Effective bridging strategies prior to infusion with tisagenlecleucel results in high response rates and long-term remission in relapsed/refractory large B-cell lymphoma: findings from a German monocentric study

Baseline characteristics

Patients’ characteristics are outlined in Table 1. Median age was 63 years (range 34–74). Twelve patients (41.4%) were female. Approximately half of the patients (48%) had a high-intermediate or high international prognostic index (IPI) at start of LD. An Eastern Cooperative Oncology Group performance status (ECOG PS) of ≥ 2 was observed in 20.7% of the patients. Prior to leukapheresis, 24 patients (83%) had received three or more lines of systemic therapy, and prior autologous stem cell transplantation (auto-SCT) had been performed in 41% (n = 12) of patients. Table S1 presents the patient characteristics of those who received bridging therapy. Specifically, it delineates patients who received radiation therapy as part of bridging (bRT) and those who did not (non-RT bridging). Notably, among patients who received radiation therapy, 82% were refractory to previous chemotherapy. Among the non-radiation therapy patients, this percentage was 50%.

Table 1 Patient characteristicsTreatment characteristics including bridging strategies

A comprehensive description of the lines of chemotherapy prior to leukapheresis, including response, is provided in table S2. At the time of leukapheresis, more than half of the patients (n = 17, 59%) were refractory to the most recent treatment, and 66% (n = 19) exhibited refractoriness to any line of therapy before leukapheresis. Prior to LD, bridging therapy was administered in 85.2% (n = 25) of patients. Bridging modalities included classical chemoimmunotherapy (52%), immunotherapy (4%), RT only (12%) and combined radio- and immunotherapy (32%) (Table 2). A detailed description of the RT parameters is provided in Table 3.

Table 2 Bridging characteristics and efficacyTable 3 Treatment characteristics and response of patients with bridging radiotherapyRadiotherapy as bridging modality

Modern state-of-the-art radiation techniques including intensity-modulated radiation therapy (IMRT), helical tomotherapy or Rapid Arc, volumetric modulated arc therapy (VMAT) with image-guidance (IGRT) were applied. The majority of patients (64%, 7/11) underwent moderately hypofractionated RT with single doses of 2.5 Gy, reaching a cumulative dose of 30.0 Gy within a time span of 2.5 weeks. The median EQD2α/β = 3 for all patients was 33.0 Gy (range, 33.0–36.0) and EQD2α/β = 10 31.3 Gy (range, 31.3 – 36.0).

The radiation field encompassed nodal involvement in 8 out of 11 patients (73%), extranodal manifestation in 3 out of 11 patients (27%), and both in one case (9%). In 7 out of 11 patients (64%), the planning target volume (PTV) comprised bulky lymph node lesion with a diameter of ≥ 5 cm.

In all 11 patients, involved site radiation was administered. The gross tumor volume (GTV) comprised solely the affected lymph node or extranodal involvement. When deemed appropriate, an internal target volume (ITV) was delineated. The safety margin was determined based on institutional positioning uncertainties and irradiation techniques (PTV, median = 1.0 cm, range, 0.2–2.0). The median total tumor volume (GTV) was 319 cc (range, 8–2049), while the median total PTV was 703 cc (range 17–3799) (Table 3). Within this cohort, the para-aortic region was the most frequently irradiated site (6/11, 55%) (Table S3). One patient had received prior irradiation at the same location (right lower leg).

Response and toxicity to radiotherapy bridging

Prior to CAR-T infusion, of 25 patients who received bridging therapy, 11 (44%) received bRT. bRT was applied mainly in patients who were refractory to prior chemotherapy (10/11), had not received radiation therapy before (10/11), and did not have 3 or more sites of lymphoma disease at the time of leukapheresis (9/11). Bridging response prior to CAR-T infusion was assessed by PET-CT and response data are provided in Table 3. In 5 out of 11 patients (45%), a transition from refractory to responsive disease was attained through bRT. Sites of progression following bRT were observed to be located outside the radiation field in all patients. Importantly, a clinically significant reduction in standardized uptake value (SUV) was achieved in 7 out of 9 evaluable patients through bRT. The median reduction in post-bRT PET-CT compared to pre-bRT PET-CT was − 7.5 (range, − 49 to + 2) or − 42% (range − 96% to + 31%) of the initial SUV (Table 3). The local response rate after bRT was notably high, with an ORR inside the radiation field of 82% (9/11). Specifically, all patients (8/8) who received moderately hypofractionated bRT in single doses of 2.5 to 3.0 Gy demonstrated a local response as defined in decrease in SUV.

Toxicity assessments following bRT demonstrated acute toxicity according to CTCAE grades 1 or 2 in 8 out of 11 patients (73%) (Table 4). No grade 3 toxicities occurred.

Table 4 Radiation induced toxicities

No significant difference in survival was noted when bRT and systemic bridging therapy was compared (data not shown).

Response to tisa-cel infusion

ORR at one month following tisa-cel infusion was 83% (24 patients), with 55% achieving a CR and 28% a PR (Table 5). Early death within 3 months after infusion was reported in four patients, all attributed to progressive disease. Among the responding patients (CR or PR) assessed at month 3 (n = 19), only one patient experienced relapse after achieving a CR at month 3 during long-term follow-up. With a median follow-up of 11.1 months for PFS and of 17.9 months for OS, Kaplan–Meier-estimated rates for the whole cohort at 12 months were 60% for PFS and 74% for OS, respectively (Table 5). When comparing PFS (Fig. 1A) and OS (Fig. 1B) of patients who received bridging therapy with or without radiation, no significant differences were observed. Using log-rank tests, significant predictors of an adverse PFS were identified by univariate analysis (refractory disease status prior to leukapheresis, ECOG PS ≥ 2, non-response to bridging and non-response at month 1 PET-CT) (Fig. 2A, B, C and D). No significant impact on PFS was observed for IPI ≥ 3 (p = 0.21) and extranodal disease (p = 0.21). An inferior OS was statistically associated with an IPI ≥ 3, PS ≥ 2, extranodal disease and non-response at month 1 PET-CT (Fig. 3A–D). No significant impact on OS was seen for refractory disease prior to leukapheresis (p = 0.22) and non-response to bridging (p = 0.09). No impact on PFS or OS was noted for LDH levels at LD (p = 0.09 or p = 0.11, respectively) and prior auto-HCT (p = 0.3 or p = 0.99, respectively).

Table 5 Outcome and survival data (total cohort)Fig. 1figure 1

Kaplan–Meier estimates for progression-free survival (A) and overall survival (B) of patients who received bridging therapy with radiation (bRT) and without radiation (non-bRT)

Fig. 2figure 2

Progression-free survival according to response to chemotherapy prior to apheresis (A), ECOG PS (B), response to bridging (C), PET-CT 1 month after CAR-T (D)

Fig. 3figure 3

Overall survival according to IPI score at CAR-T infusion (A), ECOG PS (B), presence of extranodal involvement (C), PET-CT 1 month after CAR-T (D)

Toxicity to tisa-cel infusion

CRS of any grade was observed in 27 patients (93%) patients, with CRS grade ≥ 3 in 10 patients (34%) (Table 6). In the univariate analysis, patients who had a response at month 3 showed a significantly higher incidence of Grade ≥ 3 CRS compared to those who did not respond (p = 0.044). No difference was observed when a multivariate analysis was performed (data not shown). Grade 1 ICANS was reported in 5 patients (17%), while no higher grade was identified.

Table 6 Toxicities from CAR-T therapy

Hematological toxicity was reported in 21 patients (72%) overall, primarily characterized by severe neutropenia (absolute neutrophil count, ANC < 500 cells/µl) observed in all patients who experienced hematologic toxicity (n = 21) (Table 7). According to the clinical phenotypes of recovery defined by Rejeski et al., 6 patients (29%) showed rapid neutrophil recovery without a second dip, 5 patients (24%) experienced intermittent recovery followed by a second dip below ANC < 0.5/nl after day 21, 10 patients (47%) exhibited continuous severe neutropenia beyond day 21. Prolonged neutropenia persisted in 19 patients (66%), with a median time to neutrophil recovery of 58 days (range, 22–774). It persisted in 13 patients (45%) at month one, 9 patients (30%) at month 3, and 4 patients (20%) at month 6. Two patients continued to experience prolonged neutropenia induced beyond months 9 and 12, respectively. Prolonged severe thrombocytopenia (< 20/nL or requiring transfusion for ≥ 21 days) was observed in 10 patients (34%), with a median time to platelet recovery of 74 days (range, 39–790). Prolonged severe anemia (hemoglobin < 8 g/dL or requiring transfusion) was noted in 13 patients (45%), with a median time to hemoglobin recovery of 56 days (range, 23–776).

Table 7 CAR-T cell mediated hematotoxicity

Median hospitalization time following CAR-T cell therapy was 22 (range, 18–39) days. No non-relapse deaths occurred during the follow-up period.

留言 (0)

沒有登入
gif