Next Article in Journal
Is Pulmonary Involvement a Distinct Phenotype of Post-COVID-19?
Next Article in Special Issue
Emerging and Future Targeted Therapies for Pediatric Acute Myeloid Leukemia: Targeting the Leukemia Stem Cells
Previous Article in Journal
YAP/ACSL4 Pathway-Mediated Ferroptosis Promotes Renal Fibrosis in the Presence of Kidney Stones
Previous Article in Special Issue
Harnessing the Immune System: Current and Emerging Immunotherapy Strategies for Pediatric Acute Lymphoblastic Leukemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CAR T-Cells in Acute Lymphoblastic Leukemia: Current Status and Future Prospects

by
Abdulrahman H. Almaeen
1 and
Mohamed Abouelkheir
2,3,*
1
Department of Pathology, Pathology Division, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
2
Department of Pharmacology and Therapeutics, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
3
Pharmacology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(10), 2693; https://doi.org/10.3390/biomedicines11102693
Submission received: 5 September 2023 / Revised: 28 September 2023 / Accepted: 28 September 2023 / Published: 2 October 2023
(This article belongs to the Special Issue Pathogenesis and Novel Therapies of Acute Leukemias)

Abstract

:
The currently available treatment for acute lymphoblastic leukemia (ALL) is mainly dependent on the combination of chemotherapy, steroids, and allogeneic stem cell transplantation. However, refractoriness and relapse (R/R) after initial complete remission may reach up to 20% in pediatrics. This percentage may even reach 60% in adults. To overcome R/R, a new therapeutic approach was developed using what is called chimeric antigen receptor-modified (CAR) T-cell therapy. The Food and Drug Administration (FDA) in the United States has so far approved four CAR T-cells for the treatment of ALL. Using this new therapeutic strategy has shown a remarkable success in treating R/R ALL. However, the use of CAR T-cells is expensive, has many imitations, and is associated with some adverse effects. Cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are two common examples of these adverse effects. Moreover, R/R to CAR T-cell therapy can take place during treatment. Continuous development of this therapeutic strategy is ongoing to overcome these limitations and adverse effects. The present article overviews the use of CAR T-cell in the treatment of ALL, summarizing the results of relevant clinical trials and discussing future prospects intended to improve the efficacy of this therapeutic strategy and overcome its limitations.

1. Introduction

Acute lymphoblastic leukemia (ALL) is a rare cancer in which there is an abnormal clonal proliferation of early lymphoid stem cells along with the depletion of normal bone marrow hematopoietic cells [1]. The two major types of ALL are B and T lymphoblastic leukemia. While precursor B-cell ALL (B-ALL) represents the majority of ALL cases, T-cell neoplasm tends to behave more aggressively [2]. In 2021, around 6000 cases of ALL were reported in the United States. Children and young adults’ affection represent around 80% of these cases [3]. Combination of chemotherapy, steroids, and allogeneic stem cell transplantation (Allo-SCT) is the mainstay for treating this type of leukemia [4]. While such therapeutic protocols were able to increase long-term survival rates to around 90% in pediatric ALL [5], refractoriness and relapse (R/R) after initial complete remission (CR) were reported in up to 20% of the cases [2,6]. In comparison to pediatric ALL, the figures are worse in adult leukemia. It was reported that the percentage of adults with ALL who achieved long-term disease-free survival was only 40% [7]. After relapse, the reported overall survival was only about 7% [8]. Such facts urge the need for newer therapeutic approaches to overcome R/R. One of these approaches that has been developed over the past few years is the use of chimeric antigen receptor-modified (CAR) T-cell therapy.

2. What Is CAR T-Cell Therapy?

CAR T-cells were developed and proposed as a potential therapeutic strategy by Gross and colleagues more than 3 decades ago [9]. The use of these cells entails the genetic modification of T-cells in order to enhance their ability to express cell surface synthetic receptors which are capable of interacting with specific antigens expressed on the cancer cells [10]. CARs are fusion proteins. The primary structure of a CAR consists of an extracellular tumor antigen-binding domain called the ectodomain, a transmembrane domain, and an endodomain which is an intracellular signaling domain responsible for T-cell activation [10,11]. The striking difference between CAR T-cells and unmodified T-cells is that CAR T-cells do not require human major histocompatibility complex (MHC) molecules to recognize antigens on the surface of tumor cells. CAR T-cells have the ability to directly attack cancer once these cells bind to the specified antigen located on the surface of tumor cells [10].
Currently, there are four generations of CAR T-cells. An additional fifth generation is in the pipeline. The design of the first generation of CAR T-cells was simple. The endodomain responsible for the activation of T-cells was formed of CD3ζ chain only [12]. Such a simple structure enabled only the activation of T-cells without being capable of supporting their cellular expansion. Thus, the therapeutic efficacy of this early generation was limited [9]. In fact, three signals are required to optimize T-cell activation and proliferation. Using CD3ζ chains in the first generation only ensure signal 1, which is known as T-cell receptor engagement. The other two signals are co-stimulation, which is called signal 2, and cytokine engagement, which is known as signal 3 [13]. The lack of co-stimulation (signal 2) was then addressed in the second and third generations of CAR T-cells. The addition of one or two co-stimulatory signaling domains to the endodomain of the second and third generation, respectively, fulfilled the function of signal 2 [9,14]. The use of the signaling domains of CD28, CD134, or CD137 incorporated to CD3ζ chains has significantly enhanced the activity, expansion, and survival of these modified T-cells and even stimulated interleukin (IL)-2 production [15,16,17]. The fourth generation is much more sophisticated. Modification of the structure to allow remote-controlling of CAR T-cells activity and limiting their toxicity is one example [18]. Modifications to overcome target antigen loss or down-regulation, which results in tumor escape [19,20], or to increase the CAR T-cell specificity by requiring the presence of two antigens on the same cell to allow T-cell activation [21,22], have also been reported. CAR T-cells may also be modified to enhance their ability to produce inducible cytokines, as IL-12 or interferon (IFN)-γ, which can modulate the tumor microenvironment and enhance CAR T-cells function [23,24]. These cells are known as CAR T-cells redirected for universal cytokine killings (TRUCKs), and they have demonstrated a significant ability to overcome target antigen loss/antigen-negative relapse on the surface of cancer cells [23]. The fifth generation is still in the exploratory stage. This generation is designed to cover cytokine engagement (signal 3). CAR T-cells belonging to this generation have the ability of inducing cytokine signaling upon antigen stimulation. CAR T-cells of this generation encode a truncated cytoplasmic domain from the IL-2 receptor β-chain with a transcription factor. These cells showed antigen-dependent activation and superior antitumor effects with significantly minimized toxicities. It is also expected that these cells would have a better outcome against antigen-positive relapse [25,26]. Of note, none of the third, fourth, or fifth generations of these modified T-cells have been approved yet for clinical use.

3. Manufacturing of CAR T-Cells

The manufacturing of CAR T-cells starts first with the isolation of peripheral blood mononuclear cells from the patient (autologous) or a healthy donor (allogeneic) using leukapheresis. The next step entails the use of specific antibodies to separate the target T-cell subsets, which are then activated by beads coated with anti-CD3/anti-CD28 monoclonal antibodies, anti-CD3 antibodies, or artificial antigen-presenting cells (APCs). The activation process may be enhanced by cytokines, such as IL-2. The following step is the process of gene transfer of the CAR construct to the primary T-cells. Owing to the lower risk of mutagenesis, lentiviral vectors are preferred for gene transfer over retroviral vectors [27,28]. Non-viral approaches for gene transfer have also been used in this step. The designated CAR T-cells are subjected to expansion and phenotyping, and then they will be cryopreserved for subsequent infusion [28,29,30].

4. CAR T-Cells for ALL

There are several antigens on ALL cells that have been targeted in different experimental and clinical trials on CAR T-cell therapy. CD19 has received most of the attention in B-ALL. It is constantly expressed throughout B-cell development. In addition, CD19 is not expressed on hematopoietic stem cells. This will reduce the chance of developing aplastic anemia while using this therapeutic strategy [31]. Still, targeting CD19 could possibly result in B-cell aplasia with a consequent long-lasting defect in humoral immunity and recurrent respiratory tract infections [32,33]. At the moment, all the currently FDA-approved CAR T-cells for treating ALL are targeting CD19 (Table 1).
CD20 is another target antigen under investigation as it is specific for B-cell lymphoid malignancies [38,39]. Similarly, the receptor for thymic stromal lymphopoietin (TSLPR) was also considered as a promising target for CAR T-cell therapy in selected cases of B-ALL [40,41]. For T-cell ALL, CD7-based CAR T-cells are being investigated in xenograft models, and promising results have encouraged an ongoing clinical trial [42,43]. Similarly, CD38-based CAR T-cells have been designed to target HTLV-1 + T-cell leukemia [44,45,46]. Other than targeting only one antigen on the surface of ALL cells, the use of multiple targets, as in the fourth generation of CAR T-cells, is still under investigation in order to reduce on-target, off-tumor toxicities [21,22] and prevent tumor antigen-loss relapses [19,20].

5. Limitations and Toxicities of CAR T-Cell Therapy

The first barrier to accessing CAR T-cell therapy is probably the cost. In the United States, it was estimated that one infusion of CAR T-cells can cost between USD 373,000 and 475,000 [47]. Patient care and disease monitoring can increase the number by USD 79,466 to 85,267 [48]. Moreover, the management of CAR T-cell toxicities and post-CAR T-cell therapy events can increase the total cost to exceed USD 1 million for some patients. As an average, the cost for this new therapeutic strategy can range from USD 500,000 to 600,000 [49,50]. Several technical and regulatory issues along with institutional infrastructure readiness might also be limiting factors for the use of CAR T-cell therapy [51]. Away from the high cost barrier, the two major limitations for CAR T-cell therapy are failure of treatment and toxicity.

5.1. Failure of CAR T-Cell Therapy

Treatment failure is a major limiting factor for the use of CAR T-cell therapy. The incidence of relapse among B-ALL patients who received treatment with CAR T-cells showed a wide variation in different studies and ranged between 22% and 75% [52]. Considering that only anti-CD19 CAR T-cells are the currently FDA-approved CAR T-cell therapy for B-ALL, we can classify relapse to either CD19-positive or CD19-negative relapses. Antigen-positive relapse is more common and is mainly related to T-cell potency and its in vivo expansion ability. Finding that the rate of B-ALL relapse is less in children compared to adult patients suggested that the age of T-cell might be a major determinant for antigen-positive relapse [53]. The use of co-stimulation/signaling domains and other techniques to enhance the activity, stimulate the expansion, and extend the survival of CAR T-cells has been suggested, and some have shown a promising ability to reduce antigen-positive relapse [17,26,54,55,56]. On the other hand, antigen-negative relapse or tumor antigen escape is more relevant to tumor cells. Generally, loss of CD19 antigen is responsible for about 10–20% of relapses in patients with B-ALL [57]. However, some clinical trials reported higher percentages [58]. Antigen-negative subclones might pre-exist even before starting CAR T-cell therapy and cause R/R [59]. Several mechanisms have been proposed to explain the antigen-negative relapse. While gene mutation, pre-treatment with blinatumomab, and RNA splice variants [60,61,62] can explain most of the cases of CD19-negative relapse, down-regulation of the target antigen can take place with other CAR T-cell therapies of B-ALL as anti-CD22 CAR T-cell therapy [63]. That is why the design of many of the fourth generation CAR T-cells is addressing the relapse due to antigen loss. Designing CAR T-cells with dual CD19/CD123 targeting was reported to prevent antigen-negative relapses [20]. Still, the genetic instability and heterogeneity of antigen expression on the leukemic cell surface, especially in the case of high tumor burden, can lead to another relapse even after dual-target CAR T-cell therapy [64,65,66]. Other B-cell lineage markers, as CD20 and CD22, have also been targeted, and epigenetic modifying agents have also been tested to overcome the immune evasion of tumor cells during therapy with CAR T-cells [67,68,69].

5.2. Cytokine Release Syndrome

The toxicity of CAR T-cell therapy can take many forms. One of the most important and most common toxicities related to the use of this relatively new therapeutic strategy is the cytokine release syndrome (CRS) [70]. In children with B-ALL, it was reported that the incidence of CRS can reach up to 77% [58]. In adults, figures are much higher and ranged from 85 to 93% depending on the study [53,71]. In CRS, the activation of T-cells will lead to a significant elevation of the inflammatory cytokines (e.g., IL-6, IL-10, IFN-γ, and GM-CSF) [72,73,74]. Patients are usually presented with fever, headache, myalgia, hypotension, and hypoxia. The condition may be complicated with cardio-respiratory dysfunction or even failure. The burden of the disease and the used dose of CAR T-cells are major determinant of the severity of the CRS [72,73]. The management of CRS is largely dependent on the disease grading. Anti-cytokine therapy is the mainstay for treating CRS together with proper supportive treatment for the associated hypotension and/or hypoxia. IL-6 receptor blockade via tocilizumab with or without systemic corticosteroids is usually needed in grade 2 CRS and onward. Siltuximab, an anti-IL-6 monoclonal antibody, and anakinra, an IL-1 receptor antagonist, have been suggested as alternatives to tocilizumab. Most patients respond well after therapy, yet CRS symptoms may persist in very rare cases [75,76,77].

5.3. Immune Effector Cell-Associated Neurotoxicity Syndrome

Another common toxicity associated with the use of CAR T-cells is known as immune effector cell-associated neurotoxicity syndrome (ICANS). Patients may show a wide range of presentation varying from confusion and delirium to motor dysfunction, expressive aphasia, tremor, and ataxia. Rare cases presented with myoclonic seizure and even fatal brain edema [72,73,74]. The incidences of these presentations also vary widely between studies and are largely affected by the used grading scale [78,79]. The precise mechanism of these neurologic adverse effects is unknown, but it is likely to be related to the elevated levels of different inflammatory cytokines. That is why such syndrome usually follows CRS [80]. As CD19 is expressed on the left inferior frontal gyrus, it is possible that ICANS is part of the on-target/off-tumor effect [81]. However, reporting similar neurotoxicity with CAR T-cells targeting other antigens may suggest that ICANS is not antigen-specific [82,83]. The disease is self-limited and may resolve even without any intervention [84]. Unlike CRS, tocilizumab has no role in ICANS and may even worsen the neurological manifestations. If needed, treatment with corticosteroids in addition to standard care treatment for neurological presentation, such as seizures, can reverse the disease in the majority of patients. Still, few mortalities have been reported with ICANS, especially when the cases were associated with cerebral edema [75,76,77]. Of note, both CRS and ICANS are more likely to take place when anti-CD19 CAR T-cells were used for B-ALL in comparison to their usage in other tumors [85].

5.4. Infection and Hypogammaglobulinemia

Infection is another complication to the use of CAR T-cells in leukemia. The reported incidence of infection with CAR T-cell therapy is around 43% [58]. Early infection is usually bacterial and is probably due to the immunosuppressive effect of lymphodepleting chemotherapy which is given before CAR T-cell infusion. Late infection is more relevant to CAR T-cell therapy and is more likely to be viral. This is attributed to CAR T-cell therapy itself in a phenomenon called “on-target/off-tumor recognition”. As the target antigen for CAR T-cell is shared by normal cells, it is possible that non-pathogenic immune cells expressing the target antigen (CD19) will also be affected by the used CAR T-cells. The delayed recovery of these cells will result in significant hypogammaglobulinemia [86,87,88]. The condition could be fatal and requires IgG replacement to protect against infectious complications [86,88].

5.5. Other Toxicities of CAR T-Cell Therapy

Another immunological dysfunction that might take place after CAR T-cell therapy is called hemophagocytic histiocytosis (carHLH) [75,89]. The mechanism of carHLH is unknown but involves the production of pro-inflammatory cytokines, lymphohistiocytic tissue infiltration, and immune-induced multiorgan damage and dysfunction. Until the proper patient identification and management protocols are standardized, the current management of carHLH is identical to severe CRS cases [77]. Other toxicities that have been reported with CAR T-cell therapy include cerebral edema, cardiotoxicity, allergic reaction, and tumor lysis syndrome [77].

6. Clinical Trials Using CAR T-Cells in ALL

A long list of clinical trials which are investigating the efficacy and safety of CAR T-cells in the treatment of ALL are ongoing/recruiting, completed, or even withdrawn [85,90]. We have listed the results of some of these clinical trials in Table 2. Other than clinical trials, several studies have been published to evaluate collective data obtained from related previous clinical trials. One example is the study of Pasquini et al. [91], who collected the safety and efficacy data of a CD19-targeting CAR T-cell therapy: tisagenlecleucel (tisa-cel/Kymriah). The data of 410 patients have been evaluated. There were 255 ALL patients, while the remaining 155 were non-Hodgkin lymphoma patients. The initial cure rate among ALL patients was 85.5%. Overall survival at 12 months was 77.2%. Adverse effects as grade-3 CRS accounted for 16.1% of the patients, while grade-3 ICANS was about 9.0%. These figures are much lower than those reported in some of the key studies, probably due to using different grading scales for the assessment of these adverse effects. In a parallel context, we have found one meta-analysis of several clinical trials that reported the safety and efficacy data of anti-CD19 CAR T-cells. Pooled data of 953 R/R B-ALL patients were presented, and the pooled CR was about 80%. Of note, CR was significantly improved when autologous CAR T-cells were used in comparison to allogeneic CAR T-cells. However, using autologous CAR T-cells was more likely to cause ICANS. In addition, the percentages of patients having CRS or ICANS were not significantly affected by the use of different anti-CD19 CAR T-cell constructs [92].
Of note, most of the clinical trials listed in Table 2 were using CD19-targeting CAR T-cells; few trials tested other targets. Investigating other targets would be helpful in case of relapse after using CD19-targeting CAR T-cells. In the study of Pan and his colleagues, they used CD22-targeting CAR T-cells in patients with B-ALL who failed to respond to CD19 CAR T-cell therapy. Among the 34 enrolled patients, 24 patients achieved CR or CR with incomplete count recovery after 30 days. Seven CR patients did not receive any additional treatment, and three of these patients remained in remission after 14 months. Transplantation was conducted in 11 CR patients, and 8 of them remained in remission at 4.6 to 13.3 months. Adverse effects were mild in most of the patients, and CD22 antigen mutation or loss was not reported even in patients with reported relapse [93]. Using dual-target CAR T-cells was also subjected to clinical testing to overcome relapses that might take place after the use of CD19-targeting CAR T-cell therapy. In one study, bispecific CAR T-cells targeting both CD19 and CD22 were used in patients with R/R B-ALL. All the six enrolled patients achieved CR. No neurotoxicity was reported, but one case of relapse took place 5 months later where the blast cells of the patients lost CD19 expression and exhibited diminished CD22 site density [94]. Subsequent studies enrolled more patients to enhance the reliability of the results [95,96]. A recently published meta-analysis evaluated the efficacy and safety of CD22 or CD19/CD22 CAR T-cells in the treatment of ALL and NHL. In ALL, CD19/CD22 CAR T-cells had a CR rate of about 90%. The incidences of total and severe CRS were 87% and 6%, respectively. The incidences of ICANS and severe ICANS were 16% and 3%, respectively [97].
Table 2. Selected clinical trials that investigated the safety and efficacy of using CAR T-cells in the treatment of acute lymphoblastic leukemia (ALL).
Table 2. Selected clinical trials that investigated the safety and efficacy of using CAR T-cells in the treatment of acute lymphoblastic leukemia (ALL).
Clinical TrialAge, Number, Type of LeukemiaFollow Up (Months)Used CAR T-CellT-Cell PersistenceCR, OSRELAPSE RATEReported Adverse EffectsReference
NCT01626495 and NCT010293661–24 years, 30, R/R ALL6CD19-targeting autologous CAR T-cells (CTL019)68% at
6 months
90%, 78%26%100% CRS and 27% severe CRS
43% total neurologic toxicity
[73]
NCT016264955–22 years, 39, R/R ALL CD19-targeting autologous CAR T-cells (CTL019) 46% CRS, hyperferritinemia, and organ dysfunction.
36% cardiovascular dysfunction
15% acute respiratory failure
[98]
NCT015936961–30 years, 21, R/R B-ALL or NHL10CD19-CAR incorporating an anti-CD19 single-chain variable fragment + CD3ζ + CD28 signaling domains0% at
6 months
67%, 51.6 at 10 months17%76% CRS
14% sever CRS
43% fever
43% hypokalemia
38% fever and neutropenia
[74]
NCT015936964.3–30.4 years, 50, R/R B-ALL57CD19-CAR incorporating an anti-CD19 single-chain variable fragment + CD3ζ + CD28 signaling domains 62%, Nr9.5% at 24 months [99]
NCT02030847
NCT01029366
20.6–70.4, 35, R/R B-ALL13anti-CD19 CAR T-cells therapy tisagenlecleucel (CTL019) 90%, 73% in the high dose group 90% total CRS
40% total neurologic toxicity
[100]
NCT03289455
Amelia study
1–24, 15, R/R B-ALL14Simultaneous
targeting of CD19
and CD22 (AUTO3)
86%, 60% at one year 80% mild CRS
7% encephalopathy
[95]
NCT0328945527–83, 52, R/R large B-ALL21.6Simultaneous
targeting of CD19
and CD22 (AUTO3) plus pembrolizumab
48.9% 34.6% grade 1–2 CRS
1.9% grade 3 CRS
8% neurotoxicity
[96]
NCT029756873–52 years, 20, R/R ALL10.09CD 196 months detected90% 95% CRS[101]
NCT02435849
ELIANA study
3–30, 75 + 4, R/R ALL13CTL019
Tisagenleucel (Tisacel)
An anti-CD19 single-chain variable fragment + CD3ζ +
co-stimulatory signals (4-1BB).
83% at
6 months
81%, 76% at 12 months33%77% CRS
44% sever CRS
40% ICANS
13% severe ICANS
[58,102]
Follow up of ELIANA study0–26, 185, R/R ALL11.2Tisacel 75% and 63% at 6 and 12 months
Os 85% at 6 months and 72% at 12 months
35%60% CRS of any grade
22% neurotoxicity of any grade
19% ≥ grade 3 CRS
7% ≥ grade 3 neurotoxicity
One grade 5 CRS and 1 grade 5 neurotoxicity (intracranial hemorrhage).
[103]
NCT01044069
MSKCC study
23–74, 53, R/R ALL29CD19
autologous T-cells expressing the 19-28z CAR
0% at
6 months/
83%, 12.9 months (median)39%26% severe CRS
One patient died
[53,72]
NCT020284551–26, 45, ALL9.6SCRI-CAR19v1 (a CD19 specific CAR T-cell product)~30% at 6
Months
89%, 70% at 12 months45%90% CRS
23% severe CRS
49% ICANS
23% severe ICANS
[104]
NCT018609371–22.5, 25, R/R B-ALL CD19
19-28z CAR T-cells
75%, Nr 80% CRS
16% severe CRS
28% sever neurotoxicity
[105]
NCT0186561720–47 years, 53, R/R B-ALL30.9Autologous
CD19 CAR T-cells with a 4-1BB co-stimulatory
93%
85% long term, Nr
33%23% CRS[106,107]
NCT024438310–24, 14, different hematological
malignancies
CD1978.5% at
7 months
86%, 70%40–60%93% CRS[108,109]
NCT038257311–45, 17 (4 adults, 13 pediatrics), R/R B-ALL Anti-CD19/CD22
dual CAR-T
100% CR initially, Nr50% of
patients
relapse
at 1 year
94% had grade 0–1 CRS
5.88% grade 2 CRS
[110]
NCT03330691Up to 30 years, 27, R/R ALL CD19- or CD22-specific CAR T-cell construct. Both with 4-1BB co-stimulation. 84.6%4 cases (15.4%)80% CRS (85% grade 2 or less)
38% mild neurotoxicity with a single grade 3 event.
[111]
NCT031734172–14 years (61 patients) and 15–61 years (39 patients)
R/R B-ALL, including high-risk features: BCR-ABL
fusion gene, TP53 mutation (12), extramedullary disease (including patients with central nervous system leukemia), and relapses after allogeneic stem cell transplantation (16)
8Anti-CD19 with 4-1BB and CD3ζ cytoplasmic domains 93%
OS at 12 months was 64%
OS were lower for patients
carrying the TP53 mutation or history of previous transplantation
Grade 3 and 4 CRS were 16.9% and 15.4%
Grade 2 to 3 neurological events were 12.7% and 15.4%
[112]
NCT02614066
Zuma 3
28–52, 55, R/R B-ALL16.4Autologous anti-CD19 CAR T-cell, KTE-X19 (Brexucabtagene Autoleucel): anti-CD19 single-chain variable fragment + CD3ζ + CD28 signaling domains 71% had CR/Cri
and 51% CR,
OS 12.8 months
Of grade 3 or higher:
49% anemia
36% pyrexia
25% infections
24% CRS
25% neurological events
Two grade 5 KTE-X19-related events (brain herniation and septic shock).
[113]
NCT02625480
Zuma 4
3–20, 24, R/R B-ALL36.1KTE-X19 (Brexucabtagene Autoleucel) 67% had CR/Cri, Nr Grade ≥3 adverse events in all patients:
50% hypotension
42% anemia
33% grade 3 CRS
21%, 25%, 27%, and 11% neurological events depending on the dose
[114]
ALL: acute lymphoblastic leukemia; CR: complete remission; Cri: complete remission with incomplete count recovery; CRS: cytokine release syndrome; ICANS: immune effector cell-associated neurotoxicity syndrome; NHL: non-Hodgkin lymphoma; Nr: not reported; OS: overall survival; R/R: relapsed/refractory. N.B. Whenever more than one article described the same clinical trial, we used to include the results of the latest one and point to relevant articles where preliminary data were described.

7. Future Prospects for CAR T-Cell Therapy in the Treatment of ALL

Although three decades have now lapsed since CAR T-cell therapy was first introduced as a new therapeutic modality, efforts to improve this therapeutic approach are still ongoing to overcome its limitations. One of these limitations is the time needed to manufacture personalized T-cells. Such a process can take up to 3 weeks and delay treatment [115,116]. This has led to the development of what is known as “off-the-shelf” CAR T-cells.
Off-the-shelf CAR T-cells are newly developed universal allogeneic CAR T-cells where T-cells are collected from healthy donors. This ensures that a good number of healthy T-cells are retrieved and, unlike autologous CAR T-cells, are not affected by chemotherapy or cancer cells [117,118]. It was suggested that CAR T-cells performance could be greatly affected by the quality and quantity of retrieved T-cells [53,119]. Thus, we might expect that the performance of such produced allogeneic CAR T-cells would be improved in comparison to autologous cells. Off-the-shelf CAR T-cells, in addition to ensuring good-quality cells, will also facilitate the large-scale production of T-cells, which saves both money and time. It allows the patient to have immediate access to the treatment. There is no need to wait for the 3-week interval which is usually required to manufacture personalized autologous CAR T-cells [115,118]. The term (FasTCAR) is gaining popularity and is used to indicate the next-day manufacturing of CAR T-cells using novel manufacturing platforms. The results of clinical trials on patients with relapsed ALL indicated the superior expansion capacity of these allogeneic CAR T-cells and comparable efficiency with manageable toxicity profile [120]. However, one meta-analysis reported that while neurotoxicity was reduced with allogeneic CAR T-cells, CR rates were higher when autologous CAR T-cells were used in comparison to allogeneic CAR T-cells [92]. In addition, there is another major drawback of off-the-shelf CAR T-cells. Using T-cells from a donor with MHC-mismatching can be complicated with graft rejection and the serious and life-threatening complication, graft-versus-host disease (GVHD).
In graft rejection, the remaining functioning host immune cells will attack and eliminate the transferred CAR T-cells and compromise their antitumor efficacy [121]. Rejection of allogenic CAR T-cells can be prevented in several ways. One way is to disrupt certain components of the MHC Class I of the allogeneic CAR T-cell. This might prevent the allogenic rejection by the host cells [122,123,124]. Another approach is to delete CD52 on the surface of CAR T-cells. At the same time, the anti-CD52 monoclonal antibody, alemtuzumab, is used to deplete the host T-cells before CAR T-cell infusion. Thus, the infused T-cells evaded the rejection and simultaneously will not be targeted by alemtuzumab. Such a strategy was successful but was hindered by the increased risk of cytopenia and severe viral infections [125,126]. Another way to prevent the rejection of transferred allogenic CAR T-cells is to engineer a chimeric receptor, called the alloimmune defense receptor (ADR), that enables used CAR T-cells to recognize and eliminate alloreactive lymphocytes while saving the host resting lymphocytes in peripheral blood. It was found that the co-expression of ADR and CAR on the engineered T-cells conveyed resistance to allogenic rejection, while the anti-tumor activity was not compromised [121,127].
On the other side, different strategies have been tested to bypass GVHD. Using third-party allogeneic virus-specific T-cells is one example that has been suggested based on the encouraging data when these cells were used to treat post-transplant viral infections [128,129,130,131]. Another method to overcome GVHD is the use of T-cells that are genetically modified. The genetic modification of T-cells entails the removal of endogenous molecules, as αβ T-cell receptors, in order to make the produced T-cells unable to mount the usual alloreactive immune response against the recipient’s normal tissue. Genetic modification could be achieved using different gene editing techniques, such as clustered regularly interspaced short palindromic repeat (CRISPR)-associated proteins or transcription activator-like effector nucleases (TALENS) [122,123,124,132]. Such a strategy showed some success in small clinical studies [125,126,133]. Instead of removing the αβ T-cell receptors, using a minor T-cell subset called γδ T-cells could be also promising to overcome GVHD. These cells represent only 5–10% of the total population of T-cells but have a striking feature in the way that the expression of γδ T-cell receptors on the surface of these cells is MHC-independent. This feature made this subtype of T-cells a suitable candidate for off-the-shelf CAR T-cell production with minimal risk of GVHD [134].
Another strategy to circumvent GVHD, other limitations, and the toxicities of CAR T-cells is to replace T-cells with an alternative effector cell. In this context, Natural killer (NK) cells and macrophages might be perfect targets for producing off-the-shelf CARs. These cells, unlike T-cells, are components of innate immunity, and their action is independent of MHC. Thus, it is unlikely that they may cause GVHD. Moreover, antigen escape is also unlikely to take place with NK-based CARs. These cells have the ability to recognize cancer cells independent of MHC and will keep their potency even if MHC molecules are down-regulated [135]. Indeed, CAR-NKs have shown promising results with minimal toxicities in several models of hematological and solid tumors, as well as one clinical trial [136]. Several other clinical studies have been registered, though the results are yet to be announced [137]. For macrophages, they have several advantages that can make them suitable for CARs. These cells, by selective phagocytosis, destroy cancer cells and then activate adaptive immunity by presenting the antigens to T-cells. Macrophages, by enhancing the secretion of pro-inflammatory cytokines, can also potentiate the cytotoxicity of T-cells against tumors [138,139]. In addition, these cells can easily infiltrate the tumor microenvironment and secrete cytokines that modulate the suppressive microenvironment generated by tumor cells [139]. Macrophages-based CARs showed some success in preclinical studies, and clinical trials are being conducted [138,140].
Future prospects in CAR T-cell therapy also entail the search for newer targets. As we previously discussed, all the currently available treatments for B-ALL are based on targeting CD19. Thus, tumor relapse is a common limitation for all currently available CAR T-cell therapy as a result of the down-regulation of CD19 on the surface of cancer cells [57,58]. For this reason, many of the recent studies actually focused on the search for new targets, such as CD20, CD22, and BCMA [67,69,82,141]. The success of these newer targets in comparison to targeting CD19 remains questionable. It was suggested that CD19 expression is more stable than other targets. Such stability could allow for obtaining better antitumor activity in comparison to other antigens [142]. However, one clinical trial was conducted on B-ALL patients using a relatively low dosage of CD20 CAR T-cell therapy. The study reported that targeting CD20 resulted in the induction of a high remission rate and low toxicities in patients who relapsed after CD19 CAR T-cell therapy [66]. Another new method to avoid CD19-negative relapses is to use dual-target CARs. The dual-target CARs strategy entails simultaneous targeting CD19 and another antigen. For example, the presence of either CD19 “OR” CD20 on target cells can fully induce the immune response of anti-CD20-CD19 bispecific CAR T-cells in preclinical settings [143]. Bispecific CAR T-cells could also be designed to work upon the simultaneous presence of both antigens, for example, CD19 “AND” CD22 [94,144,145]. The dual targeting of CD19 and CD123 or B-cell activating factor receptor (BAFF-R) has also been tested [146,147]. CAR T-cells which are targeting three antigens, CD19/CD20/CD22, are an even more sophisticated strategy being tested for the treatment of ALL. Initial results showed that these trivalent CAR T-cells were more effective in killing ALL cells, including CD19-negative ones [148]. In a parallel context and rather than using dual/trivalent targeting CAR T-cells, Yan et al. used a sequential infusion of two different CAR T-cells, anti-CD19 and anti-CD22, in patients with relapsed ALL after hematopoietic stem cell transplantation (HSCT). This trial has shown that the use of such a cocktail was safe and effective for relapsed cases of ALL [149].

8. CAR T-Cell Therapy for T-Cell Acute Lymphoblastic Leukemia (T-ALL)

Another indication for the use of CAR T-cell therapy is to target T-ALL. As we mentioned earlier, T-ALL is a rare but aggressive tumor with a very poor prognosis [2,150]. One major limitation that hindered the progress of developing CAR T-cells against T-ALL is that the T effector cells, which is used as a treatment, share the expression of almost all the antigens as malignant T-cells [151,152]. This will result in self-killing or what is known as the fratricide of CAR T-cells. Fratricide can reduce the proliferation of CAR T-cells in vitro and compromise their action after being infused. Several strategies have been tested to overcome this obstacle. As we discussed earlier, replacing T-cells with NK cells is one strategy. Several preclinical studies used CAR-modified NK cells to treat T-ALL, and the results were indeed encouraging to conduct clinical trials using these cells [153,154,155]. Another strategy to avoid fratricide is to find a tumor-specific antigen with a restricted expression to malignant T-cells only. As reviewed by Ren and his colleagues [156], several biomarkers have been identified and can be targeted by CAR T-cells depending on the limited expression of these biomarkers on normal T-cells. CD7 or CD38-based CAR T-cells are relevant examples [42,43,44,45,46]. While some studies showed a successful generation of anti-CD147-CAR T-cells that conveyed protection against T-ALL progression in a xenograft model [157], others have been tested clinically. For example, nanobody-derived CD7-CAR T-cells showed a remarkable ability to induce remission in one case of an 11-year-old male patient with R/R early T-ALL [158].

9. Conclusions

CAR T-cell therapy appears to be a promising new therapy for ALL, especially cases with R/R, as evident in several experimental and clinical settings. However, as with any therapeutic modality, using CAR T-cells is associated with several limitations and toxicities. In addition, the process of manufacturing these personalized T-cells is expensive and time-consuming. Continuous development of this therapeutic strategy is ongoing to overcome these limitations and adverse effects, reduce cost, and ensure the availability of this therapeutic approach in the proper time.

Author Contributions

A.H.A. and M.A. can be added for all components of the manuscript production. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Malard, F.; Mohty, M. Acute lymphoblastic leukaemia. Lancet 2020, 395, 1146–1162. [Google Scholar] [CrossRef] [PubMed]
  2. Hunger, S.P.; Mullighan, C.G. Acute lymphoblastic leukemia in children. N. Engl. J. Med. 2015, 373, 1541–1552. [Google Scholar] [CrossRef] [PubMed]
  3. Siegel, R.L.; Miller, K.D.; Fuchs, H.; Jemal, A. Cancer statistics, 2021. CA Cancer. J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  4. Terwilliger, T.; Abdul-Hay, M. Acute lymphoblastic leukemia: A comprehensive review and 2017 update. Blood Cancer J. 2017, 7, e577. [Google Scholar] [CrossRef]
  5. Pui, C.H.; Yang, J.J.; Hunger, S.P.; Pieters, R.; Schrappe, M.; Biondi, A.; Vora, A.; Baruchel, A.; Silverman, L.B.; Schmiegelow, K.; et al. Childhood acute lymphoblastic leukemia: Progress through collaboration. J. Clin. Oncol. 2015, 33, 2938–2948. [Google Scholar] [CrossRef]
  6. Hunger, S.P.; Raetz, E.A. How I treat relapsed acute lymphoblastic leukemia in the pediatric population. Blood 2020, 136, 1803–1812. [Google Scholar] [CrossRef]
  7. Kantarjian, H.; Thomas, D.; O’Brien, S.; Cortes, J.; Giles, F.; Jeha, S.; Bueso-Ramos, C.E.; Pierce, S.; Shan, J.; Koller, C.; et al. Longterm follow-up results of hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone (Hyper-CVAD), a dose-intensive regimen, in adult acute lymphocytic leukemia. Cancer 2004, 101, 2788–2801. [Google Scholar] [CrossRef]
  8. Fielding, A.K.; Richards, S.M.; Chopra, R.; Lazarus, H.M.; Litzow, M.R.; Buck, G.; Durrant, I.J.; Luger, S.M.; Marks, D.I.; Franklin, I.M.; et al. The outcome of 609 adults after relapse of acute lymphoblastic leukemia (ALL); an MRC UKALL12/ECOG 2993 study. Blood 2007, 109, 944–950. [Google Scholar] [CrossRef]
  9. Gross, G.; Waks, T.; Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl. Acad. Sci. USA 1989, 86, 10024–10028. [Google Scholar] [CrossRef]
  10. Sadelain, M.; Brentjens, R.; Rivière, I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013, 3, 388–398. [Google Scholar] [CrossRef]
  11. Chen, Y.-J.; Abila, B.; Mostafa Kamel, Y. CAR-T: What Is Next? Cancers 2023, 15, 663. [Google Scholar] [CrossRef]
  12. Schubert, M.L.; Hoffmann, J.-M.; Dreger, P.; Müller-Tidow, C.; Schmitt, M. Chimeric antigen receptor transduced T cells: Tuning up for the next generation. Int. J. Cancer. 2018, 142, 1738–1747. [Google Scholar] [CrossRef] [PubMed]
  13. Kershaw, M.H.; Westwood, J.A.; Darcy, P.K. Gene-engineered T cells for cancer therapy. Nat. Rev. Cancer 2013, 13, 525–541. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, J.; Jensen, M.; Lin, Y.; Sui, X.; Chen, E.; Lindgren, C.G.; Till, B.; Raubitschek, A.; Forman, S.J.; Qian, X.; et al. Optimizing adoptive polyclonal T cell immunotherapy of lymphomas, using a chimeric T cell receptor possessing CD28 and CD137 costimulatory domains. Hum. Gene Ther. 2007, 18, 712–725. [Google Scholar] [CrossRef] [PubMed]
  15. Finney, H.M.; Lawson, A.D.; Bebbington, C.R. Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J. Immunol. 1998, 161, 2791–2797. [Google Scholar] [CrossRef] [PubMed]
  16. Finney, H.M.; Akbar, A.N. Activation of resting human primary T cells with chimeric receptors: Costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCRζ chain. J. Immunol. 2004, 172, 104–113. [Google Scholar] [CrossRef] [PubMed]
  17. Carpenito, C.; Milone, M.C.; Hassan, R.; Simonet, J.C.; Lakhal, M.; Suhoski, M.M.; Varela-Rohena, A.; Haines, K.M.; Heitjan, D.F.; Albelda, S.M.; et al. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc. Natl. Acad. Sci. USA 2009, 106, 3360–3365. [Google Scholar] [CrossRef]
  18. Wu, C.Y.; Roybal, K.T.; Puchner, E.M.; Onuffer, J.; Lim, W.A. Remote control of therapeutic T cells through a small molecule-gated chimeric receptor. Science 2015, 350, aab4077. [Google Scholar] [CrossRef]
  19. Hegde, M.; Mukherjee, M.; Grada, Z.; Pignata, A.; Landi, D.; Navai, S.A.; Wakefield, A.; Fousek, K.; Bielamowicz, K.; Chow, K.K.; et al. Tandem CAR T cells targeting HER2 and IL13Rα2 mitigate tumor antigen escape. J. Clin. Investig. 2016, 126, 3036–3052. [Google Scholar] [CrossRef]
  20. Ruella, M.; Barrett, D.M.; Kenderian, S.S.; Shestova, O.; Hofmann, T.J.; Perazzelli, J.; Klichinsky, M.; Aikawa, V.; Nazimuddin, F.; Kozlowski, M.; et al. Dual CD19 and CD123 targeting prevents antigen-loss relapses after CD19-directed immunotherapies. J. Clin. Investig. 2016, 126, 3814–3826. [Google Scholar] [CrossRef]
  21. Lanitis, E.; Poussin, M.; Klattenhoff, A.W.; Song, D.; Sandaltzopoulos, R.; June, C.H.; Powell, D.J., Jr. Chimeric antigen receptor T cells with dissociated signaling domains exhibit focused antitumor activity with reduced potential for toxicity in Vivo. Cancer Immunol. Res. 2013, 1, 43–53. [Google Scholar] [CrossRef] [PubMed]
  22. Roybal, K.T.; Rupp, L.; Morsut, L.; Walker, W.J.; McNally, K.A.; Park, J.S.; Lim, W.A. Precision tumor recognition by T cells with combinatorial Antigen-Sensing circuits. Cell 2016, 164, 770–779. [Google Scholar] [CrossRef] [PubMed]
  23. Li, J.; Li, W.; Huang, K.; Zhang, Y.; Kupfer, G. Chimeric antigen receptor T cell (CAR-T) immunotherapy for solid tumors: Lessons learned and strategies for moving forward. J. Hematol. Oncol. 2018, 11, 22. [Google Scholar] [CrossRef] [PubMed]
  24. Akhoundi, M.; Mohammadi, M.; Sahraei, S.S.; Sheykhhasan, M.; Fayazi, N. CAR T cell therapy as a promising approach in cancer immunotherapy: Challenges and opportunities. Cell Oncol. 2021, 44, 495–523. [Google Scholar] [CrossRef] [PubMed]
  25. Kagoya, Y.; Tanaka, S.; Guo, T.; Anczurowski, M.; Wang, C.H.; Saso, K.; Butler, M.O.; Minden, M.D.; Hirano, N. A novel chimeric antigen receptor containing a JAK-STAT signaling domain mediates superior antitumor effects. Nat. Med. 2018, 24, 352–359. [Google Scholar] [CrossRef] [PubMed]
  26. Zheng, W.; O’Hear, C.E.; Alli, R.; Basham, J.H.; Abdelsamed, H.A.; Palmer, L.E.; Jones, L.L.; Youngblood, B.; Geiger, T.L. PI3K orchestration of the in vivo persistence of chimeric antigen receptor-modified T cells. Leukemia 2018, 32, 1157–1167. [Google Scholar] [CrossRef]
  27. Scholler, J.; Brady, T.L.; Binder-Scholl, G.; Hwang, W.T.; Plesa, G.; Hege, K.M.; Vogel, A.N.; Kalos, M.; Riley, J.L.; Deeks, S.G.; et al. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci. Transl. Med. 2012, 4, 132ra53. [Google Scholar] [CrossRef] [PubMed]
  28. Levine, B.L.; Miskin, J.; Wonnacott, K.; Keir, C. Global manufacturing of CAR T cell therapy. Mol. Ther. Methods. Clin. Dev. 2017, 4, 92–101. [Google Scholar] [CrossRef]
  29. Johnson, L.A.; June, C.H. Driving gene-engineered T cell immunotherapy of cancer. Cell Res. 2017, 27, 38–58. [Google Scholar] [CrossRef]
  30. Sheykhhasan, M.; Manoochehri, H.; Dama, P. Use of CAR T-cell for acute lymphoblastic leukemia (ALL) treatment: A review study. Cancer Gene Ther. 2022, 29, 1080–1096. [Google Scholar] [CrossRef]
  31. Forsberg, M.H.; Das, A.; Saha, K. The potential of CAR T therapy for relapsed or refractory pediatric and young adult B-cell ALL. Ther. Clin. Risk. Manag. 2018, 14, 1573. [Google Scholar] [CrossRef]
  32. Brudno, J.N.; Kochenderfer, J.N. Toxicities of chimeric antigen receptor T cells: Recognition and management. Blood 2016, 127, 3321–3330. [Google Scholar] [CrossRef] [PubMed]
  33. Cordeiro, A.; Bezerra, E.D.; Hill, J.A.; Turtle, C.J.; Maloney, D.G.; Bar, M. Late effects of CD19-targeted CAR-T cell therapy. Blood 2018, 132, 223. [Google Scholar] [CrossRef]
  34. FDA. Package Insert-BREYANZI. 2023. Available online: https://www.fda.gov/media/145711/download (accessed on 2 September 2023).
  35. FDA. Package Insert-KYMRIAH. 2023. Available online: https://www.fda.gov/media/107296/download (accessed on 2 September 2023).
  36. FDA. Package Insert-TECARTUS. 2023. Available online: https://www.fda.gov/media/140409/download (accessed on 2 September 2023).
  37. FDA. Package Insert-YESCARTA. 2023. Available online: https://www.fda.gov/media/108377/download (accessed on 2 September 2023).
  38. Zah, E.; Lin, M.Y.; Silva-Benedict, A.; Jensen, M.C.; Chen, Y.Y.T. Cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B cells. Cancer Immunol. Res. 2016, 4, 498–508. [Google Scholar] [CrossRef] [PubMed]
  39. Riaz, I.B.; Zahid, U.; Kamal, M.U.; Husnain, M.; McBride, A.; Hua, A.; Hamadani, A.A.; George, L.; Zeeshan, A.; Sipra, Q.R.; et al. Anti-CD 19 and anti-CD 20 CAR-modified T cells for B-cell malignancies: A systematic review and meta-analysis. Immunotherapy 2017, 9, 979–993. [Google Scholar] [CrossRef] [PubMed]
  40. Qin, H.; Cho, M.; Haso, W.; Zhang, L.; Tasian, S.K.; Oo, H.Z.; Negri, G.L.; Lin, Y.; Zou, J.; Mallon, B.S.; et al. Eradication of B-ALL using chimeric antigen receptor—Expressing T cells targeting the TSLPR oncoprotein. Blood 2015, 126, 629–639. [Google Scholar] [CrossRef]
  41. Davies, D.M.; Maher, J. TLSPR: A new CAR in the showroom for B-ALL. Blood 2015, 126, 567–569. [Google Scholar] [CrossRef]
  42. Gomes-Silva, D.; Srinivasan, M.; Sharma, S.; Lee, C.M.; Wagner, D.L.; Davis, T.H.; Rouce, R.H.; Bao, G.; Brenner, M.K.; Mamonkin, M. CD7-edited T cells expressing a CD7-specific CAR for the therapy of T-cell malignancies. Blood 2017, 130, 285–296. [Google Scholar] [CrossRef]
  43. Fleischer, L.C.; Spencer, H.T.; Raikar, S.S. Targeting T cell malignancies using CAR-based immunotherapy: Challenges and potential solutions. J. Hematol. Oncol. 2019, 12, 141. [Google Scholar] [CrossRef]
  44. Mihara, K.; Yoshida, T.; Ishida, S.; Takei, Y.; Kitanaka, A.; Shimoda, K.; Morishita, K.; Takihara, Y.; Ichinohe, T. All-trans retinoic acid and interferon-α increase CD38 expression on adult T-cell leukemia cells and sensitize them to T cells bearing anti-CD38 chimeric antigen receptors. Blood Cancer J. 2016, 6, e421. [Google Scholar] [CrossRef]
  45. Smith, A.J.; Oertle, J.; Warren, D.; Prato, D. Chimeric antigen receptor (CAR) T cell therapy for malignant cancers: Summary and perspective. J. Cell Immunother. 2016, 2, 59–68. [Google Scholar] [CrossRef]
  46. Hofmann, S.; Schubert, M.-L.; Wang, L.; He, B.; Neuber, B.; Dreger, P.; Müller-Tidow, C.; Schmitt, M. Chimeric antigen receptor (CAR) T cell therapy in acute myeloid leukemia (AML). J. Clin. Med. 2019, 8, 200. [Google Scholar] [CrossRef] [PubMed]
  47. Fiorenza, S.; Ritchie, D.S.; Ramsey, S.D.; Turtle, C.J.; Roth, J.A. Value and affordability of CAR T-cell therapy in the United States. Bone Marrow Transpl. 2020, 55, 1706–1715. [Google Scholar] [CrossRef] [PubMed]
  48. Lyman, G.H.; Nguyen, A.; Snyder, S.; Gitlin, M.; Chung, K.C. Economic evaluation of chimeric antigen receptor T-cell therapy by site of care among patients with relapsed or refractory large B-cell lymphoma. JAMA Netw. Open. 2020, 3, e202072. [Google Scholar] [CrossRef] [PubMed]
  49. Hernandez, I.; Prasad, V.; Gellad, W.F. Total costs of chimeric antigen receptor T-cell immunotherapy. JAMA Oncol. 2018, 4, 994–996. [Google Scholar] [CrossRef] [PubMed]
  50. Di, M.; Long, J.B.; Isufi, I.; Foss, F.M.; Seropian, S.; Gross, C.P.; Huntington, S.F. Total costs of care during chimeric antigen receptor T-cell therapy in patients with relapsed/refractory B cell non-Hodgkin lymphoma: A large private insurance claim-based analysis. Blood 2022, 140 (Suppl. 1), 10818–10819. [Google Scholar] [CrossRef]
  51. Gee, A.P. Manufacturing genetically modified T cells for clinical trials. Cancer Gene Ther. 2015, 22, 67–71. [Google Scholar] [CrossRef]
  52. Fabrizio, V.A.; Curran, K.J. Clinical experience of CAR T cells for B cell acute lymphoblastic leukemia. Best Pract. Res. Clin. Haematol. 2021, 34, 101305. [Google Scholar] [CrossRef]
  53. Park, J.H.; Rivière, I.; Gonen, M.; Wang, X.; Sénéchal, B.; Curran, K.J.; Sauter, C.; Wang, Y.; Santomasso, B.; Mead, E.; et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 2018, 378, 449–459. [Google Scholar] [CrossRef]
  54. Long, A.H.; Haso, W.M.; Shern, J.F.; Wanhainen, K.M.; Murgai, M.; Ingaramo, M.; Smith, J.P.; Walker, A.J.; Kohler, M.E.; Venkateshwara, V.R.; et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. 2015, 21, 581–590. [Google Scholar] [CrossRef]
  55. Ruella, M.; Kenderian, S.S.; Shestova, O.; Fraietta, J.A.; Qayyum, S.; Zhang, Q.; Maus, M.V.; Liu, X.; Nunez-Cru, 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] [PubMed]
  56. Fan, F.; Yoo, H.J.; Stock, S.; Wang, L.; Liu, Y.; Schubert, M.L.; Wang, S.; Neuber, B.; Hückelhoven-Krauss, A.; Gern, U.; et al. Ibrutinib for improved chimeric antigen receptor T-cell production for chronic lymphocytic leukemia patients. Int. J. Cancer. 2021, 148, 419–428. [Google Scholar] [CrossRef] [PubMed]
  57. Xu, X.; Sun, Q.; Liang, X.; Chen, Z.; Zhang, X.; Zhou, X.; Li, M.; Tu, H.; Liu, Y.; Tu, S.; et al. Mechanisms of relapse after CD19 CAR T-cell therapy for acute lymphoblastic leukemia and its prevention and treatment strategies. Front. Immunol. 2019, 10, 2664. [Google Scholar] [CrossRef] [PubMed]
  58. 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] [PubMed]
  59. Fischer, J.; Paret, C.; El Malki, K.; Alt, F.; Wingerter, A.; Neu, M.A.; Kron, B.; Russo, A.; Lehmann, N.; Roth, L.; et al. CD19 isoforms enabling resistance to CART-19 immunotherapy are expressed in B-ALL patients at initial diagnosis. J. Immunother. 2017, 40, 187–195. [Google Scholar] [CrossRef] [PubMed]
  60. Sotillo, E.; Barrett, D.M.; Black, K.L.; Bagashev, A.; Oldridge, D.; Wu, G.; Sussman, R.; Lanauze, C.; Ruella, M.; Gazzara, M.R.; et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 2015, 5, 1282–1295. [Google Scholar] [CrossRef] [PubMed]
  61. Orlando, E.J.; Han, X.; Tribouley, C.; Wood, P.A.; Leary, R.J.; Riester, M.; Levine, J.E.; Qayed, M.; Grupp, S.A.; Boyer, M.; et al. Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia. Nat. Med. 2018, 24, 1504–1506. [Google Scholar] [CrossRef]
  62. Pillai, V.; Muralidharan, K.; Meng, W.; Bagashev, A.; Oldridge, D.A.; Rosenthal, J.; Van Arnam, J.; Melenhorst, J.J.; Mohan, D.; DiNofia, A.M.; et al. CAR T-cell therapy is effective for CD19-dim B-lymphoblastic leukemia but is impacted by prior blinatumomab therapy. Blood Adv. 2019, 3, 3539–3549. [Google Scholar] [CrossRef]
  63. Fry, T.J.; Shah, N.N.; Orentas, R.J.; Stetler-Stevenson, M.; Yuan, C.M.; Ramakrishna, S.; Wolters, P.; Martin, S.; Delbrook, C.; Yates, B.; et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat. Med. 2018, 24, 20–28. [Google Scholar] [CrossRef]
  64. Ruella, M.; Maus, M.V. Catch me if you can: Leukemia escape after CD19-directed T cell immunotherapies. Comput. Struct. Biotechnol. J. 2016, 14, 357–362. [Google Scholar] [CrossRef]
  65. Rosenthal, J.; Naqvi, A.S.; Luo, M.; Wertheim, G.; Paessler, M.; Thomas-Tikhonenko, A.; Rheingold, S.R.; Pillai, V. Heterogeneity of surface CD19 and CD22 expression in B lymphoblastic leukemia. Am. J. Hematol. 2018, 93, E352–E355. [Google Scholar] [CrossRef]
  66. Pan, J.; Tan, Y.; Deng, B.; Tong, C.; Hua, L.; Ling, Z.; Song, W.; Xu, J.; Duan, J.; Wang, Z.; et al. Frequent occurrence of CD19-negative relapse after CD19 CAR T and consolidation therapy in 14 TP53-mutated r/r B-ALL children. Leukemia 2020, 34, 3382–3387. [Google Scholar] [CrossRef] [PubMed]
  67. Shah, N.N.; Stevenson, M.S.; Yuan, C.M.; Richards, K.; Delbrook, C.; Kreitman, R.J.; Pastan, I.; Wayne, A.S. Characterization of CD22 expression in acute lymphoblastic leukemia. Pediatr. Blood Cancer 2015, 62, 964–969. [Google Scholar] [CrossRef] [PubMed]
  68. Wang, Q.S.; Wang, Y.; Lv, H.Y.; Han, Q.W.; Fan, H.; Guo, B.; Wang, L.L.; Han, W.D. Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol. Ther. 2015, 23, 184–191. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, W.-Y.; Wang, Y.; Guo, Y.-L.; Dai, H.-R.; Yang, Q.-M.; Zhang, Y.-J.; Zhang, Y.; Chen, M.-X.; Wang, C.-M.; Feng, K.-C.; et al. Treatment of CD20-directed chimeric antigen receptor-modified T cells in patients with relapsed or refractory B-cell non-Hodgkin lymphoma: An early phase IIa trial report. Signal Transduct. Targeted Ther. 2016, 1, 16002. [Google Scholar] [CrossRef]
  70. Lee, D.W.; Gardner, R.; Porter, D.L.; Louis, C.U.; Ahmed, N.; Jensen, M.; Grupp, S.A.; Mackall, C.L. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014, 124, 188–195. [Google Scholar] [CrossRef]
  71. Shah, B.D.; Bishop, M.R.; Oluwole, O.O.; Logan, A.; Baer, M.R.; Donnellan, W.B.; Carr-O’Dwyer, K.M.; Holmes, H.; Arellano, M.L.; Ghobadi, A.; et al. End of phase I results of ZUMA-3, a phase 1/2 study of KTE-X19, anti-CD19 chimeric antigen receptor (CAR) T cell therapy, in adult patients (pts) with relapsed/refractory (R/R) acute lymphoblastic leukemia (ALL). J. Clin. Oncol. 2019, 37 (suppl. 15), 7006. [Google Scholar] [CrossRef]
  72. Davila, M.L.; Riviere, I.; Wang, X.; Bartido, S.; Park, J.; Curran, K.; Chung, S.S.; Stefanski, J.; Borquez-Ojeda, O.; Olszewska, M.; et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci. Transl. Med. 2014, 6, 224ra25. [Google Scholar] [CrossRef]
  73. Maude, S.L.; Frey, N.; Shaw, P.A.; Aplenc, R.; Barrett, D.M.; Bunin, N.J.; Chew, A.; Gonzalez, V.E.; Zheng, Z.; Lacey, S.F.; et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 2014, 371, 1507–1517. [Google Scholar] [CrossRef]
  74. 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]
  75. Neelapu, S.S.; Tummala, S.; Kebriaei, P.; Wierda, W.; Gutierrez, C.; Locke, F.L.; Komanduri, K.V.; Lin, Y.; Jain, N.; Daver, N.; et al. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat. Rev. Clin. Oncol. 2018, 15, 47–62. [Google Scholar] [CrossRef] [PubMed]
  76. Mahadeo, K.M.; Khazal, S.J.; Abdel-Azim, H.; Fitzgerald, J.C.; Taraseviciute, A.; Bollard, C.M.; Tewari, P.; Duncan, C.; Traube, C.; McCall, D.; et al. Management guidelines for paediatric patients receiving chimeric antigen receptor T cell therapy. Nat. Rev. Clin. Oncol. 2019, 16, 45–63. [Google Scholar] [CrossRef] [PubMed]
  77. Maus, M.V.; Alexander, S.; Bishop, M.R.; Brudno, J.N.; Callahan, C.; Davila, M.L.; Diamonte, C.; Dietrich, J.; Fitzgerald, J.C.; Frigault, M.J.; et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune effector cell-related adverse events. J. Immunother. Cancer 2020, 8, e001511. [Google Scholar] [CrossRef] [PubMed]
  78. Gust, J.; Hay, K.A.; Hanafi, L.A.; Li, D.; Myerson, D.; Gonzalez-Cuyar, L.F.; Yeung, C.; Liles, W.C.; Wurfel, M.; Lopez, J.A.; et al. Endothelial activation and blood-brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov. 2017, 7, 1404–1419. [Google Scholar] [CrossRef]
  79. Santomasso, B.D.; Park, J.H.; Salloum, D.; Riviere, I.; Flynn, J.; Mead, E.; Halton, E.; Wang, X.; Senechal, B.; Purdon, T.; et al. Clinical and biological correlates of neurotoxicity associated with CAR T-cell therapy in patients with B-cell acute lymphoblastic leukemia. Cancer Discov. 2018, 8, 958–971. [Google Scholar] [CrossRef]
  80. 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. Biol. Blood Marrow Transplant. 2019, 25, 625–638. [Google Scholar] [CrossRef]
  81. Kranick, S.; Phan, G.; Kochenderfer, J.; Rosenberg, S.; Nath, A. Aphasia as a complication of CD19-targeted chimeric antigen receptor immunotherapy (S52.006). Neurology 2015, 82 (Suppl. 10), S52.006. [Google Scholar]
  82. Cohen, A.D.; Garfall, A.L.; Stadtmauer, E.A.; Lacey, S.F.; Lancaster, E.; Vogl, D.T.; Dengel, K.; Ambrose, D.E.; Chen, F.; Plesa, G.; et al. B-cell maturation antigen (BCMA)-specific chimeric antigen receptor T cells (CART-BCMA) for multiple myeloma (MM): Initial safety and efficacy from a phase I study. Blood 2016, 128, 1147. [Google Scholar] [CrossRef]
  83. Shalabi, H.; Wolters, P.L.; Martin, S.; Delbrook, C.; Yates, B.; Lee, D.W.; Mackall, C.L.; Fry, T.J.; Shah, N.N. A prospective evaluation of neurocognitive function and neurologic symptoms in pediatric and young adult patients with relapsed/refractory acute lymphoblastic leukemia (ALL) undergoing anti-CD22 chimeric antigen receptor therapy. In Proceedings of the 58th ASH Annual Meeting and Exposition, San Diego, CA, USA, 5–6 March 2016. [Google Scholar] [CrossRef]
  84. Maude, S.L.; Teachey, D.T.; Porter, D.L.; Grupp, S.A. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood 2015, 125, 4017–4023. [Google Scholar] [CrossRef]
  85. Harris, K.; LaBelle, J.L.; Bishop, M.R. Current status of CAR T cell therapy for leukemias. Curr. Treat. Options. Oncol. 2021, 22, 62. [Google Scholar] [CrossRef]
  86. Kochenderfer, J.N.; Wilson, W.H.; Janik, J.E.; Dudley, M.E.; Stetler-Stevenson, M.; Feldman, S.A.; Maric, I.; Raffeld, M.; Nathan, D.A.; Lanier, B.J.; et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 2010, 116, 4099–4102. [Google Scholar] [CrossRef] [PubMed]
  87. Curran, K.J.; Pegram, H.J.; Brentjens, R.J. Chimeric antigen receptors for T cell immunotherapy: Current understanding and future directions. J. Gene Med. 2012, 14, 405–415. [Google Scholar] [CrossRef] [PubMed]
  88. Kochenderfer, J.N.; Dudley, M.E.; Feldman, S.A.; Wilson, W.H.; Spaner, D.E.; Maric, I.; Stetler-Stevenson, M.; Phan, G.Q.; Hughes, M.S.; Sherry, R.M.; et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 2012, 119, 2709–2720. [Google Scholar] [CrossRef] [PubMed]
  89. Sandler, R.D.; Tattersall, R.S.; Schoemans, H.; Greco, R.; Badoglio, M.; Labopin, M.; Alexander, T.; Kirgizov, K.; Rovira, M.; Saif, M.; et al. Diagnosis and management of secondary HLH/MAS following HSCT and CAR-T cell therapy in adults; A review of the literature and a survey of practice within EBMT centres on behalf of the Autoimmune Diseases Working Party (ADWP) and Transplant Complications Working Party (TCWP). Front. Immunol. 2020, 11, 524. [Google Scholar] [PubMed]
  90. ClinicalTrials.gov. Used Key Word: CAR-T Cells. 2023. Available online: https://clinicaltrials.gov/search?term=CAR%20T-cells%20&cond=Acute%20Lymphoblastic%20Leukemia (accessed on 2 September 2023).
  91. Pasquini, M.C.; Hu, Z.H.; Curran, K.; Laetsch, T.; Locke, F.; Rouce, R.; Pulsipher, M.A.; Phillips, C.L.; Keating, A.; Frigault, M.J.; et al. Real-world evidence of tisagenlecleucel for pediatric acute lymphoblastic leukemia and non-Hodgkin lymphoma. Blood Adv. 2020, 4, 5414–5424. [Google Scholar] [CrossRef]
  92. 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]
  93. Pan, J.; Niu, Q.; Deng, B.; Liu, S.; Wu, T.; Gao, Z.; Liu, Z.; Zhang, Y.; Qu, X.; Zhang, Y.; et al. CD22 CAR T-cell therapy in refractory or relapsed B acute lymphoblastic leukemia. Leukemia 2019, 33, 2854–2866. [Google Scholar] [CrossRef]
  94. Dai, H.; Wu, Z.; Jia, H.; Tong, C.; Guo, Y.; Ti, D.; Han, X.; Liu, Y.; Zhang, W.; Wang, C.; et al. Bispecific CAR-T cells targeting both CD19 and CD22 for therapy of adults with relapsed or refractory B cell acute lymphoblastic leukemia. J. Hematol. Oncol. 2020, 13, 30. [Google Scholar] [CrossRef]
  95. Cordoba, S.; Onuoha, S.; Thomas, S.; Pignataro, D.S.; Hough, R.; Ghorashian, S.; Vora, A.; Bonney, D.; Veys, P.; Rao, K.; et al. CAR T cells with dual targeting of CD19 and CD22 in pediatric and young adult patients with relapsed or refractory B cell acute lymphoblastic leukemia: A phase 1 trial. Nat. Med. 2021, 27, 1797–1805. [Google Scholar] [CrossRef]
  96. Roddie, C.; Lekakis, L.J.; Marzolini, M.A.V.; Ramakrishnan, A.; Zhang, Y.; Hu, Y.; Peddareddigari, V.G.R.; Khokhar, N.; Chen, R.; Basilico, S.; et al. Dual targeting of CD19 and CD22 with bicistronic CAR-T cells in patients with relapsed/refractory large B-cell lymphoma. Blood 2023, 141, 2470–2482. [Google Scholar] [CrossRef]
  97. Fergusson, N.J.; Adeel, K.; Kekre, N.; Atkins, H.; Hay, K.A. A systematic review and meta-analysis of CD22 CAR T-cells alone or in combination with CD19 CAR T-cells. Front. Immunol. 2023, 14, 1178403. [Google Scholar] [CrossRef] [PubMed]
  98. Fitzgerald, J.C.; Weiss, S.L.; Maude, S.L.; Barrett, D.M.; Lacey, S.F.; Melenhorst, J.J.; Shaw, P.; Berg, R.A.; June, C.H.; Porter, D.L.; et al. Cytokine release syndrome after chimeric antigen receptor T cell therapy for acute lymphoblastic leukemia. Crit. Care. Med. 2017, 45, e124. [Google Scholar] [CrossRef] [PubMed]
  99. Shah, N.N.; Lee, D.W.; Yates, B.; Yuan, C.M.; Shalabi, H.; Martin, S.; Wolters, P.L.; Steinberg, S.M.; Baker, E.H.; Delbrook, C.P.; et al. Long-term follow-up of CD19-CAR T-cell therapy in children and young adults with B-ALL. J. Clin. Oncol. 2021, 39, 1650–1659. [Google Scholar] [CrossRef] [PubMed]
  100. 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] [PubMed]
  101. Gu, R.; Liu, F.; Zou, D.; Xu, Y.; Lu, Y.; Liu, B.; Liu, W.; Chen, X.; Liu, K.; Guo, Y.; et al. Efficacy and safety of CD19 CAR T constructed with a new anti-CD19 chimeric antigen receptor in relapsed or refractory acute lymphoblastic leukemia. J. Hematol. Oncol. 2020, 13, 122. [Google Scholar] [CrossRef]
  102. Grupp, S.A.; Maude, S.L.; Rives, S.; Baruchel, A.; Boyer, M.W.; Bittencourt, H.; Bader, P.; Büchner, J.; Laetsch, T.W.; Stefanski, H.; et al. Updated analysis of the efficacy and safety of Tisagenlecleucel in pediatric and young adult patients with relapsed/refractory (r/r) acute lymphoblastic leukemia. Blood 2018, 132 (Suppl. 1), 895. [Google Scholar] [CrossRef]
  103. Schultz, L.M.; Baggott, C.; Prabhu, S.; Pacenta, H.; Phillips, C.L.; Rossoff, J.; Stefanski, H.; Talano, J.A.; Moskop, A.; Margossian, S.P.; et al. Disease burden impacts outcomes in pediatric and young adult B-cell acute lymphoblastic leukemia after commercial Tisagenlecleucel: Results from pediatric real-world CAR Consortium (PRWCC). Blood 2020, 136, 14–15. [Google Scholar] [CrossRef]
  104. Gardner, R.A.; Finney, O.; Annesley, C.; Brakke, H.; Summers, C.; Leger, K.; Bleakley, M.; Brown, C.; Mgebroff, S.; Kelly-Spratt, K.S.; et al. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood 2017, 129, 3322–3331. [Google Scholar] [CrossRef]
  105. Curran, K.J.; Margossian, S.; Kernan, N.A.; Silverman, L.B.; Williams, D.A.; Shukla, N.N.; Kobos, R.; Forlenza, C.J.; Steinherz, P.; Prockop, S.; et al. Toxicity and response following CD19-specific CAR T cells in pediatric/young adult relapsed/refractory B-ALL. Blood 2019, 134, 2361–2368. [Google Scholar] [CrossRef]
  106. 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]
  107. 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]
  108. Ghorashian, S.; Kramer, A.M.; Onuoha, S.; Wright, G.; Bartram, J.; Richardson, R.; Albon, S.J.; Casanovas-Company, J.; Castro, F.; Popova, B.; et al. Enhanced CAR T cell expansion and prolonged persistence in pediatric patients with ALL treated with a low-affinity CD19 CAR. Nat. Med. 2019, 25, 1408–1414. [Google Scholar] [CrossRef] [PubMed]
  109. Ghorashian, S.; Kramer, A.M.; Onuoha, S.; Wright, G.; Bartram, J.; Richardson, R.; Albon, S.J.; Casanovas-Company, J.; Castro, F.; Popova, B.; et al. Therapy of paediatric B-ALL with a fast off rate CD19 CAR leads to enhanced expansion and prolonged CAR T cell persistence in patients with low bone marrow tumour burden, and is associated with a favourable toxicity profile. Blood 2019, 134, 225. [Google Scholar] [CrossRef]
  110. Yang, J.; Jiang, P.; Zhang, X.; Zhu, X.; Dong, Q.; He, J.; Lin, N.; Wang, Z.; Cai, S.; Ye, X.; et al. Anti-CD19/CD22 dual CAR-T therapy for refractory and relapsed B-cell acute lymphoblastic leukemia. Blood 2019, 134, 284. [Google Scholar] [CrossRef]
  111. Gardner, R.A.; Annesley, C.; Wilson, A.; Summers, C.; Narayanaswamy, P.; Wu, V.; Huang, W.; Johnson, A.; Li, A.; Schultz, K.R.; et al. Efficacy of SCRI-CAR19x22 T cell product in B-ALL and persistence of anti-CD22 activity. J. Clin. Oncol. 2020, 38 (suppl. 15), 3035. [Google Scholar] [CrossRef]
  112. Zhang, X.; Lu, X.A.; Yang, J.; Zhang, G.; Li, J.; Song, L.; Su, Y.; Shi, Y.; Zhang, M.; He, J.; et al. Efficacy and safety of anti-CD19 CAR T-cell therapy in 110 patients with B-cell acute lymphoblastic leukemia with high-risk features. Blood Adv. 2020, 4, 2325–2338. [Google Scholar] [CrossRef] [PubMed]
  113. 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]
  114. Wayne, A.S.; Huynh, V.; Hijiya, N.; Rouce, R.H.; Brown, P.A.; Krueger, J.; Kitko, C.L.; Ziga, E.D.; Hermiston, M.L.; Richards, M.K.; et al. Three-year results from phase I of ZUMA-4: KTE-X19 in pediatric relapsed/refractory acute lymphoblastic leukemia. Haematologica 2023, 108, 747–760. [Google Scholar] [CrossRef]
  115. Depil, S.; Duchateau, P.; Grupp, S.A.; Mufti, G.; Poirot, L. ‘Off-the-shelf’ allogeneic CAR T cells: Development and challenges. Nat. Rev. Drug. Discov. 2020, 19, 185–199. [Google Scholar] [CrossRef]
  116. Papathanasiou, M.M.; Stamatis, C.; Lakelin, M.; Farid, S.; Titchener-Hooker, N.; Shah, N. Autologous CAR T-cell therapies supply chain: Challenges and opportunities? Cancer Gene Ther. 2020, 27, 799–809. [Google Scholar] [CrossRef]
  117. Das, R.K.; Vernau, L.; Grupp, S.A.; Barrett, D.M. Naïve T-cell deficits at diagnosis and after chemotherapy impair cell therapy potential in pediatric cancers. Cancer Discov. 2019, 9, 492–499. [Google Scholar] [CrossRef] [PubMed]
  118. Tang, T.C.Y.; Xu, N.; Nordon, R.; Haber, M.; Micklethwaite, K.; Dolnikov, A. Donor T cells for CAR T cell therapy. Biomark. Res. 2022, 10, 14. [Google Scholar] [CrossRef] [PubMed]
  119. Shah, N.N.; Fry, T.J. Mechanisms of resistance to CAR T cell therapy. Nat. Rev. Clin. Oncol. 2019, 16, 372–385. [Google Scholar] [CrossRef] [PubMed]
  120. Zhang, C.; He, J.; Liu, L.; Wang, J.; Wang, S.; Liu, L.; Ge, J.; Gao, L.; Gao, L.; Kong, P.; 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]
  121. Mo, F.; Srinivasan, M.; Heslop, H.E.; Brenner, M.K.; Mamonkin, M. Rejection-resistant off-the-shelf T cells for adoptive cell therapy. Biol. Blood Marrow Transplant. 2019, 25, S168. [Google Scholar] [CrossRef]
  122. Poirot, L.; Philip, B.; Schiffer-Mannioui, C.; Le Clerre, D.; Chion-Sotinel, I.; Derniame, S.; Potrel, P.; Bas, C.; Lemaire, L.; Galetto, R.; et al. Multiplex genome-edited T-cell manufacturing platform for “off-the-shelf” adoptive T-cell immunotherapies. Cancer Res. 2015, 75, 3853–3864. [Google Scholar] [CrossRef]
  123. Ren, J.; Liu, X.; Fang, C.; Jiang, S.; June, C.H.; Zhao, Y. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin. Cancer Res. 2017, 23, 2255–2266. [Google Scholar] [CrossRef]
  124. Cooper, M.L.; Choi, J.; Staser, K.; Ritchey, J.K.; Devenport, J.M.; Eckardt, K.; Rettig, M.P.; Wang, B.; Eissenberg, L.G.; Ghobadi, A.; et al. An “off-the-shelf” fratricide-resistant CAR-T for the treatment of T cell hematologic malignancies. Leukemia 2018, 32, 1970–1983. [Google Scholar] [CrossRef]
  125. Qasim, W.; Zhan, H.; Samarasinghe, S.; Adams, S.; Amrolia, P.; Stafford, S.; Butler, K.; Rivat, C.; Wright, G.; Somana, K.; et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci. Transl. Med. 2017, 9, eaaj2013. [Google Scholar] [CrossRef]
  126. Benjamin, R.; Graham, C.; Yallop, D.; Jozwik, A.; Mirci-Danicar, O.C.; Lucchini, G.; Pinner, D.; Jain, N.; Kantarjian, H.; Boissel, N.; et al. Genome-edited, donor-derived allogeneic anti-CD19 chimeric antigen receptor T cells in paediatric and adult B-cell acute lymphoblastic leukaemia: Results of two phase 1 studies. Lancet. 2020, 396, 1885–1894. [Google Scholar] [CrossRef]
  127. Mo, F.; Watanabe, N.; McKenna, M.K.; Hicks, M.J.; Srinivasan, M.; Gomes-Silva, D.; Atilla, E.; Smith, T.; Ataca Atilla, P.; Ma, R.; et al. Engineered off-the-shelf therapeutic T cells resist host immune rejection. Nat. Biotechnol. 2021, 39, 56–63. [Google Scholar] [CrossRef] [PubMed]
  128. Torikai, H.; Reik, A.; Liu, P.Q.; Zhou, Y.; Zhang, L.; Maiti, S.; Huls, H.; Miller, J.C.; Kebriaei, P.; Rabinovitch, B.; et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood 2012, 119, 5697–5705. [Google Scholar] [CrossRef] [PubMed]
  129. Cruz, C.R.Y.; Micklethwaite, K.P.; Savoldo, B.; Ramos, C.A.; Lam, S.; Ku, S.; Diouf, O.; Liu, E.; Barrett, A.J.; Ito, S.; et al. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: A phase 1 study. Blood 2013, 122, 2965–2973. [Google Scholar] [CrossRef]
  130. Leen, A.M.; Bollard, C.M.; Mendizabal, A.M.; Shpall, E.J.; Szabolcs, P.; Antin, J.H.; Kapoor, N.; Pai, S.Y.; Rowley, S.D.; Kebriaei, P.; et al. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood 2013, 121, 5113–5123. [Google Scholar] [CrossRef]
  131. Tzannou, I.; Papadopoulou, A.; Naik, S.; Leung, K.; Martinez, C.A.; Ramos, C.A.; Carrum, G.; Sasa, G.; Lulla, P.; Watanabe, A.; et al. Off-the-shelf virus-specific T cells to treat BK virus, human herpesvirus 6, cytomegalovirus, Epstein-Barr virus, and adenovirus infections after allogeneic hematopoietic stem-cell transplantation. J. Clin. Oncol. 2017, 35, 3547–3557. [Google Scholar] [CrossRef] [PubMed]
  132. Eyquem, J.; Mansilla-Soto, J.; Giavridis, T.; van der Stegen, S.J.; Hamieh, M.; Cunanan, K.M.; Odak, A.; Gönen, M.; Sadelain, M. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 2017, 543, 113–117. [Google Scholar] [CrossRef]
  133. Ottaviano, G.; Georgiadis, C.; Gkazi, S.A.; Syed, F.; Zhan, H.; Etuk, A.; Preece, R.; Chu, J.; Kubat, A.; Adams, S.; et al. Phase 1 clinical trial of CRISPR-engineered CAR19 universal T cells for treatment of children with refractory B cell leukemia. Sci. Transl. Med. 2022, 14, eabq3010. [Google Scholar] [CrossRef]
  134. Saura-Esteller, J.; de Jong, M.; King, L.A.; Ensing, E.; Winograd, B.; de Gruijl, T.D.; Parren, P.W.H.I.; van der Vliet, H.J. Gamma delta T-cell based cancer immunotherapy: Past-present-future. Front. Immunol. 2022, 13, 915837. [Google Scholar] [CrossRef]
  135. Mehta, R.S.; Rezvani, K. Chimeric Antigen Receptor Expressing Natural Killer Cells for the Immunotherapy of Cancer. Front. Immunol. 2018, 9, 283. [Google Scholar] [CrossRef]
  136. Liu, E.; Marin, D.; Banerjee, P.; Macapinlac, H.A.; Thompson, P.; Basar, R.; Nassif Kerbauy, L.; Overman, B.; Thall, P.; Kaplan, M.; et al. Use of CAR-transduced natural killer cells in CD19-Positive lymphoid tumors. N. Engl. J. Med. 2020, 382, 545–553. [Google Scholar] [CrossRef]
  137. ClinicalTrials.gov. Used Key Word: CAR-NK. 2023. Available online: https://clinicaltrials.gov/search?term=CAR-NK (accessed on 2 September 2023).
  138. Klichinsky, M.; Ruella, M.; Shestova, O.; Lu, X.M.; Best, A.; Zeeman, M.; Schmierer, M.; Gabrusiewicz, K.; Anderson, N.R.; Petty, N.E.; et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 2020, 38, 947–953. [Google Scholar] [CrossRef] [PubMed]
  139. Chen, Y.; Yu, Z.; Tan, X.; Jiang, H.; Xu, Z.; Fang, Y.; Han, D.; Hong, W.; Wei, W.; Tu, J. CAR-macrophage: A new immunotherapy candidate against solid tumors. Biomed. Pharmacother. 2021, 139, 111605. [Google Scholar] [CrossRef] [PubMed]
  140. ClinicalTrials.gov. Used Key Word: CAR-Macrophages. 2023. Available online: https://clinicaltrials.gov/search?term=CAR-Macrophages (accessed on 2 September 2023).
  141. Zhang, T.; Wang, T.; You, F.; Li, Z.; Chen, D.; Zhang, K.; Tian, S.; Sheng, B.; Wu, H.; Jiang, L.; et al. Nanobody-based anti-CD22-chimeric antigen receptor T cell immunotherapy exhibits improved remission against B-cell acute lymphoblastic leukemia. Transpl. Immunol. 2022, 71, 101538. [Google Scholar] [CrossRef] [PubMed]
  142. Wei, J.; Han, X.; Bo, J.; Han, W. Target selection for CAR-T therapy. J. Hematol. Oncol. 2019, 12, 62. [Google Scholar] [CrossRef] [PubMed]
  143. Martyniszyn, A.; Krahl, A.C.; André, M.C.; Hombach, A.A.; Abken, H. CD20-CD19 Bispecific CAR T Cells for the Treatment of B-Cell Malignancies. Hum. Gene. Ther. 2017, 28, 1147–1157. [Google Scholar] [CrossRef] [PubMed]
  144. Schultz, L.M.; Muffly, L.S.; Spiegel, J.Y.; Ramakrishna, S.; Hossain, N.; Baggott, C.; Sahaf, B.; Patel, S.; Craig, J.; Yoon, J.; et al. Phase I trial using CD19/CD22 bispecific CAR T cells in pediatric and adult acute lymphoblastic leukemia (ALL). In Proceedings of the 61nd ASH Annual Meeting and Exposition, Orlando, FL, USA, 7–10 December 2019. [Google Scholar]
  145. Yang, J.; Jiang, P.; Zhang, X.; Li, J.; Wu, Y.; Xu, L.; Su, Y.; Hu, X.; Zhao, X.; Dong, Q.; et al. Successful 24-hours manufacture of anti-CD19/CD22 dual chimeric antigen receptor (CAR) T cell therapy for B-cell acute lymphoblastic leukemia (B-ALL) clinically relevant abstract. In Proceedings of the 62nd ASH Annual Meeting and Exposition, San Diego, CA, USA, 5–8 December 2020. [Google Scholar]
  146. Yan, L.E.; Zhang, H.; Wada, M.; Fang, L.; Feng, J.; Zhang, W.; Chen, Q.; Cao, Y.; Pinz, K.G.; Chen, K.H.; et al. Targeting two antigens associated with B-ALL with CD19-CD123 compound Car T cell therapy. Stem. Cell. Rev. Rep. 2020, 16, 385–396. [Google Scholar] [CrossRef]
  147. Wang, X.; Dong, Z.; Awuah, D.; Chang, W.C.; Cheng, W.A.; Vyas, V.; Cha, S.C.; Anderson, A.J.; Zhang, T.; Wang, Z.; et al. CD19/BAFF-R dual-targeted CAR T cells for the treatment of mixed antigen-negative variants of acute lymphoblastic leukemia. Leukemia 2022, 36, 1015–1024. [Google Scholar] [CrossRef]
  148. Fousek, K.; Watanabe, J.; George, A.; An, X.; Samaha, H.S.; Navai, S.A.; Byrd, T.T.; Jang, A.; Kim, H.; Sujith, J.; et al. Targeting CD19-negative relapsed B-acute lymphoblastic leukemia using trivalent CAR T cells. J. Clin. Oncol. 2018, 36, 121. [Google Scholar] [CrossRef]
  149. Yan, N.; Wang, N.; Wang, G.; Huang, L.; Li, C.; Wang, D.; Wang, J.; Huang, L.; Meng, F.; Wei, J.; et al. CAR19/22 T cell cocktail therapy for B-ALL relapsed after allogeneic hematopoietic stem cell transplantation. Cytotherapy 2022, 24, 841–849. [Google Scholar] [CrossRef]
  150. Teachey, D.T.; O’Connor, D. How I treat newly diagnosed T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma in children. Blood. 2020, 135, 159–166. [Google Scholar] [CrossRef]
  151. Chen, K.H.; Wada, M.; Pinz, K.G.; Liu, H.; Lin, K.W.; Jares, A.; Firor, A.E.; Shuai, X.; Salman, H.; Golightly, M. Preclinical targeting of aggressive T-cell malignancies using anti-CD5 chimeric antigen receptor. Leukemia 2017, 31, 2151–2160. [Google Scholar] [CrossRef] [PubMed]
  152. Png, Y.T.; Vinanica, N.; Kamiya, T.; Shimasaki, N.; Coustan-Smith, E.; Campana, D. Blockade of CD7 expression in T cells for effective chimeric antigen receptor targeting of T-cell malignancies. Blood Adv. 2017, 1, 2348–2360. [Google Scholar] [CrossRef] [PubMed]
  153. Huang, J.; Alexey, S.; Li, J.; Jones, T.; Grande, G.; Douthit, L.; Xie, J.; Chen, D.; Wu, X.; Michael, M. Unique CDR3 epitope targeting by CAR-T cells is a viable approach for treating T-cell malignancies. Leukemia 2019, 33, 2315–2319. [Google Scholar] [CrossRef] [PubMed]
  154. Voynova, E.; Hawk, N.; Flomerfelt, F.A.; Telford, W.G.; Gress, R.E.; Kanakry, J.A.; Kovalovsky, D. Increased activity of a NK-specific CAR-NK framework targeting CD3 and CD5 for T-cell leukemias. Cancers 2022, 14, 524. [Google Scholar] [CrossRef] [PubMed]
  155. You, F.; Wang, Y.; Jiang, L.; Zhu, X.; Chen, D.; Yuan, L.; An, G.; Meng, H.; Yang, L. A novel CD7 chimeric antigen receptor-modified NK-92MI cell line targeting T-cell acute lymphoblastic leukemia. Am. J. Cancer Res. 2019, 9, 64–78. [Google Scholar]
  156. Ren, A.; Tong, X.; Xu, N.; Zhang, T.; Zhou, F.; Zhu, H. CAR T-cell immunotherapy treating T-ALL: Challenges and opportunities. Vaccines 2023, 11, 165. [Google Scholar] [CrossRef]
  157. Zheng, N.S.; Zhao, X.Y.; Wei, D.; Miao, J.L.; Liu, Z.K.; Yong, Y.L.; Zhang, R.Y.; Guo, Y.X.; He, L.; Wang, B.; et al. CD147-specific chimeric antigen receptor T cells effectively inhibit T cell acute lymphoblastic leukemia. Cancer Lett. 2022, 542, 215762. [Google Scholar] [CrossRef]
  158. Dai, H.P.; Cui, W.; Cui, Q.Y.; Zhu, W.J.; Meng, H.M.; Zhu, M.Q.; Zhu, X.M.; Yang, L.; Wu, D.P.; Tang, X.W. Haploidentical CD7 CAR T-cells induced remission in a patient with TP53 mutated relapsed and refractory early T-cell precursor lymphoblastic leukemia/lymphoma. Biomark. Res. 2022, 10, 6. [Google Scholar] [CrossRef]
Table 1. Currently FDA-approved CAR T-cell therapy for ALL [34,35,36,37].
Table 1. Currently FDA-approved CAR T-cell therapy for ALL [34,35,36,37].
Trade Name Generic Name FDA
Approval Date
Target AntigenIndication(s)
BREYANZI®Lisocabtagene maraleucel2021CD19
  • For adult patients with R/R large B-cell lymphoma after two or more lines of systemic therapy, including:
    Diffuse large B-cell lymphoma (DLBCL) not otherwise specified (including DLBCL arising from indolent lymphoma);
    High-grade B-cell lymphoma;
    Primary mediastinal large B-cell lymphoma;
    Follicular lymphoma (FL) grade 3B.
KYMRIAHTMTisagenlecleucel2017CD19
  • For adults with R/R DLBCL.
  • For young adult patients up to age 25 with R/R ALL.
  • Adult patients with R/R FL after two or more lines of systemic therapy.
TECARTUSTMBrexucabtagene autoleucel2020CD19
  • For adult patients with R/R B-ALL.
  • For patients with R/R mantle cell lymphoma.
YESCARTATMAxicabtagene ciloleucel2017CD19
  • For patients with the following conditions who have R/R following two or more lines of systemic therapy:
    DLBCL;
    Primary mediastinal B-cell lymphoma;
    High grade B-cell lymphoma;
    DLBCL that results from FL;
    FL.
  • N.B. None of these therapeutics could be used to treat patients with primary central nervous system lymphoma.
  • DLBCL: diffuse large B-cell lymphoma; FL: follicular lymphoma; R/R: relapsed/refractory.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Almaeen, A.H.; Abouelkheir, M. CAR T-Cells in Acute Lymphoblastic Leukemia: Current Status and Future Prospects. Biomedicines 2023, 11, 2693. https://doi.org/10.3390/biomedicines11102693

AMA Style

Almaeen AH, Abouelkheir M. CAR T-Cells in Acute Lymphoblastic Leukemia: Current Status and Future Prospects. Biomedicines. 2023; 11(10):2693. https://doi.org/10.3390/biomedicines11102693

Chicago/Turabian Style

Almaeen, Abdulrahman H., and Mohamed Abouelkheir. 2023. "CAR T-Cells in Acute Lymphoblastic Leukemia: Current Status and Future Prospects" Biomedicines 11, no. 10: 2693. https://doi.org/10.3390/biomedicines11102693

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