Next Article in Journal
Factors Associated with Axillary Lymph Node Status in Clinically Node-Negative Breast Cancer Patients Undergoing Neoadjuvant Chemotherapy
Previous Article in Journal
Extracellular Vesicles: A Novel Tool in Nanomedicine and Cancer Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical Strategies for Enhancing the Efficacy of CAR T-Cell Therapy for Hematological Malignancies

1
School of Basic Medical Sciences, Guangdong Medical University, Dongguan 523000, China
2
Department of Pathology, The First People’s Hospital of Foshan, Foshan 528000, China
3
Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang 524000, China
4
Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(18), 4452; https://doi.org/10.3390/cancers14184452
Submission received: 11 August 2022 / Revised: 31 August 2022 / Accepted: 3 September 2022 / Published: 14 September 2022

Abstract

:

Simple Summary

Cellular immunotherapy has achieved great progress in the treatment of hematological malignancies, and cell therapy may become the main force in the treatment of refractory and relapsed malignant tumors in the future. The efficacy of CAR T cells as a potential cell therapy in the treatment of hematological tumors is acknowledged. Given that clinical application strategies for optimizing CAR T-cell therapy are still immature, this article reviews the impact of various application strategies on CAR T-cell therapy.

Abstract

Chimeric antigen receptor (CAR) T cells have been successfully used for hematological malignancies, especially for relapsed/refractory B-cell acute lymphoblastic leukemia and non-Hodgkin’s lymphoma. Patients who have undergone conventional chemo-immunotherapy and have relapsed can achieve complete remission for several months with the infusion of CAR T-cells. However, side effects and short duration of response are still major barriers to further CAR T-cell therapy. To improve the efficacy, multiple targets, the discovery of new target antigens, and CAR T-cell optimization have been extensively studied. Nevertheless, the fact that the determination of the efficacy of CAR T-cell therapy is inseparable from the discussion of clinical application strategies has rarely been discussed. In this review, we will discuss some clinical application strategies, including lymphodepletion regimens, dosing strategies, combination treatment, and side effect management, which are closely related to augmenting and maximizing the efficacy of CAR T-cell therapy.

1. Introduction

Cellular immunotherapy of hematological malignancy has achieved remarkable progress in the last decade, especially that based on chimeric antigen receptor (CAR) T cells. CAR T-cells have experienced four generations (Figure 1). First-generation CAR T-cells were found to lack persistence and expansion in pre-clinical studies, while second-generation cells overcame these limitations by containing intracellular costimulatory domain (e.g., CD28 or 4-1BB), later fused to CD3zeta. Third-generation cells with two costimulatory domains of CD28 and 4-1BB showed promising results in persistence and anti-tumor activity [1], while fourth-generation CAR T-cells additionally achieved specific cytokine release through gene editing following T cell activation [2]. CD19-directed CAR T-cells as representative second-generation CAR T-cells have led to unprecedented development in the treatment of B-cell hematological malignancies, with over 90% overall survival [3,4]. It is worth mentioning that Melenhorst et al. of the University of Pennsylvania found that after infusion of second-generation CAR T-cells (CTL019) in two CLL patients, stable cloned CAR T-cells could be continuously detected in vivo, and sustained remission for 10 years was achieved, which is encouraging [5]. For patients with chemotherapy resistance and difficulty in matching hematopoietic stem cells, CAR T-cell therapy may be the salvage regimen. Several commercial products (e.g., axicabtagene ciloleucel, lisocabtagene maraleucel, tisagenlecleucel, brexucabtagene autoleucel) have been approved by the US Food and Drug Administration (FDA) for the treatment of B-cell acute lymphoblastic leukemia (B-ALL) and other relapsed or refractory (R/R) large B-cell lymphomas (Table 1) [4,6,7].
However, this emerging cellular immunotherapy faces multiple challenges in the treatment of malignancies. On the one hand, most of the patients treated with CAR T-cells suffer from cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), hematological toxicity, and grade 3/4 side effects that can threaten the life of patients [8,9,10]; on the other hand, short persistence of CAR T-cells in vivo is related with poor efficacy [11]. Therefore, some optimization schemes for CAR T-cells, such as multiple target treatment, discovery of new targets, and CAR construct optimization, have been widely studied to reduce adverse events and improve CAR T-cell efficacy [12,13,14,15,16]. However, the clinical application strategies of CAR T-cells, which have an essential role in the overall treatment process, have rarely been discussed. After determining that a patient is suitable for CAR T-cell therapy, it is necessary to determine the lymphatic clearance scheme and dosing strategy according to the patient’s basic condition, to help the patient obtain the best blood environment and maximize the utilization of CAR T-cells [17]. Furthermore, for refractory and relapsed hematological malignancies, the combined application of multiple treatments may lead to longer event-free survival for patients [18,19]. The implementation of combined strategies to increase the persistence or antitumor activity of CAR T-cells has become a research hotspot. In addition, the safe and effective implementation of a strategy is inseparable from the supervision of the disease and careful comprehensive management, which will promote patients obtaining complete remission (CR) and disease-free survival. In this review, we will discuss the lymphodepletion regimen, dosing strategy, combination treatment, and side effects management of clinical trials in order to provide new insights and directions for physicians and researchers (Figure 2).
Table 1. Pivotal clinical trials of FDA-approved CAR T-cell products.
Table 1. Pivotal clinical trials of FDA-approved CAR T-cell products.
CAR T-CellsClinical TrialsMalignancyEnrolled
Patients
Lymphodepletion
Regimens
Dosage Regimens
(CAR T Cells)
EfficacySide Effect (CRS, NE)
Axicabtagene ciloleucelNCT02348216 [20]DLBCL
PMBCL
TFL
2017
Adults
101/111
30 mg/m2 Flu and
500 mg/m2 Cy;
On days -5, -4, and -3
2 × 106 cells/kg on day 0;
a single infusion
ORR 82%
CR 54%
CRS 13%,
NE 28%
NCT02348216 [6]DLBCL
PMBCL
TFL
2019
Adults
108/119
30 mg/m2 Flu and
500 mg/m2 Cy;
On days -5, -4, and -3
2 × 106 cells/kg on day 0;
a single infusion
ORR 83%
ORR 58% c
CRS 11%,
NE 32%
NCT03105336 [21]I-NHL
2022
Adults
148/153
30 mg/m2 Flu and
500 mg/m2 Cy;
On days -5, -4, and -3
2 × 106 cells/kg on day 0;
a single infusion
OR 92%
CR 74%
CRS 7%,
NE 19%
Lisocabtagene maraleucelNCT02631044 [3]r/r LBCL
2020
Adults
269/344
30 mg/m2 Flu and
500 mg/m2 Cy
for 3 days;2–
7 days before CAR T cell infusion
50 × 106 cells (51 patients),
100 × 106 cells (177 patients),
150 × 106 cells (41 patients);
Including CD8+ and
CD4+ CAR T cells
ORR 73%
CR 53%
CRS 2%,
NE 10%
TisagenlecleucelNCT02445248 [22]r/r LBCL
2021
Adults
115/167
25 mg/m2 Flu and
250 mg/m2 Cy
for 3 days;
or bendamustine with 90 mg/m² for 2 days
0.1–6 × 108 CAR T cells;
a single infusion
(target dose: 5 × 108 CAR T cells)
OR 53%
CR 39%
CRS 23%
NCT03568461 [23]r/r FL
2022
Adults
97/98
25 mg/m2 Flu and
250 mg/m2 Cy or bendamustine with 90 mg/m² for 2 days
0.6–6 × 108 CAR T cells;
a single infusion
CR 69.1%
OR 86.2%
CRS 48.5%,
NE 37.1%
Brexucabtagene autoleucelNCT02614066 [24]r/r ALL
2021
Adults
65/71
25 mg/m2 Flu
and 900 mg/m2 Cy
days -4, -3, and -2
1 × 106 cells/kg on day 0;
a single infusion
CR 56%CRS 24%,
NE 25%
NCT02601313 [25]r/r ML
2020
Adults
71/74
30 mg/m2 Flu and
500 mg/m2 Cy
on days -5, -4, and -3
2 × 106 cells/kg on day 0;
a single infusion
ORR 93%
CR 67%
CRS 15%,
NE 31%
DLBCL = diffuse large B-cell lymphoma; CRS = cytokine release syndrome; NE = neurologic events; I-NHL = indolent non-Hodgkin lymphoma; Flu = fludarabine; Cy = cyclophosphamide; OR = overall response; r/r LBCL = relapsed or refractory large B-cell lymphomas; r/r FL = relapsed or refractory follicular lymphoma; r/rALL = relapsed/refractory acute lymphoblastic leukemia; r/r ML = relapsed or refractory mantle-cell lymphoma; PMBCL = primary mediastinal B-cell lymphoma; TFL = transformed follicular lymphoma; ORR = objective response rate; CR = complete response rate.

2. Lymphodepletion Regimen

After infusing into the blood, CAR T-cells specifically recognize the target antigen through the single-chain variable fragment (scFv) binding domain, which requires a suitable immunomodulatory environment for T cell survival and proliferation [26,27,28]. Lymphodepletion followed by CAR T-cell infusion remodels the immune environment for creating enough T cell pool, decreasing immunosuppressive cell populations, increasing the availability of cytokines and thus promoting lymphocyte proliferation and survival, and increasing tumor antigen presentation by inducing cell death [29,30,31]. Cyclophosphamide (Cy) and fludarabine (Flu) are two major lymphodepletion compositions, known as potent immunosuppressive agents. In one case, the overall response rate (ORR) without lymphodepletion was only 25%, while ORR rose to 58% after lymphodepletion that had made changes in cytokine profiles and had increased CAR T-cell durability [32,33]. However, how the lymphodepletion regimen impacts CAR T-cell efficacy is still unclear. Here, we discuss the influencing factors of chemotherapy pretreatment methods on CAR T-cell potency and durable remission based on completed clinical trials.
Generally, lymphodepletion with cyclophosphamide and/or fludarabine is implemented on -5, -4, and -3 days before CAR T-cell injection to create an immunosuppressive environment that provides CAR T-cell expansion in vivo [20,34]. Moreover, the chemotherapy pretreatment regimen adopted in most trials is a dose of fludarabine generally maintained at about 30 mg/m2·d or 25 mg/m2·d; the dose of cyclophosphamide is 200 to 3000 mg/m2·d [7,11,35,36,37], which is considered to be a relatively safe dose (Table 1). The co-administration of cyclophosphamide and fludarabine is viewed as adequate lymphodepletion. Studies have shown that a low incidence of CR and high rate of relapse may be associated with inadequate CAR T-cell persistence and expansion in vivo and cell-mediated immunosurveillance [17,38,39]. Cyclophosphamide and fludarabine-mediated lymphodepletion reshape the blood environment, prolong CAR T-cell survival time, regulate the release of activating cytokines, and improve the event-free survival time of patients [38,40,41].
Studies have shown that the effect of lymphodepletion and the reactivity of subsequent CAR T-cell therapy is dose-dependent [11]. A total of 17 patients received high-dose cyclophosphamide of 3 g/m2·day (HD-Cy), while 8 patients received low-dose cyclophosphamide at 1.5 g/m2·day (LD-Cy), with fludarabine at 25 mg/m2·day for 3 days. Results manifested that CAR T-cell expansion in the HD-Cy group was remarkably higher than that in the LD-Cy group. The disease response rate of the former was 94%, while the latter was only 38% [11]. This indicates that deeper lymphodepletion leads to higher clinical response due to thoroughly eliminating immunosuppression and reducing cytokine rejection of CAR T-cells. In addition, high-dose Cy increased the level of IL-7 and MCP-1 and was associated with CR and/or progression-free survival (PFS) [40,42]. However, the mechanism through which HD-Cy leads to a favorable cytokine profile has yet to be determined [11,43]. Nevertheless, in a previous study targeting diffuse large B-cell lymphoma (DLBCL), patients who received modulated chemotherapy with low a dose of cyclophosphamide (500 mg/m2) and fludarabine (30 mg/m2) had a higher level of cytokines, particularly IL-5 and IL-15, which are critical for T cell proliferative cytokines [34,44].
The addition of fludarabine to cyclophosphamide as a lymphodepleting regimen before CD19-directed CAR T-cell therapy significantly improved outcomes in patients treated with CAR T-cells [45]. Over 90% of patients who received fludarabine-containing lymphodepletion achieved CR and had no relapse, while 58% of patients without fludarabine suffered a relapse. In patients with CD19+ relapse, a loss of CAR T-cell in blood was detected by PCR analysis [38]. Incorporation of fludarabine into the lymphodepletion regimen, lower concentration of LDH, and higher platelet count before the lymphodepletion regimen containing fludarabine may lead to minimal residual disease-negative complete remission (MRD- CR) [32]. Compared with the lower cumulative fludarabine exposure during lymphodepletion, patients in the higher group had an 11-month improvement in leukemia-free survival, and the CD19+ recurrence rate within 1 year decreased from 100% to 27.4% [46]. However, when the disease burden is high, the effect is weakened, so the dose of fludarabine should be formulated according to the patient’s disease burden and designed individually [47]. Interestingly, for DLBCL patients who relapsed after CAR T-cell therapy, pre-existing CAR T-cells were revitalized after the second lymphatic clearance containing fludarabine and created an anti-tumor effect with a predominant grade 2 CRS [48]. This case indicates that CAR T-cells could be reactivated under circumstances by disturbing the immune equilibrium. CAR T-cell activation and cytotoxicity depend on the immune homeostasis of the blood environment, and the blood environment after lymphatic clearance seems merely to maintain for a period time, which may be one of the reasons why patients can only achieve short-term remission. At the same time, to obtain satisfactory clinical efficacy, the degree of lymphatic clearance needs to be proportional to the patient’s disease burden; thus, a universal single dosage is not suitable [11,38].

3. Dosing Regimen of CAR T-Cells

Before lymphocytes are removed, T cells will be isolated from the peripheral blood mononuclear cells (PBMC) of patients as raw materials for subsequent manufacturing of CAR T-cells [49] (Figure 2). The preparation of CAR T-cells is about 13 days, including T cells expanding for 9 days; another study shortens the preparation time to 7 days with similar effectiveness [50,51]. Surprisingly, Gracell Bio in China has developed a fabrication technique of FasTCAR, which shortens the preparation time of CAR T-cells to 1 day [52]. FasTCAR technology transforms the activation, transduction, and expansion steps into a single “concurrent activation-transduction” step, eliminating the time for in vitro expansion of CAR T-cells [53]. After obtaining CAR T-cells and achieving the standards of identity, potency, sterility, and adventitious agents, the infusion strategy of CAR T-cells can be determined by clinicians and manufacturers based on phase I clinical trials identifying the maximally tolerated dose (MTD) [54].
In CD19-directed CAR T-cell products (axicabtagene ciloleucel, lisocabtagene maraleucel, tisagenlecleucel, brexucabtagene autoleucel) that have been proved by FDA, the single infusion dose is about 1–3 × 106/kg, except that tisagenlecleucel is 0.1–6×108 viable cells, of which 5×108 viable cells are more suitable for DLBL and relapsed or refractory follicular lymphoma (FL) (Table 1) [3,4,6,7,20,21,23,34,55]. In adults, some CD19-targeted products in the treatment of different diseases in the dose range of 2 × 106/kg showed higher ORR and CR and lower incidence of high-grade CRS and neurological event (NE) side effects. Determination of a safe/efficacious dose range is based on understanding dose-response/exposure/safety analyses. A dose-escalation test showed that 1 × 106/kg is the highest single-injection tolerated dose for R/R ALL and non-Hodgkin’s lymphoma (NHL) patients; in this condition, all toxicities, including grade 4 CRS, are reversible [56]. Moreover, a lower CAR T-cell dosage of 1 × 105/kg is effective and safe for treating r/r B-ALL [57]. It can be seen that the clinical results cannot intuitively find the relationship between the dose and the efficacy of CAR T-cells, except that high doses show dose-dependent toxicity. This may be due to the initial CAR T-cell viability and different immunosuppressive gene expression of the tumor microenvironment [58].
A single injection of low-dose CAR T-cells is considered safe and effective, but not with durable remission. Some researchers have studied one-time injection or segmental injection of low-dose or high-dose CAR T-cells [37,43]. A single-arm, open-label study verified the difference between these two strategies in efficacy. The trial is divided into a high-dose single infusion (5 × 108/kg), low-dose single/fractionated infusion (5 × 107/kg), and high-dose fractionated infusion (5 × 108/kg); the numbers of patients were 6, 9, and 20, respectively [59]. The high-dose fractionated group showed the best effect, with a complete remission rate of 90% and a two-year overall survival rate of 73%, while in the high-dose single infusion group, patients suffered from refractory CRS complicated with culture-positive sepsis [59]. The low rate of adverse events of the high-dose fractionated group indicates that fractionated infusion of a high dose may alleviate immune response. Characteristics including age, disease phenotype, chemotherapy regimen received before infusion, and whether to undergo stem cell transplantation are all related to dosage selection, indicating that it is difficult to set the dosing interval to enable long-term complete remission [60]. Before receiving CAR T-cell therapy, these patients had experienced more than two lines of systemic therapy; however, at the same therapeutic dose, there were differences in the patients’ physical condition, immune homeostasis of the blood environment, disease burden, and cytokine pool after lymphatic depletion. A scoring system is set up to determine various aspects of the patient’s physical state, thereby determining the corresponding dose standard within a certain scoring range. The dosing regimen of CAR T-cells is individualized. However, studies of the clinical response of patients to CAR T-cells could provide clinicians with data for developing a personalized dosage strategy.

4. Combination Strategies

Anti-CD19 CAR T-cells show a strong short-term CR in the treatment of refractory and relapsed B-cell diseases. Its limitations include the inability to achieve long-term disease-free survival and risk of recurrence due to a lack of sustained expansion of CAR T-cells [58], and patients with a high tumor burden treated with allogeneic hematopoietic stem cell transplantation (allo-HSCT) could not achieve complete remission [61]. CAR T-cells or allo-HSCT achieved considerable short-term efficiency when used alone; researchers can explore combination to achieve long-term disease-free survival.
Allo-HSCT, as a kind of consolidation strategy, has shown a powerful capability for R/R B-cell lymphoma [61,62,63,64,65,66]. With a quick bridge to allo-HSCT after CAR T-cell therapy, the one-year event-free survival (EFS) was up to 73% compared with 50% for CAR T-cells alone, and the rate of severe grade 2–4 acute graft-versus-host disease (aGVHD) was less than 23.1% compared with 41.6% of allo-HSCT alone [7,67,68,69]. Moreover, the timing of allo-HSCT treatment affects patient prognosis. It was shown that the period within three months after treatment is considered to be a suitable time window for consolidation therapy [68]. MRD- CR patients receiving allo-HSCT showed detectable CAR T-cells for 21 months. Jiang H et al. [69] found that allo-HSCT was an independent prognostic factor for EFS and relapse-free survival (RFS) in MRD- CR patients. For patients with CD19-negative relapse, allo-HSCT may be desirable as a consolidated method because of immune reconstitution [57]. From another point of view, CAR T-cell therapy has obtained a larger range of patients for allo-HSCT [69]. In addition, CAR T-cells can be used as maintenance therapy after allo-HSCT therapy. Shi et al. found that CAR T-cells combined with lenalidomide maintenance therapy significantly prolonged PFS in patients with multiple myeloma. Lenalidomide increased the antitumor activity and persistence of CAR T-cells, which might be due to the fact that lenalidomide can maintain the long-term immune surveillance of CAR T-cells in remission, and this combined strategy promotes T cells toward the less terminally differentiated phenotype [70]. RNA sequencing and assays for transposase-accessible chromatin indicated that lenalidominde can alter T-helper response, cytokine production, T cell activation, cell-cycle control, and cytoskeletal remodeling [71,72].
CAR T-cells combined with ibrutinib, a kind of bruton’s tyrosine kinase inhibitor, improved CAR T-cells engraftment, tumor clearance, and survival in human xenograft models of resistant acute lymphocytic leukemia [73]. Ibrutinib is a crucial component of a first-line treatment option for chronic lymphocytic leukemia (CLL), and it showed superior PFS and CR in progressive CLL patients who were too frail to receive aggressive therapy [74]. Continuous use of ibrutinib leads to tolerance and tumor recurrence, and fortunately, CAR T-cells show obvious therapeutic efficacy and durability after ibrutinib failure [75]. Therefore, the combination of these two therapies may create an advance for r/r CLL patients. Compared with CLL patients treated with CAR T-cells without ibrutinib, CLL patients treated with CAR T-cells combined with ibrutinib were associated with lower CRS severity and lower serum concentrations of CRS-associated cytokines, which is consistent with pre-clinical results [76]. Moreover, one-year overall survival and PFS probabilities of a combination of CAR T-cells with ibrutinib were 86% and 59%, respectively, while anti-CD19 CAR T-cell therapy was only 38% and 50% [77]. After more than 1 year of ibrutinib therapy, the efficiency of CAR T-cell engraftment, tumor clearance, and survival were improved [73].
CAR T-cell therapy with immune checkpoint inhibitors has now moved to the clinical stage. A pre-clinical study demonstrated that CAR T-cell therapy combined with PD-1 blockade showed better tumor control, and the persistence of CAR T-cells was up to 21 days in mice [78]. Furthermore, neuroblastoma and metastatic melanoma with CAR T-cell therapy combined with PD-1 blockade indicated that PD-1 blockade can enhance cytokine production and reduce activation-induced cell death (AICD) of CAR T-cells after repeated antigen stimulation [17,79].
Moreover, the effect of the first-line chemotherapy for CLL patients can be strengthened by combining it with CAR T-cell therapy. After regular chemotherapy treatment with pentostatin, cyclophosphamide, and rituximab, patients who received CAR T-cells ranging from 3 × 106 to 3 × 107 CAR T-cells/kg were observed to have more than 28 months CR and modest CRS neurotoxicity [39]. In addition, the above methods combined with CAR T-cell therapy are moving to the clinical stage, and other molecular medicines such as immunomodulatory drugs, utomilumab, hypomethylating agents, Phosphoinositide 3-kinase (PI3K) inhibition, γ-secretase inhibitors, and fas blockade, are part of widely ongoing pre-clinical studies [80]. These combination strategies represent a new treatment paradigm that leverages the ability of genetically modified T cells to target and destroy tumor cells.

5. Occurrence and Management of Side Effects

Anti-CD19 CAR T-cell therapy led to cytokine release syndrome, hematological toxicity, and ICANS attributed to inflammatory cytokines and chemokines released by immune cells, which are the main obstacles after CAR T-cells were administrated into the blood system via intravenous injection [81]. The average onset time of CRS is 3 days after injection, and it lasts for about 7 days [82]. When the neurological event occurs, it is generally within 8 weeks and lasts for 28 days [82]. Blood events such as thrombocytopenia and neutropenia may exist throughout the whole disease event. The incidence of CRS in the milestone trial is as high as 77%, the average incidence of neurological events is more than 30%, and blood events are more than 40% [7]. Although corresponding support and drug treatments were performed during the treatment process, the final mortality rate of patients is more than 40%, accompanied by disease progression and aggravating effects of side effects [6,7,83]. Therefore, it is meaningful to review the occurrence and management of side effects. Moreover, early detection and timely control of adverse events also play important roles in the treatment of disease.
Generally, grade 1/2 CRS, mainly including fever, fatigue, nausea, mild hypoxia, or headache, is not life-threatening and is relieved after symptomatic and supportive treatments performed with close monitoring of the functional status of various organs and the level of cytokines in the blood. Grade 3/4 CRS is life-threatening to patients and is closely related to the subsequent neurotoxic events [10]. Grade 3/4 CRS is accompanied by more severe hypoxia, which requires ventilator support if necessary, and ICU care is needed in some severe cases [84]. In 2020, Pierre Sesques et al. [79,82] did a retrospective study on commercial products such as axicabtagene ciloleucel and tisagenlecleucel and found that grade 3/4 CRS and neurological events were reduced to 8% and 10% compared with the key trial in 2017, which is 11% and 32%, respectively. The lower rate of grade 3/4 side effects is due to earlier and more systematic interventions.
When CRS occurs, the levels of IL-2, IL-4, IL-6, IL-10, TNF-α, and IFN-γ in the blood are significantly increased; in particular, IL-6 is parallel to the severity of CRS. Compared with grade 1/2 CRS, grade 3/4 CRS showed a rapid increase in IL-6 in the early stage, and the patients’ fever was high and lasted for a significant period. These clinical symptoms can be used as an early prediction of severe CRS and can provide treatment strategies for adequate treatment in the later period [10,85,86]. On the other hand, the rapid expansion of infused CAR T-cells is also related to CRS [9]. At the same time, the concentration of the above cytokines also significantly increased with the expansion of CAR T-cells, which is associated with not only injection protocol but also the kind of costimulatory domain of CAR T-cells [16,36]. The IL-6 antagonist tocilizumab is the first-line treatment for CRS; however, it is usually used when supportive treatment does not work. If necessary, it can be used in combination with the immunosuppressant dexamethasone. Generally, it can provide relief after one week of treatment. However, when the tumor burden and the number of recurrences in the past are high, severe CRS requires hemodialysis or plasma exchange to overcome difficulties [10,34].
In addition, grade 3/4 CRS is related to the increased risk of subsequent infections, especially bloodstream infections that are difficult to distinguish from grade 3/4 CRS. Bloodstream infections are similar to CRS symptoms, but the treatment methods for the two are different. We can determine the infection through imaging methods and laboratory tests, such as computed tomography, bacterial examination, magnetic resonance imaging, C-reactive protein (CRP), procalcitonin, etc. When it is impossible to distinguish, clinicians should use antibiotics based on their experience [87,88]. Moreover, the incidence of disseminated intravascular coagulation (DIC) increases in patients with grade 3/4 CRS. As a result, early and proper interventions targeted to CRS-related coagulopathy greatly contribute to the control of side effects in CAR T-cell therapy [85].
Neurotoxic events occurred in the study as compared with the high incidence of CRS, but with the optimization of CAR T-cells, the incidence of CRS and neurological events decreased [37,89]. ICANS often occurs with grade 3/4 CRS. ICANS includes encephalopathy, confusion, delirium, tremor, restlessness, lethargy, and epilepsy in severe patients (Grade 3) [7]. There is no specific treatment method, other than relying on supportive treatment. Specific antibodies targeting cytokines related to neurotoxic events may be found in the future. Studies have shown that neurotoxicity is related to CAR T-cells crossing the blood–brain barrier and triggering an immune response in the brain. IFN-γ and IL-6 in the cerebrospinal fluid are nearly 40 times higher than those in the serum [90]. At the same time, the qPCR of the CAR expression indicates that it is in the cerebrospinal fluid, and the CAR copy number is three times higher [90]. Thus, the detection of components in cerebrospinal fluid can be used as a method to predict neurotoxicity [86,88,90]. Due to individual differences, CRS, hematological toxicity, and neurotoxicity may occur in patients at any time after CAR T-cell treatment. Identifying the characteristics of side effects and determining the treatment may be helpful for the positive progress of CAR T-cell therapy.

6. Discussion

With the clear superiority of CAR T-cell therapy in clinical trials, CAR T-cell therapy will become the standard therapy for hematological malignancies. However, the clinical application strategy of CAR T-cells has not yet achieved consensus, and there remain variability and uncontrollability. One of the preparations before CAR T-cell infusion is the preparation of the patient’s blood environment, including the important step of lymphodepletion. Currently, the lymphodepletion regimens used in commercial CAR T cell products are mainly cyclophosphamide and fludarabine. The main limitation is that the dose selection and combination strategies of lymphodepletion regimens are relatively single and do not take into account the individuality of the patient’s blood environment. Lymphodepletion, as an important cornerstone of CAR T-cell therapy, has direct anti-tumor cytotoxicity, activates innate immunity, enhances the production of homeostatic cytokines (MCP1, IL-7, and IL-15) [44], and reduces the immune regulation number of cells, such as myeloid-derived suppressor cells and regulatory T cells [91], and decreases indoleamine 2,3-dioxygenase (IDO) expression [92]. Therefore, close detection of lymphocyte numbers, amount of favorable cytokines, and clearance of immunosuppressive cells during lymphodepletion correlate with cytotoxic function and persistence after further CAR T-cell infusion and patient prognosis. At the same time, preclinical assessment of patients with these indicators can be used to identify chemotherapy drugs and combination strategies for lymphatic failure.
In CAR T-cell therapy procedures, the choice of therapeutic dose directly affects the prognosis of patients. CAR T-cells as a live cell drug have more variability than chemical drugs. In the previous discussion, we found that in pivotal experiments, the dose of each kind of CAR-T cell was the same in different diseases. However the relationship between dose and exposure is highly variable and may be affected by the design of the CAR, T-cell initial immune function, the proportion of CAR T-cell phenotype, disease burden, lymphoid depletion, and the subsequent effects of immunomodulatory therapy [54,93]. These factors determine that a single therapeutic dose cannot maximize efficacy for patients. In order to reduce side effects and improve the maximum utilization of CAR T-cells, dose escalation trials are underway [56]. Although low doses can reduce CRS, ICAMS, and hematological toxicity, CAR T-cells cannot sustainably expand, release toxic substances, and effectively eliminate tumor cells. The pharmacokinetic of CAR T-cells in patients is closely related to recurrence in patients. Therefore, the homeostasis of CAR T-cells in vivo should be adapted to the tumor cells per unit area. In future studies, studies of dose selection should incorporate multiple assessment metrics, such as lymphatic clearance, immune function acquired by CAR T-cells, structure and phenotype of CAR T-cells, disease burden, and patient underlying disease and tolerance ability [94].
Although CAR T-cell therapy is promising, limitations still exist, such as CAR T-cell exhaustion, tumor antigen escape, directional trafficking of CAR T-cells, the immunosuppressive microenvironment of the tumor, and the occurrence of high-grade side effects. In addition to directly killing tumor cells, the implementation of the combination therapy strategy can also modulate the immunosuppressive microenvironment, combat CAR T-cell exhaustion, and reduce immune side effects. Allo-HSCT is considered as an effective way to cure hematological malignancies. However, recurrence has become a major obstacle to Allo-HSCT therapy, mainly due to the loss and downregulation of autologous human leukocyte antigen (HLA), the upregulation of T cell inhibitory ligands, and the immunosuppressive microenvironment in the tumor [95]. CAR T-cells specifically recognize target antigens in an HLA-independent manner against the immune tolerance mechanism of tumor cells. At the same time, both CD8+ T cell subsets and CD4+ T cell subsets can be introduced into CAR to mediate cytolysis through granzyme/perforin independent of Fas or TNF-α signaling [96]. Preclinical studies have demonstrated that a variety of immunomodulatory drugs can enhance the efficacy of CAR T-cells from different perspectives. Lenalidomide increases the expression of nuclear transcription factor-κB (NF-κB) by phosphorylating the co-stimulatory domain CD28, reverses the decrease in filamentous actin (F-actin) polymerization in T cells, increases the cytokine IL-21 to change the metabolic pattern of CAR T-cells, and enhances the persistence and activity of the CD2 subset of CAR T-cells [97,98,99,100]. The rationale for ibrutinib binding to CAR T-cells is based on its direct effect on T cells and its ability to destroy the immunosuppressive TME. Ibrutinib can reduce the expression of CXC chemokine receptor 4 (CXCR4) in leukemia cells, and inhibit the signal transduction downstream of CXCR4, disrupt the homing of leukemia cells to lymphoid tissue, and promote its release into the circulation, reducing malignant CD200 (OX2) expression on cells and decreasing expression of immunosuppressive cytokines [101]. PD-1 blocking antibody increases cytokine production and restores CAR T-cell activity in vitro and in vivo [102]. Decitabine increases the activity of CD19-targeted CAR T-cells by increasing the immunogenicity of tumor cells and increases the transcription and cell surface expression of CD19 in lymphoma cell lines [103]. Preclinical studies have provided substantial evidence that combination therapy can overcome the limitations of CAR T-cells themselves and the inhibitory effect of the tumor immune microenvironment, enabling adoptive cell therapy to achieve sustained expansion and cytotoxicity. More clinical trials should be designed to determine the certainty of the efficacy of combination therapy; the development of sustainable combination therapy strategies is a direction for future research.
The success of CAR T-cell therapy lies in the timely and effective management of side effects and the ability to identify the link between the test indicators and the occurrence of side effects. In adoptive cell therapy, CRS is the most common and serious complication; it is a cytokine cascade that occurs with CAR T-cell activation. The temporal peak of this cytokine release is influenced by the co-stimulatory domain, and CRS in CAR T-cells containing CD28 structures appears early and at high levels. Cascade release of cytokines can also trigger the cytokine release of other host antigen-presenting cells and other T cells, while monocytes, macrophages, and dendritic cells are major sources of IL-6, a key player in CRS [104]. Excessive inflammation activates the vascular endothelium, increases levels of vascular permeability factors, and decreases levels of the endothelial stabilizer Ang-1, leading to loss of vascular integrity, hemodynamic instability, capillary leakage, and consumptive coagulopathy in CRS, and then triggering a series of systemic side effects [105]. Additionally, pediatric patients with B-ALL with higher tumor burden had greater CAR T-cell expansion and higher CRS severity [106]. The mechanism of CRS occurrence suggests events that need to be monitored in the management of side effects in clinical studies. The occurrence of ICANS is often secondary to severe CRS, and there is no direct relationship between the occurrence of ICANS and CAR T-cells. Preclinical studies have shown that IL-1, IL-6, and granulocyte macrophage colony stimulating factor (GMCSF) are the most important factors for the development of ICAN after CAR T-cell therapy [107]. In addition, infection events during treatment need to be closely monitored. The occurrence of infection is closely related to the patient’s blood immunodeficiency. The reasons include tumor cell suppression of normal immune cells, lymphatic depletion, and subsequent systemic reactions to serious complications [108]. The mechanism of these side effects is variable and complex, and it may require a professional team devoted to management of side effects, making it an important means to promote efficacy.

7. Conclusions

In this review, we discussed the clinical application strategies of CAR T-cells, which play a crucial role in the process of CAR T-cell therapy (Figure 2). Lymphodepletion regimen, dosing strategy, and side effect management are indispensable steps for CAR T-cell therapy. These processes should be taken into careful consideration before and during treatment to optimize the efficacy and reduce the severity of side effects. Furthermore, the development of combination therapies helps maximize CAR T-cell function, which is increasingly studied. CAR T-cells combined with allo-HSCT is a promising strategy that may cure hematological malignancy by overcoming tumor escape mechanisms and improving anti-tumor activity and persistence. In addition, the efficacy and safety of CAR T-cell application depends on the prediction and management of side effects. The monitoring of CRS and ICANS can predict the prognosis of patients for earlier intervention, especially monitoring changes in cytokines. In the future, CAR T-cells will be widely applied to cancer, including hematologic malignancy and solid tumors, due to the high efficiency of the anti-tumor effects. The exact CAR T-cell efficacy is based on the patient’s underlying disease, disease burden, and immune environment, which are all uncontrollable factors. However, when the patient is in the treatment program, ensuring that the patient obtains the longest complete remission time and disease-free survival depends on the coordination of clinical measures. As we mentioned previously, these factors are relatively controllable. More clinical trials should be investigated to explore the effect of lymphatic depletion and dose on peak CAR T-cell expansion, tumor killing, and the occurrence of side effects, which may relate to product type selection, disease burden, and modulation of the immune environment. In many ways, this can help clinicians and researchers optimize CAR T-cell therapy, expand the therapeutic window, and improve the availability of this emerging cancer immunotherapy.

Author Contributions

R.W. and W.H. contributed to the conception of the article. Q.L. wrote the first draft of the manuscript. Q.L., Z.L., R.W. and W.H. contributed to manuscript revision and reviewed and approved the submitted version. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Guangdong Basic and Applied Basic Research Foundation (No.2019A1515011854, to Rongxue Wan), National Natural Science Foundation of China (No.61427807, to Wenhua Huang), and the Introduced Major Research and Development Project Funded by Fujian Province (No. 2012I2004, to Wenhua Huang).

Acknowledgments

We would like to acknowledge the reviewers for their helpful comments on this paper.

Conflicts of Interest

The authors declare that they have no competing interests.

Abbreviations

CAR: Chimeric antigen receptor; FDA: The US Food and Drug Administration; B-ALL: B-cell acute lymphoblastic leukemia; CRS: Cytokine release syndrome; ICANS: Immune effector cell-associated neurotoxicity syndrome; CR: Complete remission; scFv: The single-chain variable fragment; Cy: Cyclophosphamide; Flu: Fludarabine; HD-Cy: High-dose cyclophosphamide; LD-Cy: Low-dose cyclophosphamide; PFS: Progression-free survival; DLBCL: Diffuse large B-cell lymphoma; MRD-CR: Minimal residual disease-negative complete remission; PBMC: The peripheral blood mononuclear cells; MTD: The maximally tolerated dose; NE: Neurological events; NHL: Non-Hodgkin’s lymphoma; allo-HSCT: Allogeneic hematopoietic stem cell transplantation; EFS: Event-free survival; aGVHD: Acute graft-versus-host disease; RFS: Relapse-free survival; CLL: Chronic lymphocytic leukemia; AICD: Activation-induced cell death; PI3K: Phosphoinositide 3-kinase; CRP: C-reactive protein; DIC: Disseminated intravascular coagulation; NF-κB: nuclear transcription factor-κB; GMCSF: Granulocyte macrophage colony stimulating factor (GMCSF).

References

  1. Enblad, G.; Karlsson, H.; Gammelgård, G.; Wenthe, J.; Lövgren, T.; Amini, R.M.; Wikstrom, K.I.; Essand, M.; Savoldo, B.; Hallböök, H.; et al. A Phase I/IIa Trial Using CD19-Targeted Third-Generation CAR T Cells for Lymphoma and Leukemia. Clin. Cancer Res. 2018, 24, 6185–6194. [Google Scholar] [CrossRef] [PubMed]
  2. Duan, D.; Wang, K.; Wei, C.; Feng, D.; Liu, Y.; He, Q.; Xu, X.; Wang, C.; Zhao, S.; Lv, L.; et al. The BCMA-Targeted Fourth-Generation CAR-T Cells Secreting IL-7 and CCL19 for Therapy of Refractory/Recurrent Multiple Myeloma. Front. Immunol. 2021, 12, 609421. [Google Scholar] [CrossRef] [PubMed]
  3. Abramson, J.S.; Palomba, M.L.; Gordon, L.I.; Lunning, M.A.; Wang, M.; Arnason, J.; Mehta, A.; Purev, E.; Maloney, D.G.; 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 2020, 396, 839–852. [Google Scholar] [CrossRef]
  4. Schuster, S.J.; Bishop, M.R.; Tam, C.S.; Waller, E.K.; Borchmann, P.; McGuirk, J.P.; Jäger, U.; Jaglowski, S.; Andreadis, C.; Westin, J.R.; et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2019, 380, 45–56. [Google Scholar] [CrossRef] [PubMed]
  5. Melenhorst, J.J.; Chen, G.M.; Wang, M.; Porter, D.L.; Chen, C.; Collins, M.A.; Gao, P.; Bandyopadhyay, S.; Sun, H.; Zhao, Z.; et al. Decade-long leukaemia remissions with persistence of CD4+ CAR T cells. Nature 2022, 602, 503–509. [Google Scholar] [CrossRef]
  6. Locke, F.L.; Ghobadi, A.; Jacobson, C.A.; Miklos, D.B.; Lekakis, L.J.; Oluwole, O.O.; Lin, Y.; Braunschweig, I.; Hill, B.T.; Timmerman, J.M.; et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): A single-arm, multicentre, phase 1–2 trial. Lancet Oncol. 2019, 20, 31–42. [Google Scholar] [CrossRef]
  7. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef]
  8. Cordeiro, A.; Bezerra, E.D.; Hirayama, A.V.; Hill, J.A.; Wu, Q.V.; Voutsinas, J.; Sorror, M.L.; Turtle, C.J.; Maloney, D.G.; Bar, M. Late Events after Treatment with CD19-Targeted Chimeric Antigen Receptor Modified T Cells. Biol. Blood Marrow Transplant. 2020, 26, 26–33. [Google Scholar] [CrossRef] [PubMed]
  9. Bao, F.; Wan, W.; He, T.; Qi, F.; Liu, G.; Hu, K.; Lu, X.-A.; Yang, P.; Dong, F.; Wang, J.; et al. Autologous CD19-directed chimeric antigen receptor-T cell is an effective and safe treatment to refractory or relapsed diffuse large B-cell lymphoma. Cancer Gene Ther. 2019, 26, 248–255. [Google Scholar] [CrossRef]
  10. Heng, G.; Jia, J.; Li, S.; Fu, G.; Wang, M.; Qin, D.; Li, Y.; Pei, L.; Tian, X.; Zhang, J.; et al. Sustained Therapeutic Efficacy of Humanized Anti-CD19 Chimeric Antigen Receptor T Cells in Relapsed/Refractory Acute Lymphoblastic Leukemia. Clin. Cancer Res. 2020, 26, 1606–1615. [Google Scholar] [CrossRef] [Green Version]
  11. Curran, K.J.; Margossian, S.P.; Kernan, N.A.; Silverman, L.B.; Williams, D.A.; Shukla, N.; Kobos, R.; Forlenza, C.J.; Steinherz, P.; Prockop, S.; et al. Toxicity and response after CD19-specific CAR T-cell therapy in pediatric/young adult relapsed/refractory B-ALL. Blood 2019, 134, 2361–2368. [Google Scholar] [CrossRef] [PubMed]
  12. 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. 2019, 6, e521–e529. [Google Scholar] [CrossRef]
  13. Pan, J.; Zuo, S.; Wang, Z. Sequential CD19-22 CAR T therapy induces sustained remission in children with r/r B-ALL. Blood 2020, 135, 387–391. [Google Scholar] [CrossRef] [PubMed]
  14. Shah, N.N.; Johnson, B.D.; Schneider, D.; Zhu, F.; Szabo, A.; Keever-Taylor, C.A.; Krueger, W.; Worden, A.A.; Kadan, M.J.; Yim, S.; et al. Bispecific anti-CD20, anti-CD19 CAR T cells for relapsed B cell malignancies: A phase 1 dose escalation and expansion trial. Nat. Med. 2020, 26, 1569–1575. [Google Scholar] [CrossRef]
  15. Rafiq, S.; Yeku, O.O.; Jackson, H.J.; Purdon, T.J.; Van Leeuwen, D.G.; Drakes, D.J.; Song, M.; Miele, M.M.; Li, Z.; Wang, P.; et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat. Biotechnol. 2018, 36, 847–856. [Google Scholar] [CrossRef]
  16. Weng, J.; Lai, P.; Qin, L.; Lai, Y.; Jiang, Z.; Luo, C.; Huang, X.; Wu, S.; Shao, D.; Deng, C.; et al. A novel generation 1928zT2 CAR T cells induce remission in extramedullary relapse of acute lymphoblastic leukemia. J. Hematol. Oncol. 2018, 11, 25. [Google Scholar] [CrossRef]
  17. Heczey, A.; Louis, C.U.; Savoldo, B.; Dakhova, O.; Durett, A.; Grilley, B.; Liu, H.; Wu, M.F.; Mei, Z.; Gee, A.; et al. CAR T Cells Administered in Combination with Lymphodepletion and PD-1 Inhibition to Patients with Neuroblastoma. Mol. Ther. 2017, 25, 2214–2224. [Google Scholar] [CrossRef]
  18. Adusumilli, P.S.; Zauderer, M.G.; Rivière, I.; Solomon, S.B.; Rusch, V.W.; O’Cearbhaill, R.E.; Zhu, A.; Cheema, W.; Chintala, N.K.; Halton, E.; et al. A Phase I Trial of Regional Mesothelin-Targeted CAR T-cell Therapy in Patients with Malignant Pleural Disease, in Combination with the Anti–PD-1 Agent Pembrolizumab. Cancer Discov. 2021, 11, 2748–2763. [Google Scholar] [CrossRef]
  19. Wei, J.; Xiao, M.; Mao, Z.; Na Wang, N.; Cao, Y.; Xiao, Y.; Meng, F.; Sun, W.; Wang, Y.; Yang, X.; et al. Outcome of aggressive B-cell lymphoma with TP53 alterations administered with CAR T-cell cocktail alone or in combination with ASCT. Signal Transduct. Target. Ther. 2022, 7, 101. [Google Scholar] [CrossRef] [PubMed]
  20. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef]
  21. Jacobson, C.A.; Chavez, J.C.; Sehgal, A.R.; William, B.M.; Munoz, J.; Salles, G.; Munshi, P.N.; Casulo, C.; Maloney, D.G.; de Vos, S.; et al. Axicabtagene ciloleucel in relapsed or refractory indolent non-Hodgkin lymphoma (ZUMA-5): A single-arm, multicentre, phase 2 trial. Lancet Oncol. 2022, 23, 91–103. [Google Scholar] [CrossRef]
  22. Schuster, S.J.; Tam, C.S.; Borchmann, P.; Worel, N.; McGuirk, J.P.; Holte, H.; Waller, E.K.; Jaglowski, S.; Bishop, M.R.; Damon, L.E.; et al. Long-term clinical outcomes of tisagenlecleucel in patients with relapsed or refractory aggressive B-cell lymphomas (JULIET): A multicentre, open-label, single-arm, phase 2 study. Lancet Oncol. 2021, 22, 1403–1415. [Google Scholar] [CrossRef]
  23. Fowler, N.H.; Dickinson, M.; Dreyling, M.; Martinez-Lopez, J.; Kolstad, A.; Butler, J.; Ghosh, M.; Popplewell, L.; Chavez, J.C.; Bachy, E.; et al. Tisagenlecleucel in adult relapsed or refractory follicular lymphoma: The phase 2 ELARA trial. Nat. Med. 2022, 28, 325–332. [Google Scholar] [CrossRef]
  24. Shah, B.D.; Bishop, M.R.; Oluwole, O.O.; Logan, A.C.; Baer, M.R.; Donnellan, W.B.; O’Dwyer, K.M.; Holmes, H.; Arellano, M.L.; Ghobadi, A.; et al. KTE-X19 anti-CD19 CAR T-cell therapy in adult relapsed/refractory acute lymphoblastic leukemia: ZUMA-3 phase 1 results. Blood 2021, 138, 11–22. [Google Scholar] [CrossRef]
  25. Wang, M.; Munoz, J.; Goy, A.; Locke, F.L.; Jacobson, C.A.; Hill, B.T.; Timmerman, J.M.; Holmes, H.; Jaglowski, S.; Flinn, I.W.; et al. KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. N. Engl. J. Med. 2020, 382, 1331–1342. [Google Scholar] [CrossRef]
  26. Mackall, C.L.; Hakim, F.T.; Gress, R.E. Restoration of T-cell homeostasis after T-cell depletion. Semin. Immunol. 1997, 9, 339–346. [Google Scholar] [CrossRef] [PubMed]
  27. Klebanoff, C.; Khong, H.T.; Antony, P.A.; Palmer, D.C.; Restifo, N.P. Sinks, suppressors and antigen presenters: How lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol. 2005, 26, 111–117. [Google Scholar] [CrossRef]
  28. Dummer, W.; Niethammer, A.G.; Baccala, R.; Lawson, B.R.; Wagner, N.; Reisfeld, R.A.; Theofilopoulos, A.N. T cell homeostatic proliferation elicits effective antitumor autoimmunity. J. Clin. Investig. 2002, 110, 185–192. [Google Scholar] [CrossRef]
  29. Muranski, P.; Boni, A.; Wrzesinski, C.; Citrin, D.; Rosenberg, S.A.; Childs, R.W.; Restifo, N.P. Increased intensity lymphodepletion and adoptive immunotherapy—how far can we go? Nat. Clin. Pract. Oncol. 2006, 3, 668–681. [Google Scholar] [CrossRef]
  30. Wrzesinski, C.; Restifo, N.P. Less is more: Lymphodepletion followed by hematopoietic stem cell transplant augments adoptive T-cell-based anti-tumor immunotherapy. Curr. Opin. Immunol. 2005, 17, 195–201. [Google Scholar] [CrossRef] [Green Version]
  31. Gattinoni, L.; Finkelstein, S.E.; Klebanoff, C.; Antony, P.A.; Palmer, D.; Spiess, P.J.; Hwang, L.N.; Yu, Z.; Wrzesinski, C.; Heimann, D.M.; et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J. Exp. Med. 2005, 202, 907–912. [Google Scholar] [CrossRef] [PubMed]
  32. Hay, K.A.; Gauthier, J.; Hirayama, A.V.; Voutsinas, J.M.; Wu, Q.; Li, D.; Gooley, T.A.; Cherian, S.; Chen, X.; Pender, B.S.; et al. Factors associated with durable EFS in adult B-cell ALL patients achieving MRD-negative CR after CD19 CAR T-cell therapy. Blood 2019, 133, 1652–1663. [Google Scholar] [CrossRef] [PubMed]
  33. Vitale, C.; Strati, P. CAR T-Cell Therapy for B-Cell non-Hodgkin Lymphoma and Chronic Lymphocytic Leukemia: Clinical Trials and Real-World Experiences. Front. Oncol. 2020, 10, 849. [Google Scholar] [CrossRef]
  34. Locke, F.L.; Neelapu, S.S.; Bartlett, N.L.; Siddiqi, T.; Chavez, J.C.; Hosing, C.M.; Ghobadi, A.; Budde, L.E.; Bot, A.; Rossi, J.M.; et al. Phase 1 Results of ZUMA-1: A Multicenter Study of KTE-C19 Anti-CD19 CAR T Cell Therapy in Refractory Aggressive Lymphoma: A Multicenter Study of KTE-C19 Anti-CD19 CAR T Cell Therapy in Refractory Aggressive Lymphoma. Mol. Ther. 2017, 25, 285–295. [Google Scholar] [CrossRef]
  35. Tang, X.-Y.; Sun, Y.; Zhang, A.; Hu, G.-L.; Cao, W.; Wang, D.-H.; Zhang, B.; Chen, H. Third-generation CD28/4-1BB chimeric antigen receptor T cells for chemotherapy relapsed or refractory acute lymphoblastic leukaemia: A non-randomised, open-label phase I trial protocol. BMJ Open 2016, 6, e013904. [Google Scholar] [CrossRef]
  36. Li, S.; Zhang, J.; Wang, M.; Fu, G.; Li, Y.; Pei, L.; Xiong, Z.; Qin, D.; Zhang, R.; Tian, X.; et al. Treatment of acute lymphoblastic leukaemia with the second generation of CD19 CAR-T containing either CD28 or 4-1BB. Br. J. Haematol. 2018, 181, 360–371. [Google Scholar] [CrossRef]
  37. Ying, Z.; Huang, X.F.; Xiang, X.; Liu, Y.; Kang, X.; Song, Y.; Guo, X.; Liu, H.; Ding, N.; Zhang, T.; et al. A safe and potent anti-CD19 CAR T cell therapy. Nat. Med. 2019, 25, 947–953. [Google Scholar] [CrossRef] [PubMed]
  38. Turtle, C.J.; Hanafi, L.-A.; Berger, C.; Gooley, T.A.; Cherian, S.; Hudecek, M.; Sommermeyer, D.; Melville, K.; Pender, B.; Budiarto, T.M.; et al. CD19 CAR–T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Investig. 2016, 126, 2123–2138. [Google Scholar] [CrossRef] [PubMed]
  39. Geyer, M.B.; Rivière, I.; Sénéchal, B.; Wang, X.; Wang, Y.; Purdon, T.J.; Hsu, M.; Devlin, S.M.; Halton, E.; Lamanna, N.; et al. Autologous CD19-Targeted CAR T Cells in Patients with Residual CLL following Initial Purine Analog-Based Therapy. Mol. Ther. 2018, 26, 1896–1905. [Google Scholar] [CrossRef]
  40. Turtle, C.J.; Hanafi, L.-A.; Berger, C.; Hudecek, M.; Pender, B.; Robinson, E.; Hawkins, R.; Chaney, C.; Cherian, S.; Chen, X.; et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8 + and CD4 + CD19-specific chimeric antigen receptor–modified T cells. Sci. Transl. Med. 2016, 8, 355ra116. [Google Scholar] [CrossRef] [Green Version]
  41. Geyer, M.B.; Rivière, I.; Sénéchal, B.; Wang, X.; Wang, Y.; Purdon, T.J.; Hsu, M.; Devlin, S.M.; Palomba, M.L.; Halton, E.; et al. Safety and tolerability of conditioning chemotherapy followed by CD19-targeted CAR T cells for relapsed/refractory CLL. JCI Insight 2019, 4, e122627. [Google Scholar] [CrossRef] [PubMed]
  42. Hirayama, A.V.; Gauthier, J.; Hay, K.A.; Voutsinas, J.M.; Wu, Q.; Gooley, T.; Li, D.; Cherian, S.; Chen, X.; Pender, B.S.; et al. The response to lymphodepletion impacts PFS in patients with aggressive non-Hodgkin lymphoma treated with CD19 CAR T cells. Blood 2019, 133, 1876–1887. [Google Scholar] [CrossRef] [PubMed]
  43. Zhao, H.; Wei, J.; Wei, G.; Luo, Y.; Shi, J.; Cui, Q.; Zhao, M.; Liang, A.; Zhang, Q.; Yang, J.; et al. Pre-transplant MRD negativity predicts favorable outcomes of CAR-T therapy followed by haploidentical HSCT for relapsed/refractory acute lymphoblastic leukemia: A multi-center retrospective study. J. Hematol. Oncol. 2020, 13, 42. [Google Scholar] [CrossRef] [PubMed]
  44. Kochenderfer, J.N.; Somerville, R.P.; Lu, T.; Shi, V.; Bot, A.; Rossi, J.; Xue, A.; Goff, S.L.; Yang, J.C.; Sherry, R.M.; et al. Lymphoma Remissions Caused by Anti-CD19 Chimeric Antigen Receptor T Cells Are Associated with High Serum Interleukin-15 Levels. J. Clin. Oncol. 2017, 35, 1803–1813. [Google Scholar] [CrossRef] [PubMed]
  45. Fabrizio, V.A.; Boelens, J.J.; Mauguen, A.; Baggott, C.; Prabhu, S.; Egeler, E.; Mavroukakis, S.; Pacenta, H.L.; Phillips, C.L.; Rossoff, J.; et al. Optimal fludarabine lymphodepletion is associated with improved outcomes after CAR T-cell therapy. Blood Adv. 2022, 6, 1961–1968. [Google Scholar] [CrossRef] [PubMed]
  46. Dekker, L.; Calkoen, F.G.; Jiang, Y.; Blok, H.; Veldkamp, S.R.; De Koning, C.C.H.; Spoon, M.; Admiraal, R.; Hoogerbrugge, P.; Vormoor, B.J.; et al. Fludarabine exposure predicts outcome after CD19 CAR T-cell therapy in children and young adults with acute leukemia. Blood Adv. 2022, 6, 1969–1976. [Google Scholar] [CrossRef] [PubMed]
  47. Dourthe, M.-E.; Rabian, F.; Yakouben, K.; Chevillon, F.; Cabannes-Hamy, A.; Méchinaud, F.; Grain, A.; Chaillou, D.; Rahal, I.; Caillat-Zucman, S.; et al. Determinants of CD19-positive vs CD19-negative relapse after tisagenlecleucel for B-cell acute lymphoblastic leukemia. Leukemia 2021, 35, 3383–3393. [Google Scholar] [CrossRef]
  48. Liang, Z.; Zhang, H.; Shao, M.; Cui, Q.; Wu, Z.; Xiao, L.; Huang, H.; Hu, Y. Lymphodepletion chemotherapy revitalizes chimeric antigen receptor T cells contributing to regression of relapsed B-cell lymphoma: A case report. Medicine 2020, 99, e22510. [Google Scholar] [CrossRef]
  49. Sadelain, M.; Rivière, I.; Riddell, S. Therapeutic T cell engineering. Nature 2017, 545, 423–431. [Google Scholar] [CrossRef]
  50. Tu, S.; Huang, R.; Guo, Z.; Deng, L.; Song, C.; Zhou, X.; Yue, C.; Zhang, L.; He, Y.; Yang, J.; et al. Shortening the ex vivo culture of CD19-specific CAR T-cells retains potent efficacy against acute lymphoblastic leukemia without CAR T-cell-related encephalopathy syndrome or severe cytokine release syndrome. Am. J. Hematol. 2019, 94, E322–E325. [Google Scholar] [CrossRef] [Green Version]
  51. Tumaini, B.; Lee, D.W.; Lin, T.; Castiello, L.; Stroncek, D.F.; Mackall, C.; Wayne, A.; Sabatino, M. Simplified process for the production of anti–CD19-CAR–engineered T cells. Cytotherapy 2013, 15, 1406–1415. [Google Scholar] [CrossRef]
  52. Zhang, C.; He, J.; Liu, L.; Wang, J.; Wang, S.; Liu, L.; Ge, J.; Gao, L.; Kong, P.; Liu, Y.; et al. Novel CD19 chimeric antigen receptor T cells manufactured next-day for acute lymphoblastic leukemia. Blood Cancer J. 2022, 12, 96. [Google Scholar] [CrossRef] [PubMed]
  53. Hong, M.; Clubb, J.D.; Chen, Y.Y. Engineering CAR-T Cells for Next-Generation Cancer Therapy. Cancer Cell 2020, 38, 473–488. [Google Scholar] [CrossRef]
  54. Li, D.H.; Whitmore, J.B.; Guo, W.; Ji, Y. Toxicity and Efficacy Probability Interval Design for Phase I Adoptive Cell Therapy Dose-Finding Clinical Trials. Clin. Cancer Res. 2017, 23, 13–20. [Google Scholar] [CrossRef] [PubMed]
  55. Shah, B.D.; Ghobadi, A.; Oluwole, O.O.; Logan, A.C.; Boissel, N.; Cassaday, R.D.; Leguay, T.; Bishop, M.R.; Topp, M.S.; Tzachanis, D.; et al. 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, 491–502. [Google Scholar] [CrossRef]
  56. Lee, D.W.; Kochenderfer, J.N.; Stetler-Stevenson, M.; Cui, Y.K.; Delbrook, C.; Feldman, S.A.; Fry, T.J.; Orentas, R.; Sabatino, M.; Shah, N.N.; 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 2015, 385, 517–528. [Google Scholar] [CrossRef]
  57. Pan, J.; Yang, J.F.; Deng, B.P.; Zhao, X.J.; Zhang, X.; Lin, Y.H.; Wu, Y.N.; Deng, Z.L.; Zhang, Y.L.; Liu, S.H.; et al. High efficacy and safety of low-dose CD19-directed CAR-T cell therapy in 51 refractory or relapsed B acute lymphoblastic leukemia patients. Leukemia 2017, 31, 2587–2593. [Google Scholar] [CrossRef]
  58. Yan, Z.-X.; Li, L.; Wang, W.; OuYang, B.-S.; Cheng, S.; Wang, L.; Wu, W.; Xu, P.-P.; Muftuoglu, M.; Hao, M.; et al. Clinical Efficacy and Tumor Microenvironment Influence in a Dose-Escalation Study of Anti-CD19 Chimeric Antigen Receptor T Cells in Refractory B-Cell Non-Hodgkin’s Lymphoma. Clin. Cancer Res. 2019, 25, 6995–7003. [Google Scholar] [CrossRef]
  59. Frey, N.V.; Shaw, P.A.; Hexner, E.O.; Pequignot, E.; Gill, S.; Luger, S.M.; Mangan, J.K.; Loren, A.W.; Perl, A.E.; Maude, S.L.; et al. Optimizing Chimeric Antigen Receptor T-Cell Therapy for Adults with Acute Lymphoblastic Leukemia. J. Clin. Oncol. 2020, 38, 415–422. [Google Scholar] [CrossRef]
  60. Anagnostou, T.; Riaz, I.B.; Hashmi, S.K.; Murad, M.H.; Kenderian, S.S. Anti-CD19 chimeric antigen receptor T-cell therapy in acute lymphocytic leukaemia: A systematic review and meta-analysis. Lancet Haematol. 2020, 7, e816–e826. [Google Scholar] [CrossRef]
  61. Ma, Y.; Zhang, S.; Fang, H.; Yu, K.; Jiang, S. A phase I study of CAR-T bridging HSCT in patients with acute CD19+ relapse/refractory B-cell leukemia. Oncol. Lett. 2020, 20, 20. [Google Scholar] [CrossRef]
  62. Li, Q.; Mu, J.; Yuan, J.; Yang, Z.; Wang, J.; Deng, Q. Low Level Donor Chimerism of CD19 CAR-T Cells Returned to Complete Donor Chimerism in Patients with Relapse After Allo-Hematopoietic Stem Cell Transplant. OncoTargets Ther. 2020, 13, 11471–11484. [Google Scholar] [CrossRef]
  63. Wen, S.; Niu, Z.; Xing, L.; Wang, Y.; Li, H.; Kuang, N.; Luo, J.; Zhang, X.; Wang, F. CAR-T bridging to allo-HSCT as a treatment strategy for relapsed adult acute B-lymphoblastic leukemia: A case report. BMC Cancer 2018, 18, 1143. [Google Scholar] [CrossRef]
  64. Hua, J.; Zhang, J.; Wu, X.; Zhou, L.; Bao, X.; Han, Y.; Miao, M.; Li, C.; Fu, Z.; Wu, D.; et al. Allogeneic Donor-Derived Anti-CD19 CAR T Cell Is a Promising Therapy for Relapsed/Refractory B-ALL After Allogeneic Hematopoietic Stem-Cell Transplantation. Clin. Lymphoma Myeloma Leuk. 2020, 20, 610–616. [Google Scholar] [CrossRef] [PubMed]
  65. Hua, J.; Zhang, J.; Zhang, X.; Wu, X.; Zhou, L.; Bao, X.; Han, Y.; Miao, M.; Li, C.; Fu, C.; et al. Donor-derived anti-CD19 CAR T cells compared with donor lymphocyte infusion for recurrent B-ALL after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2021, 56, 1056–1064. [Google Scholar] [CrossRef]
  66. González-Barca, E.; Boumendil, A.; Blaise, D.; Trněný, M.; Masszi, T.; Finel, H.; Michieli, M.; Bittenbring, J.T.; Gritti, G.; Snowden, J.A.; et al. Outcome in patients with diffuse large B-cell lymphoma who relapse after autologous stem cell transplantation and receive active therapy. A retrospective analysis of the Lymphoma Working Party of the European Society for Blood and Marrow Transplantation (EBMT). Bone Marrow Transplant. 2020, 55, 393–399. [Google Scholar] [CrossRef]
  67. Styczynski, J.; Tallamy, B.; Waxman, I.; Van De Ven, C.; Milone, M.C.; Shaw, L.M.; Harrison, L.; Morris, E.; Satwani, P.; Bhatia, M.; et al. A pilot study of reduced toxicity conditioning with BU, fludarabine and alemtuzumab before the allogeneic hematopoietic SCT in children and adolescents. Bone Marrow Transplant. 2011, 46, 790–799. [Google Scholar] [CrossRef]
  68. Zhang, Y.; Chen, H.; Song, Y.; Tan, X.; Zhao, Y.; Liu, X.; Li, Z.; Yang, F.; Jiang, M.; Gao, Z.; et al. Chimeric antigen receptor T-cell therapy as a bridge to haematopoietic stem cell transplantation for refractory/relapsed B-cell acute lymphoblastic leukemia. Br. J. Haematol. 2020, 189, 146–152. [Google Scholar] [CrossRef] [PubMed]
  69. Jiang, H.; Li, C.; Yin, P.; Guo, T.; Liu, L.; Xia, L.; Wu, Y.; Zhou, F.; Ai, L.; Shi, W.; et al. Anti-CD19 chimeric antigen receptor-modified T-cell therapy bridging to allogeneic hematopoietic stem cell transplantation for relapsed/refractory B-cell acute lymphoblastic leukemia: An open-label pragmatic clinical trial. Am. J. Hematol. 2019, 94, 1113–1122. [Google Scholar] [CrossRef] [PubMed]
  70. Shi, X.; Yan, L.; Shang, J.; Kang, L.; Yan, Z.; Jin, S.; Zhu, M.; Chang, H.; Gong, F.; Zhou, J.; et al. Anti-CD19 and anti-BCMA CAR T cell therapy followed by lenalidomide maintenance after autologous stem-cell transplantation for high-risk newly diagnosed multiple myeloma. Am. J. Hematol. 2022, 97, 537–547. [Google Scholar] [CrossRef] [PubMed]
  71. Otáhal, P.; Prukova, D.; Král, V.; Fabry, M.; Vočková, P.; Latečková, L.; Trneny, M.; Klener, P. Lenalidomide enhances antitumor functions of chimeric antigen receptor modified T cells. OncoImmunology 2016, 5, e1115940. [Google Scholar] [CrossRef] [PubMed]
  72. Works, M.; Soni, N.; Hauskins, C.; Sierra, C.; Baturevych, A.; Jones, J.C.; Curtis, W.; Carlson, P.; Johnstone, T.G.; Kugler, D.; et al. Anti–B-cell Maturation Antigen Chimeric Antigen Receptor T cell Function against Multiple Myeloma Is Enhanced in the Presence of Lenalidomide. Mol. Cancer Ther. 2019, 18, 2246–2257. [Google Scholar] [CrossRef] [PubMed]
  73. Fraietta, J.A.; Beckwith, K.A.; Patel, P.R.; Ruella, M.; Zheng, Z.; Barrett, D.M.; Lacey, S.F.; Melenhorst, J.J.; McGettigan, S.E.; Cook, D.R.; et al. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood 2016, 127, 1117–1127. [Google Scholar] [CrossRef] [PubMed]
  74. Shanafelt, T.D.; Wang, X.V.; Kay, N.E.; Hanson, C.A.; O’Brien, S.; Barrientos, J.; Jelinek, D.F.; Braggio, E.; Leis, J.F.; Zhang, C.C.; et al. Ibrutinib–Rituximab or Chemoimmunotherapy for Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2019, 381, 432–443. [Google Scholar] [CrossRef] [PubMed]
  75. Turtle, C.J.; Hay, K.; Hanafi, L.-A.; Li, D.; Cherian, S.; Chen, X.; Wood, B.; Lozanski, A.; Byrd, J.C.; Heimfeld, S.; et al. Durable Molecular Remissions in Chronic Lymphocytic Leukemia Treated with CD19-Specific Chimeric Antigen Receptor–Modified T Cells After Failure of Ibrutinib. J. Clin. Oncol. 2017, 35, 3010–3020. [Google Scholar] [CrossRef] [PubMed]
  76. Ruella, M.; Kenderian, S.S.; Shestova, O.; Fraietta, J.A.; Qayyum, S.; Zhang, Q.; Maus, M.V.; Liu, X.; Nunez-Cruz, S.; Klichinsky, M.; et al. The Addition of the BTK Inhibitor Ibrutinib to Anti-CD19 Chimeric Antigen Receptor T Cells (CART19) Improves Responses against Mantle Cell Lymphoma. Clin. Cancer Res. 2016, 22, 2684–2696. [Google Scholar] [CrossRef]
  77. Gauthier, J.; Hirayama, A.V.; Purushe, J.; Hay, K.A.; Lymp, J.; Li, D.H.; Yeung, C.C.S.; Sheih, A.; Pender, B.S.; Hawkins, R.M.; et al. Feasibility and efficacy of CD19-targeted CAR T cells with concurrent ibrutinib for CLL after ibrutinib failure. Blood 2020, 135, 1650–1660. [Google Scholar] [CrossRef]
  78. Gulati, P.; Rühl, J.; Kannan, A.; Pircher, M.; Schuberth, P.; Nytko, K.J.; Pruschy, M.N.; Sulser, S.; Haefner, M.D.; Jensen, S.M.; et al. Aberrant Lck Signal via CD28 Costimulation Augments Antigen-Specific Functionality and Tumor Control by Redirected T Cells with PD-1 Blockade in Humanized Mice. Clin. Cancer Res. 2018, 24, 3981–3993. [Google Scholar] [CrossRef]
  79. Gargett, T.; Yu, W.; Dotti, G.; Yvon, E.S.; Christo, S.N.; Hayball, J.D.; Lewis, I.D.; Brenner, M.K.; Brown, M.P. GD2-specific CAR T Cells Undergo Potent Activation and Deletion Following Antigen Encounter but can be Protected from Activation-induced Cell Death by PD-1 Blockade. Mol. Ther. 2016, 24, 1135–1149. [Google Scholar] [CrossRef]
  80. Bansal, R.; Reshef, R. Revving the CAR—Combination strategies to enhance CAR T cell effectiveness. Blood Rev. 2021, 45, 100695. [Google Scholar] [CrossRef]
  81. Chou, C.K.; Turtle, C.J. Insight into mechanisms associated with cytokine release syndrome and neurotoxicity after CD19 CAR-T cell immunotherapy. Bone Marrow Transplant. 2019, 54, 780–784. [Google Scholar] [CrossRef]
  82. Sesques, P.; Ferrant, E.; Safar, V.; Wallet, F.; Tordo, J.; Dhomps, A.; Karlin, L.; Brisou, G.; Vercasson, M.; Msc, C.H.; et al. Commercial anti-CD19 CAR T cell therapy for patients with relapsed/refractory aggressive B cell lymphoma in a European center. Am. J. Hematol. 2020, 95, 1324–1333. [Google Scholar] [CrossRef] [PubMed]
  83. Goto, H.; Makita, S.; Kato, K.; Tokushige, K.; Fujita, T.; Akashi, K.; Izutsu, K.; Teshima, T. Efficacy and safety of tisagenlecleucel in Japanese adult patients with relapsed/refractory diffuse large B-cell lymphoma. Int. J. Clin. Oncol. 2020, 25, 1736–1743. [Google Scholar] [CrossRef] [PubMed]
  84. Lee, D.W.; Santomasso, B.D.; Locke, F.L.; Ghobadi, A.; Turtle, C.J.; Brudno, J.N.; Maus, M.V.; Park, J.H.; Mead, E.; Pavletic, S.; et al. ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Immune Effector Cells. Biol. Blood Marrow Transplant. 2019, 25, 625–638. [Google Scholar] [CrossRef] [PubMed]
  85. Jiang, H.; Liu, L.; Guo, T.; Wu, Y.; Ai, L.; Deng, J.; Dong, J.; Mei, H.; Hu, Y. Improving the safety of CAR-T cell therapy by controlling CRS-related coagulopathy. Ann. Hematol. 2019, 98, 1721–1732. [Google Scholar] [CrossRef]
  86. Hu, Y.; Wu, Z.; Luo, Y.; Shi, J.; Yu, J.; Pu, C.; Liang, Z.; Wei, G.; Cui, Q.; Sun, J.; et al. Potent Anti-leukemia Activities of Chimeric Antigen Receptor–Modified T Cells against CD19 in Chinese Patients with Relapsed/Refractory Acute Lymphocytic Leukemia. Clin. Cancer Res. 2017, 23, 3297–3306. [Google Scholar] [CrossRef]
  87. Ma, F.; Ho, J.; Du, H.; Xuan, F.; Wu, X.; Wang, Q.; Wang, L.; Liu, Y.; Ba, M.; Wang, Y.; et al. Evidence of long-lasting anti-CD19 activity of engrafted CD19 chimeric antigen receptor–modified T cells in a phase I study targeting pediatrics with acute lymphoblastic leukemia. Hematol. Oncol. 2019, 37, 601–608. [Google Scholar] [CrossRef]
  88. Park, J.H.; Romero, F.A.; Taur, Y.; Sadelain, M.; Brentjens, R.J.; Hohl, T.M.; Seo, S.K. Cytokine Release Syndrome Grade as a Predictive Marker for Infections in Patients with Relapsed or Refractory B-Cell Acute Lymphoblastic Leukemia Treated with Chimeric Antigen Receptor T Cells. Clin. Infect. Dis. 2018, 67, 533–540. [Google Scholar] [CrossRef]
  89. Magnani, C.F.; Gaipa, G.; Lussana, F.; Belotti, D.; Gritti, G.; Napolitano, S.; Matera, G.; Cabiati, B.; Buracchi, C.; Borleri, G.; et al. Sleeping Beauty–engineered CAR T cells achieve antileukemic activity without severe toxicities. J. Clin. Investig. 2020, 130, 6021–6033. [Google Scholar] [CrossRef]
  90. Hu, Y.; Sun, J.; Wu, Z.; Yu, J.; Cui, Q.; Pu, C.; Liang, B.; Luo, Y.; Shi, J.; Jin, A.; et al. Predominant cerebral cytokine release syndrome in CD19-directed chimeric antigen receptor-modified T cell therapy. J. Hematol. Oncol. 2016, 9, 70. [Google Scholar] [CrossRef] [Green Version]
  91. Wrzesinski, C.; Paulos, C.M.; Kaiser, A.; Muranski, P.; Palmer, D.; Gattinoni, L.; Yu, Z.; Rosenberg, S.A.; Restifo, N.P. Increased Intensity Lymphodepletion Enhances Tumor Treatment Efficacy of Adoptively Transferred Tumor-specific T Cells. J. Immunother. 2010, 33, 1–7. [Google Scholar] [CrossRef] [PubMed]
  92. Ninomiya, S.; Narala, N.; Huye, L.; Yagyu, S.; Savoldo, B.; Dotti, G.; Heslop, H.E.; Brenner, M.K.; Rooney, C.M.; Ramos, C.A. Tumor indoleamine 2,3-dioxygenase (IDO) inhibits CD19-CAR T cells and is downregulated by lymphodepleting drugs. Blood 2015, 125, 3905–3916. [Google Scholar] [CrossRef]
  93. Ananthakrishnan, R.; Green, S.; Li, D.; LaValley, M. Extensions of the mTPI and TEQR designs to include non-monotone efficacy in addition to toxicity for optimal dose determination for early phase immunotherapy oncology trials. Contemp. Clin. Trials Commun. 2018, 10, 62–76. [Google Scholar] [CrossRef]
  94. Chiuzan, C.; Garrett-Mayer, E.; Nishimura, M.I. An Adaptive Dose-Finding Design Based on Both Safety and Immunologic Responses in Cancer Clinical Trials. Stat. Biopharm. Res. 2018, 10, 185–195. [Google Scholar] [CrossRef] [PubMed]
  95. Rovatti, P.E.; Gambacorta, V.; Lorentino, F.; Ciceri, F.; Vago, L. Mechanisms of Leukemia Immune Evasion and Their Role in Relapse After Haploidentical Hematopoietic Cell Transplantation. Front. Immunol. 2020, 11, 147. [Google Scholar] [CrossRef] [PubMed]
  96. Chmielewski, M.; Hombach, A.A.; Abken, H. Antigen-Specific T-Cell Activation Independently of the MHC: Chimeric Antigen Receptor-Redirected T Cells. Front. Immunol. 2013, 4, 371. [Google Scholar] [CrossRef]
  97. Leblanc, R.; Hideshima, T.; Catley, L.P.; Shringarpure, R.; Burger, R.; Mitsiades, N.; Mitsiades, C.; Cheema, P.; Chauhan, D.; Richardson, P.G.; et al. Immunomodulatory drug costimulates T cells via the B7-CD28 pathway. Blood 2004, 103, 1787–1790. [Google Scholar] [CrossRef]
  98. Davenport, A.J.; Cross, R.S.; Watson, K.A.; Liao, Y.; Shi, W.; Prince, H.M.; Beavis, P.A.; Trapani, J.A.; Kershaw, M.H.; Ritchie, D.S.; et al. Chimeric antigen receptor T cells form nonclassical and potent immune synapses driving rapid cytotoxicity. Proc. Natl. Acad. Sci. USA 2018, 115, E2068–E2076. [Google Scholar] [CrossRef]
  99. Loschinski, R.; Böttcher, M.; Stoll, A.; Bruns, H.; Mackensen, A.; Mougiakakos, D. IL-21 modulates memory and exhaustion phenotype of T-cells in a fatty acid oxidation-dependent manner. Oncotarget 2018, 9, 13125–13138. [Google Scholar] [CrossRef]
  100. Browning, R.L.; Byrd, W.H.; Gupta, N.; Jones, J.; Mo, X.; Hertlein, E.; Yu, L.; Muthusamy, N.; Byrd, J.C. Lenalidomide Induces Interleukin-21 Production by T Cells and Enhances IL21-Mediated Cytotoxicity in Chronic Lymphocytic Leukemia B Cells. Cancer Immunol. Res. 2016, 4, 698–707. [Google Scholar] [CrossRef] [Green Version]
  101. Chen, S.-S.; Chang, B.Y.; Chang, S.; Tong, T.; Ham, S.; Sherry, B.; Burger, J.A.; Rai, K.R.; Chiorazzi, N. BTK inhibition results in impaired CXCR4 chemokine receptor surface expression, signaling and function in chronic lymphocytic leukemia. Leukemia 2016, 30, 833–843. [Google Scholar] [CrossRef] [PubMed]
  102. Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61. [Google Scholar] [CrossRef]
  103. Li, S.; Xue, L.; Wang, M.; Qiang, P.; Xu, H.; Zhang, X.; Kang, W.; You, F.; Xu, H.; Wang, Y.; et al. Decitabine enhances cytotoxic effect of T cells with an anti-CD19 chimeric antigen receptor in treatment of lymphoma. OncoTargets Ther. 2019, 12, 5627–5638. [Google Scholar] [CrossRef] [PubMed]
  104. Titov, A.; Petukhov, A.; Staliarova, A.; Motorin, D.; Bulatov, E.; Shuvalov, O.; Soond, S.M.; Piacentini, M.; Melino, G.; Zaritskey, A.; et al. The biological basis and clinical symptoms of CAR-T therapy-associated toxicites. Cell Death Dis. 2018, 9, 897. [Google Scholar] [CrossRef] [PubMed]
  105. Obstfeld, A.E.; Frey, N.V.; Mansfield, K.; Lacey, S.F.; June, C.H.; Porter, D.L.; Melenhorst, J.J.; Wasik, M.A. Cytokine release syndrome associated with chimeric-antigen receptor T-cell therapy: Clinicopathological insights. Blood 2017, 130, 2569–2572. [Google Scholar] [CrossRef]
  106. Mueller, K.T.; Maude, S.L.; Porter, D.L.; Frey, N.; Wood, P.; Han, X.; Waldron, E.; Chakraborty, A.; Awasthi, R.; Levine, B.L.; et al. Cellular kinetics of CTL019 in relapsed/refractory B-cell acute lymphoblastic leukemia and chronic lymphocytic leukemia. Blood 2017, 130, 2317–2325. [Google Scholar] [CrossRef]
  107. Taraseviciute, A.; Tkachev, V.; Ponce, R.; Turtle, C.J.; Snyder, J.M.; Liggitt, H.D.; Myerson, D.; Gonzalez-Cuyar, L.; Baldessari, A.; English, C.; et al. Chimeric Antigen Receptor T Cell–Mediated Neurotoxicity in Nonhuman Primates. Cancer Discov. 2018, 8, 750–763. [Google Scholar] [CrossRef]
  108. Wudhikarn, K.; Perales, M.-A. Infectious complications, immune reconstitution, and infection prophylaxis after CD19 chimeric antigen receptor T-cell therapy. Bone Marrow Transplant. 2022, 1–12. [Google Scholar] [CrossRef]
Figure 1. Molecular structure of CAR in four generations of CAR T cells. The CAR structure is embedded in T cells by genetic engineering and is mainly composed of a single-chain variable fragment (scFv), a transmembrane domain, a costimulatory domain (CD28 or 4-1BB), and an intracellular activation domain CD3ζ composition. CAR T-cells utilize the specific recognition of extracellular structures and the cytotoxicity of T cells to kill target tumor cells, successfully avoiding the restrictive effect of major histocompatibility complex (MHC) class I and improving the lethality of tumor cells. The first-generation CAR T-cell scFv couples with intracellular CD3ζ to obtain specific antigen recognition ability, and the second-generation CAR T-cells add the CD28 or 4-1BB costimulatory domain structure to overcome the persistent poor activation of T cells, low activation intensity, and cytokine secretion disorder. The third-generation CAR T-cells that combine both CD28 and 4-1BB costimulatory domains have improved cytotoxicity and persistence, while fourth-generation CAR T-cells with one costimulatory domain are genetically engineered to introduce specific genes to perform cytotoxic functions and maintain T cell activation.
Figure 1. Molecular structure of CAR in four generations of CAR T cells. The CAR structure is embedded in T cells by genetic engineering and is mainly composed of a single-chain variable fragment (scFv), a transmembrane domain, a costimulatory domain (CD28 or 4-1BB), and an intracellular activation domain CD3ζ composition. CAR T-cells utilize the specific recognition of extracellular structures and the cytotoxicity of T cells to kill target tumor cells, successfully avoiding the restrictive effect of major histocompatibility complex (MHC) class I and improving the lethality of tumor cells. The first-generation CAR T-cell scFv couples with intracellular CD3ζ to obtain specific antigen recognition ability, and the second-generation CAR T-cells add the CD28 or 4-1BB costimulatory domain structure to overcome the persistent poor activation of T cells, low activation intensity, and cytokine secretion disorder. The third-generation CAR T-cells that combine both CD28 and 4-1BB costimulatory domains have improved cytotoxicity and persistence, while fourth-generation CAR T-cells with one costimulatory domain are genetically engineered to introduce specific genes to perform cytotoxic functions and maintain T cell activation.
Cancers 14 04452 g001
Figure 2. Process and clinical application strategies of CAR T-cell therapy. The preparation of CAR T-cells contains leukapheresis, T cell isolation, and stimulation and expansion in vitro; then, T cells are transduced with the CAR structure targeting a specific antigen. Then, the qualified CAR T-cells are infused into the patient via intravenous injection. In the clinical stage, lymphodepletion, dose designing of CAR T-cells and side effects management must be seriously considered during clinical treatment. If necessary, combination strategies should be taken into consideration. SI: Single Infusion; FI: Fractionated Infusion.
Figure 2. Process and clinical application strategies of CAR T-cell therapy. The preparation of CAR T-cells contains leukapheresis, T cell isolation, and stimulation and expansion in vitro; then, T cells are transduced with the CAR structure targeting a specific antigen. Then, the qualified CAR T-cells are infused into the patient via intravenous injection. In the clinical stage, lymphodepletion, dose designing of CAR T-cells and side effects management must be seriously considered during clinical treatment. If necessary, combination strategies should be taken into consideration. SI: Single Infusion; FI: Fractionated Infusion.
Cancers 14 04452 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liu, Q.; Liu, Z.; Wan, R.; Huang, W. Clinical Strategies for Enhancing the Efficacy of CAR T-Cell Therapy for Hematological Malignancies. Cancers 2022, 14, 4452. https://doi.org/10.3390/cancers14184452

AMA Style

Liu Q, Liu Z, Wan R, Huang W. Clinical Strategies for Enhancing the Efficacy of CAR T-Cell Therapy for Hematological Malignancies. Cancers. 2022; 14(18):4452. https://doi.org/10.3390/cancers14184452

Chicago/Turabian Style

Liu, Qianzhen, Zengping Liu, Rongxue Wan, and Wenhua Huang. 2022. "Clinical Strategies for Enhancing the Efficacy of CAR T-Cell Therapy for Hematological Malignancies" Cancers 14, no. 18: 4452. https://doi.org/10.3390/cancers14184452

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop