We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
ReviewOpen AccessOpen Access license

CAR-T cells and allogeneic hematopoietic stem cell transplantation for relapsed/refractory B-cell acute lymphoblastic leukemia

    Jun Liu

    Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China

    ,
    Xi Zhang

    Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China

    ,
    Jiang F Zhong

    Division of Periodontology, Diagnostic Sciences & Dental Hygiene, & Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA

    &
    Cheng Zhang

    *Author for correspondence: Tel.: +1 86 023 6877 4209;

    E-mail Address: chzhang2014@163.com

    Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China

    Published Online:https://doi.org/10.2217/imt-2017-0072

    Abstract

    Relapsed/refractory acute lymphoblastic leukemia (ALL) has a low remission rate after chemotherapy, a high relapse rate and poor long-term survival even when allogeneic hematopoietic stem cell transplantation (allo-HSCT) is performed. Chimeric antigen receptors redirected T cells (CAR-T cells) can enhance disease remission with a favorable outcome for relapsed/refractory ALL, though some cases quickly relapsed after CAR-T cell treatment. Thus, treatment with CAR-T cells followed by allo-HSCT may be the best way to treat relapsed/refractory ALL. In this review, we first discuss the different types of CAR-T cells. We then discuss the treatment of relapsed/refractory ALL using only CAR-T cells. Finally, we discuss the use of CAR-T cells, followed by allo-HSCT, for the treatment of relapsed/refractory ALL.

    The treatment of relapsed/refractory acute lymphoblastic leukemia (ALL) remains a serious challenge because of low remission rates after chemotherapy, high relapse rates and poor long-term survival even with allogeneic hematopoietic stem cell transplantation (allo-HSCT) [1–3]. The options for the treatment of relapsed/refractory ALL are especially limited [4]. Therefore, alternative therapeutic approaches are necessary to improve overall survival (OS), decrease the risk of relapse and/or minimize the toxicity of relapsed/refractory ALL.

    CD19 CARs redirected T cells can improve remission rates for patients with B-lineage hematological malignancies [5–11]. A CAR consists of an antigen-binding region of a monoclonal antibody linked to intracellular T-cell signaling domains [12]. T cells expressing CARs (CAR-T cells) can be redirected to recognize and kill the tumors without the restriction of major histocompatibility complexes. CAR-T cells can expand and effectively induce antitumor responses after transfusion into patients. Compared with therapeutic monoclonal antibodies and related approaches, a greater potency and more durable responses were achieved with CAR-T cells [13]. Although, CAR-T cells showed a favorable outcome in the treatment of relapsed/refractory ALL, the persistence is a noteworthy question and some of the cases quickly relapsed after CAR-T cell treatment [14]. Allo-HSCT is still the best way to treat refractory/relapsed ALL [15]. Thus, treatment with CAR-T cells, followed by allo-HSCT, may be the best treatment for relapsed/refractory ALL. In this review, we discuss CAR-T cells as a bridge for the treatment of relapsed/refractory ALL with allo-HSCT. We first discuss the different types of CAR-T cells. We then discuss the treatment of relapsed/refractory ALL using only CAR-T cells. Finally, we discuss the use of CAR-T cells, followed by allo-HSCT, for the treatment of relapsed/refractory ALL.

    CAR-T cells

    CARs typically consist of three parts, including an extracellular major histocompatibility antigens-independent antigen-binding domain that is usually derived from a single-chain variable fragment of a monoclonal antibody, a transmembrane-linking domain and an intracellular costimulatory T-cell signaling domain or multiple domains [16]. Some CARs also include an extracellular spacer domain (Figure 1).

    Figure 1. Composition of a chimeric antigen receptor and the generations of chimeric antigen receptors.

    A CAR is composed of an extracellular single chain variable fragment, linked to a transmembrane domain (CD8 or CD28) through a hinge (CD8 or IgG4), one or more intracellular costimulatory molecules (41-BB, CD28, CD27, ICOS and/or OX40) or cytokines (IL-12), and a CD3z signaling molecule. The ITAM and different costimulatory molecules at transmembrane region define the different generations. The first generation had a single chain antibody (CD3ζ or FcεRIγ) links the ITAM. The second generation, based on the first, added a CM1. Another CM2 was added in the third generation. The IL-12 was added to CARs from the second generation to create the fourth generation.

    CAR: Chimeric antigen receptor; CM1: Costimulatory molecule 1; CM2: Costimulatory molecule 2; ITAM: Immunoreceptor tyrosine-based activation motif.

    There have been four generations of CAR-T cells devised thus far (Figure 1). A CD3ζ intracellular signaling domain (the ‘first signal’) was utilized to activate T cells in the first generation of CARs; however, these CARs did not produce enough IL-2 and had limited activation, expansion and in vivo antitumor effects [17]. The second generation of CARs was then produced through the addition of costimulatory molecules with intracellular costimulatory signaling domains, such as CD28, CD137(4–1BB), CD134(OX40), CD27, ICOS and CD244. The second generation of CARs produced 20-times more IL-2 compared with the first generation and showed continued proliferation and killing of target cells in vivo [18–21]. The third generation of CARs was produced by adding two or more costimulatory molecules with intracellular costimulatory signaling domains. Thus, more cytokines were secreted, and killing ability was enhanced in vitro; however, better outcomes were not achieved in the clinic [22,23]. The fourth generation of CARs, knowns as T-cell redirected for universal cytokine killing, or TRUCK, were produced by adding a sequence of cytokines, such as IL-12, which can kill the tumors through releasing cytokines locally to regulate the microenvironment and recruit immune effector cells. TRUCKs can be used to treat virus infections, auto-immune diseases or metabolic disorders but not cancer [24].

    Single CAR-T cells for refractory/relapsed ALL

    Treatment of relapsed/refractory CD19+ B-cell malignancies in children, adolescents and young adults using CD19-CAR-T cells was performed in Phase I clinical trial. Retrovirally transduced CD19-CAR T cells with CD3ζ and CD28 costimulatory domains (CD19-CAR.z.28) were used. All of the patients received chemotherapy with fludarabine and cyclophosphamide prior to CD19-CAR.z.28 T-cell infusion. In total, 66.7% (14/21) achieved complication remission (CR). For the 20 patients with B-ALL, the CR rate was 70%. Molecular remissions were achieved in a total of 12 out of the 20 patients (60%) with no flow cytometric evidence of minimal residual disease (MRD < 0.01%). Two patients who underwent CAR-T cell treatment relapsed with CD19-negative B-ALL at 3 and 5 months post-treatment [25]. Davila et al. used 19–28z CAR-T cells to treat 16 adults with relapsed/refractory B-cell ALL at the Memorial Sloan-Kettering Cancer Center at a dose of 3 × 106 CAR T cells/kg. The results showed that 88% of patients reached a CR or incomplete count recovery, and 75% of the treated patients achieved MRD-negative disease status [26]. Nearly, all of the patients had a peak number of CAR-T cells within 1–2 weeks after the infusion, and these numbers decreased to low or undetectable levels by 2–3 months. At the National Cancer Institute/National Institutes of Health, CAR-T cells containing the CD28 and CD3z domains were used to treat relapsed/refractory ALL at a dose of 1 × 106/kg [25]. A variably intensive lymphodepletion prior to the treatment with CAR-T cells was used for the 21 pediatric and adult patients with either low- or high-burden disease. Fifteen of these patients reached CR.

    Lentivirally-transduced CD19-CAR-T cells with CD3ζ and 4–1BB (CD137) endodomains (CD19-CAR-CD3ζ-41BB) were also used to treat relapsed/refractory B-cell malignancies. Two children with relapsed/refractory ALL were treated with CAR-T cells at a dose of 1.4 × 106 to 1.2 × 107 per kilogram of body weight. Both patients showed CR, but while one patient was still in CR 11 months after treatment, the other relapsed within two months [27]. Maude et al. also treated 30 patients, including 18 relapsed patients after hematopoietic stem cell transplantation (HSCT), with CD19-CAR-CD3ζ-41BB cells. Out of the 30 patients, 27 (90%) achieved morphologic CR, and 22 out of the 27 also achieved molecular CR (MRD < 0.01%) 1 month after CAR-T cell treatment [28]. The 1-year disease-free survival (DFS) rate was 44%, and patients with long-term persistence of CTL019 cells experienced the longest DFS even without subsequent HSCT. However, 20 patients relapsed, and 13 of them had CD19-negative blasts [28,29]. Thirty patients with relapsed/refractory ALL were treated with CD19-CAR-CD3ζ-CD28–41BB cells. All of the patients (100%) survived for more than 21 days and had no morphologically detectable leukemia in the BM. In 27 out of 29 patients (93%), leukemia was undetectable by high-resolution flow cytometry [30].

    Allogeneic CD19-CAR-T cells have been used to treat patients with relapsed ALL after allo-HSCT [31]. In a Phase I clinical trial, the feasibility and safety of donor-derived CD19-CAR-T cells were evaluated in four patients relapsing after HSCT, six ALL patients (2 Ph+) and two patients at a high risk due to MRD+ [32]. The infusion was well-tolerated, and no patient developed graft-versus-host disease (GVHD). Six patients had CR lasting from 2 to 8 months. An 11-year-old girl with relapsed ALL after allo-HSCT was treated with CAR-T cells. After an infusion of 1 × 106/kg CAR-T cells, CAR-T-cell infusions (0.83–1.65) × 106/kg were administered three-times for maintenance. The DFS lasted for 10 months [33].

    CD19-CAR-CD3ζ-41BB cells were also used to treat nine adult patients with B-ALL and extramedullary leukemia. Six of the nine patients with definite extramedullary involvement achieved 56% 18-weeks OS. One of the two patients with partial regression of extramedullary lesions who received conditioning chemotherapy achieved a three-month-durable complete response. Four of seven patients without conditioning chemotherapy achieved dramatic regression or a mixed response in the hematopoietic system and extramedullary tissues for 2–9 months [34]. CAR-T cells are also effective against CNS leukemia. Gene marking studies revealed the presence and persistence of CAR-T cells in the cerebrospinal fluid of 46 out of 47 patients without an increased risk of encephalopathy, and four patients with CNS leukemia reached remission after CAR-T-cell treatment [29].

    In a Phase II multicenter clinical trial in China, 50 refractory/relapsed ALL patients were treated with the CD19scFv/CD28/CD137/CD27/CD3ζ-iCasp9 (4SCAR-19) lentivector. Out of the patients with blasts < 50% (group 1), 94.3% reached CR and of the patients with blast >50% (group 2), 66.7% (10/15) reached CR [35]. The 120-days LFS for patients in group 1 and group 2 were 86 and 44.4%, respectively. The rates of 10-month OS for patients in group 1 and group 2 were 82 and 36%, respectively.

    Recently, a meta-analysis investigated the efficacy of CD19-CAR-T cells in refractory B-cell malignances in Phase I clinical trials. Data from 1991 to 2014 were collected from PubMed and the Web of Science. A response rate evaluation was analyzed from fourteen clinical trials and included data from 119 patients. Progression-free survival (PFS) analysis was performed on data from 62 patients in 12 clinical trials. The overall pooled response rate of CD19-CAR-T cells was 73%. ALL patients had higher response rates than chronic lymphocytic leukemia and lymphoma patients. Lymphodepletion and the absence of IL-2-administrating T cells were two key factors associated with a better clinical response. The 6-month and 1-year PFS rates for the 62 patients were 80.0 and 76.3%, respectively. The median interval of PFS was 7.0 months. Higher numbers of infused CAR-T cells and lymphodepletion were associated with a better prognosis [36].

    Altogether, approximately 40–90% of patients with refractory/relapsed ALL can reach complete molecular or cellular remission through treatment with CAR-T cells; however, because CAR-T cells survived in the body for only a short time, some of the patients relapsed quickly, which may be related to the different in the costimulatory molecular or vectors used, or variation in the disease burden across treated individuals [36,37] (Table 1). Thus, other treatments should be performed to improve outcomes in the treatment of refractory/relapsed ALL.

    Table 1. Selected summary of the reported clinical trials of chimeric antigen receptor-T-cell therapy in B-acute lymphoblastic leukemia.
    CenterTargetNo.CARVectorDiseaseResultsRole of HSCTChemotherapyNotesRef.
    NCICD192028-ζRVChildren and young adult patientsCR 61%, OS 51% at 6 months, DFS 78.8% at 6 months17/20 patients with MRD negative disease went to HSCTFlu/CyNo relapse after HSCT, 2/3 patients with no HSCT relapsed with CD19-negative disease[25]
    MSKCCCD191628-ζRVAdults ALL88% CR or CRi, 75% achieved an MRD-negative disease status7 of the 16 (44%) patients eligible for an allo-SCT treated for an allo-SCTN/ANo relapses for these patients underwent allo-HSCT after CAR-T therapy[26]
    UPenn/CHOPCD1930CD28-4-1BB-ζLVChildren and adults90% CR, 22/27 patients with MRD negative CR3 patients underwent allo-HSCTVariable3 patients underwent allo-HSCT remained in remission 7 to 12 months after the CAR-T infusion[28]
    UPenn/CHOPCD19534-1BB-ζLVChildren and yong adult patientsCR 50/53(94%). EFS: 70% at 6 months and 45% at 12 months; OS: 78 % at 12 months.6/29 patients in CR received HSCT. Of 20 relapsed patients, 3 has HSCT after CAR-T treatmentFlu/Cy20 relapsed, 13/20 (65%) with CD19-negative disease. CAR-T cells persisted for 3-39 months. 4 patients with leukemic meningitis reached remission after CAR-T-cell treatment[29]
    FHCRCCD1930CD28-4-1BB-ζLVALLCR 30/30 (100%), 27/29(93%) at MRD negative CRN/AFlu/Cy or Cy/VPFirst CAR with defined CD4+ and CD8+ T cell subsets[30]
    BCMCD19628-ζRVALLCR 100%N/AN/ACR lasting from 2 months to 8 months[32]
    China (multicenter)CD195028-4-1BB-27-ζLVALLCR 94.3% when balsts <50%, 66.7% when blasts >50%N/AVariableFirst 4th-G CAR[35]
    MSKCCCD194628-ζRVAdult ALL83% CR rate12 patients underwent HSCTCyNo differences in outcomes whether or not HSCT was done. The OS at 6 months was 76 and 14% in the MRD-CR cohort and in the MRD+ CR cohort, respectively[39]
    MDACCCD194228-ζSBAdjuvant post-allo-HSCT(10), relapse (8)Adjuvant trial: CR 3/10(30%); Relapse trial: CR 3/13(23%, for all disease)N/AN/A [70]

    4th-G: Fourth-generation; 4-1BB is alternatively known as CD137 or TNFRSF9 (TNF receptor superfamily member 9); ALL: Acute lymphoblastic leukemia; Allo-HSCT: Allogeneic hematopoietic stem cell transplantation; BCM: Baylor College of Medicine; CAR: Chimeric antigen receptor; CAR-T cell: Chimeric antigen receptors redirected T cell; CHOP: Children's Hospital of Philadelphia; CR: Complete remission; FHCRC: Fred Hutchinson Cancer Research Center; HSCT: Hematopoietic stem cell transplantation; LV: Lentiviral; MDACC: MD Anderson Cancer Center; MRD: Minimal residual leukemia; MSKCC: Memorial Sloan-Kettering Cancer Center; NCI: National Cancer Institute, RV: Retroviral; SB: Sleeping beauty; UPenn: University of Pennsylvania.

    CAR-T cells followed by allo-HSCT for refractory/relapsed ALL

    At present, allo-HSCT is still the best way to treat refractory/relapsed ALL [15]. However, allo-HCT is largely ineffective for refractory/relapsed ALL patients at the time of transplantation. Although, the initial outcomes are inspiring for refractory/relapsed ALL treated with CAR-T cells, some of the cases relapsed within 3–6 months, which represents a serious obstacle for CAR-T cells treatment that must be overcome if it is to be widely adopted [38]. We suggest that the best way to treat refractory/relapsed ALL may be to perform CAR-T-cell treatment followed by allo-HSCT to improve survival rates for patients.

    Lee et al. reported on 10 patients with refractory/relapsed ALL at the NIH who achieved MRD-negative disease after CAR-T cells infusion and then underwent allo-HSCT while in CAR-induce MRD-negative CR. These patients remain disease-free with no unexpected peritransplant toxicities and no subsequent relapses [25]. However, in the same study, two patients who achieved MRD-negative CR were judged ineligible by their treating physicians for allo-HSCT, and both relapsed with CD19-negative leukemia at 3 and 5 months, respectively.

    Brentjens et al. reported four patients that were treated with CAR-T cells, followed by allo-HSCT [7]. One patient died of a suspected pulmonary embolus 2 months after the allo-HSCT while in CR, with no evidence of any disease. The other three patients had no significant complications and remained in MRD-negative CR from 3 to 6 months after allo-HSCT. However, one patient who was ineligible for allo-HSCT relapsed 3 months after CAR-T-cell treatment. Maude et al. reported that three patients in remission were treated with CAR-T cells followed by allo-HSCT, and they remained in remission 7–12 months after CAR-T-cell treatment [30].

    Davila et al. reported that no relapses were happened for seven of the 16 (44%) refractory/relapsed ALL patients treated with CAR-T cells followed by allo-HSCT therapy [26]. In another study, adults with refractory/relapsed ALL were treated with CD19-CAR-T cells. After achieving a CR, 13 of 37 patients underwent allo-HSCT. No significant difference was found between patients who underwent allo-HSCT and those who did not (79 vs 80%) in terms 6-month OS [39]. However, it is difficult to estimate whether there was a greater risk of relapse for the patients who underwent consolidative allo-HSCT because it was not a randomized trial. Recently, the absence of post-treatment MRD was used as a predictive marker of survival and the effect of allo-HSCT after CAR-T-cell therapy was studied in refractory/relapsed ALL patients [39]. The OS rate at 6 months was 76% in the MRD CR cohort and 14% in the MRD+ CR cohort. Further, post-treatment MRD status emerged as a strong predictive marker of OS. Further analysis showed that the OS rates at 6 months were 70% in patients who underwent post-CAR allo-HSCT and 64% in patients who did not get allo-HSCT after CAR-T cells therapy; this difference was statistically insignificant, but suggestive. The follow-up time may have been too short and the number of cases too small to detect a statistically significant difference.

    Although, other studies have treated refractory/relapsed ALL patients with CAR-T cells followed by allo-HSCT, the number of cases in these studies were too few to draw definitive conclusions, and the authors did not report the outcomes of CAR-T treatment with and without allo-HSCT separately. The question of whether consolidation allo-HSCT is required to maintain remission induced by CAR-T cells in ALL remains largely unanswered, but can and should be tested in clinical trials. Given the exciting clinical outcomes reported for the treatment of refractory/relapsed ALL with CAR-T cells followed by allo-HSCT in the phase I setting, a Phase II trial (ChiCTR-OOC-16008448) already underway at our center will formally assess the efficacy of this experimental treatment in appropriately powered cohorts of patients with relapsed/refractory B-ALL. However, it is very important to identify some key factors in treatment success that will help to prevent recurrence for those patients who really need an allograft as consolidation therapy.

    Toxicities

    Cytokine-release syndrome (CRS), neurotoxicity and B-cell aplasia are the major toxic effects for CAR-T-cell treatment [40]. The most common and potentially severe toxic effect is CRS [18,32]. The risk of severe CRS may be related to the tumor burden [23]. The level of sIL2Ra, IFN-γ, IL-10 and IL-6 are remarkably elevated in CRS and are accompanied by the mild flu-like symptoms to, in the most severe cases, multisystem organ failure and shock [2,18]. More attention should be paid to the rates of severe CRS associated with CARs with different costimulatory molecules. More serious CRS appears to occur with the 4–1BB CAR-T cells compare with other costimulatory molecules [41–43].

    The most common neurologic side effect associated with CAR-T treatment is global encephalopathy, which may be brief and self-limited, but can lead to death [4]. Chronic B-cell aplasia and resultant hypogammaglobulinemia can also occur. Pneumonitis, fever, tumor lysis syndrome, hypotension, hypoxia, tachycardia and fatigue are also associated with CAR-T-cell therapy. However, most of these complications can be controlled.

    The additional cost for CAR-T therapy is also high, which may prevent its more widespread use in the near future. Allo-HSCT is good option for some patients because the morbidity and mortality from allo-HSCT is not insignificant, however, we must pay attention to the potential GVHD complications [44].

    Conclusion & future perspective

    CAR-T-cell therapy represents the beginning of a new era of using engineered T cells for cancer therapy. Although, it has shown its impressive effect in clinical studies, many obstacles preventing its broader application, including the complicated genetic technology required for manufacturing CAR-T cells, tumor escape, inadequate toxicity management, relatively fast relapse of the disease, a lack of persistence and the need to standardize CAR-T-cell treatment [45].

    The technology for manufacturing genetically modified T cells is complex and patient-specific. It will take time to build the cell-processing infrastructure to deliver this therapy to large numbers of patients outside major academic centers. However, the US FDA has approved the Novartis biologics license application for CART-19 in pediatric ALL. The Kite's BLA for CART-19 in diffuse large cell B lymphoma is also under consideration. The choice of effector T cells and CAR design must be optimized to improve effectiveness and limit toxicities [46]. It is difficult to compare across clinical trials because of the differences in conditioning chemotherapy, CAR constructs, CAR-T-cell production methods, toxicity management and the size of the tumor burdens.

    Another important aspect of CAR-T generation is the means of delivering the foreign gene into human cells. For CAR-T cells, viral system is the most commonly used tool for achieving this. Retroviruses (including lentivirus), adenovirus and adeno-associated virus are the main virus vectors. Among these, engineered retroviruses are the most popular tools for gene delivery [27]. But the insertion mutation used to induce the immune reaction can lead to tumorigenesis and toxicity, the carrier capacity is limited and the titer achieved is not high enough [47,48]. The safest vector that has been developed thus far may be the third-generation minimal lentiviral vector [49]. Thus, perhaps CAR-T cell treatment should be standardized to use this vector [50]. Formulating strategies that can improve cell isolation and culture methods and allow large-scale automation of CAR-modified T-cell production should also be considered, as this could facilitate their utilization in the clinic and reduce the cost of engineered T-cell therapies [24,51]. The accessibility and survival issues of CAR-T cells should also be resolved in the future.

    Multispecific CARs could be used to avoid tumor escape. The use of CARs whose activity can be tuned by administration of small molecule pharmaceuticals or incorporation of suicide genes might allow the control of adverse events. T cells that are resistant to hostile microenvironments or have enhanced persistence, such as those from the bone marrow, could be produced [52,53]. The combination of CAR-T cells with other immunotherapeutics, such as oncolytic viruses or checkpoint blockade antibodies, should also be explored, as this can increase efficacy [54].

    The management of toxicity is the central question for CAR-T-cell treatment. How toxicities could be solved without interfering with CAR-T-cell expansion, and whether conditioning chemotherapy is necessary should also be determined [13,55]. Many CAR-T-cell therapies used on patients with B-ALL have consisted of CD4+ and CD8+ T cells at a fixed 1:1 ratio [56]. It is unknown whether the ratio of T-cell subset is important in CAR-T cell therapy, so further study is needed to explore the optimum subset ratio [57]. Another major challenge will be to identify unique tumor antigens that can be targeted with selective T-cell therapy. Preclinical and early-phase clinical studies have shown that targeting multiple antigens in CAR-T-cell therapy can prevent antigen loss relapses. For example, CAR-T cells targeting CD19/CD20, CD19/CD22, or CD19/CD123, can also improve the extremely limited options and poor outcomes of patients with CD19-negative relapses [58–60]. Attention should also be paid to the uncontrolled proliferation of CAR-T-cell therapies. In order to specifically deplete CAR-T cells in a controlled manner, a suicide switch could be incorporated into the CAR construct, although this may also weaken persistence and therefore the ability of CAR-T cells to fight disease long-term [61–64]. Ultimately, cellular therapies should be integrated into the routine practice and standard of care for ALL.

    CAR-T cells with prolonged persistence may prevent relapses through long-term immunosurveillance. CAR-T cells generated using a lentiviral vector and 4–1BB costimulation seems to be more persistent than those made using a retroviral vector and CD28, however, there have been no direct comparisons [64]. Some experiments showed that the CAR-T cells with CD28 molecules can cause constitutive stimulation, proliferation and growth [65]. However, the 4–1BB molecules can induce early exhaustion of CAR-T cells, which may limit antitumor efficacy [66,67]. The high short-term potency in some situations may give rise to deep remissions, which may make up for the lack of long-term persistence [26]. Thus, presently the optimal persistence time remains unclear, although it seems that the persistence of CAR-T-cell therapy is critical.

    Donor lymphocyte infusion showed limited efficacy in producing a long-lasting graft-versus-leukemia effect or preventing a life-threatening GVHD associated with the infusion of a high number of T cells in ALL for the prevention of relapse or the treatment of relapse after allo-HSCT [68,69]. Patients treated with allogeneic CD19-CAR-T cells used for B-cell malignancies that progressed after allo-HSCT received a single infusion of CAR-T cells obtained from each recipient's allo-HSCT donor. No chemotherapy or other therapies were administered for these patients. eight out of 20 patients obtained remission, with six obtaining CR and two obtaining partial remissions. The response rate was highest for ALL, with four of five patients obtaining MRD-negative CR. No GVHD was observed [70]. Donor-derived allogeneic anti-CD19-CAR-T cells can cause regression of B-cell malignancies resistant to standard donor lymphocyte infusions without causing GVHD [9]. It is therefore a safe and feasible approach for patients to receive donor-derived CAR-T cell therapy for relapse after allo-HSCT [71,72]. Thus, early infusion of CAR-T cells could be used for the prevention of relapse for high-risk cases or when molecular relapse is detected. Presently, our center is performing a clinical trial (ChiCTR-OOC-16008447) in which CAR-T cells are used to prevent the relapse of B-ALL after allo-HSCT when molecular relapse is detected.

    Allogeneic CAR-T cells are beneficial for patients with relapsed B-cell malignancies after allo-HSCT, and have low toxicities and complications [68–73]. Recently, coinfusion of haploidentical donor-derived CD19- CAR-T cells and mobilized peripheral blood stem cells following induction chemotherapy was performed in a 71-year-old female with relapsed and refractory ALL. The patient achieved CR with undetectable MRD and full donor cell engraftment was observed with acute GVHD [74]. Therefore, CAR-T cells may be used as a part of conditioning regimen in allo-HSCT for refractory ALL. However, the time and numbers of CAR-T-cell infusion should be researched in this model.

    Altogether, CAR-T cells are an extremely promising area of research that holds endless potential for future treatment of ALL. However, we have only seen the tip of the iceberg, and many crucial questions have yet to be answered.

    Executive summary

    Background

    • The treatment of relapsed/refractory acute lymphoblastic leukemia (ALL) is still a serious challenge.

    • Although, CAR-T cells showed favorable outcomes in the treatment of relapsed/refractory ALL, the persistence is a noteworthy question and some of the cases quickly relapsed after CAR-T-cell treatment.

    • Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is still the best way to treat refractory/relapsed ALL at present.

    • Treatment with CAR-T cells, followed by allo-HSCT, may be the best treatment for relapsed/refractory ALL.

    CAR-T cells

    • CARs typically consist of three parts, including an extracellular major histocompatibility antigens-independent antigen-binding domain, a transmembrane-linking domain and an intracellular costimulatory T-cell signaling domain or multiple domains.

    • Four generations of CAR-T cells have been devised thus far.

    Single CAR-T cells for refractory/relapsed ALL

    • Most patients with refractory/relapsed ALL can reach complete molecular or cellular remission through treatment with CAR-T cells.

    • Some of the patients who receive CAR-T cells treatment relapse quickly.

    CAR-T cells followed by allo-HSCT for refractory/relapsed ALL

    • Good outcomes were observed for patients with refractory/relapsed ALL who treated by CAR-T cells followed by allo-HSCT.

    • whether consolidation allo-HSCT is required to maintain remission after CAR-T cells treatment in refractory/relapsed ALL remains largely unanswered.

    • It is very important to identify some key factors to prevent recurrence for those patients who really need an allograft as consolidation therapy after CAR-T-cell treatment.

    Toxicities

    • Cytokine release syndrome, neurotoxicity and B-cell aplasia are the major toxic effects of CAR-T-cell treatment.

    • Most of the toxicities can be controlled.

    Future Perspective

    • Many questions remain to be resolved before CAR-T cells can be put into broader use.

    • CAR-T cells can be as a conditioning regimen in allo-HSCT for refractory/relapsed ALL.

    • With the development of commercial manufacture, CAR-T cells will be more widely available as a drug to treat cancer in the future, which can benefit to many patients.

    • The combination of CAR-T cells with allo-HSCT has promising therapeutic potential.

    Financial & competing interests disclosure

    This study was funded by the clinical key foundation of Xinqiao Hospital of Third Military Medical University (No. 2016YLC03). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1 Bhojwani D, Pui C-H. Relapsed childhood acute lymphoblastic leukaemia. Lancet Oncol. 14(6), e205–e217 (2013).
    • 2 Raetz EA, Bhatla T. Where do we stand in the treatment of relapsed acute lymphoblastic leukemia? Hematology Am. Soc. Hematol. Educ. Program 129–136 (2012).
    • 3 Forman SJ, Rowe JM. The myth of the second remission of acute leukemia in the adult. Blood 121(7), 1077–1082 (2013).
    • 4 Schrappe M, Hunger SP, Pui CH et al. Outcomes after induction failure in childhood acute lymphoblastic leukemia. N. Engl. J. Med. 366(15), 1371–1381 (2012).
    • 5 Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365(8), 725–733 (2011).
    • 6 Kalos M, Levine BL, Porter DL et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci. Transl. Med. 3(95), 95–73 (2011).
    • 7 Brentjens RJ, Davila ML, Riviere I et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci. Transl. Med. 5(177), 177ra38 (2013).
    • 8 Kochenderfer JN, Yu Z, Frasheri D, Restifo NP, Rosenberg SA. Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. Blood 116(19), 3875–3886 (2010).
    • 9 Kochenderfer JN, Dudley ME, Carpenter RO et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 122(25), 4129–4139 (2013).
    • 10 Kochenderfer JN, Dudley ME, Kassim SH et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J. Clin. Oncol. 33(6), 540–549 (2015).
    • 11 Savoldo B, Ramos CA, Liu E et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J. Clin. Invest. 121(15), 1822–1826 (2011).
    • 12 Kershaw MH, Westwood JA, Darcy PK. Gene-engineered T cells for cancer therapy. Nat. Rev. Cancer 13(8), 525–541 (2013).
    • 13 Amrolia PJ, Pule M. Chimeric antigen receptor T cells for ALL. Lancet 385(9967), 488–490 (2015).
    • 14 Maude SL, Teachey DT, Porter DL, Grupp SA. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood 125(26), 4017–4023 (2015).
    • 15 Chen XH, Zhang C, Zhang X et al. Role of antithymocyte globulin and granulocyte-colony stimulating factor-mobilized bone marrow in allogeneic transplantation for patients with hematologic malignancies. Biol. Blood Marrow Transplant 15(2), 266–273 (2009).
    • 16 Tasian SK, Gardner RA. CD19-redirected chimeric antigen receptor-modified T cells: a promising immunotherapy for children and adults with B-cell acute lymphoblastic leukemia (ALL). Ther. Adv. Hematol. 6(5), 228–241 (2015).
    • 17 Brocker T. Chimeric Fv-zeta or Fv-epsilon receptors are not sufficient to induce activation or cytokine production in peripheral T cells. Blood 96, 1999–2001 (2000).
    • 18 Kowolik CM, Topp MS, Gonzalez S et al. CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Res. 66(22), 10995–1004 (2006).
    • 19 Milone MC, Fish JD, Carpenito C et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol. Ther. 17(8), 1453–1464 (2009).
    • 20 Song DG, Ye Q, Poussin M, Harms GM, Figini M, Powell DJ Jr. CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo. Blood 119(3), 696–706 (2012).
    • 21 Guedan S, Chen X, Madar A et al. ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cells. Blood 124(7), 1070–1080 (2014).
    • 22 Marin V, Pizzitola I, Agostoni V et al. Cytokine-induced killer cells for cell therapy of acute myeloid leukemia: improvement of their immune activity by expression of CD33-specific chimeric receptors. Haematologica 95(12), 2144–2152 (2010).
    • 23 Till BG, Jensen MC, Wang J et al. CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4–1BB domains: pilot clinical trial results. Blood 119(17), 3940–3950 (2012).
    • 24 Chmielewski M, Abken H. TRUCKs: the fourth generation of CARs. Expert Opin. Biol. Ther. 15(8), 1145–1154 (2015).
    • 25 Lee D, Kochenderfer J, Stetler-Stevenson M et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a Phase 1 dose-escalation trial. Lancet 385(9967), 517–528 (2014). •• The paper found that no relapse was observed for 17/20 patients with minimal residual disease (MRD) negative acute lymphoblastic leukemia (ALL) after CAR-T cells treatment and hematopoietic stem cell transplantation.
    • 26 Davila ML, Riviere I, Wang X et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci. Transl. Med. 6(224), 224ra25 (2014).
    • 27 Grupp S, Kalos M, Barrett D et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 368, 1509–1158 (2013).
    • 28 Maude S, Frey N, Shaw PA et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 371(16), 1507–1517 (2014).
    • 29 Grupp SA, Maude SL, Shaw PA et al. Durable remissions in children with relapsed/refractory ALL treated with T cells engineered with a CD19-targeted chimeric antigen receptor (CTL019). Blood 126, 681 (2015).
    • 30 Turtle CJ, Hanafi LA, Berger C et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Invest. 126(6), 2123–3218 (2016). •• This is the first CAR with defined CD4+ and CD8+ T-cell subsets.
    • 31 Magnani CF, Biondi A, Biagi E. Donor-derived CD19-targeted T cells in allogeneic transplants. Curr. Opin. Hematol. 22(6), 497–502 (2015).
    • 32 Cruz CR, Micklethwaite KP, Savoldo B 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 122(17), 2965–2973 (2013).
    • 33 Zuo YX, Wang JB, Lu AD et al. Chimeric antigen receptors T cells in treatment of a relapsed pediatric acute lymphoblastic leukemia, relapse after allogenetic hematopoietic stem cell transplantation: case report and review of literature review. Zhonghua Xue Ye Xue Za Zhi 37(2), 115–118 (2016).
    • 34 Dai H, Zhang W, Li X et al. Tolerance and efficacy of autologous or donor-derived T cells expressing CD19 chimeric antigen receptors in adult B-ALL with extramedullary leukemia. Oncoimmunology 4(11), e1027469 (2015).
    • 35 Dong L, Chang LJ, Gao Z et al. Chimeric antigen receptor 4SCAR19-Modified T cells in acute lymphoid leukemia: a Phase II multi-center clinical trial in china. Blood 126, 3774 (2015). • This is first report on the use of fourth-generation CAR-T cells for the treatment of refractory/relapsed ALL.
    • 36 Zhang T, Cao L, Xie J et al. Chimeric antigen receptor-modified T cells for treatment of B cell malignancies in Phase I clinical trials: a meta-analysis. Oncotarget 6(32), 33961–33971 (2015).
    • 37 Maude S, Barrett DM. Current status of chimeric antigen receptor therapy for haematological malignancies. Br. J. Haematol. 172(1), 11–22 (2016).
    • 38 Maude SL, Shpall EJ, Grupp SA. Chimeric antigen receptor T-cell therapy for ALL. Hematology Am. Soc. Hematol. Educ. Program 2014, 559–564 (2014).
    • 39 Park JH, Riviere I, Wang X et al. Implications of minimal residual disease negative complete remission (MRD-CR) and allogeneic stem cell transplant on safety and clinical outcome of CD19-targeted 19–28z CAR modified T cells in adult patients with relapsed, refractory B-cell ALL. Blood 126, 682 (2015). •• This paper studied MRD status post-CAR-T treatment as a factor in the success of allogeneic hematopoietic stem cell transplantation (allo-HSCT) after CAR-T-cell therapy for refractory/relapsed ALL patients. The results showed no differences in outcomes between the MRD− CR and MRD+ CR cohorts regardless of whether they received allo-HSCT. However, there were only a small number of cases and the follow-up time was short.
    • 40 Zhang C, Liu J, Zhong JF, Zhang X. Engineering CAR-T cells. Biomarker Res. 5, 22 (2017).
    • 41 Long AH, Haso WM, Shern JF et al. 4–1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. 21(6), 581–590 (2015).
    • 42 Song DG, Ye Q, Carpenito C et al. In vivo persistence, tumor localization, and antitumor activity of CAR-engineered T cells is enhanced by costimulatory signaling through CD137 (4–1BB). Cancer Res. 71(13), 461746–27 (2011). •• Compares the in vivo persistence, tumor localization and antitumor activity of CAR-T cells with different costimulatory signaling.
    • 43 Teachey DT, Lacey SF, Shaw PA et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov. 6(6), 664–679 (2016). •• A model that can accurately predict which patients are likely to develop severe cytokine-release syndrome before they become critically ill, which may guide future cytokine-directed therapy.
    • 44 Tanaka Y, Kurosawa S, Tajima K et al. Analysis of non-relapse mortality and causes of death over 15 years following allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 51(4), 553–559 (2016).
    • 45 Wang Z, Wu Z, Liu Y, Han W. New development in CAR-T cell therapy. J. Hematol. Oncol. 10(1), 53 (2017).
    • 46 Sommermeyer D, Hudecek M, Kosasih PL et al. Chimeric antigen receptor-modified T cells derived from defined CD8(+) and CD4(+) subsets confer superior antitumor reactivity in vivo. Leukemia 30(2), 492–500 (2016).
    • 47 Hu WS, Pathak VK. Design of retroviral vectors and helper cells for gene therapy. Pharmacol. Rev. 52(4), 493–511 (2000).
    • 48 Wang GP, Garrigue A, Ciuffi A et al. DNA bar coding and pyrosequencing to analyze adverse events in therapeutic gene transfer. Nucleic Acids Res. 36(9), e49 (2008).
    • 49 Dull T, Zufferey R, Kelly M et al. A third-generation lentivirus vector with a conditional packaging system. J. Virol. 72(11), 8463–8471 (1998).
    • 50 Riet T, Abken H. Chimeric antigen receptor T cells: power tools to wipe out leukemia and lymphoma. Expert Rev. Hematol. 8(4), 383–385 (2015).
    • 51 Levine BL. Performance-enhancing drugs: design and production of redirected chimeric antigen receptor (CAR) T cells. Cancer Gene Ther. 22(2), 79–84 (2015).
    • 52 Dolnikov A, Shen S, Klamer G et al. Antileukemic potency of CD19-specific T cells against chemoresistant pediatric acute lymphoblastic leukemia. Exp. Hematol. 43(2), 1001–1014 (2015).
    • 53 Maus MV, June CH. Making better chimeric antigen receptors for adoptive T-cell therapy. Clin. Cancer Res. 22(8), 1875–1884 (2016).
    • 54 Enblad G, Karlsson H, Loskog AS. CAR T-cell therapy: the role of physical barriers and immunosuppression in lymphoma. Hum. Gene Ther. 26(8), 498–505 (2015).
    • 55 Turtle CJ. Chimeric antigen receptor modified T cell therapy for B cell malignancies. Int. J. Hematol. 99(2), 132–140 (2014).
    • 56 Turtle CJ, Berger C, Sommermeyer D et al. Immunotherapy with CD19-specific chimeric antigen receptor (CAR)-modified T cells of defined subset composition. J. Clin. Oncol. 33, 3006 (2015).
    • 57 Levine BL, Miskin J, Wonnacott K, Keir C. Global manufacturing of CAR-T cell therapy. Mol. Ther. Methods Clin. Dev. 4, 92–101 (2016).
    • 58 Ruella M, Barrett DM, Kenderian SS et al. Combination of anti-CD123 and anti-CD19 chimeric antigen receptor T cells for the treatment and prevention of antigen-loss relapses occurring after CD19-targeted immunotherapies. Blood 126, 2523 (2015).
    • 59 Qin H, Haso W, Nguyen SM, Fry TJ. Preclinical development of bispecific chimeric antigen receptor targeting both CD19 and CD22. Blood 126, 4427 (2015).
    • 60 Zah E, Lin MY, Silva-Benedict A, Jensen MC, Chen YY. T cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B cells. Cancer Immunol. Res. 4(6), 498–508 (2016).
    • 61 Budde LE, Berger C, Lin Y et al. Combining a CD20 chimeric antigen receptor and an inducible caspase 9 suicide switch to improve the efficacy and safety of T cell adoptive immunotherapy for lymphoma. PLoS ONE 8(12), e82742.
    • 62 Di Stasi A, Tey SK, Dotti G et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 365(18), 1673–1683 (2011).
    • 63 Kloss CC, Condomines M, Cartellieri M, Bachmann M, Sadelain M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat. Biotechnol. 31(1), 71–75 (2013).
    • 64 Porter DL, Hwang WT, Frey NV et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci. Transl. Med. 7(303), 303ra139 (2015).
    • 65 Frigault MJ, Lee J, Basil MC et al. Identification of chimeric antigen receptors that mediate constitutive or inducible proliferation of T cells. Cancer Immunol. Res. 3(4), 356–367 (2015).
    • 66 Long AH, Haso WM, Shern JF et al. 4–1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. 21(6), 581–590 (2015).
    • 67 Kawalekar OU, O'Connor RS, Fraietta JA et al. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity 44(2), 380–390 (2016).
    • 68 Porter DL, Alyea EP, Antin JH et al. NCI first international workshop on the biology, prevention, and treatment of relapse after allogeneic hematopoietic stem cell transplantation: report from the committee on treatment of relapse after allogeneic hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 16(11), 1467–503 (2010).
    • 69 Deol A, Lum LG. Role of donor lymphocyte infusions in relapsed hematological malignancies after stem cell transplantation revisited. Cancer Treat. Rev. 36(7), 528–538 (2010).
    • 70 Brudno JN, Somerville RP, Shi V et al. Allogeneic T cells that express an anti-CD19 chimeric antigen receptor induce remissions of B-cell malignancies that progress after allogeneic hematopoietic stem-cell transplantation without causing graft-versus-host disease. J. Clin. Oncol. 34(10), 1112–1121 (2016).
    • 71 Liu J, Zhong JF, Zhang X, Zhang C. Allogeneic CD19-CAR-T cell infusion after allogeneic hematopoietic stem cell transplantation in B cell malignancies. J. Hematol. Oncol. 10(1), 35 (2017). •• The paper systematically reviewed the data and found that infusion of allogeneic CAR-T cells was safe and effective for treating relapsed ALL after allo-HSCT.
    • 72 Hu Y, Sun J, Wu Z et al. Predominant cerebral cytokine release syndrome in CD19-directed chimeric antigen receptor-modified T cell therapy. J. Hematol. Oncol. 9(1), 70 (2016). • The paper first reported predominant cerebral cytokine-release syndrome after CAR-T cells for refractory/relapsed ALL after allo-HSCT.
    • 73 Anwer F, Shaukat AA, Zahid U et al. Donor origin CAR T cells: graft versus malignancy effect without GVHD, a systematic review. Immunotherapy 9(2), 123–130 (2017). • The paper systematically reviewed the data and found that only 5 out of 72 patients from 7 studies treated with donor-derived CAR T cells (6.9%) developed moderate graft-versus-host disease.
    • 74 Cai B, Guo M, Wang Y et al. Co-infusion of haplo-identical CD19-chimeric antigen receptor T cells and stem cells achieved full donor engraftment in refractory acute lymphoblastic leukemia. J. Hematol. Oncol. 9(1), 131 (2016). •• The paper first showed that it was safe and effective for CAR-T cells to be coinfused with hematopoietic stem cell transplantation in the treatment of refractory/relapsed ALL.