Abstract
Here we present the 3-year results of ZUMA-4, a phase I/II multicenter study evaluating the safety and efficacy of KTEX19, an autologous anti-CD19 chimeric antigen receptor (CAR) T-cell therapy, in pediatric/adolescent patients with relapsed/refractory B-cell acute lymphoblastic leukemia. Phase I explored two dose levels and formulations. The primary endpoint was the incidence of dose-limiting toxicities. Thirty-one patients were enrolled; KTE-X19 was administered to 24 patients (median age 13.5 years, range 3-20; median follow-up 36.1 months). No dose-limiting toxicities were observed. All treated patients had grade ≥3 adverse events, commonly hypotension (50%) and anemia (42%). Grade 3 cytokine release syndrome rates were 33% in all treated patients, 75% in patients given the dose of 2×106 CAR T cells/kg, 27% in patients given the dose of 1×106 cells/kg in the 68 mL formulation, and 22% in patients given the dose of 1×106 cells/kg in the 40 mL formulation; the percentages of patients experiencing grade ≥3 neurologic events were 21%, 25%, 27%, and 11% respectively. Overall complete remission rates (including complete remission with incomplete hematologic recovery) were 67% in all treated patients, 75% in patients given 2×106 CAR T cells/kg, 64% in patients given 1×106 cells/kg in the 68 mL formulation, and 67% in patients given 1×106 cells/kg in the 40 mL formulation. Overall minimal residual diseasenegativity rates were 100% among responders; 88% of responders underwent subsequent allogeneic stem-cell transplantation. In the 1×106 (40 mL) group (recommended phase II dose), the median duration of remission censored at allogeneic stem-cell transplantation and median overall survival were not reached. Pediatric/adolescent patients with relapsed/refractory B-cell acute lymphoblastic leukemia achieved high minimal residual disease-negative remission rates with a manageable safety profile after a single dose of KTE-X19. Phase II of the study is ongoing at the dose of 1×106 CAR T cells/kg in the 40 mL formulation. ClinicalTrials.gov: NCT02625480.
Introduction
Acute lymphoblastic leukemia (ALL) is the most common childhood malignancy, representing approximately 75% of childhood leukemias and 25% of all childhood cancers; 85% of these cases are B-cell precursor ALL (B-ALL).1-3 Although most children with B-ALL achieve durable complete remissions (CR) after initial treatment, approximately 10-20% develop relapsed/refractory (R/R) B-ALL with reported 2-year event-free survival rates of 35-46% after a second salvage attempt.2,3 While allogeneic stem-cell transplant (alloSCT) is a standard treatment option for many patients who relapse after first-line chemotherapy, some do not qualify for or are not indicated for alloSCT because of an inability to achieve CR, lack of a suitable donor, comorbidities, or late bone marrow relapse.4-8 Rates of relapse following alloSCT remain high despite treatment advances and survival rates are low2,9-11 especially for those with residual disease,12,13 highlighting the need for more effective therapies in pediatric R/R B-ALL.
The CD19-targeting immunotherapeutic agent blinatumomab, approved for the treatment of R/R B-ALL in adults and children, has shown efficacy in pediatric R/R B-ALL with CR rates of 39-63%, though the median overall survival (OS) was 7.5-14.6 months with more favorable survival in patients who proceeded to alloSCT.14-18 Tisagenlecleucel, an anti-CD19 chimeric antigen receptor (CAR) T-cell therapy approved for the treatment of R/R B-ALL in patients ≤25 years of age,19,20 led to more favorable remission rates in a phase II study than those previously reported with blinatumomab, although patients who had received prior anti-CD19 therapies were excluded from that study.21,22
An anti-CD19 CAR T-cell therapy containing a CD3ζ and CD28 co-stimulatory domain, developed at the National Cancer Institute,23,24 resulted in a 70% complete remission rate (CR+CR with incomplete hematologic recovery [CRi]) in children and adults ≤30 years of age with R/R B-ALL.25 KTE-X19, an autologous anti-CD19 CAR T-cell therapy with a CD3ζ and CD28 co-stimulatory domain,26,27 is approved by the Food and Drug Administration for the treatment of adults with R/R B-ALL.26,28 The manufacturing process for KTE-X19 removes leukemic blasts, as the presence of blasts may result in manufacturing failures and exhaustion of anti-CD19 CAR T cells during ex vivo manufacturing.29,30 KTE-X19 is manufactured at a centralized facility with worldwide shipment allowing for a fast turnaround time, which is critical for patients with rapidly proliferating disease and high tumor burden.28,30,31
Here we report the long-term results of phase I of the multicenter, single-arm, open-label, ZUMA-4 study evaluating the safety and efficacy of KTE-X19 in children and adolescents with R/R B-ALL.
Methods
Patients
In phase I of ZUMA-4, eligible patients were ≤21 years of age with a body weight of ≥6 kg and had R/R B-ALL, defined as refractory to first-line therapy, R/R after two or more lines of systemic therapy, or R/R after alloSCT if the patient was ≥100 days from alloSCT at the time of enrollment and off immunosuppressive medications for ≥4 weeks prior to enrollment. Prior treatment with blinatumomab was allowed (see Online Supplementary Methods for detailed eligibility criteria).
Study design and treatment
The phase I portion of ZUMA-4 was conducted at ten sites in the USA and one in Canada (Online Supplementary Table S1). The Institutional Review Board of each study site approved the study protocol. All patients or legally acceptable representatives (e.g., parent, legal guardian) provided written, informed assent/consent to participation in the study, which was conducted in accordance with the principles of the Declaration of Helsinki. This trial is registered at www.ClinicalTrials.gov (NCT02625480).
The objective of phase I was to evaluate the safety of KTE-X19 and determine the recommended phase 2 dose (RP2D) of KTE-X19 based on the incidence of dose-limiting toxicities (DLT; defined in the Online Supplementary Methods and Online Supplementary Table S2) and the overall safety profile. DLT were evaluated in the first three patients treated at the starting dose of 2×106 CAR T cells/kg. One additional patient was enrolled to receive 2×106 CAR T cells/kg. A Safety Review Team analyzed safety data after these patients had been followed for 28 days post-infusion, and subsequent patients received 1×106 CAR T cells/kg to further evaluate the potential to mitigate the risk of cytokine release syndrome (CRS) and neurologic events and thereby improve the risk:benefit ratio. The 1×106 CAR T cells/kg dosing formulation was modified from 68 mL to 40 mL for patients in a second cohort to achieve a higher final product cell density as part of product optimization to increase cell viability during cryopreservation and thawing.
Patients underwent leukapheresis to obtain cells for CAR T-cell manufacturing, followed by subsequent conditioning chemotherapy with fludarabine 25 mg/m2/day on days -4, -3, and -2, and cyclophosphamide 900 mg/m2 on day -2. Fresh leukapheresis material was used for CAR T-cell manufacturing; the manufactured CAR T cells were cryopreserved for shipment to the sites and thawed prior to infusion. Specified bridging chemotherapy was permitted between leukapheresis and conditioning chemotherapy (Online Supplementary Methods; Online Supplementary Table S3). KTE-X19 was administered on day 0 at the target dose of 2×106 or 1×106 CAR T cells/kg (68 mL or 40 mL formulation). Hospitalization was required for a minimum of 7 days after the infusion, followed by response assessments at prespecified time-points (Online Supplementary Methods).
Patients receiving 1×106 CAR T cells/kg (68 mL) were treated under original toxicity management guidelines, which included administration of tocilizumab for neurologic events only in the context of concurrent CRS and initiation of steroids for grade 2 neurologic events; patients receiving 1×106 CAR T cells/kg (40 mL) were treated under revised toxicity management guidelines according to which steroids were initiated for grade 1 neurologic events (Online Supplementary Table S4).
Outcomes and assessments
The primary endpoint of the phase I part of the study was the incidence of DLT in the set of patients evaluable for DLT, which included the first three patients treated with KTE-X19 at the 2×106 CAR T cells/kg dose. Secondary end-points included safety, overall CR rate (CR+CRi), duration of remission (DOR), minimal residual disease (MRD)-negativity rate, alloSCT rate, OS, and relapse-free survival (RFS). CRS was graded according to the 2014 modified criteria of Lee et al.32 Bone marrow evaluations and response assessments were conducted at day 28 and months 2 and 3 during the post-treatment period, and months 6, 9, 12, 15, 18, and 24 during the long-term follow-up period. MRD was tested in bone marrow using flow cytometry (Neogenomics, Fort Myers, FL, USA; sensitivity 0.01%). Additional endpoint and disease assessments are detailed in the Online Supplementary Methods.
Statistical analysis
The safety and efficacy analyses included all patients who received any dose of KTE-X19. Data are reported as of September 9, 2020. Additional statistical methods are described in the Online Supplementary Methods.
Results
Patients
Between February 17, 2016 and August 1, 2018, 31 patients were enrolled and underwent leukapheresis. The median time from leukapheresis to KTE-X19 product release was 14.0 days (range, 9.0-20.0) for all treated patients, 16.5 days (range, 12.0-23.0) from leukapheresis to delivery to study site, and 27.0 days (range, 18.0-41.0) from leukapheresis to infusion. Of the 31 enrolled patients, 24 (77%) received conditioning chemotherapy and were subsequently given KTE-X19. Seven patients were not given KTE-X19 for the following reasons: adverse event (n=1), unsuccessful product manufacture (n=3), ineligible due to adverse event (n=1), unsuccessful product manufacture and ineligible (n=1), and death (n=1) (Figure 1). Twenty-four patients received conditioning chemotherapy followed by KTE-X19; four patients received the 2×106 CAR T cells/kg dose, 11 received the 1×106 CAR T cells/kg (68 mL) dose formulation, and nine received the 1×106 CAR T cells/kg (40 mL) dose formulation. The median follow-up for all treated patients was 36.1 months (range, 24.0-53.9). The median age of treated patients was 13.5 years. Forty-two percent of patients had received three or more prior lines of therapy, including six patients (25%) who had previously undergone alloSCT, eight (33%) who had previously received blinatumomab, and one (4%) who had been treated with prior inotuzumab ozogamicin (Table 1). One patient (4%) had non-central nervous system extramedullary disease (Online Supplementary Results). Some patients had high-risk cytogenetics, including the Philadelphia chromosome t(9;22) mutation (n=4 [17%]), MLL translocation t(4;11) t(8;14) (n=1 [4%]), complex karyotype (≥5 abnormalities, n=4 [17%]), low hypodiploidy (30-39 chromosomes, n=1 [4%]), and near triploidy (60-78 chromosomes, n=2 [8%]). Of the 31 enrolled patients, 30 (97%) received bridging therapy per protocol with new baseline disease assessments performed just prior to conditioning chemotherapy.
Safety
Among the three DLT-evaluable patients receiving 2×106 CAR T cells/kg, no DLT were observed. All treated patients (n=24) experienced at least one grade ≥3 adverse event, most commonly hypotension (50%) and anemia (42%) (Table 2; Online Supplementary Table S5). Serious adverse events of any grade occurred in 71% of patients (Online Supplementary Table S6). Grade ≥3 infections occurred in 42% of patients (Online Supplementary Table S7).
CRS was reported in 21 of the 24 treated patients (88%), with eight (33%) experiencing grade ≥3 CRS (Table 3) according to modified Lee grading criteria.32 No grade 4 or grade 5 CRS events occurred. The most common grade ≥3 CRS symptoms were hypotension (50%) and pyrexia (25%).
Any-grade and grade ≥3 hypoxia was observed in 13% and 8% of patients, respectively. The median time to the onset of CRS and duration after KTE-X19 infusion was 5 days (range, 1-14) and 7 days, respectively, with all events resolved.
Among all treated patients, any-grade neurologic events were reported in 16 patients (67%), and grade ≥3 events occurred in five patients (21%), with encephalopathy (13%) being the most common grade ≥3 event (Table 3). One grade 4, fully reversible neurologic event (brain edema) occurred in a patient who received 1×106 CAR T cells/kg (68 mL); for the management of this event, the patient was treated with dexamethasone, mannitol, sodium chloride, and tocilizumab. There were no grade 5 neurologic events. Overall, the median time to onset of neurologic events was 9.5 days (range, 3-60) after infusion, the median time from resolution of the first CRS to onset of the first neurologic event was 4 days (range, -3 to 52 [the first CRS resolved after the onset of the first neurologic event in 4 patients]), and the median duration of neurologic events was 8 days. Neurologic events resolved in 14 of 16 patients (88%). The neurologic events of the remaining two patients were ongoing at the time of death, which was due to an adverse event (n=1) or progressive disease (n=1). Ten of 16 patients (63%) who experienced neurologic events had concurrent CRS.
Among all treated patients, 42% received steroids, 63% received tocilizumab, and 46% received vasopressors (Table 3). Improved overall safety was observed in the nine patients treated with the 1×106 CAR T cells/kg (40 mL) dose under revised toxicity management, relative to the four patients treated with 2×106 CAR T cells/kg and the 11 patients treated with 1×106 CAR T cells/kg (68 mL) under the original guidelines. Of the patients receiving 2×106 CAR T cells/kg, 75% experienced grade ≥3 CRS, compared with 27% and 22% of patients receiving 1×106 CAR T cells/kg (68 mL and 40 mL, respectively). Grade ≥3 neurologic events were observed in 25% of patients who received 2×106 CAR T cells/kg and 27% of patients who received 1×106 CAR T cells/kg (68 mL) but were lowest (11%) in patients who received 1×106 CAR T cells/kg (40 mL). In addition, the median time to onset of neurologic events, as well as CRS, appeared to be delayed in the 1×106 CAR T cells/kg dose cohorts compared with the 2×106 CAR T cells/kg dose cohort (Table 3).
Among the eight patients (33%) who died on study, six died from progressive disease (median, 190.5 days after KTE-X19 infusion), and two patients died from adverse events considered unrelated to KTE-X19, including disseminated mucormycosis (n=1, day 15 after KTE-X19 infusion) and Escherichia sepsis (n=1, day 409 after KTE-X19 infusion). Of those who died, three patients had received 2×106 CAR T cells/kg, four had received 1×106 CAR T cells/kg (68 mL), and one had received 1×106 CAR T cells/kg (40 mL). No patient tested positive for replication-competent retrovirus or antibodies to anti-CD19 CAR at any time.
Efficacy
With a median follow-up of 36.1 months (range, 24.0-53.9), all treated patients (n=24) were evaluable for efficacy. The overall remission rate by investigator assessment was 67%, with 29% of patients (n=7) achieving CR and 38% achieving CRi (n=9) (Table 4). In the 2×106, 1×106 (68 mL), and 1×106 (40 mL) CAR T cells/kg dose groups, the CR+CRi rate was 75%, 64%, and 67%, respectively. Prespecified subgroup analyses of CR+CRi are reported in Online Supplementary Figure S1. Of eight patients who had received prior blinatumomab therapy, three (38%) achieved CR+CRi (Online Supplementary Figure S1, Online Supplementary Results). The median time from infusion to first CR+CRi across dose levels was 30 days (range, 26-113 days). The overall MRD-negativity rate was 100% among the 16 patients with CR+CRi. Sixteen patients overall (67%) underwent alloSCT as subsequent consolidative therapy, including two, eight and six patients in the 2×106, 1×106 (68 mL), and 1×106 (40 mL) CAR T cells/kg dose groups, respectively. These included 14 of the 16 patients (88%) who achieved CR+CRi, the patient who achieved CR with partial hematologic recovery, and the patient with blastfree hypoplastic/aplastic bone marrow; the latter two subsequently achieved CR. Of all 16 transplanted patients, the median time to subsequent alloSCT was 2.3 months (range, 1.4-24.9) after KTE-X19; five of the 16 patients had received a prior transplant. Of the two patients who achieved CR+CRi but did not undergo a subsequent alloSCT, one died due to progressive disease, and one was lost to follow-up.
The median DOR for the 16 patients who achieved CR+CRi after KTE-X19 was 7.2 months (95% confidence interval [95% CI]: 4.1 months-not estimable) after censoring for subsequent alloSCT, and was 4.1 months, 10.7 months, and not reached in the 2×106, 1×106 (68 mL), and 1×106 (40 mL) CAR T cells/kg dose groups, respectively (Figure 2A). The median DOR was 14.2 months (95% CI: 3.9 months-not estimable) without censoring for subsequent alloSCT (Online Supplementary Figure S2A). The median DOR among the 14 patients with CR+CRi who underwent a subsequent alloSCT after KTE-X19 was 10.7 months (95% CI: 7.2 months-not estimable). The median RFS for all treated patients (n=24) after censoring for subsequent alloSCT was 5.2 months (95% CI: 0.03 months-not estimable). The median RFS for the group that received 1×106 CAR T cells/kg (40 mL) was not reached and was 5.2 months (95% CI: 0.03 months-not estimable) and 9.1 months (95% CI: 0.03 months-not estimable) in the 2×106 and 1×106 (68 mL) cells/kg cohorts, respectively (Figure 2B). The median RFS was 7.4 months (95% CI: 0.03 months-not estimable) without censoring for subsequent alloSCT (Online Supplementary Figure S2B). For the 16 patients who proceeded to subsequent alloSCT, the median RFS was 9.1 months (95% CI: 9.1 months-not estimable). The median RFS of patients in the intention-to-treat group (i.e., all those enrolled) with and without censoring for subsequent alloSCT (Online Supplementary Figure S3A, B) was 6.1 months (95% CI: 0.03 months-not estimable) and 6.2 months (95% CI: 0.03 months-not estimable), respectively. The median OS was not reached among all treated patients and in both 1×106 CAR T cells/kg dose groups and was 8.0 months for the 2×106 CAR T cells/kg dose group (Figure 2C). The 24-month OS rate was 87.5% (95% CI: 38.7-98.1%) for the 1×106 cells/kg (40 mL) dose and 72.7% (95% CI; 37.1-90.3%) for the 1×106 cells/kg (68 mL) dose. In the intention-to-treat group, the median OS was not reached (Online Supplementary Figure S3C). Overall, as of the data cutoff, eight of 24 treated patients (33%) had died, one had discontinued the study due to withdrawal of consent, and one was lost to follow-up. The remaining 14 patients (58%) were still alive and in continued follow-up as of the data cutoff; all of these patients underwent subsequent alloSCT after the administration of KTE-X19.
Based on the safety and efficacy data analysis, the RP2D was 1×106 KTE-X19 cells/kg (40 mL formulation) with revised toxicity management.
Translational analysis
CAR T-cell expansion in peripheral blood measured by droplet digital polymerase chain reaction and expressed as the number of CAR gene copies/mg DNA in blood was observed across dose groups with peak CAR T-cell levels reached by day 14 followed by a subsequent CAR T-cell contraction to baseline (Figure 3A; Online Supplementary Table S8). Median CAR T-cell levels were undetectable in blood by droplet digital polymerase chain reaction across all dose groups at 3 months after KTE-X19 infusion (Online Supplementary Table S8). Median peak CAR gene copies/mg DNA in blood were similar between the 1×106 CAR T cells/kg dose cohorts but were higher in the 2×106 CAR T cells/kg cohort (Figure 3B; Online Supplementary Figure S4A). Patients achieving CR+CRi trended toward higher peak blood CAR gene copies/mg DNA in blood than non-responders, as did patients who were MRD negative compared to those who were MRD positive (Figure 3C, D; Online Supplementary Figure S4B, C). CAR gene copies/µg DNA in blood trended higher in patients who had grade ≥3 neurologic events compared with those who had grade ≤2 neurologic events (Figure 3E; Online Supplementary Figure S4D), while there was no apparent difference in CAR gene copies/mg DNA in blood for the small numbers of patients with either high- or low-grade CRS (Figure 3F; Online Supplementary Figure S4E). The median peak CAR gene copies/mg DNA in blood was 5.16×104 (range, 0-2.40×105) in the 16 patients who had not received prior blinatumomab therapy, and was 6.15×103 (range, 0-2.49×105) in the eight patients who had received prior blinatumomab.
Peak levels of multiple key serum cytokines, chemokines, and pro-inflammatory biomarkers occurred by day 7. Commensurate with peak CAR expansion, some serum analytes trended higher in patients dosed with 2×106 compared with 1×106 CAR T cells/kg (interleukin [IL]-2, IL-5, IL-6, IL-8, IL-10, IL-15, IL-16, ferritin, granzyme B, intercellular adhesion molecule 1, interferon-γ, and tumor necrosis factor-a) (Figure 4; Online Supplementary Figure S5; Online Supplementary Table S9).
Peak serum levels of vascular cell adhesion molecule-1 and IL-16 were associated with grade ≥3 CRS. Such associations were not observed in patients with grade ≥3 neurologic events, which may have been due to the small number of patients with such events (Online Supplementary Table S10). Product characteristics were similar across dose levels (Online Supplementary Table S11). Proportions of less differentiated CCR7+ T cells in products trended higher in patients with CR+CRi and MRD negativity (Online Supplementary Table S12). This product profile also appeared to trend with higher levels of neurotoxicity but was not associated with CRS. The ratio of CD4 to CD8 T cells was not associated with response or toxicity.
Discussion
In phase I of ZUMA-4, no DLT were observed with KTE-X19 among the DLT-evaluable pediatric patients with R/R BALL. Although no DLT were observed at the initial dose of 2×106 CAR T cells/kg, a lower dose of 1×106 CAR T cells/kg with a 68 mL formulation was explored in a second cohort of patients in an effort to further improve the risk:benefit ratio, and dosing and toxicity management were further optimized in a third cohort at 1×106 CAR T cells/kg with a 40 mL formulation and revised toxicity management. This led to a more optimal risk:benefit ratio for the 1×106 CAR T cells/kg (40 mL) dose level with improvements for CRS and neurologic events. In addition, while MRD-negativity rates were ≥73% for all formulations, rates of MRD negativity and CR alone were highest in patients who received 1×106 CAR T cells/kg (40 mL). Importantly, the medians for DOR, RFS, and OS were not reached among the nine patients in the 1×106 CAR T cells/kg (40 mL) cohort, with most responders (5/6 [83%]) proceeding to subsequent alloSCT. Recognizing the limitations of a small cohort, nevertheless the 24-month OS rate in this group was 87.5%. These results suggest a meaningful durability of response with optimized dosing/formulation of KTE-X19 followed by subsequent alloSCT in pediatric/adolescent patients with R/R B-ALL.
The role of alloSCT following anti-CD19 CAR T-cell therapy in pediatric/adolescent patients with R/R B-ALL is still not well defined; studies in adult populations have provided somewhat conflicting results.33,34 In the present study, the medians for DOR censored at subsequent alloSCT and OS were not reached in patients treated at the RP2D of 1×106 CAR T cells/kg (40 mL). Fourteen of the 16 patients (88%) who achieved CR+CRi, including five treated at the RP2D, underwent alloSCT as subsequent therapy. AlloSCT was not required per protocol but was allowed at the investigators’ discretion. ZUMA-4 was not designed to assess outcomes after subsequent therapies; however, most responding patients proceeded to alloSCT after KTE-X19 as per investigators’ decision.
An evaluation of DOR in ZUMA-4 without censoring for subsequent alloSCT revealed a favorable median of 14.2 months. Additionally, the median RFS with censoring for subsequent alloSCT was 5.2 months, but was 7.4 months without censoring. It has recently been reported that pediatric and young adults with R/R CD19+ ALL who had no history of alloSCT, but who received consolidative alloSCT following anti-CD19 CAR T-cell therapy, trended toward improved leukemia-free survival with ≥1 year follow-up.35 In a recently published phase I study of anti-CD19 CAR T-cell therapy in children and young adults with R/R B-ALL with 75% of MRD-negative responding patients proceeding to alloSCT, the median OS at 4.8 years follow-up was 70.2 months following alloSCT, the 5-year event-free survival following alloSCT was 61.9%, and the cumulative incidence of relapse following alloSCT was only 9.5% at 24 months.36 Interestingly, a retrospective review of pediatric and young adult patients found that CD34-selected T-cell depleted alloSCT following CAR T-cell therapy may result in improved transplant-related mortality and OS versus that with unmodified alloSCT.37
It is difficult to draw conclusions from ZUMA-4 about the association between CAR T-cell persistence and durability of response given the low number of patients and the high rate of subsequent alloSCT. The median CAR T-cell levels in the blood of ZUMA-4 patients were undetectable across all doses at 3 months after the infusion, with the median time to alloSCT being 2.3 months and alloSCT likely eliminating remaining CAR T cells. Similarly, subsequent alloSCT precludes the assessment of B-cell aplasia in ZUMA-4. In studies with tisagenlecleucel, and in contrast to our study, subsequent alloSCT was performed in a minority of responding patients (12% to 16%).38,39 With a short median follow-up of only 13.1 months approximately 40% of patients with a complete response to tisagenlecleucel relapsed, mostly with CD19– leukemia despite persistent CAR T cells.21 After a median 24-month follow-up in that study, the 18-month OS rate was 70%,38 whereas the 24-month OS rate in ZUMA-4 for patients treated at the RP2D was 87.5%. Data presented herein support the promising potential role for KTE-X19 in extending response durability and survival in pediatric/adolescent patients with R/R B-ALL, particularly if followed by alloSCT.
While differences in trial designs and patient populations preclude direct trial-to-trial comparisons, recent studies with blinatumomab, which also targets CD19, indicate a median OS of just 7.5 months in pediatric R/R B-ALL,14 similar to results in adult ALL.40 Also for blinatumomab, consolidation with subsequent alloSCT has shown improved outcomes (87% vs. 29% 1-year OS probability for patients with vs. without subsequent alloSCT, respectively).17 Additionally, remission rates with blinatumomab were higher among pediatric patients with lower baseline tumor burden (<50% blasts; 56% CR) than in those with higher tumor burden (≥50% blasts; 33% CR).14
Data from ZUMA-4 suggest that KTE-X19 has the potential to offer more favorable efficacy in patients with high disease burden compared to results reported with blinatumomab. In ZUMA-4, a clear association between remission rates and bone marrow blasts prior to conditioning chemotherapy was not apparent, as CR rates were 83%, 50%, 80%, 50%, and 60% in patients with ≤5%, >5 to ≤25%, >25 to ≤50%, >50 to ≤75%, and >75 to 100% blasts at baseline, respectively. However, the small number of patients in each quartile, as well as the relatively high median tumor burden at baseline, limits interpretation (Online Supplementary Figure S1). This is in line with the findings of another pediatric and young adult study using CD19-directed CAR T-cell therapy in which no difference was observed in response rates based on disease burden.41 Notably, however, a large retrospective study of pediatric and young adult patients with ALL found that pre-treatment disease burden was independently associated with poorer survival after CD19 CAR T-cell therapy.42
While CR+CRi rates appeared lower in patients who had received prior blinatumomab therapy in ZUMA-4, conclusions are limited due to the small number of patients. There are conflicting reports on the impact of prior anti-CD19 therapies, such as blinatumomab, in patients who later receive anti-CD19 CAR T-cell therapy. In a single institution study it was observed that prior blinatumomab therapy was associated with a significantly higher rate of failure to achieve MRD-negative remission and also subsequent loss of remission with antigen escape after tisagenlecleucel in pediatric and adult R/R ALL.43 In contrast, a large, multicenter retrospective study of CD19 CAR T-cell therapy in pediatric and young adult patients with R/R ALL found no difference in outcomes in regard to prior blinatumomab exposure, with the exception that non-response to blinatumomab was independently associated with lower CR, RFS, and event-free survival rates.42 In ZUMA-4, one of three patients who had had a prior non-response to blinatumomab achieved a CR; however, the small numbers limit interpretation of these data.
The adverse event profile in ZUMA-4 was consistent with that in prior studies of anti-CD19 CAR T-cell therapies. For the patients who received KTE-X19, the median time from leukapheresis to product delivery to the study site was 16.5 days. In comparison, for the first 37 commercially manufactured tisagenlecleucel products for patients with B-ALL, the reported median throughput time was 23 days from receipt of the leukapheresed product to delivery to the clinical site.44 The rapid turnaround time for treated patients in ZUMA-4 supports the feasibility in the setting of rapidly proliferating ALL. With the RP2D established, ZUMA-4 has transitioned into the phase II portion of the study.
We observed higher proportions of less differentiated CCR7+ T cells in products in patients with CR+CRi and a trend in MRD-negative patients, as well as a trend toward higher peak CAR T-cell expansion in patients achieving CR/CRi and MRD negativity. These findings are consistent with a report that the frequency of CCR7+ T cells in anti-CD19 CAR T-cell products correlates with CAR T-cell expansion.45
ZUMA-4 was limited by the small number of patients treated at each dose level; as such, the study was not powered to assess the contribution of various patients’ characteristics to the outcomes observed. The durable outcomes reported herein are encouraging, although it is challenging to assess the long-term efficacy of KTE-X19 alone given that most responding patients proceeded to subsequent alloSCT. Future studies are warranted to determine which patients might benefit the most from KTE-X19 followed by alloSCT.
The unmet medical need in R/R pediatric ALL is greatest for patients who relapse early or have primary refractory disease with a 5-year OS rate of 21-28%.2,4,8,46-48 In addition, the risk of treatment-related morbidity and mortality is 3-5 times greater in patients who have MRD-positive disease at the end of initial and later lines of therapy than in patients who have undetectable MRD.3 To address this evolving unmet medical need, ZUMA-4 was further amended to broaden the eligibility criteria to include patients with MRD-positive disease and patients with early first relapse (≤18 months). Additionally, a second cohort was opened for pediatric patients with R/R non-Hodgkin lymphoma (diffuse large B-cell lymphoma, Burkitt lymphoma, and primary mediastinal B-cell lymphoma).
Footnotes
- Received March 10, 2022
- Accepted October 14, 2022
Correspondence
Disclosures
ASW reports research funding from Kite, a Gilead Company, Servier, and Institut de Recherches Internationales Servier. VH reports research funding from Servier; consultancy or an advisory role for Jazz, Gilead, and Servier; and speakers' bureau participation for Servier. NH reports consultancy or an advisory role for Stemline Therapeutics; honoraria from Incyte and Stemline Therapeutics; and research funding from Pfizer and Novartis. RHR reports honoraria from Novartis; consultancy or an advisory role for Novartis; and research funding from Tessa. PAB reports consultancy or an advisory role for Novartis, Kite, a Gilead Company, Jazz, Servier, and Janssen. JK reports consultancy or an advisory role for Kite, a Gilead Company, and Novartis; and employment with Parexel, ICON, and Syneos. CLK, EDZ, and MKR have no relevant financial relationships to disclose. MLH reports consultancy or an advisory role for Sobi Pharmaceuticals; spouse employment with GLAdiator Biosciences and Coagulant Therapeutics Corporation; spouse with leadership role at KaliVir Immunotherapeutics; spouse with stock or other ownership in GLAdiator Biosciences, Coagulant Therapeutics Corporation, and KaliVir Immunotherapeutics; and spouse with patents, royalties, or other intellectual property from GLAdiator Biosciences and Coagulant Therapeutics Corporation. AB reports honoraria from Novartis, Jazz, Servier, and Janssen; consultancy or an advisory role for AstraZeneca, Novartis, Jazz, Celgene, and Servier; research funding from Servier; and travel support from Novartis, Servier, and Jazz. PCS reports employment with and travel support from Kite, a Gilead Company; and stock or other ownership in Gilead Sciences. JR reports former employment with Kite, a Gilead Company; and stock or other ownership in Gilead Sciences. LZ reports employment with Kite, a Gilead Company, and stock or other ownership in AbbVie within the past 2 years. LG reports former employment with and stock or other ownership in Kite, a Gilead Company. RJ reports employment with Vida Ventures and former employment with Kite, a Gilead company; stock or other ownership in Gilead and Amgen; consultancy or an advisory role for Capstan; and travel support from Vida Ventures. RV reports former employment with Kite, a Gilead Company, and stock options with Gilead Sciences. BKM reports employment with, stock, and travel support from Kite, a Gilead Company; and stock options with Lava. DWL reports consultancy or an advisory role for Amgen, Bristol Myers Squibb, and Harpoon Therapeutics; research funding from Kite, a Gilead Company; a patent related to CAR T cells; and spouse employment with and stock or other ownership in Karyopharm Therapeutics.
Contributions
ASW, RV, RJ, and DWL designed the study. ASW, VH, NH, RHR, PAB, JK, MR, CLK, EDZ, MLH, MKR, AB, and DWL enrolled and treated patients and gathered data. PCS, JR, LZ, LG, BKM, RJ, and RV contributed to the verification, analysis, and interpretation of the data. All authors participated in writing the manuscript, had full access to the data, and approved the final submitted version.
Data-sharing statement
Kite is committed to sharing clinical trial data with external medical experts and scientific researchers in the interest of advancing public health, and access can be requested by contacting
Funding
This study was supported by Kite, a Gilead Company. Medical writing support was provided by Nexus Global Group Science, funded by Kite, a Gilead Company.
Acknowledgments
The authors would like to thank the patients who participated in the study and their families, friends, and caregivers; the study staf and health care providers at all the study sites; Christine Wang, PhD, Martha Sensel, PhD, MBA, Daniela van Eickels MD, MPH, and Daniel C. Lee, MD of Kite, a Gilead Company, for supporting the development of the manuscript.
References
- DasGupta RK, Marini BL, Rudoni J, Perissinotti AJ. A review of CD19-targeted immunotherapies for relapsed or refractory acute lymphoblastic leukemia. J Oncol Pharm Pract. 2018; 24(6):453-467. https://doi.org/10.1177/1078155217713363PubMedGoogle Scholar
- Sun W, Malvar J, Sposto R. Outcome of children with multiply relapsed B-cell acute lymphoblastic leukemia: a Therapeutic Advances in Childhood Leukemia & Lymphoma study. Leukemia. 2018; 32(11):2316-2325. https://doi.org/10.1038/s41375-018-0094-0PubMedPubMed CentralGoogle Scholar
- Hunger SP, Mullighan CG. Acute lymphoblastic leukemia in children. N Engl J Med. 2015; 373(16):1541-1552. https://doi.org/10.1056/NEJMra1400972PubMedGoogle Scholar
- Crotta A, Zhang J, Keir C. Survival after stem-cell transplant in pediatric and young-adult patients with relapsed and refractory B-cell acute lymphoblastic leukemia. Curr Med Res Opin. 2018; 34(3):435-440. https://doi.org/10.1080/03007995.2017.1384373PubMedGoogle Scholar
- Cooper SL, Brown PA. Treatment of pediatric acute lymphoblastic leukemia. Pediatr Clin North Am. 2015; 62(1):61-73. https://doi.org/10.1016/j.pcl.2014.09.006PubMedPubMed CentralGoogle Scholar
- Gaynon PS, Qu RP, Chappell RJ. Survival after relapse in childhood acute lymphoblastic leukemia. Cancer. 1998; 82(7):1387-1395. https://doi.org/10.1002/(SICI)1097-0142(19980401)82:7<1387::AID-CNCR24>3.0.CO;2-1Google Scholar
- Malempati S, Gaynon PS, Sather H, La MK Stork LC, Children's Oncology Group. Outcome after relapse among children with standard-risk acute lymphoblastic leukemia: Children's Oncology Group study CCG-1952. J Clin Oncol. 2007; 25(36):5800-5807. https://doi.org/10.1200/JCO.2007.10.7508PubMedGoogle Scholar
- Nguyen K, Devidas M, Cheng SC. Factors influencing survival after relapse from acute lymphoblastic leukemia: a Children's Oncology Group study. Leukemia. 2008; 22(12):2142-2150. https://doi.org/10.1038/leu.2008.251PubMedPubMed CentralGoogle Scholar
- Gassas A, Sung L, Saunders EF, Doyle J. Graft-versus-leukemia effect in hematopoietic stem cell transplantation for pediatric acute lymphoblastic leukemia: significantly lower relapse rate in unrelated transplantations. Bone Marrow Transplant. 2007; 40(10):951-955. https://doi.org/10.1038/sj.bmt.1705853PubMedGoogle Scholar
- Kato M, Horikoshi Y, Okamoto Y. Second allogeneic hematopoietic SCT for relapsed ALL in children. Bone Marrow Transplant. 2012; 47(10):1307-1311. https://doi.org/10.1038/bmt.2012.29PubMedGoogle Scholar
- Locatelli F, Zecca M, Messina C. Improvement over time in outcome for children with acute lymphoblastic leukemia in second remission given hematopoietic stem cell transplantation from unrelated donors. Leukemia. 2002; 16(11):2228-2237. https://doi.org/10.1038/sj.leu.2402690PubMedGoogle Scholar
- Bader P, Salzmann-Manrique E, Balduzzi A. More precisely defining risk peri-HCT in pediatric ALL: pre- vs post-MRD measures, serial positivity, and risk modeling. Blood Adv. 2019; 3(21):3393-3405. https://doi.org/10.1182/bloodadvances.2019000449PubMedPubMed CentralGoogle Scholar
- Pulsipher MA, Carlson C, Langholz B. IgH-V(D)J NGS-MRD measurement pre- and early post-allotransplant defines very low- and very high-risk ALL patients. Blood. 2015; 125(22):3501-3508. https://doi.org/10.1182/blood-2014-12-615757PubMedPubMed CentralGoogle Scholar
- von Stackelberg A, Locatelli F, Zugmaier G. Phase I/phase II study of blinatumomab in pediatric patients with relapsed/refractory acute lymphoblastic leukemia. J Clin Oncol. 2016; 34(36):4381-4389. https://doi.org/10.1200/JCO.2016.67.3301PubMedGoogle Scholar
- BLINCYTO (blinatumomab) [package insert]. 2022. Google Scholar
- BLINCYTO (blinatumomab) [Summary of Product Characteristics]. 2022. Google Scholar
- Locatelli F, Zugmaier G, Mergen N. Blinatumomab in pediatric relapsed/refractory B-cell acute lymphoblastic leukemia: RIALTO expanded access study final analysis. Blood Adv. 2022; 6(3):1004-1014. https://doi.org/10.1182/bloodadvances.2021005579PubMedPubMed CentralGoogle Scholar
- Locatelli F, Zugmaier G, Mergen N. Blinatumomab in pediatric patients with relapsed/refractory acute lymphoblastic leukemia: results of the RIALTO trial, an expanded access study. Blood Cancer J. 2020; 10(7):77. https://doi.org/10.1038/s41408-020-00342-xPubMedPubMed CentralGoogle Scholar
- KYMRIAH (tisagenlecleucel ) [Summary of Product Characteristics]. 2022. Google Scholar
- KYMRIAH (tisagenlecleucel) [package insert]. 2022. Google Scholar
- Maude SL, Laetsch TW, Buechner J. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018; 378(5):439-448. https://doi.org/10.1056/NEJMoa1709866PubMedPubMed CentralGoogle Scholar
- Rives S, Maude SL, Hiramatsu H. Tisagenlecleucel in pediatric and young adult patients (pts) with relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL): final analyses from the ELIANA study. Hemasphere. 2022; 6(Suppl 3):27-28. https://doi.org/10.1097/01.HS9.0000843344.19780.98Google Scholar
- Kochenderfer JN, Feldman SA, Zhao Y. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J Immunother. 2009; 32(7):689-702. https://doi.org/10.1097/CJI.0b013e3181ac6138PubMedPubMed CentralGoogle Scholar
- Kochenderfer JN, Wilson WH, Janik JE. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood. 2010; 116(20):4099-4102. https://doi.org/10.1182/blood-2010-04-281931PubMedPubMed CentralGoogle Scholar
- Lee DW, Kochenderfer JN, Stetler-Stevenson M. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015; 385(9967):517-528. https://doi.org/10.1016/S0140-6736(14)61403-3PubMedPubMed CentralGoogle Scholar
- TECARTUS® (brexucabtagene autoleucel) [prescribing information]. 2021. Google Scholar
- TECARTUS® (autologous anti-CD19-transduced CD3+ cells) [Summary of Product Characteristics]. 2021. Google Scholar
- Shah BD, Ghobadi A, Oluwole OO. KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet. 2021; 398(10299):491-502. https://doi.org/10.1016/S0140-6736(21)01222-8PubMedGoogle Scholar
- Sabatino M, Choi K, Chiruvolu V, Better M. Production of anti-CD19 CAR T cells for ZUMA-3 and -4: phase 1/2 multicenter studies evaluating KTE-C19 in patients with relapsed/refractory B-precursor acute lymphoblastic leukemia (R/R ALL). Blood. 2016; 128(22):1227. https://doi.org/10.1182/blood.V128.22.1227.1227Google Scholar
- Shah BD, Bishop MR, Oluwole OO. KTE-X19 anti-CD19 CAR T-cell therapy in adult relapsed/refractory acute lymphoblastic leukemia: ZUMA-3 phase 1 results. Blood. 2021; 138(1):11-22. https://doi.org/10.1182/blood.2020009098PubMedGoogle Scholar
- Wayne AS, Michel G, Lee DW. ZUMA-4: A phase 1/2 multicenter study of KTE-X19 in pediatric and adolescent patients with relapsed/refractory B cell acute lymphoblastic leukemia or non-Hodgkin lymphoma. Blood. 2020; 136(Suppl 1):42. https://doi.org/10.1182/blood-2020-137233Google Scholar
- Lee DW, Gardner R, Porter DL. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014; 124(2):188-195. https://doi.org/10.1182/blood-2014-05-552729PubMedPubMed CentralGoogle Scholar
- Park JH, Riviere I, Gonen M. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018; 378(5):449-459. https://doi.org/10.1056/NEJMoa1709919PubMedPubMed CentralGoogle Scholar
- Hay KA, Gauthier J, Hirayama AV. Factors associated with durable EFS in adult B-cell ALL patients achieving MRD-negative CR after CD19 CAR T-cell therapy. Blood. 2019; 133(15):1652-1663. https://doi.org/10.1182/blood-2018-11-883710PubMedPubMed CentralGoogle Scholar
- Summers C, Wu QV, Annesley C. Hematopoietic cell transplantation after CD19 chimeric antigen receptor T cell-induced acute lymphoblastic lymphoma remission confers a leukemia-free survival advantage. Transplant Cell Ther. 2022; 28(1):21-29. https://doi.org/10.1016/j.jtct.2021.10.003PubMedGoogle Scholar
- Shah NN, Lee DW, Yates B. Long-term follow-up of CD19-CAR T-cell therapy in children and young adults with B-ALL. J Clin Oncol. 2021; 39(15):1650-1659. https://doi.org/10.1200/JCO.20.02262PubMedPubMed CentralGoogle Scholar
- Fabrizio VA, Kernan NA, Boulad F. Low toxicity and favorable overall survival in relapsed/refractory B-ALL following CAR T cells and CD34-selected T-cell depleted allogeneic hematopoietic cell transplant. Bone Marrow Transplant. 2020; 55(11):2160-2169. https://doi.org/10.1038/s41409-020-0926-1PubMedPubMed CentralGoogle Scholar
- Grupp SA, Maude SL, Rives S. Updated analysis of the efficacy and safety of tisagenlecleucel in pediatric and young adult patients with relapsed/refractory (r/r) acute lymphoblastic leukemia. Blood. 2018; 132(Suppl 1):895. https://doi.org/10.1182/blood-2018-99-112599Google Scholar
- Pasquini MC, Hu ZH, Curran K. Real-world evidence of tisagenlecleucel for pediatric acute lymphoblastic leukemia and non-Hodgkin lymphoma. Blood Adv. 2020; 4(21):5414-5424. https://doi.org/10.1182/bloodadvances.2020003092PubMedPubMed CentralGoogle Scholar
- Kantarjian H, Stein A, Gökbuget N. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med. 2017; 376(9):836-847. https://doi.org/10.1056/NEJMoa1609783PubMedPubMed CentralGoogle Scholar
- Gardner RA, Finney O, Annesley C. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood. 2017; 129(25):3322-3331. https://doi.org/10.1182/blood-2017-02-769208PubMedPubMed CentralGoogle Scholar
- Myers RM, Taraseviciute A, Steinberg SM. Blinatumomab nonresponse and high-disease burden are associated with inferior outcomes after CD19-CAR for B-ALL. J Clin Oncol. 2022; 40(9):932-944. https://doi.org/10.1200/JCO.21.01405PubMedPubMed CentralGoogle Scholar
- Pillai V, Muralidharan K, Meng W. CAR T-cell therapy is effective for CD19-dim B-lymphoblastic leukemia but is impacted by prior blinatumomab therapy. Blood Adv. 2019; 3(22):3539-3549. https://doi.org/10.1182/bloodadvances.2019000692PubMedPubMed CentralGoogle Scholar
- Tyagarajan S, Spencer T, Smith J. Optimizing CAR-T cell manufacturing processes during pivotal clinical trials. Mol Ther Methods Clin Dev. 2019; 16:136-144. https://doi.org/10.1016/j.omtm.2019.11.018PubMedPubMed CentralGoogle Scholar
- Xu Y, Zhang M, Ramos CA. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood. 2014; 123(24):3750-3759. https://doi.org/10.1182/blood-2014-01-552174PubMedPubMed CentralGoogle Scholar
- Rheingold SR, Ji L, Xu X. Prognostic factors for survival after relapsed acute lymphoblastic leukemia (ALL): a Children’s Oncology Group (COG) study. J Clin Oncol. 2019; 37(15_suppl):10008. https://doi.org/10.1200/JCO.2019.37.15_suppl.10008Google Scholar
- Oskarsson T, Söderhäll S, Arvidson J. Relapsed childhood acute lymphoblastic leukemia in the Nordic countries: prognostic factors, treatment and outcome. Haematologica. 2016; 101(1):68-76. https://doi.org/10.3324/haematol.2015.131680PubMedPubMed CentralGoogle Scholar
- Schrappe M, Hunger SP, Pui CH. Outcomes after induction failure in childhood acute lymphoblastic leukemia. N Engl J Med. 2012; 366(15):1371-1381. https://doi.org/10.1056/NEJMoa1110169PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.