Cytopenia after CAR‑T cell therapy: Analysis of 63 patients with relapsed and refractory B‑cell non‑Hodgkin lymphoma

  • Authors:
    • Tingting Qiu
    • Luan Hu
    • Yongtian Zhang
    • Ying Wang
    • Sha Ma
    • Depeng Li
    • Zhenyu Li
    • Kailin Xu
  • View Affiliations

  • Published online on: June 22, 2023     https://doi.org/10.3892/ol.2023.13924
  • Article Number: 338
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to determine the clinical characteristics of cytopenia in patients with relapsed and refractory B‑cell non‑Hodgkin lymphoma (B‑NHL) who were treated with chimeric antigen receptor T‑cell (CAR‑T) therapy. Thus, a total of 63 patients with relapsed and refractory B‑NHL who underwent CAR‑T therapy between March 2017 and October 2021 were retrospectively selected for analysis. Neutropenia, anemia and thrombocytopenia at grade ≥3 occurred in 48 (76.19%), 16 (25.39%) and 15 (23.80%) cases, respectively. The results of a multivariate analysis demonstrated that the baseline absolute neutrophil count (ANC) and hemoglobin concentration were independent risk factors for grade ≥3 cytopenia. A total of 3 patients died early and were therefore excluded from the present study. Furthermore, cell recovery was examined at day +28 after infusion; 21 patients (35%) did not recover from cytopenia and 39 patients (65%) recovered. A multivariate analysis demonstrated that the baseline ANC <2.29x109/l, baseline hemoglobin <114.50 g/l and baseline IL‑6 >21.43 pg/l were independent risk factors affecting hemocyte recovery. In conclusion, patients with relapsed and refractory B‑NHL exhibited an increased incidence of grade ≥3 hematologic toxicity following CAR‑T cell therapy, while baseline blood cell and IL‑6 levels are independent risk factors for hemocyte recovery.

Introduction

Chimeric antigen receptor T-cells (CAR-T) recognize tumor antigen-specific receptors through gene recombination (1). CAR-T therapy produced encouraging results in relapsed and refractory hematological malignancies, including B-cell acute lymphoblastic leukemia (B-ALL) (2,3), non-Hodgkin lymphoma (NHL) (4,5) and multiple myeloma (MM) (6,7). Although clinical trials of CAR-T therapy have demonstrated marked efficacy, patients may experience associated adverse events, including cytokine release syndrome (CRS), neurological toxicity (NT) and macrophage activation syndrome. Of note, cytopenia is common in patients following CAR-T therapy and numerous patients experience cytopenia prior to CAR-T cell infusion. Furthermore, blood cells are further reduced following CAR-T therapy and patients are prone to infection, bleeding and other complications that may be life-threatening. At present, large-scale reports of cytopenia in patients with B-cell NHL (B-NHL) who underwent CAR-T therapy are lacking. Thus, the present study aimed to determine potential changes in cytopenia in patients with relapsed and refractory B-NHL following CAR-T therapy and provide a theoretical basis for early intervention in patients with CAR-T-associated complications.

Materials and methods

Patients

A retrospective analysis was performed using patients with relapsed and refractory B-NHL. All patients had been admitted to the Hematology Department of The Affiliated Hospital of Xuzhou Medical University (Xuzhou, China) between March 2017 and October 2021 and had complete clinical data. All patients were enrolled in the CAR-T cell clinical trials (NCT02782351, NCT02794961, NCT02903810 and NCT03207178). Patients who met the following criteria were included in the present study: i) A confirmed diagnosis of relapsed and refractory B-NHL; ii) an expected survival of >12 weeks; iii) provided written informed consent. Patients were excluded for the following reasons: i) Other major systemic disease, such as severe respiratory failure, heart function grade III to IV or uremic phase; ii) complications with severe and uncontrollable present infection, severe coagulation dysfunction, disseminated intravascular coagulation or severe thrombosis; iii) active viral hepatitis B or C; iv) autoimmune diseases or other chronic diseases that depend on immunosuppressants or hormone therapy; v) psychological diseases that do not allow for coordination with treatment or efficacy evaluations; and vi) pregnancy or lactation. The present study was approved by the Medical Ethics Committee of the Affiliated Hospital of Xuzhou Medical University (Xuzhou, China).

Manufacture of CAR-T cells

Appropriate amounts of mononuclear cells (usually 1×109) were sorted from the peripheral blood of patients and prepared by a third-party company in vitro. Genetic engineering technology was used to form second generation CAR-T cells expressing target antibody fragments and the transmembrane-activated signaling domain of the T-cell receptor, intracellular linking 4-1BB or CD28 co-stimulatory molecules were fused (8). Strict quality control and aseptic technique were performed during CAR-T cell culture.

CAR-T therapy and monitoring

CAR-T cell infusion was performed on Day 0 and each patient was administered fludarabine (35 mg/m2 on day −5 to −3) and cyclophosphamide (750 mg/m2 on day −5) as pre-treatment. Enrolled patients were transfused with a total of 1×106/kg CAR-T cells and vital signs and patient condition were closely observed during infusion. Specific factors were monitored from enrollment to the end of follow-up after CAR-T cell therapy, including: i) Vital signs, such as body temperature, heart rate and blood pressure, examined 1–2 times per day prior to CAR-T cell infusion, following CAR-T cell infusion and at each follow-up after discharge; and ii) routine blood examination, including liver and kidney function, C-reactive protein (CRP), ferritin and IL-6. CRS, NT grading and efficacy evaluation were performed according to a previously published procedure (9). All factors were examined prior to pre-treatment and CAR-T cell infusion, and periodically following CAR-T cell infusion. Follow-up was completed in both inpatient and outpatient settings.

Cytopenia classification and hematologic recovery

Cytopenia was graded according to the Common Terminology Criteria for Adverse Events (version, 4.0) (10). Grade 3 or higher cytopenia was defined as absolute neutrophil count (ANC) <1.0×109/l, hemoglobin <80 g/l and platelet count <50×109/l. Hematologic recovery was defined as ANC ≥1.0×109/l, hemoglobin ≥80 g/l and platelet count ≥50×109/l for three consecutive days without granulocyte-stimulating factors or blood transfusions. According to the hematologic recovery observed on day +28, patients were divided into recovered and unrecovered groups. Of note, any series of blood cells that did not recover were classified as the unrecovered group.

Statistical analysis

Data were analyzed using SPSS (version, 26.0; IBM Corp.). The single-sample Wilcoxon signed-rank test was used to test whether a normal distribution was present. Continuous variables of non-normal distribution were described using the median (range) and the Mann-Whitney U-test was used for intergroup comparisons. Categorical variables were evaluated using the Chi-squared test or Fisher's exact test. A logistic regression model was used to analyze univariate and multivariate influencing factors. Receiver operating characteristic (ROC) curve analysis was used to obtain the optimal cut-off values. P<0.05 was considered to indicate a statistically significant difference.

Results

Patient characteristics

According to the inclusion and exclusion criteria, a total of 63 patients with relapsed and refractory B-NHL were enrolled in the present study, with a median age of 46 years (range, 21–72 years), including 45 males (71.42%) and 18 females (28.57%), with a male-to-female ratio of 2.5:1. Of note, 47 cases (74.60%) were primary type diffuse large B-cell lymphoma (DLBCL), including 4 cases of follicular cell lymphoma. The present study also included 4 cases (6.34%) of primary central DLBCL, 2 cases (3.17%) of primary mediastinal DLBCL, 5 cases (7.93%) of follicular cell lymphoma, 4 cases (6.34%) of B-cell lymphoma leukemia and 1 case (1.58%) of mantle cell lymphoma. In addition to 4 patients with B-cell lymphoma leukemia, 4 patients exhibited bone marrow involvement. The median number of prior chemotherapies was 8 (range, 3-26), and 4 patients (6.34%) received autologous hematopoietic stem cell transplantation (HSCT). Types of CAR-T infusion included 11 cases (17.46%) of CD19, 9 cases (14.28%) of CD20, 23 cases (36.50%) of CD19-CD20, 19 cases (30.15%) of CD19-CD22 and 1 case (1.58%) of CD20-NK. Bridging therapy was permitted for the patients before CAR-T treatment. Only nine patients received bridging therapy, mainly low-dose chemotherapy including CHOP (cyclophosphamide, doxorubicin, vincristine and prednisolone), reduced ESHAP (etoposide, cisplatin, cytarabine and methylprednisolone), and one patient received lenalidomide and zanubrutinib. A total of 2 patients who were administered anti-CD19 and CD22 CAR cells received CD20 monoclonal antibody in addition. Following CAR-T cell infusion, 44 (69.84%) patients experienced CRS. Of these, 4 patients (6.34%) developed grade ≥3 CRS and 40 patients (63.49%) developed grade 1–2 CRS. In addition, 3 patients (4.76%) developed grade 3 NT and presented with recurrent seizures, and a total of 7 patients (11.11%) developed grade 1 NT and presented with headaches. Efficacy was evaluated 30 days following infusion and 3 patients who died early were excluded. Results of the present study demonstrated that the overall response rate was 65%, including 11 cases (18.3%) with complete response, 28 cases (46.7%) with partial response, 12 cases (20%) with stable disease and 9 cases (15%) with progressive disease (Table SI).

Occurrence of cytopenia

A total of 32 out of 63 patients (50.79%) with B-NHL experienced at least one series of baseline cytopenia prior to CAR-T therapy, including 18 patients (28.57%) with a median ANC of 3.15×109/l (0.61-11.35 ×109/l), 22 patients (34.92%) with a median hemoglobin concentration of 114 g/l (51–165 g/l) and 9 patients (14.28%) with a median platelet count of 178×109/l (43–447×109/l). Following CAR-T therapy, 61 patients (96.82%) experienced neutropenia, 46 patients (73.01%) experienced anemia and 17 patients (26.98%) experienced thrombocytopenia. Of these, 48 patients (76.19%) experienced grade ≥3 neutropenia, 16 patients (25.39%) experienced grade ≥3 anemia and 15 patients (23.80%) experienced grade ≥3 thrombocytopenia. The median lowest values of ANC, hemoglobin concentration and platelet count were 0.64×109/l (0.02-2.31×109/l), 100 g/l (32–137 g/l) and 121×109/l (2–307×109/l), respectively. Patients achieved the lowest ANC at a median time of day 5 (range, 0–27 days), the lowest hemoglobin at a median time of day 8 (range, −5–28 days) and the lowest platelet count at a median time of day 10 (range, 0–28 days) (data not shown).

Factors associated with grade ≥3 cytopenia

To determine the risk factors associated with grade ≥3 cytopenia following CAR-T therapy, a univariate risk factor analysis was carried out. The results indicated that grade ≥3 cytopenia was not associated with sex, age, different CAR-T targets, times of prior chemotherapies, prior autologous HSCT, bone marrow involvement, CRS or NT. Of note, grade ≥3 cytopenia was associated with the Eastern Cooperative Oncology Group (ECOG) score, baseline ANC and hemoglobin concentration, and baseline ferritin and IL-6 levels. Grade ≥3 neutropenia was significantly associated with the ECOG score, baseline ANC and hemoglobin, and baseline IL-6 and ferritin. Anemia grade >3 was significantly associated with the ECOG score, baseline hemoglobin and platelet count, baseline IL-6 and ferritin, and baseline CRP. Thrombocytopenia grade >3 was significantly associated with the ECOG score, baseline hemoglobin and platelet count, baseline IL-6 and ferritin, baseline CRP and the severity of CRS (Table SII, Table SIII, Table IV). The incidence of thrombocytopenia in patients with grade 1–2 CRS was 32.50% (13 cases) and this was significantly higher than that in patients without CRS (5.26%; 1 case). The incidence of thrombocytopenia in patients with grade ≥3 CRS was 25% (1 case) and this was higher than that in patients without CRS (5.26%), but the difference between groups was not statistically significant (Table SV). A binary logistic regression model was applied for multivariate risk factor analysis and the results indicated that only baseline ANC and hemoglobin levels were independently associated with grade ≥3 cytopenia (Table I).

Table I.

Analysis of risk factors for cytopenia above grade 3.

Table I.

Analysis of risk factors for cytopenia above grade 3.

Multivariate analysis
Characteristic [normal ranges]Cytopenia groupNo cytopenia groupUnivariate analysis P
OR (95% CI)P-value
Gender 0.195
  Male32 (66.66)13 (86.67)
  Female16 (33.34)2 (13.33)
Age, years47 (21–72)45 (24–70)0.600
ECOG score2 (0–3)1 (0–3)0.0261.935 (0.761–4.919)0.166
Number of prior chemotherapies, times8 (3–24)10 (6–26)0.147
Prior autologous HSCT 1.000
  Yes3 (6.25)1 (6.67)
  No45 (93.75)14 (93.33)
Bone marrow involvement 0.667
  Yes7 (14.5)1 (6)
  No41 (85.5)14 (94)
Targets of CAR-T cell 0.503
  CD198 (16.67)3 (20)
  CD207 (14.58)2 (13.33)
  CD19-CD2017 (35.41)6 (40)
  CD19-CD2216 (33.33)3 (20)
  CD19-NK0 (0)1 (6.67)
CRS 0.380
  None16 (33.33)3 (20)
  Grade 1-228 (58.33)12 (80)
  Grade ≥34 (8.33)0 (0)
NT 1.000
  None40 (83.33)13 (86.67)
  Grade 1-25 (10.41)2 (13.33)
  Grade ≥33 (6.25)0 (0)
Baseline ANC, ×109/l [1.80–6.30]2.86 (0.61–11.35)4.31 (1.55–7.01)0.0050.504 (0.297–0.865)0.011
Baseline hemoglobin, g/l [115–150]106 (51–149)127 (103–165)<0.0010.947 (0.899–0.998)0.041
Baseline platelet count, ×109/l [125–350]157 (23–359)212 (127–447)0.158
Baseline CRP, mg/l [0.0–5.0]18.25 (0.40–200)5.8 (0.4–114.70)0.113
Baseline ferritin, ng/ml [30–400]613.60 (20–25822)324.1 (33.10–830)0.0141.001 (0.998–1.004)0.362
Baseline IL-6, pg/l [0.01–7.0]12.31 (1.50–882)4.24 (1.50–25)0.004  1.06 (0.967–1.162)0.211

[i] Values are expressed as n (%) or the median (range). CRP, C-reactive protein; CAR-T, chimeric antigen receptor T-cell; ANC, absolute neutrophil count; CRS, cytokine release syndrome; NT, neurological toxicity; NK, natural killer; OR, odds ratio; HSCT, hematopoietic stem cell transplantation; ECOG, Eastern Cooperative Oncology Group.

Analysis of factors associated with hematologic recovery

A total of 60 patients were included in the follow-up at 28 days following CAR-T cell infusion, following the exclusion of 3 patients who died early. Patients were divided into recovered and unrecovered groups, according to whether the peripheral blood cell count was recovered. Among them, 21 patients (35%) exhibited no hematologic recovery and 39 patients (65%) exhibited hematologic recovery. Day +28 hemocyte recovery was analyzed in all patients and the results suggested that patients in the recovered group exhibited higher baseline blood cell counts, baseline IL-6, ferritin and baseline CRP levels. Of note, the peak levels of IL-6, ferritin and CRP also affected blood cell recovery. Univariate analysis demonstrated that sex, age, disease stage, different CAR-T targets, number of prior chemotherapies, autologous HSCT, bone marrow involvement, CRS and NT were not associated with hemocyte recovery at day +28 (Table II). ROC curve analysis was used to obtain the optimal cut-off values of baseline ANC, hemoglobin, baseline IL-6, baseline ferritin and peak ferritin (area under the curve, 0.7-0.9) (Fig. SI). Cut-off values were used to convert continuous variables into categorical variables. Univariate analysis indicated that the factors affecting hemocyte recovery were baseline ANC, baseline hemoglobin, baseline IL-6, baseline ferritin and peak values of ferritin. Furthermore, the results of the multivariate analysis suggested that baseline ANC <2.29×109/l, baseline hemoglobin <114.5 g/l and baseline IL-6 >21.43 pg/l were independent influencing factors on hematologic recovery (Table III).

Table II.

Clinical characteristics of the hematologic recovery group and the unrecovered group at day +28.

Table II.

Clinical characteristics of the hematologic recovery group and the unrecovered group at day +28.

CharacteristicRecovery groupUnrecovered groupP-value
Gender 1.0
  Male27 (69.23)15 (71.42)
  Female12 (30.76)6 (28.57)
Age, years47 (23–72)42 (21–68)0.172
ECOG score1 (0–3)2 (0–3)0.349
Number of prior chemotherapies, times8 (3–26)9 (3–25)0.493
Prior autologous HSCT 0.278
  Yes1 (2.56)2 (9.52)
  No38 (97.43)19 (90.47)
Bone marrow involvement 0.114
  Yes3 (7.7)5 (23.8)
  No36 (92.3)16 (76.2)
Targets of CAR-T cells 0.599
  CD198 (20.5)2 (9.5)
  CD205 (12.8)2 (9.5)
  CD19-CD2015 (38.5)8 (38.1)
  CD19-CD2210 (25.6)9 (42.9)
  CD19-NK1 (2.6)0 (0)
CRS 0.796
  None12 (30.77)6 (28.57)
  Grade 1-225 (64.10)15 (71.43)
  Grade ≥32 (5.12)0 (0)
NT 0.193
  None34 (87.18)17 (80.95)
  Grade 1-25 (12.82)2 (9.52)
  Grade ≥30 (0)2 (9.52)
Baseline ANC, ×109/l3.61 (0.61-7.01)1.77 (0.71-11.35)0.004
Baseline hemoglobin, g/l119 (77–165)90 (51–147)<0.001
Baseline platelet count, ×109/l200 (54–447)151 (23–321)0.016
Baseline CRP, mg/l7.2 (0.40-128.90)28.5 (1–200)0.022
Baseline ferritin, ng/ml426 (20–3,323)774 (92.13–25,822)0.012
Baseline IL-6, pg/l5.88 (1.50–57)22.25 (1.50–149.10)0.003
Peak CRP, mg/l63.30 (1.30–200)91.3 (1.50–1,636)0.048
Peak ferritin, ng/ml542.40 (25.12–7,000)1,396 (122.60–25,822)0.003
Peak IL-6, pg/l21.35 (4.06–102)47.35 (5.02–1,453)0.040

[i] Values are expressed as n (%) or the median (range). CRP, C-reactive protein; CAR-T, chimeric antigen receptor T-cell; ANC, absolute neutrophil count; CRS, cytokine release syndrome; NT, neurological toxicity; NK, natural killer; HSCT, hematopoietic stem cell transplantation; ECOG, Eastern Cooperative Oncology Group.

Table III.

Univariate and multivariate analysis of hematologic recovery at day +28.

Table III.

Univariate and multivariate analysis of hematologic recovery at day +28.

Univariate analysisMultivariate analysis


VariableOR (95%CI)P-valueOR (95%CI)P-value
Baseline ANC <2.29×109/l     11 (3.130–38.660)<0.00114.834 (2.391–92.051)0.004
Baseline hemoglobin <114.50 g/l8.500 (2.371–30.466)   0.001  7.963 (1.323–47.921)0.023
Baseline IL-6 >21.43 pg/l0.107 (0.028–0.410)   0.0010.091 (0.100–0.802)0.031
Baseline ferritin >558.65 ng/ml0.250 (0.081–0.770)   0.0160.966 (0.098–9.525)0.976
Peak ferritin >1195 ng/ml0.110 (0.031–0.395)   0.0010.456 (0.036–5.851)0.546

[i] ANC, absolute neutrophil count; OR, odds ratio.

Discussion

As a novel cellular immunotherapy, CAR-T cells have achieved high response and remission rates in patients with relapsed and refractory hematological malignancies, including B-ALL, B-NHL and MM. Of note, numerous clinical guidelines recommend CAR-T cell therapy. Common adverse events associated with CAR-T therapy, including CRS and NT, are detailed in the previous literature (11,12). However, studies exploring potential factors associated with hematological toxicity, particularly in relapsed and refractory B-NHL, are lacking. The present study retrospectively analyzed the occurrence, recovery and risk factors of cytopenia in patients with relapsed and refractory B-NHL following CAR-T therapy.

In the phase II study ZUMA-1 (11), the incidence of grade >3 neutropenia, anemia and thrombocytopenia following CAR-T cell infusion was 78, 43 and 38%, respectively, in 111 patients with relapsed and refractory B-NHL. Fried et al (13) reported that the incidence of grade ≥3 neutropenia and thrombocytopenia was 72 and 28% in 38 patients with relapsed and refractory B-NHL and ALL, respectively, following CD19 CAR-T cell infusion. The present study suggested that patients with relapsed and refractory B-NHL experienced cytopenia following CAR-T cell infusion and the incidence of grade ≥3 neutropenia, anemia and thrombocytopenia was 76.19, 25.3 and 23.80%, respectively. The incidence of grade ≥3 neutropenia was comparable to the incidence observed in previous studies, while the incidence of grade ≥3 anemia and thrombocytopenia was lower. Cytopenia following CAR-T cell infusion is common and patients exhibit the highest incidence of grade ≥3 neutropenia. Thus, clinicians should closely monitor cytopenia following CAR-T cell infusion and treatment should be used to prevent infection, bleeding or anemia.

Results of previous studies suggested that cytopenia following the infusion of CAR-T cells is biphasic or triphasic, with the first phase occurring within 3–4 weeks. Such early cytopenia may be attributed to lymphodepletion regimens, bridging chemotherapy or radiotherapy carried out prior to CAR-T cell infusion, or severe CRS (14,15). Results of the present study demonstrated that the lowest time to cytopenia occurrence was 5–10 days following infusion. Furthermore, associated risk factors were analyzed in the present study and the results indicated that grade ≥3 cytopenia was not associated with primary disease stage, the number of prior chemotherapies, prior autologous HSCT, CRS or NT. However, grade ≥3 cytopenia was associated with the ECOG score, baseline blood cell levels and baseline inflammatory factor levels. The present study also determined the risk factors associated with grade ≥3 neutropenia, anemia and thrombocytopenia and the results demonstrated that the severity of CRS was associated with grade ≥3 thrombocytopenia. Through univariate analysis, Fried et al (13) revealed that the only risk factor associated with grade ≥3 anemia and thrombocytopenia was autologous HSCT within 1 year prior to CAR-T cell infusion. Patients with grade ≥2 CRS and low baseline blood cell levels exhibited a higher rate of grade ≥3 thrombocytopenia; however, these results were not statistically significant. Although the potential association between severe CRS and grade ≥3 cytopenia was not consistently reported among studies (13,14), close monitoring of patients and timely intervention are essential for clinicians.

To date, research has focused on the duration and mechanisms of cytopenia and results of previous studies demonstrated that prolonged cytopenia is associated with prognosis. However, the definition of prolonged cytopenia differed among studies. In the majority of previous studies (14,16), day +28 to day +42 following CAR-T cell infusion was usually set as a time node; however, other studies extended the time node to day +80 or day +90. The TRANSCEND NHL-001 trial (16) included 269 patients with relapsed and refractory B-NHL, 37% of whom experienced persistent cytopenia at day +29 following CAR-T cell infusion. Hockings et al (14) analyzed 39 patients with relapsed and refractory B-NHL and highlighted that 43% experienced persistent cytopenia at day +28 following CAR-T cell infusion. Furthermore, Nagle et al (17) reported that the 1-year overall survival (OS) rate of 31 patients with relapsed and refractory DLBCL who had experienced persistent cytopenia on day +30 following CAR-T cell infusion was 36%, while the 1-year OS rate of patients without prolonged cytopenia was 81% and this difference was statistically significant. Results of the present study demonstrated that 35% of patients with relapsed and refractory B-NHL experienced prolonged cytopenia at day +28 following CAR-T cell infusion. Collectively, these results demonstrated that cytopenia may not be a result of pre-treatment-mediated myelotoxicity. However, the duration of clinical observations of persistent cytopenia following CAR-T cell infusion differed among the previously studies.

Risk factors reported for prolonged cytopenia associated with CAR-T cell therapy are varied, including a higher number of previous treatment lines, an increased median age, bone marrow reserves with baseline cell defects, severity of CRS, baseline and peak levels of various inflammatory cytokines and potential clonal hematopoiesis. Of note, research has predominantly focused on the severity of CRS, baseline bone marrow reserve, and baseline and peak levels of inflammatory cytokines. Strati et al (18) analyzed the risk factors associated with cytopenia at day +30 in 31 patients with relapsed and refractory B-NHL enrolled in the ZUMA-1 and ZUMA-9 trials. Of note, the results of this previous study demonstrated no association between the incidence of CRS and grade >3 NT. The present study did not indicate any association between severe CRS or NT with prolonged cytopenia, either; however, baseline blood count levels, baseline IL-6, ferritin, baseline CRP levels and the corresponding peak values were associated with prolonged cytopenia. In a multicenter retrospective study, Rejeski et al (19) analyzed hematological toxicity following CAR-T therapy in 258 patients with relapsed and refractory B-NHL, using day +60 as the study endpoint. The results of this previous study indicated that the incidence and severity of CRS and NT, the peak levels of cytokines and the number of previous treatments were not associated with prolonged cytopenia, while baseline thrombocytopenia and methemoglobinemia were risk factors. These results suggested that bone marrow reserves and inflammatory state prior to CAR-T therapy are key features of prolonged cytopenia. Of note, bone marrow reserves may be impacted by local inflammatory factors, such as IL-6 signaling involved in hematopoietic stem cell development.

The present study is limited by being a retrospective single-center study with incomplete data, including a lack of the expansion of CAR-T cells. Furthermore, only the changes and recovery of blood cells at day +28 were analyzed, and they were not observed for a longer period of time. In addition, the relationship between cytopenia and clinical prognosis was not analysed. Considering the influence of basic cytokines on blood cell recovery, the mechanism of their influence may be further explored and the prediction model may be established.

Collectively, the results of the present study suggested that prolonged cytopenia occurred in patients following CAR-T cell therapy and patients also experienced a high incidence of CAR-T-associated hematologic toxicity. Furthermore, the risk factors associated with prolonged cytopenia were bone marrow reserves and baseline levels of inflammatory factors.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

The authors would like to thank Professor Wei Sang, Professor Jiang Cao, Professor Zhi-Ling Yan and Professor Feng Zhu (all from the Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China) for their clinical assistance to patients included in this study.

Funding

This work was supported by grants from the National Nature Science Foundation of China (grant nos. 81400129, 81800124 and 81700177).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

TQ designed the study and wrote the manuscript. LH and YZ primarily wrote the manuscript and prepared the tables. LH, YZ, YW and SM made substantial contributions to the acquisition and analysis of data. DL, ZL and KX made contributions to the analysis and interpretation of data and revised the manuscript. TQ and LH checked and confirmed the authenticity of the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Ethical approval for this study was obtained from the Medical Ethics Committee of the Affiliated Hospital of Xuzhou Medical University (Xuzhou, China; nos. XYFY2017-KL009-01, XYFY2018-KL013-01 and XYFY2020-KL080-01). Patients provided written informed consent prior to sample collection.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Muhammad N, Mao Q and Xia H: CAR T-cells for cancer therapy. Biotechnol Genet Eng Rev. 33:190–226. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, Fry TJ, Orentas R, Sabatino M, Shah NN, et al: T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: A phase 1 dose-escalation trial. Lancet. 385:517–528. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, Bader P, Verneris MR, Stefanski HE, Myers GD, et al: Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 378:439–448. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Kochenderfer JN, Dudley ME, Kassim SH, Somerville RPT, Carpenter RO, Stetler-Stevenson M, Yang JC, Phan GQ, Hughes MS, Sherry RM, et al: Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol. 33:540–549. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Kochenderfer JN, Somerville RPT, Lu T, Shi V, Bot A, Rossi J, Xue A, Goff SL, Yang JC, Sherry RM, et al: Lymphoma remissions caused by anti-CD19 chimeric antigen receptor T cells are associated with high serum interleukin-15 levels. J Clin Oncol. 35:1803–1813. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Mikkilineni L and Kochenderfer JN: Chimeric antigen receptor T-cell therapies for multiple myeloma. Blood. 130:2594–2602. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Yan Z, Cao J, Cheng H, Qiao J, Zhang H, Wang Y, Shi M, Lan J, Fei X, Jin L, et al: A combination of humanised anti-CD19 and anti-BCMA CAR T cells in patients with relapsed or refractory multiple myeloma: A single-arm, phase 2 trial. Lancet Haematol. 6:e521–e529. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Qian L, Li D, Ma L, He T, Qi F, Shen J and Lu XA: The novel anti-CD19 chimeric antigen receptors with humanized scFv (single-chain variable fragment) trigger leukemia cell killing. Cell Immunol. 304–305. 49–54. 2016.PubMed/NCBI

9 

Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, Grupp SA and Mackall CL: Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 124:188–195. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Chen AP, Setser A, Anadkat MJ, Cotliar J, Olsen EA, Garden BC and Lacouture ME: Grading dermatologic adverse events of cancer treatments: The common terminology criteria for adverse events version 4.0. J Am Acad Dermatol. 67:1025–1039. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, Braunschweig I, Oluwole OO, Siddiqi T, Lin Y, et al: Axicabtagene Ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 37:2531–2544. 2017. View Article : Google Scholar

12 

Hirayama AV, Gauthier J, Hay KA, Voutsinas JM, Wu Q, Pender BS, Hawkins RM, Vakil A, Steinmetz RN, Riddell SR, et al: High rate of durable complete remission in follicular lymphoma after CD19 CAR-T cell immunotherapy. Blood. 134:636–640. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Fried S, Avigdor A, Bielorai B, Meir A, Besser MJ, Schachter J, Shimoni A, Nagler A, Toren A and Jacoby E: Early and late hematologic toxicity following CD19 CAR-T cells. Bone Marrow Transplant. 54:1643–1650. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Hockings C, Kuhnl A, Wong S, Shah M, Inam S, Patten P, Cuthill K, Pagliuca T, Yallop D, Benjamin R, et al: Characterisation of early and late cytopenias in lymphoma patients following treatment with antiCD19 CAR-T therapy. In Bone Marrow Transplant. 55:Springernature. (London). 238–239. 2020.

15 

Cordeiro A, Bezerra ED, Hirayama AV, Hill JA, Wu QV, Voutsinas J, Sorror ML, Turtle CJ, Maloney DG and Bar M: Late events after treatment with CD19-targeted chimeric antigen receptor modified T cells. Biol Blood Marrow Transplant. 26:26–33. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Abramson JS, Palomba ML, Gordon LI, Lunning MA, Wang M, Arnason J, Mehta A, Purev E, Maloney DG, Andreadis C, et al: Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): A multicentre seamless design study. Lancet. 396:839–852. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Nagle SJ, Murphree C, Raess PW, Schachter L, Chen A, Hayes-Lattin B, Nemecek E and Maziarz RT: Prolonged hematologic toxicity following treatment with chimeric antigen receptor T cells in patients with hematologic malignancies. Am J Hematol. 96:455–461. 2021. View Article : Google Scholar : PubMed/NCBI

18 

Strati P, Varma A, Adkins S, Nastoupil LJ, Westin J, Hagemeister FB, Fowler NH, Lee HJ, Fayad LE, Samaniego F, et al: Hematopoietic recovery and immune reconstitution after axicabtagene ciloleucel in patients with large B-cell lymphoma. Haematologica. 106:2667–2672. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Rejeski K, Perez A, Sesques P, Hoster E, Berger C, Jentzsch L, Mougiakakos D, Frölich L, Ackermann J, Bücklein V, et al: CAR-HEMATOTOX: A model for CAR T-cell related hematological toxicity in relapsed/refractory large B-cell lymphoma. Blood. 138:2499–2513. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2023
Volume 26 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Qiu T, Hu L, Zhang Y, Wang Y, Ma S, Li D, Li Z and Xu K: Cytopenia after CAR‑T cell therapy: Analysis of 63 patients with relapsed and refractory B‑cell non‑Hodgkin lymphoma. Oncol Lett 26: 338, 2023
APA
Qiu, T., Hu, L., Zhang, Y., Wang, Y., Ma, S., Li, D. ... Xu, K. (2023). Cytopenia after CAR‑T cell therapy: Analysis of 63 patients with relapsed and refractory B‑cell non‑Hodgkin lymphoma. Oncology Letters, 26, 338. https://doi.org/10.3892/ol.2023.13924
MLA
Qiu, T., Hu, L., Zhang, Y., Wang, Y., Ma, S., Li, D., Li, Z., Xu, K."Cytopenia after CAR‑T cell therapy: Analysis of 63 patients with relapsed and refractory B‑cell non‑Hodgkin lymphoma". Oncology Letters 26.2 (2023): 338.
Chicago
Qiu, T., Hu, L., Zhang, Y., Wang, Y., Ma, S., Li, D., Li, Z., Xu, K."Cytopenia after CAR‑T cell therapy: Analysis of 63 patients with relapsed and refractory B‑cell non‑Hodgkin lymphoma". Oncology Letters 26, no. 2 (2023): 338. https://doi.org/10.3892/ol.2023.13924