi CART19, ELIANA, ENSIGN, and 16CT022 received tisagenlecleucel and 41 patients from the 13BT022 trial received the humanised CD19-directed CAR, huCART19. We categorised patients into two strata on the basis of CNS status at relapse or within the 12 months preceding CAR T-cell infusion-either CNS-positive or CNS-negative disease. Patients with CNS-positive disease were further divided on the basis of morphological bone marrow involvement-either combined bone marrow and CNS involvement, or isolated CNS involvement. Endpoints were the proportion of patients with complete response at 28 days after infusion, Kaplan-Meier analysis of relapse-free survival and overall survival, and the incidence of cytokine release syndrome and neurotoxicity. FINDINGS: Of all 195 patients, 66 (34%) were categorised as having CNS-positive disease and 129 (66%) as having CNS-negative disease, and 43 (22%) were categorised as having isolated CNS involvement. The median length of follow-up was 39 months (IQR 25-49) in the CNS-positive stratum and 36 months (18-49) in the CNS-negative stratum. The proportion of patients in the CNS-positive stratum with a complete response at 28 days after infusion was similar to that in the CNS-negative stratum (64 [97%] of 66 vs 121 [94%] of 129; p=0·74), with no significant difference in relapse-free survival (60% [95% CI 49-74] vs 60% [51-71]; p=0·50) or overall survival (83% [75-93] vs 71% [64-79]; p=0·39) at 2 years between the two groups. Overall survival at 2 years was significantly higher in patients with isolated CNS involvement compared with those with bone marrow involvement (91% [82-100] vs 71% [64-78]; p=0·046). The incidence and severity of neurotoxicity (any grade, 53 [41%] vs 38 [58%]; grade 1, 24 [19%] vs 20 [30%]; grade 2, 14 [11%] vs 10 [15%]; grade 3, 12 [9%] vs 6 [9%], and grade 4, 3 [2%] vs 2 [3%]; p=0·20) and cytokine release syndrome (any grade, 110 [85%] vs 53 [80%]; grade 1, 12 [9%] vs 2 [3%]; grade 2, 61 [47%] vs 38 [58%]; grade 3, 18 [14%] vs 7 [11%] and grade 4, 19 [15%] vs 6 [9%]; p=0·26) did not differ between the CNS-negative and the CNS-positive disease strata. INTERPRETATION: Tisagenlecleucel and huCART19 are active at clearing CNS disease and maintaining durable remissions in children and young adults with CNS relapsed or refractory B-cell acute lymphocytic leukaemia or lymphocytic lymphoma, without increasing the risk of severe neurotoxicity; although care should be taken in the timing of therapy and disease control to mitigate this risk. These preliminary findings support the use of these CAR T-cell therapies for patients with CNS relapsed or refractory B-cell acute lymphocytic leukaemia. FUNDING: Children's Hospital of Philadelphia Frontier Program.


Subject(s)
Antigens, CD19/immunology , Central Nervous System Neoplasms/therapy , Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Central Nervous System Neoplasms/immunology , Child, Preschool , Female , Humans , Infant , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, Chimeric Antigen/immunology , Recurrence , Young Adult
11.
J Clin Oncol ; 39(8): 920-930, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33417474

ABSTRACT

PURPOSE: To prospectively evaluate the effectiveness of risk-adapted preemptive tocilizumab (PT) administration in preventing severe cytokine release syndrome (CRS) after CTL019, a CD19 chimeric antigen receptor T-cell therapy. METHODS: Children and young adults with CD19-positive relapsed or refractory B-cell acute lymphoblastic leukemia were assigned to high- (≥ 40%) or low- (< 40%) tumor burden cohorts (HTBC or LTBC) based on a bone marrow aspirate or biopsy before infusion. HTBC patients received a single dose of tocilizumab (8-12 mg/kg) after development of high, persistent fevers. LTBC patients received standard CRS management. The primary end point was the frequency of grade 4 CRS (Penn scale), with an observed rate of ≤ 5 of 15 patients in the HTBC pre-defined as clinically meaningful. In post hoc analyses, the HTBC was compared with a historical cohort of high-tumor burden patients from the initial phase I CTL019 trial. RESULTS: The primary end point was met. Seventy patients were infused with CTL019, 15 in the HTBC and 55 in the LTBC. All HTBC patients received the PT intervention. The incidence of grade 4 CRS was 27% (95% CI, 8 to 55) in the HTBC and 3.6% (95% CI, 0.4 to 13) in the LTBC. The best overall response rate was 87% in the HTBC and 100% in the LTBC. Initial CTL019 expansion was greater in the HTBC than the LTBC (P < .001), but persistence was not different (P = .73). Event-free and overall survival were worse in the HTBC (P = .004, P < .001, respectively). In the post hoc analysis, grade 4 CRS was observed in 27% versus 50% of patients in the PT and prior phase I cohorts, respectively (P = .18). CONCLUSION: Risk-adapted PT administration resulted in a decrease in the expected incidence of grade 4 CRS, meeting the study end point, without adversely impacting the antitumor efficacy or safety of CTL019.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, CD19/immunology , Cytokine Release Syndrome/drug therapy , Drug Resistance, Neoplasm , Immunotherapy, Adoptive/adverse effects , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Salvage Therapy , Adolescent , Adult , Child , Child, Preschool , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/pathology , Female , Follow-Up Studies , Humans , Infant , Male , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Pilot Projects , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Prospective Studies , Retrospective Studies , Risk Factors , Survival Rate , Young Adult
12.
J Clin Oncol ; 39(27): 3044-3055, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34156874

ABSTRACT

PURPOSE: CD19-targeted chimeric antigen receptor (CAR)-modified T cells demonstrate unprecedented responses in B-cell acute lymphoblastic leukemia (B-ALL); however, relapse remains a substantial challenge. Short CAR T-cell persistence contributes to this risk; therefore, strategies to improve persistence are needed. METHODS: We conducted a pilot clinical trial of a humanized CD19 CAR T-cell product (huCART19) in children and young adults with relapsed or refractory B-ALL (n = 72) or B-lymphoblastic lymphoma (n = 2), treated in two cohorts: with (retreatment, n = 33) or without (CAR-naive, n = 41) prior CAR exposure. Patients were monitored for toxicity, response, and persistence of huCART19. RESULTS: Seventy-four patients 1-29 years of age received huCART19. Cytokine release syndrome developed in 62 (84%) patients and was grade 4 in five (6.8%). Neurologic toxicities were reported in 29 (39%), three (4%) grade 3 or 4, and fully resolved in all cases. The overall response rate at 1 month after infusion was 98% (100% in B-ALL) in the CAR-naive cohort and 64% in the retreatment cohort. At 6 months, the probability of losing huCART19 persistence was 27% (95% CI, 14 to 41) for CAR-naive and 48% (95% CI, 30 to 64) for retreatment patients, whereas the incidence of B-cell recovery was 15% (95% CI, 6 to 28) and 58% (95% CI, 33 to 77), respectively. Relapse-free survival at 12 and 24 months, respectively, was 84% (95% CI, 72 to 97) and 74% (95% CI, 60 to 90) in CAR-naive and 74% (95% CI, 56 to 97) and 58% (95% CI, 37 to 90) in retreatment cohorts. CONCLUSION: HuCART19 achieved durable remissions with long-term persistence in children and young adults with relapsed or refractory B-ALL, including after failure of prior CAR T-cell therapy.


Subject(s)
Antigens, CD19/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Male , Pilot Projects , Young Adult
13.
J Immunother Cancer ; 8(2)2020 12.
Article in English | MEDLINE | ID: mdl-33335028

ABSTRACT

Immune effector cell (IEC) therapies offer durable and sustained remissions in significant numbers of patients with hematological cancers. While these unique immunotherapies have improved outcomes for pediatric and adult patients in a number of disease states, as 'living drugs,' their toxicity profiles, including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), differ markedly from conventional cancer therapeutics. At the time of article preparation, the US Food and Drug Administration (FDA) has approved tisagenlecleucel, axicabtagene ciloleucel, and brexucabtagene autoleucel, all of which are IEC therapies based on genetically modified T cells engineered to express chimeric antigen receptors (CARs), and additional products are expected to reach marketing authorization soon and to enter clinical development in due course. As IEC therapies, especially CAR T cell therapies, enter more widespread clinical use, there is a need for clear, cohesive recommendations on toxicity management, motivating the Society for Immunotherapy of Cancer (SITC) to convene an expert panel to develop a clinical practice guideline. The panel discussed the recognition and management of common toxicities in the context of IEC treatment, including baseline laboratory parameters for monitoring, timing to onset, and pharmacological interventions, ultimately forming evidence- and consensus-based recommendations to assist medical professionals in decision-making and to improve outcomes for patients.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/immunology , Immunologic Factors/immunology , Immunotherapy/methods , Guidelines as Topic , Humans , Retrospective Studies
14.
Clin J Oncol Nurs ; 23(2): 35-41, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30880817

ABSTRACT

BACKGROUND: This article presents an overview of pediatric relapsed and refractory acute lymphoblastic leukemia (ALL) and chimeric antigen receptor (CAR) T-cell therapy in pediatric patients. OBJECTIVES: Acute and chronic post-CAR T-cell effects and considerations are discussed, along with survivorship considerations. METHODS: A case study illustrates the identification and management of physiologic and psychosocial sequelae. FINDINGS: B-cell aplasia, hypogammaglobulinemia, infections, and cumulative effects of CAR T-cell therapy and other treatments are a concern in the pediatric population. Unique to pediatric and young adult survivors of childbearing potential are implications for post-treatment fertility. Financial toxicities and psychosocial needs require a family-centered approach to interventions that address the impact of CAR T-cell therapy not only on the patient, but also on caregivers and siblings.


Subject(s)
Immunotherapy, Adoptive , Pediatrics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Survival Rate , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/psychology
15.
Clin J Oncol Nurs ; 21(2 Suppl): 22-28, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28315553

ABSTRACT

BACKGROUND: Immunotherapy provides a promising treatment option for children and adolescents with refractory or relapsed acute lymphoblastic leukemia (ALL). 
. OBJECTIVES: This article presents a hospital's experience with providing chimeric antigen receptor (CAR) T-cell therapy, followed by a detailed discussion of the trajectory of treatment provided for pediatric patients and their families.
. METHODS: Clinical experience in delivering care to pediatric patients undergoing CAR T-cell therapy is described. Care coordination, patient and family assessment and education, and post-CAR T-cell infusion monitoring are presented. 
. FINDINGS: Of 59 patients having been treated with CAR T-cell therapy at the authors' institution, 93% had a complete response at day 28. The 12-month relapse-free survival rate is 55%. A multidisciplinary team of skilled clinicians is recommended to support patient and family needs throughout screening, treatment, and follow-up while coordinating care with the referring oncologist.


Subject(s)
Cell- and Tissue-Based Therapy , Chimera/immunology , Immunotherapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/therapeutic use , T-Lymphocytes/immunology , Adolescent , Child , Education, Nursing, Continuing , Female , Humans , Male , Receptors, Antigen, T-Cell/immunology , Recurrence
16.
J Adv Pract Oncol ; 8(1): 44-55, 2017.
Article in English | MEDLINE | ID: mdl-29900016

ABSTRACT

Despite more intensive therapy, high-risk neuroblastoma continues to be a challenging disease to treat. Postconsolidation immunotherapy has been studied for many years and has proven to be effective in clinical trials. Immunotherapy has become the standard of care for patients with high-risk disease, and many institutions across the country are providing this therapy. The care of these patients is complex and often associated with many side effects. The purpose of this article is to review the most common side effects seen in clinical practice and examine their management. Furthermore, this article will discuss the need for a consistent and educated multidisciplinary front-line team to care for these patients, with advanced practitioners playing a lead role to provide the care and attention needed for this patient population.

17.
Clin J Oncol Nurs ; 21(2 Suppl): 13-21, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28315552

ABSTRACT

BACKGROUND: Significant research progress has been made in immunotherapies since the mid-1990s, and this rapid evolution necessitates evidence-based education on immunotherapies, their pathophysiology, and their toxicities to provide safe, effective care.
. OBJECTIVES: The aim of this article is to provide an evidence-based overview, with implications for practice, of checkpoint inhibitors, monoclonal antibodies, oncolytic viral therapies, and chimeric antigen receptor T-cell therapies.
. METHODS: Each immunotherapy category is presented according to the pathophysiology of its immune modulation, the classes of agents within each category, evidence-based toxicities associated with each class, and implications for practice.
. FINDINGS: Immunotherapies vary in their pathophysiology and offer potential to be highly effective for the management of a wide array of cancer types. Understanding the unique pathophysiology and toxicities is necessary to assess, manage, and provide safe, effective patient-focused care.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Evidence-Based Nursing/standards , Immunotherapy/standards , Melanoma/immunology , Melanoma/therapy , Patient-Centered Care/standards , Practice Guidelines as Topic , Adult , Aged , Aged, 80 and over , Education, Nursing, Continuing , Evidence-Based Nursing/education , Female , Humans , Male , Middle Aged
18.
Cancer Discov ; 6(6): 664-79, 2016 06.
Article in English | MEDLINE | ID: mdl-27076371

ABSTRACT

UNLABELLED: Chimeric antigen receptor (CAR)-modified T cells with anti-CD19 specificity are a highly effective novel immune therapy for relapsed/refractory acute lymphoblastic leukemia. Cytokine release syndrome (CRS) is the most significant and life-threatening toxicity. To improve understanding of CRS, we measured cytokines and clinical biomarkers in 51 CTL019-treated patients. Peak levels of 24 cytokines, including IFNγ, IL6, sgp130, and sIL6R, in the first month after infusion were highly associated with severe CRS. Using regression modeling, we could accurately predict which patients would develop severe CRS with a signature composed of three cytokines. Results were validated in an independent cohort. Changes in serum biochemical markers, including C-reactive protein and ferritin, were associated with CRS but failed to predict development of severe CRS. These comprehensive profiling data provide novel insights into CRS biology and, importantly, represent the first data that can accurately predict which patients have a high probability of becoming critically ill. SIGNIFICANCE: CRS is the most common severe toxicity seen after CAR T-cell treatment. We developed models that can accurately predict which patients are likely to develop severe CRS before they become critically ill, which improves understanding of CRS biology and may guide future cytokine-directed therapy. Cancer Discov; 6(6); 664-79. ©2016 AACR.See related commentary by Rouce and Heslop, p. 579This article is highlighted in the In This Issue feature, p. 561.


Subject(s)
Biomarkers , Cell- and Tissue-Based Therapy/adverse effects , Cytokines/metabolism , Immunotherapy/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Tumor Lysis Syndrome/etiology , Tumor Lysis Syndrome/metabolism , Adolescent , Adult , Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, CD19/immunology , Antigens, CD19/metabolism , Cell- and Tissue-Based Therapy/methods , Child , Child, Preschool , Female , Humans , Immunotherapy/methods , Male , Middle Aged , Phenotype , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Prognosis , ROC Curve , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Severity of Illness Index , Treatment Outcome , Tumor Lysis Syndrome/diagnosis , Tumor Lysis Syndrome/drug therapy , Young Adult
19.
Clin Cancer Res ; 18(24): 6732-41, 2012 12 15.
Article in English | MEDLINE | ID: mdl-23092876

ABSTRACT

PURPOSE: Children with high-risk neuroblastoma have a poor prognosis with chemotherapy alone, and hematopoietic stem cell transplantation offers improved survival. As a dose-escalation strategy, tandem transplants have been used, but are associated with persistent immunocompromise. This study evaluated the provision of an autologous costimulated, activated T-cell product to support immunologic function. EXPERIMENTAL DESIGN: Nineteen subjects with high-risk neuroblastoma were enrolled in a pilot phase and 23 subjects were entered in to the randomized study. Immunologic reconstitution was defined by flow cytometric and functional assays. Next-generation sequencing was conducted to identify changes to the T-cell repertoire. Twenty-two patients were vaccinated to define effects on antibody responses. RESULTS: Subjects who received their autologous costimulated T-cell product on day 2 had significantly superior T-cell counts and T-cell proliferation compared with those who received T cells on day 90. Early administration of autologous T cells suppressed oligoclonality and enhanced repertoire diversity. The subjects who received the day 2 T-cell product also had better responses to the pneumococcal vaccine. CONCLUSIONS: The infusion of activated T cells can improve immunologic function especially when given early after transplant. This study showed the benefit of providing cell therapies during periods of maximum lymphopenia.


Subject(s)
Adoptive Transfer , B-Lymphocytes/immunology , Neuroblastoma/therapy , T-Lymphocytes/transplantation , Antigens, Bacterial/immunology , Cell Proliferation , Child, Preschool , Female , Humans , Immunologic Memory , Immunotherapy, Adoptive , Infant , Influenza Vaccines/immunology , Linear Models , Lymphocyte Count , Male , Neuroblastoma/immunology , Pilot Projects , Statistics, Nonparametric , T-Lymphocytes/immunology , Transplantation, Autologous , Treatment Outcome
20.
J Pediatr Oncol Nurs ; 21(1): 16-21, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15058402

ABSTRACT

Central lines are commonly placed in pediatric oncology patients during their therapy. Advanced practice nurses (APNs) play an important role in the coordination and facilitation of line placements so that they may be inserted in an expeditious manner Collaboration between the Divisions of Oncology and Surgery is often key to attaining this goal. At the Children's Hospital of Philadelphia, the APNs in Oncology and Surgery have developed a collaborative approach to the placement of central lines in oncology patients. Many positive changes ensued between these divisions after collaboration occurred and a system for line placements was put into place. A well-defined process for central line placement is currently being utilized that meets the needs of the patients. Central lines are being placed in a timelier manner, procedures are coordinated, and treatment is started earlier, which has increased the satisfaction of patients, families, and health care providers. Finally, this collaboration led to an improved relationship and better communication between the Divisions of Oncology and Surgery.


Subject(s)
Catheterization, Central Venous/nursing , Models, Nursing , Nurse Clinicians , Oncology Nursing/organization & administration , Pediatric Nursing/organization & administration , Perioperative Nursing/organization & administration , Algorithms , Catheterization, Central Venous/methods , Catheterization, Central Venous/psychology , Cooperative Behavior , Decision Trees , Hospitals, Pediatric , Humans , Interdepartmental Relations , Interprofessional Relations , Nurse Clinicians/organization & administration , Nurse Clinicians/psychology , Nurse's Role , Organizational Innovation , Patient Satisfaction , Patient Selection , Philadelphia , Referral and Consultation/organization & administration , Time Factors , Total Quality Management/organization & administration
SELECTION OF CITATIONS
SEARCH DETAIL