624
views
2
recommends
+1 Recommend
1 collections
    5
    shares

      Interested in becoming a HOD published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      See further information on submitting a paper at https://hod-journal.org/submit-a-paper/

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Chimeric antigen receptor engineered T-cell therapy for central nervous system lymphoma

      Published
      review-article
      Bookmark

            Abstract

            Central nervous system lymphoma (CNSL) includes primary and secondary subtypes. It is associated with poor prognosis even after aggressive therapies. Primary CNSL involves mainly the brain, eyes, leptomeninges and spinal cord, without evidence of systemic non-Hodgkin’s lymphoma (NHL). Secondary CNSL refers to involvement of the CNS secondary to systemic NHL. Chimeric antigen receptor T (CAR-T) cells are genetically engineered T-cells directed against tumor target antigens. CAR-T-cells have shown encouraging results in treating B-cell malignancies. Clinical data on CAR-T-cells in CNSL treatment are limited, because of concerns regarding the immunoprivileged status of the CNS and the possibility of immune effector cell-associated neurotoxicity syndrome. Clinical trials on CAR-T therapy for CNSL are increasingly being conducted to evaluate its efficiency and safety since CAR-T-cells have been detected in the cerebrospinal fluid from a patient with PMBCL who received CAR-T-cell therapy. Current data suggest that CAR-T-cells are an emerging therapeutic modality for CNSL with clinical benefits and acceptable adverse effects. However, whether CAR-T therapy may be a promising therapeutic avenue remains controversial, because evidence from large-scale randomized clinical trials remains lacking. Herein, we provide a review of existing clinical data on CAR-T-cell therapy for CNSL, discuss the limitations of CAR-T-cells in CNSL treatment and hypothesize strategies to overcome these challenges.

            Main article text

            1. INTRODUCTION

            Central nervous system lymphoma (CNSL) is an uncommon malignant tumor of the CNS, comprising 1%–6% of CNS tumors [1, 2]. CNSL is divided into primary central nervous system lymphoma (PCNSL) and secondary central nervous system lymphoma (SCNSL). PCNSL is a rare type of diffuse large B-cell lymphoma (DLBCL) originating in the brain parenchyma, spinal cord, eyes or meninges, without evidence of systemic dissemination [3]. It accounts for 3% of primary CNS tumors and 1% of non-Hodgkin’s lymphoma (NHL) in adults [4, 5]. The 5-year survival rate for PCNSL is 30.1% [68]. Approximately 16%–26% of PCNSL cases do not initially respond to high-dose methotrexate [9, 10], but most patients relapse within 5 years of treatment [6]. SCNSL is defined by CNS involvement secondary to recurrent or progressive systemic lymphoma outside the CNS [11]. It also refers to isolated CNS relapse after primary systemic lymphoma remission [11]. CNS invasion occurs in 10%–30% of DLBCL [11] and 0.2%–0.6% of Hodgkin’s lymphoma cases [1214]. The median survival time after diagnosis of SCNSL is approximately 2–6 months [1520]. In patients with CNSL, high-dose methotrexate based chemotherapeutic regimens followed by autologous stem cell transplantation (ASCT) or radiotherapy have been found to significantly improve survival rates [10, 11, 2126]. However, only a minority of patients can achieve long-term progression-free survival (PFS) and overall survival (OS) [11, 20, 27, 28]. Although novel agents such as Bruton tyrosine kinase inhibitors, immunomodulatory drugs, PI3K/AKT/mTOR inhibitors and checkpoint inhibitors have exhibited promising efficacy, the outcomes for these patients remain dismal [2932]. Novel, efficacious and safe therapies are urgently required for relapsed or refractory (R/R) CNSL.

            Chimeric antigen receptor (CAR) is a synthetic protein with major components: an antigen-recognition moiety, a T-cell signaling domain and a costimulatory domain [33, 34]. In cancer immunotherapy, CAR is expressed in genetically modified T-cells that specifically recognize tumor-specific antigens, thereby inducing an acquired antitumor immune response [35, 36]. CAR-T-cell therapy has shown encouraging clinical responses in R/R B cell malignancies by targeting CD19, CD20 or CD22 [3744]. In 2014, CD19 CAR-T-cells administered intravenously were first detected in cerebrospinal fluid (CSF) from two patients with B-cell lymphoma with CAR-T-associated neurotoxic effects [42], thus suggesting that CAR-T-cells effectively penetrate the CNS. Therefore, a strong biologic rationale exists for treating R/R CNSL with CD19/CD20/CD22-specific CAR-T-cells. However, patients with CNSL have been excluded from most of the major trials because of poor response and the possibility of immune effector cell-associated neurotoxicity syndrome (ICANS) after CAR-T-cell treatment. Given the limited available clinical trial data for patients with CNSL receiving CAR-T-cell therapy, no definitive conclusion has been reached regarding whether CAR-T-cell therapy is a safe, efficacious and long-lasting therapeutic avenue. In this review, we summarize the most important clinical data on CAR-T-cell treatment for CNSL, discuss the potential mechanisms responsible for resistance, and discuss strategies to enhance its efficacy and to optimize its role in the therapeutic armamentarium for CNSL.

            2. CLINICAL DATA

            2.1 Clinical data on CD19 CAR-T therapy for CNSL

            A preponderance of evidence has demonstrated that CD19-targeted CAR-T-cell therapy significantly improves CNSL prognosis. Simultaneously targeting other tumor antigens, including CD20, CD22 and CD70, is also a therapeutic option. To date, CAR-T-cells specific for multiple antigens have been developed, at least three of which have been evaluated in patients with CNSL in clinical trials. Given the rarity and poor prognosis of CNSL, each clinical trial has usually enrolled fewer than twenty patients with R/R CNSL. All the clinical trials described in this review are summarized in Table 1 .

            Table 1 |

            Published studies on CAR-T-cells for treatment of primary and secondary CNS lymphomas.

            AuthorNCT/ChiCTRCAR-T-cell doseStudy designStudy populationConditioning regimenConcomitant maintenanceToxicityOutcome
            Li et al. [44]ChiCTR-OPN-16008526CD19 CAR-T-cells (2.0–7.0×106/kg, n = 5)
            CD22 CAR-T-cells (3.0–7.0×106/kg, n = 1)
            Phase 1 clinical trialPCNSL-DLBCL (n = 1)
            SCNSL-DLBCL (n = 4)
            Flu/Cy (n = 5)Radiotherapy
            PD-1 inhibitor
            ASCT
            Grade 1 CRS (n = 4); Grade 2 CRS (n = 1); Grade 1 NT (n = 1); Grade 4 NT (n = 1)60-day assessment
            CR (n = 1)
            PR (n = 4)
            Wu et al. [45]ChiCTR-OPN-16009847CD19 CAR-T-cells (2.0–9.2×106/kg, n = 13)
            CD22 CAR-T-cells (2.6–8.4×106/kg, n = 13)
            Phase 1 clinical trialPCNSL-DLBCL (n = 4)
            SCNSL-DLBCL (n = 9)
            Dox+BEAM (n = 5)
            TBC (n = 6)
            TBCF (n = 2)
            Auto-HSCT(n = 13)Grade 1 CRS (n = 9); Grade 2 CRS (n = 2); Grade 1 NT (n = 2); Grade 3 NT (n = 1)CR (n = 8)
            PR (n = 3)
            PD (n = 2)
            Tu. et al. [46]NCT03125577CD19 CAR-T-cells 1×108; CD70 CAR-T-cells 8.2×107 Case report on a patient enrolled in a phase 1 clinical trialPCNSL-DLBCL (n = 1)Flu/CyNoneNone30-day assessment: CR
            17-month assessment: CR
            Abramson et al. [47]NCT02631044CD19 CAR-T-cells
            NP
            Case report on a patient enrolled in a phase 1 clinical trialSCNSL-DLBCL (n = 1)Flu/CyNoneNone1-month assessment: CR
            Frigault et al. [48]NCT04134117Tisagenlecleucel (0.6–6.0 ×108)Retrospective cohort studySCNSL-DLBCL (n = 5)
            SCNSL-HGBCL (n = 2)
            SCNSL-PMBCL(n = 1)
            Flu/CyIbrutinib (n = 2)Grade 1 CRS (n = 7); Grade 1 NT (n = 4)28-day assessment CR (n = 2)
            PR (n = 2)
            PD (n = 2)
            Deceased due to PD (n = 2)
            90-day assessment
            CR (n = 2)
            PR (n = 1)
            PD (n = 2)
            Frigault et al. [49]NCT02445248Tisagenlecleucel (0.6–6.0 ×108)Phase I/II clinical trialPCNSL-DLBCL (n = 12)Flu/CySteroid (n = 4)Grade 1 CRS (n = 7); Grade 1 NT (n = 3); Grade 2 NT (n = 2); Grade 3 NT (n = 1)CR (n = 6)
            PR (n = 1)
            Siddiqi et al. [50]NCT02153580CD19 CAR-T-cells (1.15–6.0×108, n = 7)Retrospective cohort studyPCNSL-DLBCL (n = 5)NPNPGrade 1 CRS (n = 3); Grade 2 CRS (n = 2); Grade 1 NT (n = 3); Grade 2 NT (n = 1); Grade 3 NT (n = 1)28-day assessment
            CR (n = 3)
            SD (n = 2)
            Ghafouri et al. [51]NPAxicabtagene CiloleucelRetrospective cohort studySCNSL-DLBCL (n = 2)
            SCNSL-HGBCL (n = 2)
            SCNSL-PMBCL (n = 1)
            NPNPGrade 1 CRS (n = 1); Grade 2 CRS (n = 1); Grade 3 NT (n = 1); Grade 4 NT (n = 1)28-day assessment
            CR (n = 3)
            SD (n = 1)
            PD (n = 1)
            208-day assessment
            Deceased due to PD (n = 4)
            CR (n = 1)

            Abbreviations: NCT: national clinical trial identifier. ChiCTR: Chinese clinical trial register. PCNSL: primary central nervous system lymphoma. SCNSL: secondary central nervous system lymphoma. DLBCL: diffuse large B-cell lymphoma. HGBCL: high-grade B-cell lymphoma. PMBCL: primary mediastinal B-cell lymphoma. Flu: fludarabine. Cy: cyclophosphamide. DOX: doxorubicin. TBC: thiotepa, busulfan, cyclophosphamide. TBCF: thiotepa, busulfan, cyclophosphamide, fludarabine. BEAM: carmustine, etoposide, cytarabine, melphalan. AST: autologous stem cell transplantation. Auto-HSCT: autologous hematopoietic stem cell transplantation. NP: not provided. CRS: cytokine release syndrome. NT: neurotoxicity. CR: complete response. PR: partial response. PD: progressive disease.

            Study design, study population, route of CAR-T-cell delivery, antigens, toxicity, patient outcome, and NCT/ChiCTR are indicated.

            Maximum CRS and NT were graded according to ASTCT [23]. Abbreviations: ASTCT: American Society for Transplantation and Cellular Therapy. ChiCTR: Chinese clinical trial register. CNS: central nervous system. CR: complete response. CRS: cytokine release syndrome.

            NCT: national clinical trial identifier. NT: neurotoxicity. PD: progressive disease. PR: partial response.

            CD19-targeted receptors are currently the most investigated CAR-T-cell product. The US Food and Drug Administration has approved axicabtagene ciloleucel (Axi-Cel), tisagenlecleucel (Tisa-Cel) and lisocabtagene maraleucel (Liso-Cel) for B-cell malignancies. CD19 CAR-T-cells were successfully applied in SCNSL for the first time in 2017 [47]. A patient with primary refractory DLBCL with CNS involvement has been reported to achieve complete remission (CR) at the site of cerebral lymphoma with Liso-Cel. The remission lasted 12 months, and no CRS or ICANS was observed. CD19 CAR-T-cells were identified in the CSF, thus confirming the ability of these cells to migrate from the periphery into the CNS and subsequently mediate anti-tumor effects.

            Tisa-Cel has also shown promising efficacy and manageable adverse reactions in CNSL. Frigault et al. [48] (2019.12) have conducted a retrospective study in eight patients with secondary systemic large B-cell lymphoma with CNS involvement, who were treated with Tisa-Cel. Four patients showed an early response to Tisa-Cel at day +28 (two CRs and two partial responses (PRs)). Two patients died because of disease progression within 30 days after CAR-T-cell infusion. Among the four patients who initially responded to treatment, responses were ongoing in three patients at day +90, and only one patient achieved a second CR with radiotherapy after systemic relapse. No patient experienced grade ≥2 CRS or ICANS. In this cohort, active systemic disease was not a prerequisite for CAR-T-cell expansion and disease response, thus suggesting that sufficient intravenously infused CAR-T-cells reached the CNS by crossing the blood-brain barrier (BBB).

            A phase I/II study of Tisa-Cel in patients with relapsed PCNSL has recently been reported [49] (2022.2). A total of twelve patients with relapsed PCNSL received Tisa-Cel treatment, with a median follow up of 12.2 months (range, 3.64–23.5 months). Six patients demonstrated CR, one patient demonstrated PR, and three patients had sustained CR at the end of follow-up. Seven patients developed grade 1 CRS, two patients experienced grade 1 ICANS, three patients had grade 2 ICANS, and one patient presented grade 3 ICANS. CD19 CAR-T-cells were expanded in the peripheral blood and CSF.

            Ghafouri et al. [51] (2020.11) subsequently demonstrated the feasibility and safety of Axi-Cel in five R/R NHL patients with CNS invasion, three of whom received bridging therapy. The 28-day post-treatment evaluation showed that three patients attainted CR, one patient had stable disease (SD), and one patient experienced disease progression. Of the four responders (three with CR and one with SD), two died of disease progression, and one died of cardiopulmonary failure within 208 days after administration of CAR-T-cells. The remaining responder who underwent ASCT after CAR-T therapy had sustained remission at the end of follow-up. The median PFS and OS of the four responders were 134.2 days and 155.0 days (range, 86–208), respectively. Two patients experienced grade 1 and grade 2 CRS, and supportive care and tocilizumab were provided, respectively. Two patients had grade 3 or grade 4 ICANS and were well managed with supportive care and steroids.

            Siddiqi et al. [50] have reported that patients with PCNSL show good tolerance to another studied CD19 CAR-T-cell product engineered to express epidermal growth factor receptor. These CAR-T-cells can be eliminated through targeting epidermal growth factor receptor in the event of severe CAR-T associated toxicity. Of a total of five patients with PCNSL have received these specfic CD19 CAR-T-cells, three achieved CR, and two had SD on day 28 postinfusion. The durations of response for the three patients with CR were 43 days, 273 days and 520 days. All patients developed no greater than grade 2 CRS, and one patient developed grade 3 ICANS. Tocilizumab and steroids were necessary for two patients with severe toxicity, which was reversible and tolerable. Exploratory analyses revealed the presence of CAR-T-cells in the CSF in the absence of systemic lymphoma.

            To our knowledge, Xu et al. [52] have conducted the largest clinical trial to date exploring the efficacy and safety of CD19 CAR-T-cell therapy for R/R B-cell acute lymphoblastic leukemia (ALL) with CNS invasion. Severe CRS (grade ≥3) and ICANS (grade ≥3) were observed in 9 (18.8%) and 11 patients (22.9%), respectively. All treatment-associated toxicity symptoms were controllable. CAR-T-cells performed better in CNS than in BM. This study has provided strong evidence supporting the therapeutic potential of CD19 CAR-T-cells in CNS.

            2.2 Clinical data on dual CAR-T therapy for CNSL

            Despite major advances in CD19 CAR-T therapy in clinical trials, the rates of long-term PFS for patients with R/R CNSL are low. Consequently, dual CAR-T-cells (separate infusions of two different CAR-T-cell products) have been increasingly used in patients with CNSL to improve poor outcomes.

            Tu et al. [46] have reported a patient with R/R PCNSL receiving CD19 and CD70 CAR-T-cell infusion, who achieved CR after 1 month. The patient sustained CR for more than 17 months without experiencing CRS or ICANS. Both CD19 and CD70 CAR-T-cells were detectable at the 10th month after infusion.

            To date, CD 19 specific and CD22 specific CAR-T therapies in CNSL have been evaluated in two clinical trials. In 2020, Li et al. [44] reported on four patients with R/R SCNSL and one patient with R/R SCNSL receiving CAR19 and CAR22 T-cell cocktail therapy and follow-up for 6–16 months. In the 2nd month after infusion of CAR19 and CAR22 T-cells, CR and PR were achieved in one and four patients, respectively. PD was observed in three patients at the 3rd month, and relapse occurred in one patient at the 8th month; the remaining patient received CAR-T-cell infusion after ASCT, and remission lasted for 14 months. The median PFS was only 3 months. No patient experienced greater than grade 2 CRS. Grade 1 and grade 4 ICANS were observed in one patient each. ICANS was completely reversible by glucocorticoid and plasma exchange. All patients had CAR-T-cell expansion in CSF. CAR-T-cells targeting a single antigen were effective, but the reasponse was not durable for patients with CNSL. Hence, Wu et al. [45] have explored the efficacy, perisitence and safety of CD19/22 CAR-T-cells administered after ASCT in four patients with PCNSL and nine patients with SCNSL, including two patients with CR at enrollment. Among the remaining 11 patients, 6 attained CR, and 3 attained PR within 3 months; the median duration of response was 14.03 months. The overall response rate and complete remission rate were 81.81% and 54.55%, respectively. The estimated 1-year PFS and OS rates were 74.59% and 82.50%, respectively. Two patients did not respond to this therapy and died because of PD, with a median survival time of 2.33 months. No patient experienced grade 3 or 4 CRS, and only one patient experienced grade 3 ICANS. The novel treatment with sequential CD19/22 CAR-T-cell therapy after ASCT for patients with CNSL appeared to have encouraging long-term efficacy with controllable adverse effects.

            3. POSSIBLE MECHANISMS UNDERLYING THE EFFECTS OF CAR-T-CELL THERAPY FOR CNSL

            The BBB is an important physiological barrier that separates the CNS from the peripheral circulation, and regulates cellular and molecular exchange between the blood vessels and brain parenchyma. It is a crucial obstacle in the delivery of drugs into the CNS [53]. Moreover, the selective properties of the BBB and the blood-CSF barrier strictly limit the entry of immune cells into the CNS [53]. Recently, impressive clinical regression of CNS tumors has been achieved with engineered CAR-T-cells in many clinical trials. After systemic administration, CAR-T-cells traffic to the CSF and mediate anti-tumor effects without direct neurotoxicity [54]. The exact mechanism through which CAR-T-cells cross the BBB remains unclear.

            3.1 Endothelial activation and BBB disruption

            Peripheral inflammation mediates BBB disruption through multiple pathways [53]. Gust et al. [55] have revealed that in CAR-T therapy-associated ICANS, high levels of IL-6, IFN-γ and TNF-α activate endothelial cells, thus increasing BBB permeability. A patient with fatal neurotoxicity has presented endothelial activation and multifocal vascular disruption in the brain. Preclinical data have demonstrated that CAR-T-cells delivered intraventricularly are detectable in the peripheral blood of mice for more than 300 days, even without detectable lymphoma [56]. Single-cell RNA sequencing analysis has indicated that CD19 expressed in human and mouse brain mural cells is highly important for the integrity of the BBB [57]. Administration of CD19-specific CAR-T-cells can cause BBB disruption and pericyte depletion in mice lacking B cells [57]. CAR-T-cells have been postulated to penetrate the BBB, enter the CNS, and mediate anti-tumor effects through activating endothelial cells and disrupting the BBB. The detailed pathophysiology remains poorly understood.

            3.2 The cerebroventricular environment enhances CAR-T-cell potency

            CAR-T-cell delivery into resection cavities or administration into the CSF are feasible in glioblastoma [58]. Regional intraventricular (ICV) injection of CAR-T-cells for CNSL has not yet been conducted. In a murine lymphoma model, Wang et al. [56] have observed that CAR-T-cells infused ICV not only eradicate CNSL, but also migrate to the periphery, home to systemic tumors and expand in vivo, thus completely eliminating systemic lymphoma. They have further found that CAR-T-cells exposed to the CSF in the ICV environment exhibit superior anti-lymphoma activity and memory function [56], thus suggesting that the cerebroventricular environment may improve the efficacy of CAR-T-cells. However, similar phenomena have not yet been confirmed in immunocompetent animal models.

            4. CURRENT CHALLENGES AND POTENTIAL STRATEGIES FOR CAR-T-CELL THERAPY IN CNSL

            Although several studies have reported the anti-tumor effects of CAR-T-cells, the rates of long-term PFS in patients receiving CAR-T-cell treatment remain low. Morever, the concurrent toxicity limits clinical applications. A summary of possible reasons for the poor efficacy of CAR-T-cells is as follows.

            4.1 Low CAR-T-cell persistence

            The short lifespan of CAR-T-cells critically impairs the efficacy of CAR-T therapies [5961]. CAR construction, ex vivo manipulation and T-cell exhaustion may contribute to the low persistence of CAR-T-cells.

            Compared with murine CAR-T-cells, humanized CAR-T-cells show enhanced persistence and diminished T-cell depletion, owing to lower immunogenicity and less antigen-independent tonic signaling [6264]. CD28-CAR enhances T-cell effector funtion but has limited effects on durability, whereas 4-1BB-CAR and ICOS-CAR exhibit opposite functions [6569]. Drent et al. [70] have shown that, in contrast to a single 4-1BB domain in CAR, CAR-T-cells with both CD28 and 4-1BB domains have superior efficacy and persistence. Beyond improvements in the design of CAR-T-cells, CAR-engineered NK cells and macrophages have been found to have anti-tumor effects [7174] and thus are likely to be promising strategies for CNSL treatment. However, experience with CAR-NK cells and CAR-macrophages is restricted mainly to pre-clinical investigations. Dual targeting CAR-T-cells are also a novel therapy for CNSL [4446].

            The type of T-cells used for infusion critically affects the success of CAR-T therapy. Previous chemotherapeutic strategies containing clofarabine or doxorubicin may result in lymphopenia or lead to poor quality of the final CAR-T-cell products. Improved persistence and efficacy of CAR-T-cell treatment can be achieved with early lineage cells with enriched T-cell populations [75, 76]. Before CAR-T-cell infusion, the most common lymphodepleting chemotherapy regimen containing cyclophosphamide (Cy), fludarabine (Flu), and bendamustine (Ben) was used to eradicate regulatory T-cells (Tregs) and other immunosuppressive cells, thus increasing CAR-T-cell expansion and prolonging their persistence [77, 78]. Hirayama et al. [79] have used high dose cy-flu lymphodepletion for patients with aggressive B-NHL and found that patients with favorable cytokine profiles have longer PFS. Therefore, enhancing the efficacy of CAR-T-cell therapy is more dependent on biological effects steming from lymphodepletion therapy than on the intensity of lymphodepleting treatments. Adequate lymphodepletion is essential for obtaining optimal clinical benefits from CAR-T-cell treatment.

            The tumor microenvironment impairs T-cell function and number, thereby influencing the persistence of CAR-T-cells in tumors [8082]. CAR-T-cells also progressively languish under persist chronic antigen exposure [83]. In an acute myeloid leukemia model, the PI3K/AKT pathway leads to low persistence of CD33-specific CAR-T-cells, whereas PI3K inhibitor treatment increases the durability and prolongs the efficacy of CAR-T-cells [84]. Exhausted T-cells overexpress inhibitory receptors, such as Programmed Cell Death Protein 1 (PD-1), T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) [8587]. Thus, increased anti-tumor effects have been observed in brain tumor models when a PD1-directed domain was incorporated into CARs or CAR-T-cells were used in combination with checkpoint-blocking antibodies [88]. PD1 inhibitors have been demonstrated to prolong the action of CAR-T-cells and enhance their anti-tumor efficacy in B-cell lymphoma and ALL [89, 90]. Blockade of TIM-3 or CTLA-4 has been hypothesized to improve the efficacy of CAR-T therapy. In addition, many studies have revealed the anti-lymphoma effects of Bcl-2 CAR-T-cells and confirmed the rationale for combining CAR-T-cells and Bruton tyrosine kinase inhibitors/PI3K inhibitors/HDAC inhibitors/rituximab [9199]. To date, combination therapy for blockade of multiple immune checkpoints/targeted agents and CAR-T-cells for CNSL has not been reported. Thus, further studies remain needed to confirm the efficacy and safety of different combination therapies.

            4.2 The delivery route of CAR-T-cells

            The traditional delivery route of CAR-T-cells is intravenous administration. To improve treatment efficacy, CD19 CAR-T-cells were first administered ICV to NOD-scid IL2R γ-/- mice with CNS and/or systemic lymphoma [56]. The ICV-administered CAR-T-cells eradicated CNSL more efficiently than IV infused CAR-T-cells [56]. Agarwalla et al. [100] have recently generated an implantable multifunctional alginate scaffold for T-cell engineering and release (MASTER), which provides a new control interface for viral vector-mediated gene transfer. It shortens the time required to prepare CAR-T-cells and regulates their functions in mice. In comparison to conventional CAR-T-cells, CAR-T-cells generated in vivo, induced by MASTER, have better persistence in a mouse lymphoma model. Altering the route of administration may be a valuable new strategy to enhance the efficacy of CAR-T-cells.

            4.3 Antigen escape

            After CAR-T-cell therapy, tumoral target antigen escape may contribute to relapse. Maude et al. [101] have reported that 15 patients with recurrent B-cell ALL after Tisa-Cel infusion displayed complete loss of CD19 expression. Downregulation/loss of CD19 antigen was observed in 30%–70% of patients with ALL who had recurrent disease after CD19 CAR-T-cell treatment [102, 103]. Antigen escape may also be involved in the recurrence of CNSL after CAR-T infusion [104]. However, few large-scale high-quality data are available regarding antigen escape in CNSL. A strategy to decrease the relapse rate caused by antigen escape or loss is concomitant targeting of multiple target tumor antigens. CD19-CD20/CD19-CD22 bispecific CAR-T-cells or dual-targeted (CD19/CD22) CAR-T-cells have demonstrated promising results [105108]. Further exploration of optimal target antigens is necessary to prevent CNSL relapse, through improving the anti-tumor response and decreasing antigen escape.

            4.4 CAR-T-cell associated toxicity

            CAR-T-cell therapies are associated with unique acute toxicity of CRS and ICANS. Delayed toxicity, including prolonged cytopenias and a risk of opportunistic infections, is also increasingly being recognized.

            CRS is characterized by fever, hypotension and respiratory insufficiency, and is the most common acute toxicity in CAR-T-cell therapy. CRS is triggered by cytokine release after CAR-T-cells recognize and engage with the corresponding target antigen [109, 110]. Patients with high disease burden, a high number of administered CAR-T-cells, a high peak of CAR-T-cell expansion and endothelial activation before CAR-T treatment are at high risk of CRS [111113]. Because different risk factors and CRS grading systems have been used, the reported incidence of CRS in patients with CNSL ranges from 40% to 100% [44, 45, 4851].

            ICANS is another common acute toxicity with an incidence of 20%–100% for CNSL in CAR-T-cell clinical trials [44, 45, 4851]. Several risk factors are associated with ICANS, including severe CRS, elevated pre-treatment lactate dehydrogenase, decreased platelets and endothelial growth factor levels, an elevated serum Ang-2/Ang-1 ratio, increased ferritin on day 3 after CAR-T-cell administration and pre-existing neurologic comorbidities [54, 55, 114]. The extent of CAR-T-cell activation and toxicity are partially associated with the affinity of the antigen binding domain toward its target epitope and the costimulatory elements of CAR.

            Severe CRS and ICANS are life-threatening, and most acute toxicity is reversible in response to successful high dose glucocorticoid-based treatment. Approximately 27%–50% of patients with NHL treated with Axi-Cel develop high-grade CRS and ICANS, and require hormonal therapy [37, 115, 116]. The duration and cumulative dose of glucocorticoid depend on the severity of CRS/ICANS. Currently, several guidelines for CAR-T-cell related toxicity management recommend that patients with grade 2/3 ICANS receive 10 mg dexamethasone intravenously every 6–8 h, and that patients with grade 4 ICANS be administered 1000 mg methylprednisolone for 3 days [110, 117119]. However, Neill et al. [120] have reported that patients receiving long-term intensive hormone therapy for severe CRS/ICANS have a higher risk of infection. Another study has found that high dose steroids provide rapid relief of severe CRS in B-ALL, but impair the expansion and persistence of CAR-T-cells [121]. In contrast, other studies have found no association between glucocorticoid administration and poor performance of CAR-T-cells [114, 122]. Liu et al. [123] have reported no differences in the existence of CAR-T-cells in BM and CSF between hormonal and non-hormonal therapeutic groups. In addition, prophylactic application of glucocorticoids has been demonstrated to decrease the incidence of severe CRS/ICANS without exacerbating neurologic toxicity and impairing the function of CAR-T-cells [124126]. Because the above-mentioned conclusions are based on a small number of patients infused with CAR-T-cells, further studies remain necessary to explore the appropriate initial time, cumulative dose and duration of glucocorticoid administration for toxicity management.

            Grade 3 or 4 cytopenias can persist for more than 30 days after CAR-T-cell infusion, and severe hematological toxicity is observed in approximately 30% of patients with B-cell hematologic malignancies receiving Axi-Cel or Tisa-Cel treatment [37, 101, 127, 128]. Prolonged severe neutropenia and lymphopenia are likely to be associated with ongoing CAR-T-cell activity and hematopoiesis disruption [128], thereby increasing the risk of viral, bacterial or fungal infections [129131]. Thus, a risk-adapted dose or fractionated administration of CAR-T-cells may have the potential to avoid severe hematological toxicity [39, 54, 132].

            5. CONCLUSIONS

            CAR-T-cell therapy appears to be a novel and promising strategy for CNSL treatment. Limited clinical trials have reported that CAR-T-cell treatment is feasible without excessive toxicity, although its anti-tumor effects are not persistent. Large-scale studies are urgently needed to further confirm the anti-tumor effects and elucidate the underlying mechanisms of CAR-T-cells in CNSL therapy. Moreover, developing strategies to enhance the efficacy of CAR-T-cells will be critical, including modifing CAR design, optimizing the dose and route of CAR-T-cell administration, minimizing CAR-T associated toxicity, circumventing antigen escape and optimizing the combination of CAR-T-cells with other therapeutic approaches.

            ACKNOWLEDGEMENTS

            This work was supported by funding from the National Natural Science Foundation of China (82070211 to L.H.; 81800115 to T.S.).

            CONFLICTS OF INTEREST

            The authors declare no competing financial interests.

            REFERENCES

            1. Ling SM, Roach M, Larson DA, Wara WM. Radiotherapy of primary central nervous system lymphoma in patients with and without human immunodeficiency virus. Ten years of treatment experience at the University of California San Francisco. Cancer. 1994. Vol. 73:2570–82. 817405510.1002/1097-0142(19940515)73:10<2570::aid-cncr2820731019>3.0.co;2-1

            2. Ferreri AJ, Marturano E. Primary CNS lymphoma. Best Pract Res Clin Haematol. 2012. Vol. 25:119–30. 2240982810.1016/j.beha.2011.12.001

            3. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, et al.. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol. 2016. Vol. 131:803–20. 2715793110.1007/s00401-016-1545-1

            4. Ostrom QT, Gittleman H, Farah P, Ondracek A, Chen Y, et al.. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2006-2010. Neuro Oncol. 2013. Vol. 15:p. ii1–56. 2413701510.1093/neuonc/not151

            5. Ricard D, Idbaih A, Ducray F, Lahutte M, Hoang-Xuan K, et al.. Primary brain tumours in adults. Lancet. 2012. Vol. 379:1984–96. 2251039810.1016/S0140-6736(11)61346-9

            6. Chukwueke UN, Nayak L. Central nervous system lymphoma. Hematol Oncol Clin North Am. 2019. Vol. 33:597–611. 3122915710.1016/j.hoc.2019.03.008

            7. Grommes C, DeAngelis LM. Primary CNS lymphoma. J Clin Oncol. 2017. Vol. 35:2410–8. 2864070110.1200/JCO.2017.72.7602

            8. Ferreri AJM, Holdhoff M, Nayak L, Rubenstein JL. Evolving treatments for primary central nervous system lymphoma. Am Soc Clin Oncol Educ Book. 2019. Vol. 39:454–66. 3109961410.1200/EDBK_242547

            9. Houillier C, Soussain C, Ghesquières H, Soubeyran P, Chinot O, et al.. Management and outcome of primary CNS lymphoma in the modern era: an LOC network study. Neurology. 2020. Vol. 94:e1027–39. 3190728910.1212/WNL.0000000000008900

            10. Ferreri AJ, Cwynarski K, Pulczynski E, Ponzoni M, Deckert M, et al.. Chemoimmunotherapy with methotrexate, cytarabine, thiotepa, and rituximab (MATRix regimen) in patients with primary CNS lymphoma: results of the first randomisation of the International Extranodal Lymphoma Study Group-32 (IELSG32) phase 2 trial. Lancet Haematol. 2016. Vol. 3:e217–27. 2713269610.1016/S2352-3026(16)00036-3

            11. Tomita N, Kodama F, Kanamori H, Motomura S, Ishigatsubo Y. Secondary central nervous system lymphoma. Int J Hematol. 2006. Vol. 84:128–35. 1692613410.1532/IJH97.06091

            12. Hirmiz K, Foyle A, Wilke D, Burrell S, Brownstone R, et al.. Intracranial presentation of systemic Hodgkin’s disease. Leuk Lymphoma. 2004. Vol. 45:1667–71. 1537022210.1080/10428190410001673409

            13. Dujovny M, McBride D, Segal R. Intracranial manifestations of Hodgkin’s disease. Surg Neurol. 1980. Vol. 13:258–65. 7376061

            14. Sapozink MD, Kaplan HS. Intracranial Hodgkin’s disease. A report of 12 cases and review of the literature. Cancer. 1983. Vol. 52:p. 1301–7. 688329110.1002/1097-0142(19831001)52:7<1301::aid-cncr2820520728>3.0.co;2-5

            15. Bashir RM, Bierman PJ, Vose JM, Weisenburger DD, Armitage JO. Central nervous system involvement in patients with diffuse aggressive non-Hodgkin’s lymphoma. Am J Clin Oncol. 1991. Vol. 14:478–82. 172027810.1097/00000421-199112000-00004

            16. Tomita N, Kodama F, Sakai R, Koharasawa H, Hattori M, et al.. Predictive factors for central nervous system involvement in non-Hodgkin’s lymphoma: significance of very high serum LDH concentrations. Leuk Lymphoma. 2000. Vol. 38:335–43. 1083074010.3109/10428190009087024

            17. Recht L, Straus DJ, Cirrincione C, Thaler HT, Posner JB. Central nervous system metastases from non-Hodgkin’s lymphoma: treatment and prophylaxis. Am J Med. 1988. Vol. 84:425–35. 334824510.1016/0002-9343(88)90262-8

            18. Zinzani PL, Magagnoli M, Frezza G, Prologo G, Gherlinzoni F, et al.. Isolated central nervous system relapse in aggressive non-Hodgkin’s lymphoma: the Bologna experience. Leuk Lymphoma. 1999. Vol. 32:571–6. 1004843010.3109/10428199909058415

            19. Haioun C, Besson C, Lepage E, Thieblemont C, Simon D, et al.. Incidence and risk factors of central nervous system relapse in histologically aggressive non-Hodgkin’s lymphoma uniformly treated and receiving intrathecal central nervous system prophylaxis: a GELA study on 974 patients. Groupe d’Etudes des Lymphomes de l’Adulte. Ann Oncol. 2000. Vol. 11:685–90. 1094205610.1023/a:1008394827806

            20. El-Galaly TC, Cheah CY, Bendtsen MD, Nowakowski GS, Kansara R, et al.. Treatment strategies, outcomes and prognostic factors in 291 patients with secondary CNS involvement by diffuse large B-cell lymphoma. Eur J Cancer. 2018. Vol. 93:57–68. 2947710210.1016/j.ejca.2018.01.073

            21. Batchelor T, Carson K, O’Neill A, Grossman SA, Alavi J, et al.. Treatment of primary CNS lymphoma with methotrexate and deferred radiotherapy: a report of NABTT 96-07. J Clin Oncol. 2003. Vol. 21:p. 1044–9. 1263746910.1200/JCO.2003.03.036

            22. Herrlinger U, Küker W, Uhl M, Blaicher HP, Karnath HO, et al.. NOA-03 trial of high-dose methotrexate in primary central nervous system lymphoma: final report. Ann Neurol. 2005. Vol. 57:p. 843–7. 1592903410.1002/ana.20495

            23. Ferreri AJ, Reni M, Foppoli M, Martelli M, Pangalis GA, et al.. High-dose cytarabine plus high-dose methotrexate versus high-dose methotrexate alone in patients with primary CNS lymphoma: a randomised phase 2 trial. Lancet. 2009. Vol. 374:1512–20. 1976708910.1016/S0140-6736(09)61416-1

            24. Morris PG, Correa DD, Yahalom J, Raizer JJ, Schiff D, et al.. Rituximab, methotrexate, procarbazine, and vincristine followed by consolidation reduced-dose whole-brain radiotherapy and cytarabine in newly diagnosed primary CNS lymphoma: final results and long-term outcome. J Clin Oncol. 2013. Vol. 31:3971–9. 2410103810.1200/JCO.2013.50.4910

            25. Omuro A, Correa DD, DeAngelis LM, Moskowitz CH, Matasar MJ, et al.. R-MPV followed by high-dose chemotherapy with TBC and autologous stem-cell transplant for newly diagnosed primary CNS lymphoma. Blood. 2015. Vol. 125:1403–10. 2556834710.1182/blood-2014-10-604561

            26. Kiefer T, Hirt C, Späth C, Schüler F, Al-Ali HK, et al.. Long-term follow-up of high-dose chemotherapy with autologous stem-cell transplantation and response-adapted whole-brain radiotherapy for newly diagnosed primary CNS lymphoma: results of the multicenter Ostdeutsche Studiengruppe Hamatologie und Onkologie OSHO-53 phase II study. Ann Oncol. 2012. Vol. 23:1809–12. 2211592710.1093/annonc/mdr553

            27. Langner-Lemercier S, Houillier C, Soussain C, Ghesquières H, Chinot O, et al.. Primary CNS lymphoma at first relapse/progression: characteristics, management, and outcome of 256 patients from the French LOC network. Neuro Oncol. 2016. Vol. 18:1297–303. 2695138210.1093/neuonc/now033

            28. Han CH, Batchelor TT. Diagnosis and management of primary central nervous system lymphoma. Cancer. 2017. Vol. 123:4314–24. 2895040510.1002/cncr.30965

            29. Nayak L, Iwamoto FM, LaCasce A, Mukundan S, Roemer MGM, et al.. PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma. Blood. 2017. Vol. 129:3071–3. 2835624710.1182/blood-2017-01-764209

            30. Grommes C, Pastore A, Palaskas N, Tang SS, Campos C, et al.. Ibrutinib unmasks critical role of bruton tyrosine kinase in primary CNS lymphoma. Cancer Discov. 2017. Vol. 7:1018–29. 2861998110.1158/2159-8290.CD-17-0613

            31. Rubenstein JL, Geng H, Fraser EJ, Formaker P, Chen L, et al.. Phase 1 investigation of lenalidomide/rituximab plus outcomes of lenalidomide maintenance in relapsed CNS lymphoma. Blood Adv. 2018. Vol. 2:1595–607. 2998685210.1182/bloodadvances.2017014845

            32. Lionakis MS, Dunleavy K, Roschewski M, Widemann BC, Butman JA, et al.. Inhibition of B cell receptor signaling by ibrutinib in primary CNS lymphoma. Cancer Cell. 2017. Vol. 31:833–43 e5. 2855232710.1016/j.ccell.2017.04.012

            33. Sadelain M, Brentjens R, Riviere I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013. Vol. 3:388–98. 2355014710.1158/2159-8290.CD-12-0548

            34. Wang Z, Wu Z, Liu Y, Han W. New development in CAR-T cell therapy. J Hematol Oncol. 2017. Vol. 10:53 2822279610.1186/s13045-017-0423-1

            35. Sadelain M, Riviere I, Brentjens R. Targeting tumours with genetically enhanced T lymphocytes. Nat Rev Cancer. 2003. Vol. 3:35–45. 1250976510.1038/nrc971

            36. Ho WY, Blattman JN, Dossett ML, Yee C, Greenberg PD. Adoptive immunotherapy: engineering T cell responses as biologic weapons for tumor mass destruction. Cancer Cell. 2003. Vol. 3:431–7. 1278136010.1016/s1535-6108(03)00113-2

            37. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, et al.. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017. Vol. 377:2531–44. 2922679710.1056/NEJMoa1707447

            38. Schuster SJ, Svoboda J, Chong EA, Nasta SD, Mato AR, et al.. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med. 2017. Vol. 377:2545–54. 2922676410.1056/NEJMoa1708566

            39. Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, et al.. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. 2016. Vol. 126:2123–38. 2711123510.1172/JCI85309

            40. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, et al.. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014. Vol. 371:1507–17. 2531787010.1056/NEJMoa1407222

            41. Turtle CJ, Hanafi LA, Berger C, Hudecek M, Pender B, 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. Vol. 8:355ra116 2760555110.1126/scitranslmed.aaf8621

            42. Kochenderfer JN, Dudley ME, Kassim SH, Somerville RPT, Carpenter RO, et al.. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol. 2015. Vol. 33:540–9. 2515482010.1200/JCO.2014.56.2025

            43. van de Donk NW, Moreau P, Plesner T, Palumbo A, Gay F, et al.. Clinical efficacy and management of monoclonal antibodies targeting CD38 and SLAMF7 in multiple myeloma. Blood. 2016. Vol. 127:681–95. 2663111410.1182/blood-2015-10-646810

            44. Li T, Zhao L, Zhang Y, Xiao Y, Wang D, et al.. CAR T-cell therapy is effective but not long-lasting in B-cell lymphoma of the brain. Front Oncol. 2020. Vol. 10:1306. 3290386610.3389/fonc.2020.01306

            45. Wu J, Meng F, Cao Y, Zhang Y, Zhu X, et al.. Sequential CD19/22 CAR T-cell immunotherapy following autologous stem cell transplantation for central nervous system lymphoma. Blood Cancer J. 2021. Vol. 11:131 3426718710.1038/s41408-021-00523-2

            46. Tu S, Zhou X, Guo Z, Huang R, Yue C, et al.. CD19 and CD70 dual-target chimeric antigen receptor T-cell therapy for the treatment of relapsed and refractory primary central nervous system diffuse large B-cell lymphoma. Front Oncol. 2019. Vol. 9:1350. 3186727510.3389/fonc.2019.01350

            47. Abramson JS, McGree B, Noyes S, Plummer S, Wong C, et al.. Anti-CD19 CAR T cells in CNS diffuse large-B-cell lymphoma. N Engl J Med. 2017. Vol. 377:783–4. 2883448610.1056/NEJMc1704610

            48. Frigault MJ, Dietrich J, Martinez-Lage M, Leick M, Choi BD, et al.. Tisagenlecleucel CAR T-cell therapy in secondary CNS lymphoma. Blood. 2019. Vol. 134:860–6. 3132038010.1182/blood.2019001694

            49. Frigault MJ, Dietrich J, Gallagher K, Roschewski M, Jordan JT, et al.. Safety and efficacy of tisagenlecleucel in primary CNS lymphoma: a phase I/II clinical trial. Blood. 2022. Vol. 139:2306–15. 3516765510.1182/blood.2021014738

            50. Siddiqi T, Wang X, Blanchard MS, Wagner JR, Popplewell LL, et al.. CD19-directed CAR T-cell therapy for treatment of primary CNS lymphoma. Blood Adv. 2021. Vol. 5:4059–63. 3449270310.1182/bloodadvances.2020004106

            51. Ghafouri S, Timmerman J, Larson S, Mead MD. Axicabtagene Ciloleucel CAR T-cell therapy for relapsed/refractory secondary CNS non-Hodgkin lymphoma: comparable outcomes and toxicities, but shorter remissions may warrant alternative consolidative strategies? Bone Marrow Transplant. 2021. Vol. 56:974–7. 3316893310.1038/s41409-020-01099-4

            52. Qi Y, Zhao M, Hu Y, Wang Y, Li P, et al.. Efficacy and safety of CD19-specific CAR T-cell-based therapy in B-cell acute lymphoblastic leukemia patients with CNSL. Blood. 2022. Vol. 139:3376–86. 3533877310.1182/blood.2021013733

            53. Obermeier B, Daneman R, Ransohoff RM. Development, maintenance and disruption of the blood-brain barrier. Nat Med. 2013. Vol. 19:1584–96. 2430966210.1038/nm.3407

            54. Santomasso BD, Park JH, Salloum D, Riviere I, Flynn J, 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. Vol. 8:958–71. 2988058410.1158/2159-8290.CD-17-1319

            55. Gust J, Hay KA, Hanafi LA, Li D, Myerson D, et al.. Endothelial activation and blood-brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov. 2017. Vol. 7:1404–19. 2902577110.1158/2159-8290.CD-17-0698

            56. Wang X, Huynh C, Urak R, Weng L, Walter M, et al.. The cerebroventricular environment modifies CAR T cells for potent activity against both central nervous system and systemic lymphoma. Cancer Immunol Res. 2021. Vol. 9:75–88. 3309321710.1158/2326-6066.CIR-20-0236

            57. Parker KR, Migliorini D, Perkey E, Yost KE, Bhaduri A, et al.. Single-cell analyses identify brain mural cells expressing CD19 as potential off-tumor targets for CAR-T immunotherapies. Cell. 2020. Vol. 183:126–42 e17. 3296113110.1016/j.cell.2020.08.022

            58. Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, et al.. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med. 2016. Vol. 375:2561–9. 2802992710.1056/NEJMoa1610497

            59. Xu X, Sun Q, Liang X, Chen Z, Zhang X, et al.. Mechanisms of relapse after CD19 CAR T-cell therapy for acute lymphoblastic leukemia and its prevention and treatment strategies. Front Immunol. 2019. Vol. 10:2664. 3179859010.3389/fimmu.2019.02664

            60. Hucks G, Rheingold SR. The journey to CAR T cell therapy: the pediatric and young adult experience with relapsed or refractory B-ALL. Blood Cancer J. 2019. Vol. 9:10 10.1038/s41408-018-0164-6

            61. Nie Y, Lu W, Chen D, Tu H, Guo Z, et al.. Mechanisms underlying CD19-positive ALL relapse after anti-CD19 CAR T cell therapy and associated strategies. Biomark Res. 2020. Vol. 8:18 3251435110.1186/s40364-020-00197-1

            62. Wagner DL, Fritsche E, Pulsipher MA, Ahmed N, Hamieh M, et al.. Immunogenicity of CAR T cells in cancer therapy. Nat Rev Clin Oncol. 2021. Vol. 18:379–93. 3363336110.1038/s41571-021-00476-2

            63. Mirzaei HR, Jamali A, Jafarzadeh L, Masoumi E, Alishah K, et al.. Construction and functional characterization of a fully human anti-CD19 chimeric antigen receptor (huCAR)-expressing primary human T cells. J Cell Physiol. 2019. Vol. 234:9207–15. 3036258610.1002/jcp.27599

            64. Jafarzadeh L, Masoumi E, Alishah K, Mirzaei HR, Jamali A, et al.. Construction and functional characterization of a fully human anti-mesothelin chimeric antigen receptor (CAR) expressing T cell. Iran J Allergy Asthma Immunol. 2020. Vol. 19:264–75. 3261566010.18502/ijaai.v19i3.3454

            65. Dai Q, Han P, Qi X, Li F, Li M, et al.. 4-1BB Signaling boosts the anti-tumor activity of CD28-incorporated 2(nd) generation chimeric antigen receptor-modified T cells. Front Immunol. 2020. Vol. 11:539654. 3328180910.3389/fimmu.2020.539654

            66. Milone MC, Fish JD, Carpenito C, Carroll RG, Binder GK, et al.. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther. 2009. Vol. 17:1453–64. 1938429110.1038/mt.2009.83

            67. Cheng Z, Wei R, Ma Q, Shi L, He F, et al.. In vivo expansion and antitumor activity of coinfused CD28- and 4-1BB-engineered CAR-T cells in patients with B cell leukemia. Mol Ther. 2018. Vol. 26:976–85. 2950320410.1016/j.ymthe.2018.01.022

            68. Li G, Boucher JC, Kotani H, Park K, Zhang Y, et al.. 4-1BB enhancement of CAR T function requires NF-κB and TRAFs. JCI Insight. 2018. Vol. 3:e121322. 3023228110.1172/jci.insight.121322

            69. Guedan S, Madar A, Casado-Medrano V, Shaw C, Wing A, et al.. Single residue in CD28-costimulated CAR-T cells limits long-term persistence and antitumor durability. J Clin Invest. 2020. Vol. 130:3087–97. 3206926810.1172/JCI133215

            70. Drent E, Poels R, Ruiter R, van de Donk NWCJ, Zweegman S, et al.. Combined CD28 and 4-1BB costimulation potentiates affinity-tuned chimeric antigen receptor-engineered T cells. Clin Cancer Res. 2019. Vol. 25:4014–25. 3097973510.1158/1078-0432.CCR-18-2559

            71. Oei VYS, Siernicka M, Graczyk-Jarzynka A, Hoel HA, Yang W, et al.. Intrinsic functional potential of NK-cell subsets constrains retargeting driven by chimeric antigen receptors. Cancer Immunol Res. 2018. Vol. 6:467–80. 2945947710.1158/2326-6066.CIR-17-0207

            72. Liu E, Marin D, Banerjee P, Macapinlac HA, Thompson P, et al.. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N Engl J Med. 2020. Vol. 382:545–53. 3202337410.1056/NEJMoa1910607

            73. Burger MC, Zhang C, Harter PN, Romanski A, Strassheimer F, et al.. CAR-Engineered NK cells for the treatment of glioblastoma: turning innate effectors into precision tools for cancer immunotherapy. Front Immunol. 2019. Vol. 10:2683. 3179859510.3389/fimmu.2019.02683

            74. Klichinsky M, Ruella M, Shestova O, Lu XM, Best A, et al.. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol. 2020. Vol. 38:947–53. 3236171310.1038/s41587-020-0462-y

            75. Singh N, Perazzelli J, Grupp SA, Barrett DM. Early memory phenotypes drive T cell proliferation in patients with pediatric malignancies. Sci Transl Med. 2016. Vol. 8:320ra3 2673879610.1126/scitranslmed.aad5222

            76. Das RK, Storm J, Barrett DMJCR. Abstract 1631: T cell dysfunction in pediatric cancer patients at diagnosis and after chemotherapy can limit chimeric antigen receptor potential. 2018.

            77. Turtle CJ, Hanafi LA, Berger C, Sommermeyer D, Pender B, et al.. Addition of fludarabine to cyclophosphamide lymphodepletion improves in vivo expansion of CD19 chimeric antigen receptor-modified T cells and clinical outcome in adults with B cell acute lymphoblastic leukemia. Blood. 2015. Vol. 126:3773. 10.1182/blood.V126.23.3773.3773

            78. Turtle CJ, Berger C, Sommermeyer D, Hanafi LA, Pender B, et al.. Anti-CD19 chimeric antigen receptor-modified T cell therapy for B cell non-hodgkin lymphoma and chronic lymphocytic leukemia: fludarabine and cyclophosphamide lymphodepletion improves in vivo expansion and persistence of CAR-T cells and clinical outcomes. Blood. 2015. Vol. 126:184. 10.1182/blood.V126.23.184.184

            79. Hirayama AV, Gauthier J, Hay KA, Voutsinas JM, Wu Q, et al.. The response to lymphodepletion impacts PFS in patients with aggressive non-Hodgkin lymphoma treated with CD19 CAR T cells. Blood. 2019. Vol. 133:1876–87. 3078261110.1182/blood-2018-11-887067

            80. Lim AR, Rathmell WK, Rathmell JC. The tumor microenvironment as a metabolic barrier to effector T cells and immunotherapy. Elife. 2020. Vol. 9:e55185. 3236780310.7554/eLife.55185

            81. Pietrobon V, Marincola FM. Hypoxia and the phenomenon of immune exclusion. J Transl Med. 2021. Vol. 19:9 3340761310.1186/s12967-020-02667-4

            82. Pai SI, Cesano A, Marincola FM. The paradox of cancer immune exclusion: immune oncology next frontier. Cancer Treat Res. 2020. Vol. 180:173–95. 3221587010.1007/978-3-030-38862-1_6

            83. Gennert DG, Lynn RC, Granja JM, Weber EW, Mumbach MR, et al.. Dynamic chromatin regulatory landscape of human CAR T cell exhaustion. Proc Natl Acad Sci U S A. 2021. Vol. 118:e2104758118. 3428507710.1073/pnas.2104758118

            84. Zheng W, O’Hear CE, Alli R, Basham JH, Abdelsamed HA, et al.. PI3K orchestration of the in vivo persistence of chimeric antigen receptor-modified T cells. Leukemia. 2018. Vol. 32:1157–67. 2947906510.1038/s41375-017-0008-6

            85. Hsu CL, Ou DL, Bai LY, Chen CW, Lin L, et al.. Exploring markers of exhausted CD8 T cells to predict response to immune checkpoint inhibitor therapy for hepatocellular carcinoma. Liver Cancer. 2021. Vol. 10:346–59. 3441412210.1159/000515305

            86. Baitsch L, Baumgaertner P, Devêvre E, Raghav SK, Legat A, et al.. Exhaustion of tumor-specific CD8(+) T cells in metastases from melanoma patients. J Clin Invest. 2011. Vol. 121:2350–60. 2155585110.1172/JCI46102

            87. Fourcade J, Sun Z, Benallaoua M, Guillaume P, Luescher IF, et al.. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med. 2010. Vol. 207:2175–86. 2081992310.1084/jem.20100637

            88. Shen SH, Woroniecka K, Barbour AB, Fecci PE, Sanchez-Perez L, et al.. CAR T cells and checkpoint inhibition for the treatment of glioblastoma. Expert Opin Biol Ther. 2020. Vol. 20:579–91. 3202753610.1080/14712598.2020.1727436

            89. Cherkassky L, Morello A, Villena-Vargas J, Feng Y, Dimitrov DS, et al.. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J Clin Invest. 2016. Vol. 126:3130–44. 2745429710.1172/JCI83092

            90. Zhu HB, Deng Q, Zhang R, Jiang YY, Meng JX, et al.. [Effect of PD-1 inhibitor Nivolumab on the proliferation and cytotoxicity of anti-CD19 chimeric antigen receptor T cells]. Zhonghua Xue Ye Xue Za Zhi. 2018. Vol. 39:584–8. 3012201910.3760/cma.j.issn.0253-2727.2018.07.011

            91. Wang H, Han P, Qi X, Li F, Li M, et al.. Bcl-2 Enhances chimeric antigen receptor T cell persistence by reducing activation-induced apoptosis. Cancers (Basel). 2021. Vol. 13:197. 3342984510.3390/cancers13020197

            92. Ruella M, Kenderian SS, Shestova O, Fraietta JA, Qayyum S, 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. Vol. 22:2684–96. 2681945310.1158/1078-0432.CCR-15-1527

            93. Schubert ML, Hückelhoven A, Hoffmann JM, Schmitt A, Wuchter P, et al.. Chimeric antigen receptor T cell therapy targeting CD19-positive leukemia and lymphoma in the context of stem cell transplantation. Hum Gene Ther. 2016. Vol. 27:758–71. 2747923310.1089/hum.2016.097

            94. Stock S, Übelhart R, Schubert ML, Fan F, He B, et al.. Idelalisib for optimized CD19-specific chimeric antigen receptor T cells in chronic lymphocytic leukemia patients. Int J Cancer. 2019. Vol. 145:1312–24. 3073778810.1002/ijc.32201

            95. Xu Y, Li S, Wang Y, Liu J, Mao X, et al.. Induced CD20 expression on B-cell malignant cells heightened the cytotoxic activity of chimeric antigen receptor engineered T cells. Hum Gene Ther. 2019. Vol. 30:497–510. 3038196610.1089/hum.2018.119

            96. Chu Y, Yahr A, Huang B, Ayello J, Barth M, et al.. Romidepsin alone or in combination with anti-CD20 chimeric antigen receptor expanded natural killer cells targeting Burkitt lymphoma in vitro and in immunodeficient mice. Oncoimmunology. 2017. Vol. 6:e1341031. 2893264410.1080/2162402X.2017.1341031

            97. Rufener GA, Press OW, Olsen P, Lee SY, Jensen MC, et al.. Preserved activity of CD20-specific chimeric antigen receptor-expressing T cells in the presence of rituximab. Cancer Immunol Res. 2016. Vol. 4:509–19. 2719706810.1158/2326-6066.CIR-15-0276

            98. James SE, Orgun NN, Tedder TF, Shlomchik MJ, Jensen MC, et al.. Antibody-mediated B-cell depletion before adoptive immunotherapy with T cells expressing CD20-specific chimeric T-cell receptors facilitates eradication of leukemia in immunocompetent mice. Blood. 2009. Vol. 114:5454–63. 1988048910.1182/blood-2009-08-232967

            99. Mihara K, Yanagihara K, Takigahira M, Kitanaka A, Imai C, et al.. Synergistic and persistent effect of T-cell immunotherapy with anti-CD19 or anti-CD38 chimeric receptor in conjunction with rituximab on B-cell non-Hodgkin lymphoma. Br J Haematol. 2010. Vol. 151:37–46. 2067816010.1111/j.1365-2141.2010.08297.x

            100. Agarwalla P, Ogunnaike EA, Ahn S, Froehlich KA, Jansson A, et al.. Bioinstructive implantable scaffolds for rapid in vivo manufacture and release of CAR-T cells. Nat Biotechnol. 2022. 3533233910.1038/s41587-022-01245-x

            101. Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, et al.. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018. Vol. 378:439–48. 2938537010.1056/NEJMoa1709866

            102. Majzner RG, Mackall CL. Tumor antigen escape from CAR T-cell therapy. Cancer Discov. 2018. Vol. 8:1219–26. 3013517610.1158/2159-8290.CD-18-0442

            103. Maude SL, Teachey DT, Porter DL, Grupp SA. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood. 2015. Vol. 125:4017–23. 2599945510.1182/blood-2014-12-580068

            104. Nayyar N, White MD, Gill CM, Lastrapes M, Bertalan M, et al.. MYD88 L265P mutation and CDKN2A loss are early mutational events in primary central nervous system diffuse large B-cell lymphomas. Blood Adv. 2019. Vol. 3:375–83. 3072311210.1182/bloodadvances.2018027672

            105. Rafiq S, Hackett CS, Brentjens RJ. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol. 2020. Vol. 17:147–67. 3184846010.1038/s41571-019-0297-y

            106. Dai H, Wu Z, Jia H, Tong C, Guo Y, 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. Vol. 13:30 3224550210.1186/s13045-020-00856-8

            107. Wei G, Zhang Y, Zhao H, Wang Y, Liu Y, et al.. CD19/CD22 dual-targeted CAR T-cell therapy for relapsed/refractory aggressive B-cell lymphoma: a safety and efficacy study. Cancer Immunol Res. 2021. Vol. 9:1061–70. 3429004810.1158/2326-6066.CIR-20-0675

            108. Martyniszyn A, Krahl AC, André MC, Hombach AA, Abken H. CD20-CD19 bispecific CAR T cells for the treatment of B-cell malignancies. Hum Gene Ther. 2017. Vol. 28:1147–57. 2920787810.1089/hum.2017.126

            109. Titov A, Petukhov A, Staliarova A, Motorin D, Bulatov E, et al.. The biological basis and clinical symptoms of CAR-T therapy-associated toxicites. Cell Death Dis. 2018. Vol. 9:897 3018158110.1038/s41419-018-0918-x

            110. Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, et al.. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014. Vol. 124:188–95. 2487656310.1182/blood-2014-05-552729

            111. Cao JX, Wang H, Gao WJ, You J, Wu LH, et al.. The incidence of cytokine release syndrome and neurotoxicity of CD19 chimeric antigen receptor-T cell therapy in the patient with acute lymphoblastic leukemia and lymphoma. Cytotherapy. 2020. Vol. 22:214–26. 3230511310.1016/j.jcyt.2020.01.015

            112. Hay KA, Hanafi LA, Li D, Gust J, Liles WC, et al.. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood. 2017. Vol. 130:2295–306. 2892401910.1182/blood-2017-06-793141

            113. Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, 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. Vol. 385:517–28. 2531950110.1016/S0140-6736(14)61403-3

            114. Karschnia P, Jordan JT, Forst DA, Arrillaga-Romany IC, Batchelor TT, et al.. Clinical presentation, management, and biomarkers of neurotoxicity after adoptive immunotherapy with CAR T cells. Blood. 2019. Vol. 133:2212–21. 3080863410.1182/blood-2018-12-893396

            115. Jacobson CA, Hunter BD, Redd R, Rodig SJ, Chen PH, et al.. Axicabtagene ciloleucel in the non-trial setting: outcomes and correlates of response, resistance, and toxicity. J Clin Oncol. 2020. Vol. 38:3095–106. 3266783110.1200/JCO.19.02103

            116. Nastoupil LJ, Jain MD, Feng L, Spiegel JY, Ghobadi A, et al.. Standard-of-care axicabtagene ciloleucel for relapsed or refractory large B-cell lymphoma: results from the US lymphoma CAR T consortium. J Clin Oncol. 2020. Vol. 38:3119–28. 3240163410.1200/JCO.19.02104

            117. Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, et al.. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol. 2018. Vol. 15:47–62. 2892599410.1038/nrclinonc.2017.148

            118. Dholaria BR, Bachmeier CA, Locke F. Mechanisms and management of chimeric antigen receptor T-cell therapy-related toxicities. BioDrugs. 2019. Vol. 33:45–60. 3056041310.1007/s40259-018-0324-z

            119. Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood. 2016. Vol. 127:3321–30. 2720779910.1182/blood-2016-04-703751

            120. Neill L, Mackenzie SC, Marzolini MAV, Townsend W, Ardeshna KM, et al.. Steroid use, advanced stage disease and ≥3 lines of prior chemotherapy are associated with a higher risk of infection following CD19 CAR T-cell therapy for B-NHL: real world data from a large UK center. Blood. 2020. Vol. 136:20–1

            121. Davila ML, Riviere I, Wang X, Bartido S, Park J, et al.. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014. Vol. 6:224ra25 2455338610.1126/scitranslmed.3008226

            122. Sun Z, Xun R, Liu M, Wu X, Qu H. The association between glucocorticoid administration and the risk of impaired efficacy of axicabtagene ciloleucel treatment: a systematic review. Front Immunol. 2021. Vol. 12:646450. 3395912810.3389/fimmu.2021.646450

            123. Liu S, Deng B, Yin Z, Pan J, Lin Y, et al.. Corticosteroids do not influence the efficacy and kinetics of CAR-T cells for B-cell acute lymphoblastic leukemia. Blood Cancer J. 2020. Vol. 10:15 10.1038/s41408-020-0280-y

            124. Gardner RA, Ceppi F, Rivers J, Annesley C, Summers C, et al.. Preemptive mitigation of CD19 CAR T-cell cytokine release syndrome without attenuation of antileukemic efficacy. Blood. 2019. Vol. 134:2149–58. 3169782610.1182/blood.2019001463

            125. Kadauke S, Myers RM, Li Y, Aplenc R, Baniewicz D, et al.. Risk-adapted preemptive tocilizumab to prevent severe cytokine release syndrome after CTL019 for pediatric B-cell acute lymphoblastic leukemia: a prospective clinical trial. J Clin Oncol. 2021. Vol. 39:920–30. 3341747410.1200/JCO.20.02477

            126. Topp MS, van Meerten T, Houot R, Minnema M, Milpied N, et al.. Earlier steroid use with axicabtagene ciloleucel (Axi-Cel) in patients with relapsed/refractory large B cell lymphoma (R/R LBCL). 2020. Vol. 26: 10.1016/j.bbmt.2019.12.603

            127. Schuster SJ, Bishop MR, Tam CS, Waller EK, Borchmann P, et al.. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med. 2019. Vol. 380:45–56. 3050149010.1056/NEJMoa1804980

            128. Locke FL, Ghobadi A, Jacobson CA, Miklos DB, Lekakis LJ, 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. Vol. 20:31–42. 3051850210.1016/S1470-2045(18)30864-7

            129. Cordeiro A, Bezerra ED, Hirayama AV, Hill JA, Wu QV, et al.. Late events after treatment with CD19-targeted chimeric antigen receptor modified T cells. Biol Blood Marrow Transplant. 2020. Vol. 26:26–33. 3141956810.1016/j.bbmt.2019.08.003

            130. Logue JM, Zucchetti E, Bachmeier CA, Krivenko GS, Larson V, et al.. Immune reconstitution and associated infections following axicabtagene ciloleucel in relapsed or refractory large B-cell lymphoma. Haematologica. 2021. Vol. 106:978–86. 3232750410.3324/haematol.2019.238634

            131. Hill JA, Li D, Hay KA, Green ML, Cherian S, et al.. Infectious complications of CD19-targeted chimeric antigen receptor-modified T-cell immunotherapy. Blood. 2018. Vol. 131:121–30. 2903833810.1182/blood-2017-07-793760

            132. Frey NV, Shaw PA, Hexner EO, Pequignot E, Gill S, et al.. Optimizing chimeric antigen receptor T-cell therapy for adults with acute lymphoblastic leukemia. J Clin Oncol. 2020. Vol. 38:415–22. 3181557910.1200/JCO.19.01892

            Author and article information

            Journal
            hod
            Hematology and Oncology Discovery
            Compuscript (Ireland )
            2811-5619
            19 July 2022
            : 1
            : 1
            : 20220001
            Affiliations
            [a ]Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
            [b ]Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
            Author notes
            *Correspondence: lhuang@ 123456tjh.tjmu.edu.cn (L. Huang)
            Article
            10.15212/HOD-2022-0001
            4a0b7aa5-398a-4536-bb4b-c68aedd755f8
            Copyright © 2022 The Authors.

            This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 International.

            History
            : 05 May 2022
            : 07 June 2022
            : 22 June 2022
            Page count
            Tables: 1, References: 132, Pages: 13
            Categories
            Review

            Medicine,Hematology
            Central nervous system lymphoma,immune effector cell-associated neurotoxicity syndrome,Chimeric antigen receptor T-cell

            Comments

            Comment on this article