Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Adoptive cellular immunotherapy with chimeric antigen receptor (CAR) T cell has changed the treatment landscape of B-cell non-Hodgkin's lymphoma (NHL), especially for aggressive B-cell lymphomas. Single-center and multicenter clinical trials with anti-CD19 CAR T-cell therapy have shown great activity and long-term remissions in poor-risk diffuse large B-cell lymphoma (DLBCL) when no other effective treatment options are available. Two CAR T-cell products [axicabtagene ciloleucel (axi-cel) and tisagenlecleucel] have obtained US Food and Drug Administration approval for the treatment of refractory DLBCL after two lines of therapy. A third product, liso-cel, is currently being evaluated in clinical trials and preliminary results appear very promising. CAR T-cell-related toxicity with cytokine-release syndrome and neurotoxicity remain important potential complications of this therapy. Here, we review the s biology, structure, clinical trial results and toxicity of two commercially approved CAR T-cell products and others currently being studied in multicenter clinical trials in B-cell NHLs.

Free full text 


Logo of tahLink to Publisher's site
Ther Adv Hematol. 2019; 10: 2040620719841581.
Published online 2019 Apr 15. https://doi.org/10.1177/2040620719841581
PMCID: PMC6466472
PMID: 31019670

CAR T-cell therapy for B-cell lymphomas: clinical trial results of available products

Abstract

Adoptive cellular immunotherapy with chimeric antigen receptor (CAR) T cell has changed the treatment landscape of B-cell non-Hodgkin’s lymphoma (NHL), especially for aggressive B-cell lymphomas. Single-center and multicenter clinical trials with anti-CD19 CAR T-cell therapy have shown great activity and long-term remissions in poor-risk diffuse large B-cell lymphoma (DLBCL) when no other effective treatment options are available. Two CAR T-cell products [axicabtagene ciloleucel (axi-cel) and tisagenlecleucel] have obtained US Food and Drug Administration approval for the treatment of refractory DLBCL after two lines of therapy. A third product, liso-cel, is currently being evaluated in clinical trials and preliminary results appear very promising. CAR T-cell-related toxicity with cytokine-release syndrome and neurotoxicity remain important potential complications of this therapy. Here, we review the s biology, structure, clinical trial results and toxicity of two commercially approved CAR T-cell products and others currently being studied in multicenter clinical trials in B-cell NHLs.

Keywords: B-cell lymphoma, chimeric antigen receptor, cytokine-release syndrome, immunotherapy, refractory

Introduction

The use of chimeric antigen receptor (CAR) modified T cells targeting specific tumor-cell antigens has certainly changed the landscape of immunotherapy in cancer. This technology involves harnessing cytotoxic immune T cells in order to target specific tumor-cell antigens. In non-Hodgkin’s lymphomas (NHLs), specifically in diffuse large B-cell lymphoma (DLBCL), targeting CD19+ malignant B cells has proven highly efficacious in the refractory-disease setting when no other available treatment options exist. As a result, two CAR T-cell products are nowadays approved for refractory DLBCL. Here, we extensively review the mechanism of action, efficacy and toxicity(ies) of available CAR T-cell products currently in clinical use for B-cell NHLs.

Treatment overview of selected B-cell lymphomas

It is estimated that in 2019, there will be 74,200 diagnosed cases of NHL with approximately 19,970 disease-specific related deaths.1 NHL is the seventh leading cause of new cancer cases and accounts for approximately 3% of cancer-related deaths in the United States.1 Among all NHLs, DLBCL is the most common lymphoma subtype comprising 32.5% of all newly diagnosed cases, followed by follicular lymphoma (FL) with 17.1%, and mantle-cell lymphoma (MCL) representing 3–5%.2

Diffuse large B-cell lymphoma

Over 25,000 new cases of DLBCL are diagnosed annually in the United States, representing an incidence rate of 6.9 per 100,000.3 Addition of the anti-CD20 monoclonal antibody, namely rituximab, to the standard chemotherapy, R-CHOP, resulted in significant improvement in complete response (CR) rates, event-free (EFS) and overall (OS) survival in DLBCL.4 Unfortunately, approximately 30–40% of cases relapse or progress after R-CHOP.5 There are specific subgroups of patients who will have poor responses and outcomes to standard R-CHOP such as MYC-rearranged DLBCL, high-grade B-cell lymphomas with MYC, BCL2 or BCL rearrangements, activated B-cell (ABC) DLBCL that could benefit from novel approaches.611

High-dose therapy followed by autologous hematopoietic cell transplantation (auto-HCT) is considered standard of care in patients with relapsed DLBCL that is sensitive to salvage chemoimmunotherapy, typically a platinum-containing base regimen.12,13 Randomized controlled studies and registry data have shown better survival with auto-HCT vis-à-vis standard chemotherapy or chemoimmunotherapy.12,14 Nonetheless, 40–50% of the cases will not be eligible for auto-HCT due to chemorefractory disease, and the other 50% who undergo the procedure are at risk of disease relapse postautografting.12,14,15 Unfortunately, salvage therapies have limited efficacy in some relapsed/refractory settings such as primary progression, stable disease after frontline therapy and relapsed disease within 12 months from diagnosis, showing short-lasting objective response rates of only 26% (complete response rate of 7%) and an overall survival (OS) of 6.3 months.16,17 In patients who ultimately receive an allogeneic HCT (allo-HCT), the 5-year OS ranges from 18–37%, based on two registry studies from the Center for International Blood and Marrow Transplant Research (CIBMTR).1820 This limited efficacy of allo-HCT is in large part due to the high nonrelapse mortality (NRM), which may exceed 40%, mainly when using myeloablative conditioning (MAC) regimens.18,21,22

Follicular lymphoma

FL is a biologically heterogeneous disease that represents the most common type of indolent NHL in the Western world.23,24 There are several prognostic tools or models that integrate clinical data, laboratory studies and even molecular data that stratify the disease in different risk subgroups with specific outcomes.2527

Combination of conventional chemotherapy plus rituximab is considered the standard frontline treatment of patients with FL and other indolent lymphomas.28 Treatment response is an important determinant of outcomes in patients with lymphomas, including FL subtype. Trotman and colleagues, in a pooled analysis from three multicenter studies evaluating six cycles of frontline rituximab-based chemotherapy for high-tumor-burden FL prior to response assessment with conventional contrast-enhanced computed tomography (CT) and positron emission tomography (PET) low-dose CT, demonstrated that achievement of CR was associated with good prognosis.2932 Duration of first remission (CR1) has shown as prognostic in a landmark study that used data from the National LymphoCare Study (NLCS) that showed disease progression within 2 years from initial therapy was associated with inferior 5-year OS (50% versus 90%) in patients with stage 2–4 FL treated with R-CHOP as frontline regimen.33 A combined observational study from the NLCS and CIBMTR showed that early use of auto-HCT (defined as within 1 year of frontline induction failure) was associated with significantly reduced mortality [hazard ratio = 0.63; 95% confidence interval (CI) = 0.42–0.94, p = 0.02].34

Patients with FL relapsing after multiple lines of therapy are offered an allo-HCT with curative intent if deemed eligible for the procedure. Use of MAC regimens have been associated with high NRM exceeding 40%.35,36 Availability of reduced-intensity conditioning regimens have expanded allo-HCT to patients with FL owing to a more favorable toxicity profile, a lower risk of NRM of 16% and encouraging 3-year OS exceeding 80%.37,38 Although impressive, there are several limitations to universally offering allo-HCT to FL patients due to the fact that these patients tend to, generally, be of more advanced age and have associated comorbidities that may disqualify them from receiving the procedure.

Mantle-cell lymphoma

MCL is a relatively rare entity accounting for approximately 3–5% of all NHL cases.39,40 It is a distinct subtype of B-cell lymphoma which is diagnosed by detection of cyclin D1, immunophenotyping of cell surface antigens (CD5+, CD20+, CD23−), and molecular testing for the t(11;14) (q13;q32) by fluorescence in situ hybridization.39 In line with prognostic tools available for other NHLs, the MCL International Prognostic Index (IPI; MIPI) has been developed.41 MIPI segregates MCL patients into three distinct prognostic risk subgroups: low, intermediate, and high, with anticipated median OS of not reached, 51 months, and 29 months, respectively.41

High-dose therapy followed by auto-HCT is considered an optimal treatment strategy as frontline consolidation for chemosensitive disease, particularly younger patients or even for older patients who have adequate organ function and good performance status. The Nordic MCL trial treated 160 consecutive patients, treatment naïve, younger than 66 years, in a phase II protocol with dose-intensified induction R-CHOP, alternating with rituximab plus high-dose cytarabine. Authors reported excellent outcomes with long-term efficacy.42 For patients of more advanced age with or without associated comorbidities and poor performance status, practicing hematologists generally prescribe R-CHOP as the preferred frontline treatment choice; however, other regimens such as bendamustine and rituximab (BR) are also offered.43,44

For relapsed/refractory MCL, either ibrutinib or acalabrutinib have elicited excellent responses but cures are not anticipated and patients will eventually relapse.45,46 Prognosis of relapsed/refractory MCL is generally poor after failing an auto-HCT. An analysis from the European Society for Blood and Marrow Transplantation (EBMT) showed a 5-year OS of 34% in patients who receive an allo-HCT at the expense of an NRM of 30%.47 Patients who received an allo-HCT after a late relapse (defined as > 12 months) from auto-HCT had superior OS when compared with those with earlier progression after autografting.47 Newer and more effective therapies are needed for patients with relapsed/refractory MCL.

Rationale for CAR T-cell therapy in B-cell lymphomas

The basic anatomy of a CAR structure consists of an antigen-recognition domain, usually a single-chain variable fragment (scFv) derived from a monoclonal antibody targeting the selected antigen (i.e. CD19); a hinge [usually derived from CD8 or immunoglobulin 4 (Ig4) molecules] that links the recognition site to the transmembrane domain which bridges the membrane; and finally, the intracellular domain that typically contains a CD3ζ chain critical for T-cell receptor (TCR) signaling. Second-generation CAR molecules contain a second costimulatory-signaling molecule, such as CD28 or 4-BB, that enhances T-cell activation and antitumor potency.4851 CD19 is a transmembrane glycoprotein involved in regulating activation of B cells in an antigen–receptor-dependent manner. CD19 is uniformly expressed at all stages of B-cell differentiation and it is carried during B-cell malignant transformation.52 CD19 is expressed in over 95% of B-cell malignancies, such as chronic lymphocytic leukemia (CLL), B-cell NHL, and B-cell acute lymphoblastic leukemia (ALL). Although CD19 is expressed on normal, nonmalignant B cells, it is well established that patients can survive depleted B-cell levels resulting from chemotherapy or chemoimmunotherapy.

All these factors make CD19 an attractive target for immunotherapeutic approaches. Several companies and academic institutions have developed and continue developing pivotal trials with anti-CD19 CAR T-cell-directed therapies

Overview of CAR T-cell products and manufacturing process

CAR T-cell biology

CAR T-cells represent an autologous cellular immunotherapy using gene transfer to reprogram T cells to recognize and eliminate cancerous cells by targeting tumor-associated antigens. Although CAR T-cell therapies have been recently approved for wide commercial use, this is hardly a new concept, as earlier reports showed the feasibility of combining a monoclonal antibody originally developed the idea engineering T-cell-derived scFv region with TCR-associated activation domains from CD3ζ or CD3γ. This strategy combines antibody specificity with the homing, tissue penetration and target-cell destruction mediated by T lymphocytes.53 The first-regeneration CARs delivered activated T cells against specific tumor specific antigens but demonstrated limited persistence and weak proliferation, leading to limited antitumor activity.54 According to the known two-step process for T-cell activation, costimulation is necessary for complete stimulation; therefore, second-generation CAR T cells included costimulatory domains that led to a significant improvement in signaling strength, expansion and persistence.55 The most widely used costimulatory domains are CD28 and 4-1BB, but other molecules such as OX40 and CD27 have also shown enhanced CAR T-cell function.56 Second-generation CAR T cells, as we know them today, contain three components: an extracellular antigen-recognition domain, a transmembrane domain and an intracellular signaling domain (as discussed above).50 The majority of the trials are utilizing second-generation CARs. In general, CD28-based CARs have a greater expansion but less persistence in contrast to 4-1BB based CARs which appear to have longer persistence. It remains to be seen whether these properties have clinical implications pertaining to efficacy.57

Axi-cel (KTE-019) was approved by the US Food and Drug Administration (FDA) in October 2017 for treatment of adult patients with refractory/relapsed (R/R) large B-cell lymphoma after two or more lines of systemic therapy (including DLBCL not otherwise specified, primary mediastinal large B-cell lymphoma, high-grade B-cell lymphoma and DLBCL arising from FL). Tisagenlecleucel (also approved for patients up to 25 years of age with B-cell precursor ALL) was also approved in May 2018 for adult patients with R/R large B-cell lymphoma after two or more lines of systemic therapy. As opposed to axi-cel, tisagenlecleucel is not approved for primary mediastinal lymphoma. Another CAR T-cell product, liso-cel (JCAR017) is currently being studied in clinical trials with promising efficacy.

CAR T-cell manufacturing

The CAR T-cell manufacturing process begins with T-cell harvesting by collecting peripheral mononuclear cells (PMBCs) through leukapheresis. The product is transferred to a good manufacturing practice facility where CD3+ T cells are separated (in other products, no CD3+-based separation occurs), then expanded and activated. Then, CAR gene transduction into the T cells ensues through a vector, typically using a replication-defective virus (lentivirus or retrovirus). The CAR T cells are expanded in vitro and then infused back to the patient.50,58 Although all three aforementioned anti-CD19 CAR products use the same scFv region, FMC63; there are several differences, but it is unclear whether these variances affect function, safety and clinical efficacy. As mentioned, axi-cel contains a CD28 costimulatory domain, while tisagenlecleucel and liso-cel, contain a 41-BB costimulatory domain. Liso-cel is the only product manufactured in a controlled process that enables administration of a fixed ratio of CD4 and CD8 CAR T cells. The lower variability and defined cellular composition may lead to lower rates of toxicity; however, this remains to be elucidated59 (Table 1).

Table 1.

Characteristics of selected second-generation CAR products studied in B-cell lymphomas.

Axicabtagene ciloleucelTisagenlecleucelLisocabtagene maraleucel
US FDA indication for lymphomaAdult DLBCLAdult DLBCLNot applicable**
Costimulatory domainCD284-1BB (CD 137)4-1BB (CD 137)
scFvFMC63FMC63FMC63
Vector deliveryRetrovirusLentivirusLentivirus
Defined cellsNoNoYes, CD4:CD8 fixed ratio
Lymphodepleting chemotherapy
(×3 days)
Cy 500 mg/m2
Flu 30 mg/m2
Cy 250 mg/m2
Flu 25 mg/m2*
Cy 300 mg/m2
Flu 30 mg/m2
*An alternate lymphodepletion regimen can be given prior to tisagenlecleucel consisting of: bendamustine 90 mg/m2 IV daily for 2 days if a patient previously experienced grade 4 hemorrhagic cystitis or demonstrates resistance to a previous Cy-containing regimen.
**Lisocabtagene maraleucel is not approved for commercial use at the present time.

CAR, chimeric antigen receptor; Cy, cyclophosphamide; DLBCL, diffuse large B-cell lymphoma; FDA, US Food and Drug Administration; Flu, fludarabine; IV, intravenous; scFv, single-chain variable fragment.

Pharmacokinetics and persistence

In order to achieve antitumor efficacy, CAR T cells must reach tumor cells, interact with their intended antigen, proliferate, kill tumor cells, attempt to escape inhibitory immune mechanisms and a hostile tumor microenvironment and persist over time in order to ensure durable tumor control.60 The pharmacokinetics (PK) of CAR T cells usually refers to maximum concentration (Cmax) or peak, area under the curve and persistence. The PK of CAR T cells may differ across CAR constructs. In general, within hours after CAR T-cell infusion there is a rapid initial decline that seems to be related to redistribution to tissues. This is followed by the development of a Cmax (peak) and expansion that usually occur 1–2 weeks after infusion; the peak and expansion are clinically related to response. This is followed by a phase of slower decline in the number of CAR T cells that can last over a period of weeks to even years.61 In general, the expansion and persistence of CAR T cells are considered essential for its antitumor efficacy, thus key predictors of clinical response.6265 The upregulation of the inflammatory cytokines interleukin 15 (IL-15) and the granulocyte/macrophage colony-stimulating factor (GM-CSF) have been shown to contribute with CAR T-cell expansion and persistence. These cytokines rise a few days after lymphodepleting chemotherapy (especially when fludarabine and cyclophosphamide are used).63,66 There are other cytokines that appear to mediate the cytotoxic effect of T cells such as IL-6, IL-10 and granzyme B.66

Clinical efficacy of CAR T-cell therapy

Early studies in B-cell lymphomas

Initial clinical trials of anti-CD19 CAR T cells for B-cell lymphoma were carried out in single institutions. These included a diverse population of refractory B-cell NHLs, including DLBCL, FL, primary mediastinal B-cell lymphoma (PMBCL), marginal-zone lymphomas (MZL) and transformed follicular lymphomas (TFLs). Two early reports of anti-CD19 CART cells were described in patients with indolent NHL.67,68

The NCI conducted the first CAR T-cell study, which demonstrated clinical activity in DLBCL using the CD3ζ-CD28 CAR T construct (later licensed as axi-cel by Kite Pharma, a Gilead Company). The prescribed lymphodepleting regimen consisted of a combination of cyclophosphamide (total dose of 60 mg/kg) followed by fludarabine 25 mg/m2 daily for 5 days.62 This study included nine patients with refractory CD19+ B-cell lymphoma [DLBCL (four), PMBCL (four) and DLBCL transformed from CLL (one)] CAR T-cell manufacturing was successful in all patients. There were five CRs and two partial responses (PRs) out of the seven evaluable patients. Three patients still had ongoing CR at the last reported follow up.62 The duration of response (DoR) ranged from 38 to 56 months, in patients with ongoing responses in a long-term follow-up report.69

A larger report from the NCI included 22 aggressive B-cell lymphoma patients (DLBCL = 13, TFL = 3, PMBCL = 2, FL = 2, MCL = 1 and Richter transformation (RT) = 1). In this study, a low-dose conditioning chemotherapy (cyclophosphamide 300–500 mg/m2 and fludarabine 30 mg for 3 days) was considered to have a lymphodepleting action and was associated with less hematologic and nonhematologic toxicity. In DLBCL patients, the overall response rate (ORR) and CR rates were 68% and 47%, respectively. The median duration of remission was 12.5 months and the 12-months progression-free survival (PFS) was 63.3%.63

Investigators at the Fred Hutchinson Cancer Research Center (FHCRC) developed CAR T cells using a 4-1BB as costimulatory domain. A phase I clinical trial using this CAR construct, and a predefined 1:1 CD4:CD8 ratio was conducted based upon strong preclinical data. Specifically, CAR T cells manufactured using purified CD4+ or CD8+ central memory (CM) or naïve (N) T cells in a specific 1:1 CD4:CD8 ratio were more potent in eliminating CD19+ tumor cells as compared with those manufactured from effector memory (EM) T cells in mouse models. Thirty-four patients with various refractory or relapsed B-cell NHLs including de novo DLBCL (11), TFL (11), MCL (4), and FL (6) were treated.70 Patients with relapsed postauto- and postallo-HCT were also included. Results were encouraging, with ORR and CR rates for the whole group of 63% and 33%, respectively. In a subgroup of aggressive lymphomas (DLBCL and TFL) the ORR and CR rates were 67% and 38%, respectively. This CAR construct is now licensed by JUNO Therapeutics for development as JCAR017.

Another 4-1BB CART (CTL019) construct with significant antilymphoma action was developed at the University of Pennsylvania. Preliminary results confirmed its efficacy in patients with a variety of B-cell NHLs, including DLBCL, FL, and MCL.71,72 The updated analysis included 38 patients with DLBCL (n = 23) and FL (n = 15); however, 10 DLBCL patients could not be infused for a variety of reasons (rapid disease progression = 4, inability to manufacture CAR T cells = 5, and consent withdrawal = 1). The lymphodepleting chemotherapy included several regimens that were chosen as per physician discretion. The median CTL019 dose was 5.79 × 106 (range: 3.08–8.87 × 106) CAR T cells/kg. Among DLBCL patients, the ORR and CR rates were 50% and 43%, respectively. The median PFS was 3.2 months; and the PFS at last follow up (median 28.6 months) was 43%. There were no significant differences in outcomes between GCB/non-GC, double-hit status or transformed FL subgroups.72,73 The median DoR was not reached with 86% of responding DLBCL patients maintaining an ongoing response at the time of the last follow up.

Multicenter studies in aggressive B-cell lymphomas

The early single-center studies showed significant antilymphoma activity in aggressive B-cell NHLs and led the design of multicenter studies that included several academic institutions in association with pharmaceutical companies.

Axicabtagene ciloleucel (KTE-C19)

The first multicenter trial to evaluate CAR T-cell therapy for refractory DLBCL used the NCI CD3ζ/CD28 CAR construct (KTE-19, now axi-cel) with a streamlined closed-manufacturing process. The ZUMA-1 clinical trial consisted of a phase I and a phase II portion that evaluated the efficacy of axi-cel in refractory high-grade B-cell lymphoma. The cell dose and conditioning chemotherapy previously tested at the NCI were confirmed safe in seven patients with refractory DLBCL (as defined per SCHOLAR-1: best response as SD to last systemic therapy or progressed within 12 months of prior autologous transplant).16 No bridging chemotherapy was allowed (prior to conditioning chemotherapy or prior to CAR T-cell infusion). The lymphodepleting regimen entailed cyclophosphamide 500 mg/m2 and fludarabine 30 mg/m2 × 3 days followed by infusion of axi-cel at a dose of 1–2 × 106 CAR T cells/kg16,74 The objective response was 71% with four patients achieving CR (57%) at 1 month evaluation. Three patients had ongoing CR at 12 months post axi-cel infusion. Reversible grade 3 neurotoxicity (NT) and cytokine-release syndrome (CRS) were reported among this cohort. One fatality occurred in a patient who experienced grade 4 CRS and grade 4 encephalopathy, and died of intracranial bleeding, which was considered unrelated to axi-cel. This patient appeared to have had a high inflammatory state prior to chemotherapy and CAR T-cell infusion. For the phase II portion of the trial, changes in the safety evaluation were made and included baseline C-reactive protein (CRP) assessment and delaying CAR T-cell infusion in patients with fevers until appropriate work-up was completed.

The pivotal phase II portion of the ZUMA-1 had similar eligibility criteria as the phase I, with two cohorts: cohort 1 for DLBCL and cohort 2 for PMBCL and TFL.75 The primary endpoint was ORR in patients with more than 6 months follow up postaxi-cel infusion, as compared with historical controls. Secondary endpoints were DoR, OS, safety, and levels of CAR T cells and cytokines. A total of 111 patients were enrolled. Seventy percent were refractory to at least three lines of therapy and 21% relapsed within 12 months of auto-HCT. Ten patients could not receive axi-cel for various reasons [serious adverse events (SAEs) prior to conditioning regimen = five, nonmeasurable disease = two, no product available = one, and SAE postconditioning regimen = two].

The 101 patients that received axi-cel infusion were the prespecified intent-to-treat analysis cohort. The CAR T-cell manufacturing success was 99%. The median time from apheresis to axi-cel delivery was 17 days. The study met the primary endpoint compared with the historical cohort (SCHOLAR-1) with an ORR of 83% and CR of 54% (in comparison with the a prespecified ORR of 20%, p < 0.0001) representing a eightfold higher CR rate in comparison with SCHOLAR-1. The latest data with a median follow up of 27.1 months was presented at ASH, 2018.76 The ongoing ORRs and CRs were 39 and 37%, respectively. Overall objective responses remained consistent across patient and disease-specific variables, such as advanced stage, age, bulky disease, high IPI score or refractory subgroups [R/R postautohematopoietic stem-cell transplantation (HSCT) or higher than second line of therapy]. The median DoR was 11.1 months in all responders and was not reached in those achieving CR. The PFS at 12,18 and 24 months was 44%, 40%, and 39%, respectively. The 12, 18 and 24-month OS was 60%, 53% and 51%, respectively. An initial PFS plateau was seen at 6 months postaxi-cel infusion; however, there were 10 patients that exhibited disease progression beyond 6 months. The median PFS and OS were 5.9 months and not reached, respectively.76 Of note, 23 out of 61 patients with either PR (11) or SD (12) converted into CR with no additional intervention. The median time of conversion of PR to CR was 64 days (49–424).76,77 Based on these results, axi-cel was approved by the FDA for R/R high-grade B-cell lymphoma, TFL and PMBCL after two preceding lines of therapy.

Tisagenlecleucel (CTL019)

The JULIET trial is a phase II multicenter global study in patients with refractory DLBCL utilizing CTL019, the anti-CD19 CAR using a 4-1BB costimulatory domain developed by scientists from the University of Pennsylvania and was initially studied in single-center trials.73 Interim results were presented at the American Society of Hematology 59th annual meeting in 2017 and the European Hematology Association meeting in 2018.7880 CAR T cells were manufactured centrally; however, in contrast to ZUMA-1, cryopreserved apheresis products were utilized and bridging chemotherapy was allowed per clinician discretion for patients with rapidly progressive disease. There were two regimens utilized, as lymphodepleting chemotherapy consisted of fludarabine 25 mg/m2 and cyclophosphamide 250 mg/m2 for 3 days or bendamustine 90 mg/m2 for 2 days. Key eligibility criteria included aggressive B-cell lymphoma (DLBCL or TFL), relapse after autologous HSCT or ineligible for HSCT, or refractory after two lines of therapy. Similar to the ZUMA-1 trial, the primary endpoint was ORR and CR rates.

The update analysis had a data cutoff of 21 May 2018. A total of 167 patients were enrolled and 115 patients were infused with tisagenlecleucel (4 patients were not infused by data cutoff).81,82 Fifty patients could not be infused with CTL019 due to inability to manufacture CAR T cells (n = 12) and change in disease/patient status (n = 38). The median age of study subjects was 56 (22–76) years. The median dose of transduced cells was 3.0 × 108 (0.1–6 × 108). In this study, 51% had refractory disease with at least three lines of therapy and 49% had prior auto-HCT. A total of 92% patients received bridging chemotherapy. The median time from infusion to data cutoff was 19.3 months. In the 99 evaluable patients ([gt-or-equal, slanted]3 months of follow up) the best ORRs and CRs were 54% and 40%, respectively. The 12- and 18-month relapse-free survival was 64 %. The 12- and 18-month OS in all patients were 48% and 43%, respectively. The median DoR in responders was not reached. The median OS for all patients and CR patients was 11.1 months and not reached, respectively. Responses were similar across different subgroups (postauto- HCT, double-hit lymphoma, refractory/relapsed status, age, etc.). Similar to ZUMA-1, conversion into CR was observed in 15/28 (54%) patients who originally achieved PR.80,82 Outpatient infusion of CTL019 was feasible and was given to 26 patients, and 20 (77%) of those remained as outpatients for more than 3 days.78 No deaths were attributed to CLT019, but three patients died within 30 days of infusion (all due to disease progression).

Lisocabtagene maraleucel (JCAR017)

The 4-1BB CAR T-cell construct using a defined CD4:CD8 T-cell ratio and developed at the FHCCR was tested in the multicenter TRANSCEND-001 study.83,84 This study was divided in two groups: the FULL and CORE cohorts. The FULL cohort included patients with R/R DLBCL, TFL, FL grade 3b, MCL, RT, DLBCL arising from MZL and PMBCL. The CORE dataset included only R/R DLBCL and TFL. The initial analysis included three cohorts with different dose levels (DL): DL-1S was 5 × 107, and DL-2S was 1 × 108. A small cohort of patients received double dose (n = 6) of JCAR017 at 5 × 107 that was administered 14 days apart (cohort no longer open). The conditioning regimen consisted of fludarabine 30 mg/m2 and cyclophosphamide 300 mg/m2 daily for 3 days. Bridging therapy was allowed for disease control. After the preliminary analysis, the DL-2S was determined for the expansion phase.85,86

In the updated analysis, 134 patients were enrolled and 114 patients infused with liso-cel. Twenty patients were not infused due to rapid disease progression/death (n = 13), consent withdrawal (n = 5) or inability to manufacture (n = 2). Out of the infused patients, 12 had a nonconforming product, thus 102 patients were evaluable in the FULL cohort (CORE cohort = 73).87 In the CORE cohort, the median age was 60 (20–82) years and, at least 50% were DLBCL cases refractory to three or more lines of therapy, and 38% had failed a prior auto-HCT. Preliminary analysis was reported previously.84,85 The updated analysis has a median follow up of 12 months reporting the best ORR (CR) rates in the FULL and CORE cohorts of 75% (55%) and 80% (59%), respectively. The 6-month ORR and CR rates in the CORE cohort were 47% and 41%, respectively. Responses rates were not affected by high-risk DLBCL characteristics such as double-hit lymphoma status, chemorefractory disease or prior auto-HCT failure. The median DoR was not reached in CR patients in the FULL and CORE cohorts, confirming findings of prior reports.84,85 The 12-month OS was 63% in all responders and 89% those achieving CR. A total of 93% patients with CR as best response at 6 months had ongoing response at data cutoff (Tables 2 and and33).87

Table 2.

Patient characteristics in the three anti-CD19 CAR T-cell therapy multicenter trials in aggressive B-cell NHLs.

CharacteristicsZUMA-1
(Neelapu et al.77)
JULIET
(Borchmann et al.80)
TRANSCEND1
(Abramson et al.87)
Patients enrolled (infused), n111 (101)165 (111)134 (114)2
CORE: 73
Evaluable patients, n10193102 (CORE: 73)
Median age (range), years58 (23–76)56 (22–76)60 (20–82)
Age [gt-or-equal, slanted] 65 years24%23%33 %
Lymphoma subtypesDLBCL, TFL, PMBCLDLBCL, TFLDLBCL, TFL (CORE)3
Double-hit lymphomaNR27%22%
[gt-or-equal, slanted] 3 lines of therapy69%51%50%
Primary refractoriness26%NR49%
Refractory to last therapy77%54%67%
Prior autologous HCT21%49%38%
1Data presented from the CORE cohort.
2Twelve patients had a nonconforming product.
3The FULL cohort included: DLBCL transformed from CLL (Richter transformation) and MZL, PMBCL and follicular lymphoma 3B. CORE included only DLBCL and TFL.

CLL, chronic lymphoblastic leukemia; DLBCL, diffuse large B-cell lymphoma; FL, follicular lymphoma; HCT, hematopoietic cell transplantation; MZL, marginal-zone lymphoma; NR, not reported; PMBCL, primary mediastinal B-cell lymphoma; TFL, transformed lymphoma.

Table 3.

Multicenter studies with autologous anti-CD19 CAR T-cell therapy for aggressive B-cell lymphomas.

VariablesZUMA-1 (Locke et al.76)JULIET (Schuster et al.82)TRANSCEND (Abramson et al.87)
Patients enrolled (treated), n111 (101)165 (111)134 (114)
73 in CORE
Median follow up27.1 months19.3 months112 months
Costimulatory domainCD284-1BB4-1BB
CAR T dose (range)2.0 × 106 cells/kgMedian, 3.1 × 108 cellsDL1 5.0 × 107 cells2
DL2 1.0 × 108 cells
Lymphodepleting regimenFlu 30 mg/m2 × 3 days
Cy 500 mg/m2 × 3 days
Flu 25/m2x 3 days
Cy 250 mg/m2 x3 d or B 90 mg/m2 × 2 days
Flu 30 mg/m2 × 3 days
Cy 300 mg/m2 × 3 days
Efficacy
 Best ORR (CR)82% (54%)52% (40%)80% (59%)
 6-month ORR (CR)41% (36%)33% (29%)47% (41%)
 Ongoing ORR (CR)39% (37%)NRNR
 mDOR11.1 monthsNot reached9.2 months
12-month PFS44%66%NR
 18-month PFS40%64%NR
 12-month OS59%49%63%
 18- month OS53%43%NR
1Median time from infusion to data cutoff.
2Six patients received double dose of DL1.

B, bendamustine; CR, complete response; Cy, cyclophosphamide; Flu, fludarabine; mDOR, median duration of response; NR, not reported, ORR, overall response rate.

Clinical activity of CAR T-cell therapy for DLBCL outside clinical trials

With the approval of axi-cel and tisagenlecleucel for the treatment of refractory DLBCL, there was growing interest in reporting the efficacy of this therapy in real clinical practice and outside clinical trials. In an effort to replicate the results of the ZUMA-1 trial with axi-cel, an extraordinary effort was carried out by 23 US cancer centers with experience and with certification to treat patients with CAR T-cell therapy. To date, there are no reports of tisagenlecleucel in DLBCL outside clinical trials.

The first study was reported by Nastoupil and colleagues88 and included 295 patients, with 274 patients treated. The final CAR T-cell product did not meet FDA specifications in seven patients. The median time of axi-cell manufacturing was 21.5 days. As opposed to ZUMA-1 trial, around 55% patients received any form of bridging therapy (chemotherapy, targeted therapy or radiation). General characteristics included a median age of 60 (21–83), stage III/IV in 83%, performance status (PS) 0–1 in 81%, three or more lines of therapy in 75% of cases and relapsed postauto-HCT in 33%. Interestingly, 43% of patients (124/286) would have not been eligible for the ZUMA-1 trial (such as platelets < 75 000, ejection fraction < 50%, prior allo-HCT, among other factors). The overall efficacy was similar as to the ZUMA-1 trial with 3-month ORR and CR rates of 81% and 57%, respectively.

The second study, presented by Jacobson and colleagues,89 included 108 patients infused with axi-cel; of those, 104 were evaluable for efficacy. The median age was 63.8, PS 0–1 in 90% of cases, prior auto-HCT in 27%, prior allo-HCT in 3%. About 52% of the evaluable patients received bridging chemotherapy after apheresis; 60% of patients would have not met criteria for the ZUMA-1 clinical trial. In the 95 patients evaluable for response, the best ORR and CR rates were 71 and 44%, respectively. Similarly, about 50% of patients who initially had a PR, achieved CR at a later time.

These two reports concluded that the efficacy of axi-cel in refractory disease could be replicated outside the strict eligibility criteria of clinical trials. It should be highlighted that this therapy needs to be offered in centers with experience and capability of administering high-risk immunotherapy and cellular therapies (Table 4).

Table 4.

Comparison of efficacy and toxicity of patients treated with axi-cel outside clinical trials (‘real-world experience’) with ZUMA-1.

CharacteristicsZUMA-1
(Locke et al. 2018)
Nastoupil et al.88
(ASH 2018)
Jacobson et al.89
(ASH 2018)
Patients enrolled (infused), n111 (101)295 (274)NR (104)
Median age (range), years58 (23–76)60 (21–83)63.8 (21–80)
Median follow up27.1 months3.9 months5.6 months
Double-hit lymphomaNR23%24%
[gt-or-equal, slanted] 3 lines of therapy69%75%NR
Primary refractoriness26%35%NR
Refractory to last therapy77%42%91%
Prior autologous HCT21%33%27%
Bridging chemotherapy055%40%
Efficacy
 Best ORR (CR)82% (58%)81% (57%)71% (44%)
 Median PFS5.9 months6.18 months5.6 months
 6-month OS78%72%NR
Toxicity
 CRS all grades (3–4)93% (13%)92% (7%)94% (16%)
 Neurotoxicity all grades (3–4)65% (31%)69% (33%)76% (39%)
 Tocilizumab use45%63%67%
 Steroids use29%55%64%
 Grade 5 AEs4%3%17%2
1A total of 7 nonrelapse mortalities due to: infection (n = 5); hemophagocytic lymphohistiocytosis (n = 1); cerebral edema (n = 1).
2A total of 7 nonrelapse mortalities due to: CRS (n = 2); neurotoxicity (n = 1); infection (n = 2); cardiovascular (n = 2).

AE, adverse event; ASH 2018, 60th Annual Meeting of the American Society of Hematology; axi-cel, axicabtagene ciloleucel; CR, complete response; CRS, cytokine-releasing syndrome; HCT, hematopoietic cell transplantation; NR, not reported; ORR, overall response rate; OS, overall survival; PFS, progression-free survival.

CAR T-cell therapy for indolent lymphomas

Although trials using anti-CD19 CAR T cells focus mainly on aggressive B-cell lymphomas, the first patients to receive this type of therapy were those having indolent NHLs. The first-generation anti-CD19 CAR T-cells (without costimulation) reported no clinical efficacy in FL cases.67 The first case successfully treated with anti-CD19 CAR T cells with CD28 as the costimulatory domain was reported by the NCI in a refractory FL patient achieving long-term remission.68 A subsequent NCI study with CD28 anti-CD19 CAR T cells reported PR of 100% in five indolent NHL patients (four FL and one MZL) with 75% having ongoing responses at the time the study was published.90 These patients received a conditioning regimen consisting of cyclophosphamide 60 mg/kg × 2 days and fludarabine 25 mg/m2 × 5 days.90

In the 32 patients treated with the 4-1BB CAR T-cell construct (1:1 CD4/CD8 ratio) from FHCRC, there were 5 evaluable FL patients and the reported ORR and CR rates were 80% (4/5) and 40% (2/5), respectively.70

The largest data in FL to date come from the CTL019 CAR T cell from the University of Pennsylvania that included 14 FLs. These FL patients had relapsed within 24 months of initial diagnosis and remained refractory to least two lines of therapy.71,91 Patients received a variety of conditioning regimens such as bendamustine 70 mg/m2 × 2 days, cyclophosphamide, radiation plus cyclophosphamide and fludarabine–cyclophosphamide. This trial included FL patients with poor prognosis features, including prior multiple therapies (median of five), relapsed postauto-HCT (21%) and allo-HCT (one patient). The updated analysis showed a 3-month ORR and CR of 79% (11/14) and 71% (10/14), respectively. The median PFS was not reached and 70% of FL patients were disease free at a median follow up of 28.6 months.73,91

These data support anti-CD19 CAR T-cell therapy as a promising alternative therapy in poor-risk FL, despite the low number of patients; and it may have a curative potential given the long-term ongoing responses. The ZUMA-5 is a dedicated indolent B-cell NHL trial that is currently enrolling patients [ClinicalTrials.gov identifier: NCT03105336].

CAR T-cell therapy for MCL

The experience with autologous anti-CD19 CAR T-cell therapy in MCL is limited to a few cases in single-center clinical studies. The 4-1BB CAR T-cell trial at the FHCRC with fixed CD4:CD8 ratio included four patients with MCL that received doses of 2 × 105–107 CARs/kg. The ORR was 25% (no CR).70 The NCI-based CD28-CAR T-cell trial reported a long-term CR (>17 months) in an MCL.63 The University of Pennsylvania (U Penn)-based CTL019 CAR T-cell trial included two patients with MCL having 50% ORR (no documented CR).71 Albeit limited experience, there seems to be promising activity of anti-CD19 CAR T cells in refractory MCL. We are eagerly awaiting the preliminary results of the ZUMA-2 clinical trial of axi-cel in patients with ibrutinib-refractory MCL [ClinicalTrials.gov identifier: NCT02601313].

CAR T-cell-therapy-related toxicity

The toxicities related to CAR T-cell therapy were initially described in the earlier studies in B-cell lymphomas and ALL.62,70,92,93 There are two main categories of toxicity: CRS and neurotoxicity or CAR T-cell-related encephalopathy syndrome (CRES). Organ damage can accompany CRS (renal failure, cardiac dysfunction, liver dysfunction, etc.).94,95

Cytokine-release syndrome

CRS is an excessive inflammatory response caused by overactivation of immune-effector cells that leads to significant release and elevation of inflammatory cytokines such as IL-1, IL-2, IL-6, IL-10, IL-15, interferon (IFN)-γ and tumor necrosis factor (TNF). This occurs typically in patients that receive CAR T-cell therapy.9496 Patients, with this inflammatory response, present with a variety of symptoms, such as fevers, general malaise, hypotension, and hypoxia. In severe cases, irreversible organ damage and death can occur.95,96

The most important factors for successful treatment of CRS are early identification and accurate grading in order to guide optimal management. Grading is based on hemodynamic instability, degree of hypoxemia, organ damage and presence of comorbidities. Patients with grade 3 and 4 CRS (and sometimes grade 2 in patients with important comorbidities) usually require aggressive measures, such as vasopressors, management in the intensive care unit, anticytokine therapy and steroids. As IL-6 is a key player in the etiology of CRS, the administration of tocilizumab (anti-IL-6 receptor) and siltuximab (anti-IL-6 antibody) have become standard approaches for the CRS management.92,95,96

There are clinical factors that correlate with the development of CRS, such as disease burden (specifically in ALL) and dose of CAR T cells.62,70,80,92,93 Increased levels of TNF (TNF-alpha), IL-2R, IL-6, IFN-γ, IL-10, IL-15, and ferritin have demonstrated association with severity of CRS.62,70 Peak CRP levels have been shown to directly correlate with CRS severity and can be used as a surrogate marker for early treatment/supportive care.92 Recent preclinical work in mouse models helped clarify further the potential etiology of CRS and NT. In two separate studies, the role of monocytes/macrophages and cytokine production upon interaction with CAR T cells, especially as a source of inflammatory cytokine production and kinetics (notably, IL-1 elevation preceded the IL-6 rise), showed the role of IL-1 blockade in potentially preventing CRS and NT.97,98

Neurotoxicity

NT is another common complication of CAR T-cell therapy that is less understood than CRS. Symptoms of NT can sometimes overlap to those seen in CRS. Patients have a variety of symptoms such as confusion, obtundation, tremors and headaches. Other symptoms such as aphasia, cranial nerve abnormalities and seizures have been described. As in CRS, early identification and adequate grading is strongly recommended. Tools such as mini mental-status evaluation have been used commonly to grade NT. A more specific (and simplified) defined criterion for measurement of NT, also known as CRES, was recently developed.94 The role of anti-IL-6 therapy is unclear and does not seem to have a beneficial role in treatment of NT, thus the mainstay treatment of CRES is steroids. Neurology evaluation with brain imaging, cerebrospinal fluid (CSF) examination and electroencephalogram (EEG) assessments is usually recommended to rule out other causes.

Additionally, NT appears to be cytokine driven.62 The ZUMA-1 described how elevated levels of IL-2, ferritin and GM-CSF were significantly associated grade [gt-or-equal, slanted] 3 NT.99 Baseline higher tumor burden, elevated lactate dehydrogenase (LDH), CRP and ferritin were associated with neurotoxicity and CRS in the JULIET trial.80 The initial report of biomarkers in the TRANSCEND study demonstrated an eightfold increased risk of CRS and neurotoxicity with elevated LDH (>500/µl) and tumor burden (>50 cm2).100 Disruption of the blood–brain barrier (BBB), endothelial activation and increased IL-1 levels have been recently described as potential drivers of NT.98,101

In general, the vast majority of CAR T-cell-related toxicities resolve within few weeks as reported in both single-center and pivotal multicenter studies; however, CAR T-cell fatalities have also been reported.62,70,74,90 While, the symptoms and signs of each type of toxicity may overlap, it is important for the clinician to recognize these potential complications, as they could be life threatening and, in certain circumstances, lead to death. The diagnosis and management of CRS and CRES have been extensively discussed in other publications.9496

The multicenter ZUMA-1, JULIET and TRANSCEND trials have reported CAR T-related toxicity with some differences in frequency, timing and severity. These variations are possibly due to differences in patient population, disease subtypes (and clinical presentation), use of bridging chemotherapy, use of different toxicity grading systems and differences in CAR T-cell constructs. The Lee criteria96 were used to grade CRS in ZUMA-1 and TRANSCEND studies while the U Penn Criteria were used for severity stratification of CRS in the JULIET trial.102 Table 5 describes the frequency and features of CAR T-related toxicities in the three multicenter studies in NHL.

Table 5.

Toxicities in the three largest multicenter studies with anti-CD19 CAR T-cell therapy for aggressive B-cell lymphomas.

StudyZUMA-1
(Locke, 2018)
JULIET (Schuster)82TRANSCEND1 (Abramson et al.)87
No patients enrolled (treated)111 (101)165 (111)134 (114)
Cytokine-release syndrome 2
Time to onset, median, range
Duration, median, range2 days (1–12)3 days (1–9)5 days (2–12)
 Grade (all)
 Grade 3 or 48 days (NR)7 days (2–30)5 days (NR)
 Tocilizumab use93%58%37%
 Vasopressors use13%23%1%
 Steroid treatment43%16%21%
 ICU admission17%6% (high dose)NR
27%11%17%
NRNRNR
Infections
 All grades35%334%NR
 Grade 3 or 431%320%NR
Neurotoxicity 2
Time to onset, median (range)5 days (1–17)NR10 days (3–23)
Duration, median, range17 days (NR)NR11 days (NR)
 All grades64%20%23%
 Grade 3 or 428%11%13%
1Reported from the full cohort data.
2Grading was performed using the Penn criteria.
3Febrile neutropenia.

ICU, intensive care unit; NR, not reported.

Toxicities such as CRS and NT were also described in the outside clinical trials (real clinical experience). In general, the rates of toxicity were somewhat similar to what it was seen in clinical trials, except for one of the reports that showed lower CRS grade [gt-or-equal, slanted] 3. Another interesting finding was that higher rates of tocilizumab and steroid use were reported. We believe that this underscores the fact that centers may treat CRS and NT more aggressively or that there is a better knowledge that steroids or tocilizumab does not affect efficacy of CAR T cells. Table 4 describes a comparison of toxicities seen in ZUMA-1 and outside clinical trial experience.

Challenges in CAR T-cell therapy: potential interventions

Overcoming resistance

Availability of CAR T-cell therapy has changed the treatment landscape of refractory DLBCL. Unfortunately, about 50–60% of patients will not achieve a CR or will relapse after CAR T-cell therapy. Thus, understanding the mechanism of relapse after CAR T-cell treatment is paramount. One mechanism is CD19 immunological antigen escape, as CD19-negative B-cell malignancy relapses have been reported.103 Inability to express CD19 in B-cell malignancies due to epitope/antigen loss of the CD19 through splicing/mutation mechanisms have been described as resistance mechanism or relapse in patients receiving CD19-directed therapy (such as CARs or bispecific antibodies).77,104,105 Another potential mechanism is increased activation of the programmed cell-death 1/programmed cell-death ligand 1 (PD-1/PD-L1) pathway that has been seen in relapsed DLBCL post CAR T-cell therapy.77

One approach to overcome resistance is by targeting a different antigen. Anti-CD20 CAR T cells showed modest activity in earlier studies but with greater efficacy once costimulation with 4-1BB was added.67,106 An ORR and CR rate of 80–83% and 17–50% were described in two different trials, respectively.107,108 Another target, CD22, has shown antilymphoma activity in preclinical studies.109 Although these results are focused on CD22+ B-cell ALL, there are ongoing trials for refractory CD22+ B-cell NHLs [ClinicalTrials.gov identifiers: NCT02315612, NCT02794961].110 Bispecific CARs targeting CD19 and CD20 antigens for B-cell malignancies have been developed (CD19-OR-CD20 CAR) with significant preclinical activity, even in CD19-negative tumor cells.111 Hossain and colleagues,112 from Stanford University, presented preliminary data on CD19/CD22 bispecific CAR T cells in nine patients (five DLBCL and four B-cell ALL) that showed clinical activity (one CR and two PRs in the DLBCL cohort) and a tolerable toxicity profile. PD-1 inhibition has become an attractive approach, with reported success in case reports.113,114 A trial of atezolizumab (anti PD-L1 inhibitor) plus axi-cel (ZUMA-6) was presented and included 12 patients with three different cohorts. The clinical activity was promising with an ORR of 92% (CR 58%). Of note, albeit in its early phase, there was a higher grade 3 NT (50%) in comparison with the reported NT of ZUMA-1.115,116 Other checkpoint inhibitors such as pembrolizumab and durvalumab are being studied [ClinicalTrials.gov identifiers: NCT03310619, NCT03630159]. Other agents with the potential to improve activation, expansion, and persistence such as utomilumab (4-1BB agonist), ibrutinib and avadomide (CC-122, an immunomodulator) are also being tested in combination with different CAR T-cell products [ClinicalTrials.gov identifiers: NCT03331198, NCT03310619, NCT03704298].

Patient selection/timing

With two available CAR T-cell products for refractory DLBCL after at least two preceding lines of therapy (excluding patients with primary central nervous system lymphoma) there is no guidance on the FDA label with regards to (a) specific condition(s) where one product is to be favored over another. Early referral to centers certified to prescribe CAR T-cell therapies is strongly encouraged to avoid toxicities from ineffective chemoimmunotherapies.117 Another issue is the time from apheresis to infusion of CAR T cells that can range between 2–5 weeks, depending of the CAR construct and preauthorization (by private insurances). This time could be critical for patients with an otherwise aggressive and refractory disease. Readily available CAR products are donor-derived CAR T cells (allogeneic) that seem feasible and safe.118,119 Donor-derived CAR T cells were initially reported by the NCI in CD19+ B-cell malignancies, including DLBCL, ALL and CLL with clinical efficacy. The CAR T-cell production took 8 days. Interestingly, no cases of acute graft versus host disease (GVHD) were reported but two cases of chronic GVHD were.120 In order to further minimize the risk of GVHD, another approach is to suppress the TCRs by genome editing: by disrupting the expression of the alpha or beta TCR chains using different technologies; the transcription-activator-like effector nucleases being one of the best known methodologies.121

Cost/Financial toxicity

The excitement of this promising therapy for DLBCL has been tempered by its hefty cost of $375,000 for the CAR T-cell product alone without accounting for cost of hospitalization and treatment of complications such as CRS and CRES, which could amount to additional hundreds of thousands of dollars. It is important to consider these factors for pharmacoeconomic analysis in order to determine pricing-, coverage- and outcome-based reimbursement, as well as to the added value to society.122

Conclusions

CD19- targeted CAR T-cells represent the new standard of care for patients with DLBCL that are refractory to at least two prior lines of therapy. While this represents a significant addition to the treatment armamentarium of DLBCL, approximately 50% of cases will continue to succumb to their disease. As a result, future research must focus on identifying disease-, treatment- or patient-related factors that can help successfully predict treatment outcomes. For patients who fail to achieve early CR (defined as within 90 days), early therapeutic interventions with immune modulators or checkpoint inhibitors, or others, represent interesting questions that will need to be studied in ongoing and future clinical trials.

Footnotes

Funding: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Conflict of interest statement: The authors declare that there is no conflict of interest.

ORCID iD: Julio C. Chavez An external file that holds a picture, illustration, etc.
Object name is 10.1177_2040620719841581-img1.jpg https://orcid.org/0000-0002-2045-6238

Contributor Information

Julio C. Chavez, Department of Malignant Hematology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.

Christina Bachmeier, Department of Blood and Marrow Transplantation, Moffitt Cancer Center, Tampa, FL, USA.

Mohamed A. Kharfan-Dabaja, Division of Hematology/Oncology, Mayo Clinic, Jacksonville, FL, USA.

References

1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin 2018; 68: 7–30. [Abstract] [Google Scholar]
2. Al-Hamadani M, Habermann TM, Cerhan JR, et al. Non-Hodgkin lymphoma subtype distribution, geodemographic patterns, and survival in the US: a longitudinal analysis of the National Cancer Data Base from 1998 to 2011. Am J Hematol 2015; 90: 790–795. [Abstract] [Google Scholar]
3. Teras LR, DeSantis CE, Cerhan JR, et al. 2016 US lymphoid malignancy statistics by World Health Organization subtypes. CA Cancer J Clin 2016; 66: 443–459. [Abstract] [Google Scholar]
4. Coiffier B, Thieblemont C, Van Den Neste E, et al. Long-term outcome of patients in the LNH-98.5 trial, the first randomized study comparing rituximab-CHOP to standard CHOP chemotherapy in DLBCL patients: a study by the Groupe d’Etudes des Lymphomes de l’Adulte. Blood 2010; 116: 2040–2045. [Europe PMC free article] [Abstract] [Google Scholar]
5. Coiffier B, Sarkozy C. Diffuse large B-cell lymphoma: R-CHOP failure-what to do? Hematology Am Soc Hematol Educ Program 2016; 2016: 366–378. [Europe PMC free article] [Abstract] [Google Scholar]
6. Barrans S, Crouch S, Smith A, et al. Rearrangement of MYC is associated with poor prognosis in patients with diffuse large B-cell lymphoma treated in the era of rituximab. J Clin Oncol 2010; 28: 3360–3365. [Abstract] [Google Scholar]
7. Johnson NA, Slack GW, Savage KJ, et al. Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J Clin Oncol 2012; 30: 3452–3459. [Europe PMC free article] [Abstract] [Google Scholar]
8. Petrich AM, Gandhi M, Jovanovic B, et al. Impact of induction regimen and stem cell transplantation on outcomes in double-hit lymphoma: a multicenter retrospective analysis. Blood 2014; 124: 2354–2361. [Abstract] [Google Scholar]
9. Alizadeh AA, Eisen MB, Davis RE, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 2000; 403: 503–511. [Abstract] [Google Scholar]
10. Lenz G, Wright G, Dave SS, et al. Stromal gene signatures in large-B-cell lymphomas. N Engl J Med 2008; 359: 2313–2323. [Europe PMC free article] [Abstract] [Google Scholar]
11. Cuccuini W, Briere J, Mounier N, et al. MYC+ diffuse large B-cell lymphoma is not salvaged by classical R-ICE or R-DHAP followed by BEAM plus autologous stem cell transplantation. Blood 2012; 119: 4619–4624. [Europe PMC free article] [Abstract] [Google Scholar]
12. Philip T, Guglielmi C, Hagenbeek A, et al. Autologous bone marrow transplantation as compared with salvage chemotherapy in relapses of chemotherapy-sensitive non-Hodgkin’s lymphoma. N Engl J Med 1995; 333: 1540–1545. [Abstract] [Google Scholar]
13. Gisselbrecht C, Schmitz N, Mounier N, et al. Rituximab maintenance therapy after autologous stem-cell transplantation in patients with relapsed CD20(+) diffuse large B-cell lymphoma: final analysis of the collaborative trial in relapsed aggressive lymphoma. J Clin Oncol 2012; 30: 4462–4469. [Europe PMC free article] [Abstract] [Google Scholar]
14. Mounier N, Canals C, Gisselbrecht C, et al. High-dose therapy and autologous stem cell transplantation in first relapse for diffuse large B cell lymphoma in the rituximab era: an analysis based on data from the European Blood and Marrow Transplantation Registry. Biol Blood Marrow Transplant 2012; 18: 788–793. [Abstract] [Google Scholar]
15. Gisselbrecht C, Van Den Neste E. How I manage patients with relapsed/refractory diffuse large B cell lymphoma. Br J Haematol 2018; 182: 633–643. [Europe PMC free article] [Abstract] [Google Scholar]
16. Crump M, Neelapu SS, Farooq U, et al. Outcomes in refractory diffuse large B-cell lymphoma: results from the international SCHOLAR-1 study. Blood 2017; 130: 1800–1808. [Europe PMC free article] [Abstract] [Google Scholar]
17. Costa LJ, Maddocks K, Epperla N, et al. Diffuse large B-cell lymphoma with primary treatment failure: ultra-high risk features and benchmarking for experimental therapies. Am J Hematol 2017; 92: 161–170. [Europe PMC free article] [Abstract] [Google Scholar]
18. Bacher U, Klyuchnikov E, Le-Rademacher J, et al. Conditioning regimens for allotransplants for diffuse large B-cell lymphoma: myeloablative or reduced intensity? Blood 2012; 120: 4256–4562. [Europe PMC free article] [Abstract] [Google Scholar]
19. Fenske TS, Ahn KW, Graff TM, et al. Allogeneic transplantation provides durable remission in a subset of DLBCL patients relapsing after autologous transplantation. Br J Haematol 2016; 174: 235–248. [Europe PMC free article] [Abstract] [Google Scholar]
20. Kharfan-Dabaja MA, El-Jurdi N, Ayala E, et al. Is myeloablative dose intensity necessary in allogeneic hematopoietic cell transplantation for lymphomas? Bone Marrow Transplant 2017; 52: 1487–1494. [Abstract] [Google Scholar]
21. Van Kampen RJ, Canals C, Schouten HC, et al. Allogeneic stem-cell transplantation as salvage therapy for patients with diffuse large B-cell non-Hodgkin’s lymphoma relapsing after an autologous stem-cell transplantation: an analysis of the European Group for Blood and Marrow Transplantation Registry. J Clin Oncol 2011; 29: 1342–1348. [Abstract] [Google Scholar]
22. Hamadani M, Saber W, Ahn KW, et al. Impact of pretransplantation conditioning regimens on outcomes of allogeneic transplantation for chemotherapy-unresponsive diffuse large B cell lymphoma and grade III follicular lymphoma. Biol Blood Marrow Transplant 2013; 19: 746–753. [Europe PMC free article] [Abstract] [Google Scholar]
23. Morton LM, Wang SS, Devesa SS, et al. Lymphoma incidence patterns by WHO subtype in the United States, 1992–2001. Blood 2006; 107: 265–276. [Europe PMC free article] [Abstract] [Google Scholar]
24. Kahl BS. Follicular lymphoma: are we ready for a risk-adapted approach? Hematology Am Soc Hematol Educ Program 2017; 2017: 358–364. [Europe PMC free article] [Abstract] [Google Scholar]
25. Solal-Celigny P, Roy P, Colombat P, et al. Follicular lymphoma international prognostic index. Blood 2004; 104: 1258–1265. [Abstract] [Google Scholar]
26. Federico M, Bellei M, Marcheselli L, et al. Follicular lymphoma international prognostic index 2: a new prognostic index for follicular lymphoma developed by the international follicular lymphoma prognostic factor project. J Clin Oncol 2009; 27: 4555–4562. [Abstract] [Google Scholar]
27. Pastore A, Jurinovic V, Kridel R, et al. Integration of gene mutations in risk prognostication for patients receiving first-line immunochemotherapy for follicular lymphoma: a retrospective analysis of a prospective clinical trial and validation in a population-based registry. Lancet Oncol 2015; 16: 1111–1122. [Abstract] [Google Scholar]
28. Rummel MJ, Niederle N, Maschmeyer G, et al. Bendamustine plus rituximab versus CHOP plus rituximab as first-line treatment for patients with indolent and mantle-cell lymphomas: an open-label, multicentre, randomised, phase 3 non-inferiority trial. Lancet 2013; 381: 1203–1210. [Abstract] [Google Scholar]
29. Salles G, Seymour JF, Offner F, et al. Rituximab maintenance for 2 years in patients with high tumour burden follicular lymphoma responding to rituximab plus chemotherapy (PRIMA): a phase 3, randomised controlled trial. Lancet 2011; 377: 42–51. [Abstract] [Google Scholar]
30. Dupuis J, Berriolo-Riedinger A, Julian A, et al. Impact of [(18)F]fluorodeoxyglucose positron emission tomography response evaluation in patients with high-tumor burden follicular lymphoma treated with immunochemotherapy: a prospective study from the Groupe d’Etudes des Lymphomes de l’Adulte and GOELAMS. J Clin Oncol 2012; 30: 4317–4322. [Abstract] [Google Scholar]
31. Federico M, Luminari S, Dondi A, et al. R-CVP versus R-CHOP versus R-FM for the initial treatment of patients with advanced-stage follicular lymphoma: results of the FOLL05 trial conducted by the Fondazione Italiana Linfomi. J Clin Oncol 2013; 31: 1506–1513. [Abstract] [Google Scholar]
32. Trotman J, Luminari S, Boussetta S, et al. Prognostic value of PET-CT after first-line therapy in patients with follicular lymphoma: a pooled analysis of central scan review in three multicentre studies. Lancet Haematol 2014; 1: e17–27. [Abstract] [Google Scholar]
33. Casulo C, Byrtek M, Dawson KL, et al. Early relapse of follicular lymphoma after rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone defines patients at high risk for death: an analysis from the National LymphoCare study. J Clin Oncol 2015; 33: 2516–2522. [Europe PMC free article] [Abstract] [Google Scholar]
34. Casulo C, Friedberg JW, Ahn KW, et al. Autologous transplantation in follicular lymphoma with early therapy failure: a National LymphoCare study and center for international blood and marrow transplant research analysis. Biol Blood Marrow Transplant 2018; 24: 1163–1171. [Europe PMC free article] [Abstract] [Google Scholar]
35. Avivi I, Montoto S, Canals C, et al. Matched unrelated donor stem cell transplant in 131 patients with follicular lymphoma: an analysis from the Lymphoma Working Party of the European Group for Blood and Marrow Transplantation. Br J Haematol 2009; 147: 719–728. [Abstract] [Google Scholar]
36. van Besien K, Sobocinski KA, Rowlings PA, et al. Allogeneic bone marrow transplantation for low-grade lymphoma. Blood 1998; 92: 1832–1836. [Abstract] [Google Scholar]
37. Khouri IF, McLaughlin P, Saliba RM, et al. Eight-year experience with allogeneic stem cell transplantation for relapsed follicular lymphoma after nonmyeloablative conditioning with fludarabine, cyclophosphamide, and rituximab. Blood 2008; 111: 5530–5536. [Europe PMC free article] [Abstract] [Google Scholar]
38. Laport GG, Wu J, Logan B, et al. Reduced-intensity conditioning with fludarabine, cyclophosphamide, and high-dose rituximab for allogeneic hematopoietic cell transplantation for follicular lymphoma: a phase two multicenter trial from the blood and marrow transplant clinical trials network. Biol Blood Marrow Transplant 2016; 22: 1440–1448. [Europe PMC free article] [Abstract] [Google Scholar]
39. Vose JM. Mantle cell lymphoma: 2015 update on diagnosis, risk-stratification, and clinical management. Am J Hematol 2015; 90: 739–745. [Abstract] [Google Scholar]
40. Andersen NS, Jensen MK, de Nully Brown P, et al. A Danish population-based analysis of 105 mantle cell lymphoma patients: incidences, clinical features, response, survival and prognostic factors. Eur J Cancer 2002; 38: 401–408. [Abstract] [Google Scholar]
41. Hoster E, Dreyling M, Klapper W, et al. A new prognostic index (MIPI) for patients with advanced-stage mantle cell lymphoma. Blood 2008; 111: 558–565. [Abstract] [Google Scholar]
42. Geisler CH, Kolstad A, Laurell A, et al. Long-term progression-free survival of mantle cell lymphoma after intensive front-line immunochemotherapy with in vivo-purged stem cell rescue: a nonrandomized phase 2 multicenter study by the Nordic Lymphoma Group. Blood 2008; 112: 2687–2693. [Europe PMC free article] [Abstract] [Google Scholar]
43. Flinn IW, van der Jagt R, Kahl BS, et al. Randomized trial of bendamustine-rituximab or R-CHOP/R-CVP in first-line treatment of indolent NHL or MCL: the BRIGHT study. Blood 2014; 123: 2944–2952. [Europe PMC free article] [Abstract] [Google Scholar]
44. Kluin-Nelemans HC, Hoster E, Hermine O, et al. Treatment of older patients with mantle-cell lymphoma. N Engl J Med 2012; 367: 520–531. [Abstract] [Google Scholar]
45. Wang ML, Rule S, Martin P, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med 2013; 369: 507–516. [Europe PMC free article] [Abstract] [Google Scholar]
46. Wang M, Rule S, Zinzani PL, et al. Acalabrutinib in relapsed or refractory mantle cell lymphoma (ACE-LY-004): a single-arm, multicentre, phase 2 trial. Lancet 2018; 391: 659–667. [Europe PMC free article] [Abstract] [Google Scholar]
47. Dietrich S, Boumendil A, Finel H, et al. Outcome and prognostic factors in patients with mantle-cell lymphoma relapsing after autologous stem-cell transplantation: a retrospective study of the European Group for Blood and Marrow Transplantation (EBMT). Ann Oncol 2014; 25: 1053–1058. [Abstract] [Google Scholar]
48. Brentjens RJ, Latouche JB, Santos E, et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes costimulated by CD80 and interleukin-15. Nat Med 2003; 9: 279–286. [Abstract] [Google Scholar]
49. Geldres C, Savoldo B, Dotti G. Chimeric antigen receptor-redirected T cells return to the bench. Semin Immunol 2016; 28: 3–9. [Europe PMC free article] [Abstract] [Google Scholar]
50. Kochenderfer JN, Feldman SA, Zhao Y, et al. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J Immunother 2009; 32: 689–702. [Europe PMC free article] [Abstract] [Google Scholar]
51. Sadelain M. CAR therapy: the CD19 paradigm. J Clin Invest 2015; 125: 3392–3400. [Europe PMC free article] [Abstract] [Google Scholar]
52. Scheuermann RH, Racila E. CD19 antigen in leukemia and lymphoma diagnosis and immunotherapy. Leuk Lymphoma 1995; 18: 385–397. [Abstract] [Google Scholar]
53. Eshhar Z, Waks T, Gross G, et al. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci USA 1993; 90: 720–724. [Europe PMC free article] [Abstract] [Google Scholar]
54. Brocker T, Karjalainen K. Signals through T cell receptor-zeta chain alone are insufficient to prime resting T lymphocytes. J Exp Med 1995; 181: 1653–1659. [Europe PMC free article] [Abstract] [Google Scholar]
55. 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 2011; 121: 1822–1826. [Europe PMC free article] [Abstract] [Google Scholar]
56. Song DG, Ye Q, Poussin M, et al. CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo. Blood 2012; 119: 696–706. [Abstract] [Google Scholar]
57. Fesnak AD, June CH, Levine BL. Engineered T cells: the promise and challenges of cancer immunotherapy. Nat Rev Cancer 2016; 16: 566–581. [Europe PMC free article] [Abstract] [Google Scholar]
58. Wang X, Riviere I. Clinical manufacturing of CAR T cells: foundation of a promising therapy. Mol Ther Oncolytics 2016; 3: 16015. [Europe PMC free article] [Abstract] [Google Scholar]
59. Ramsborg CG, Guptill P, Weber C, et al. JCAR017 is a defined composition CAR T cell product with product and process controls that deliver precise doses of CD4 and CD8 CAR T cell to patients with NHL. Blood 2017; 130: 4471. [Google Scholar]
60. Van Der Stegen SJ, Hamieh M, Sadelain M. The pharmacology of second-generation chimeric antigen receptors. Nat Rev Drug Discov 2015; 14: 499–509. [Europe PMC free article] [Abstract] [Google Scholar]
61. Milone MC, Bhoj VG. The pharmacology of T cell therapies. Mol Ther Methods Clin Dev 2018; 8: 210–221. [Europe PMC free article] [Abstract] [Google Scholar]
62. 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 2015; 33: 540–549. [Europe PMC free article] [Abstract] [Google Scholar]
63. Kochenderfer JN, Somerville RPT, Lu T, et al. Lymphoma remissions caused by Anti-CD19 chimeric antigen receptor T Cells are associated with high serum interleukin-15 levels. J Clin Oncol 2017; 35: 1803–1813. [Europe PMC free article] [Abstract] [Google Scholar]
64. Porter DL, Levine BL, Kalos M, et al. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 2011; 365: 725–733. [Europe PMC free article] [Abstract] [Google Scholar]
65. 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 2015; 7: 303ra139. [Europe PMC free article] [Abstract] [Google Scholar]
66. Bot A, Rossi JM, Jiang Y, et al. Cyclophosphamide and fludarabine conditioning chemotherapy induces a key homeostatic cytokine profile in patients prior to CAR T cell therapy. Blood 2015; 126: 4426. [Google Scholar]
67. Jensen MC, Popplewell L, Cooper LJ, et al. Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specific chimeric antigen receptor redirected T cells in humans. Biol Blood Marrow Transplant 2010; 16: 1245–1256. [Europe PMC free article] [Abstract] [Google Scholar]
68. Kochenderfer JN, Wilson WH, Janik JE, et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 2010; 116: 4099–4102. [Europe PMC free article] [Abstract] [Google Scholar]
69. Kochenderfer JN, Somerville RPT, Lu T, et al. Long-duration complete remissions of diffuse large B cell lymphoma after anti-CD19 chimeric antigen receptor T cell therapy. Mol Ther 2017; 25: 2245–2253. [Europe PMC free article] [Abstract] [Google Scholar]
70. Turtle CJ, Hanafi LA, Berger C, et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med 2016; 8: 355ra116. [Europe PMC free article] [Abstract] [Google Scholar]
71. Schuster SJ, Svoboda J, Dwivedy Nasta S, et al. Sustained remissions following chimeric antigen receptor modified T cells directed against CD19 (CTL019) in patients with relapsed or refractory CD19+ lymphomas. Blood 2015; 126: 183. [Google Scholar]
72. Schuster SJ, Svoboda J, Nasta SD, et al. Treatment with chimeric antigen receptor modified T cells directed against CD19 (CTL019) results in durable remissions in patients with relapsed or refractory diffuse large B cell lymphomas of germinal center and non-germinal center origin, “Double Hit” diffuse large B Cell lymphomas, and transformed follicular to diffuse large B cell lymphomas. Blood 2016; 128: 3026. [Abstract] [Google Scholar]
73. Schuster SJ, Svoboda J, Chong EA, et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med 2017; 377: 2545–2554. [Europe PMC free article] [Abstract] [Google Scholar]
74. Locke FL, Neelapu SS, Bartlett NL, et al. Phase 1 results of ZUMA-1: a multicenter study of KTE-C19 Anti-CD19 CAR T cell therapy in refractory aggressive lymphoma. Mol Ther 2017; 25: 285–295. [Europe PMC free article] [Abstract] [Google Scholar]
75. Locke FL, Neelapu SS, Bartlett NL, et al. Clinical and biologic covariates of outcomes in ZUMA-1: a pivotal trial of axicabtagene ciloleucel (axi-cel; KTE-C19) in patients with refractory aggressive non-Hodgkin lymphoma (r-NHL). J Clin Oncol 2017; 35(Suppl.): 7512. [Google Scholar]
76. Locke FL, Ghobadi A, Jacobson CA, et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1–2 trial. Lancet Oncol 2019; 20: 31–42. [Europe PMC free article] [Abstract] [Google Scholar]
77. Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med 2017; 377: 2531–2544. [Europe PMC free article] [Abstract] [Google Scholar]
78. Schuster S, Bishop MR, Tam C, et al. Global pivotal phase 2 trial of the CD19-targeted therapy CTL019 in adult patients with relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL)—an interim analysis. Hematolog Oncol 2017; 35: 27. [Google Scholar]
79. Schuster SJ, Bishop MR, Tam CS, et al. Primary analysis of Juliet: a global, pivotal, phase 2 trial of CTL019 in adult patients with relapsed or refractory diffuse large B-cell lymphoma. Blood 2017; 130(Suppl. 1): 577. [Google Scholar]
80. Borchmann P, Tam C, Jager U. (eds). An updated analysis of JULIET, a global pivotal Phase 2 trial of tisagenlecleucel in adult patients with relapsed or refractory (r/r) diffuse large b-cell lymphoma (DLBCL). The 23rd Congress of EHA; June, 2018. Stockholm, Sweden. [Google Scholar]
81. Schuster SJ, Bishop MR, Tam CS, et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med 2019; 380: 45–56. [Abstract] [Google Scholar]
82. Schuster SJ, Bishop MR, Tam C, et al. Sustained disease control for adult patients with relapsed or refractory diffuse large B-Cell lymphoma: an updated analysis of Juliet, a global pivotal phase 2 trial of tisagenlecleucel. Blood 2018; 132: 1684. [Google Scholar]
83. Abramson JS, Palomba ML, Gordon LI, et al. CR rates in relapsed/refractory (R/R) aggressive B-NHL treated with the CD19-directed CAR T-cell product JCAR017 (TRANSCEND NHL 001). Am Soc Clin Oncol 2017; 35: 7513–7513. [Google Scholar]
84. Abramson JS, Palomba ML, Gordon LI, et al. High durable CR rates in relapsed/refractory (R/R) aggressive B-NHL treated with the CD19-directed CAR T cell product JCAR017 (TRANSCEND NHL 001): defined composition allows for dose-finding and definition of pivotal Cohort. Blood 2017; 130(Suppl. 1): 581. [Abstract] [Google Scholar]
85. Abramson J, Palomba ML, Gordon L, et al. High CR rates in relapsed/refractory (R/R) aggressive B-NHL treated with the CD19-directed CAR T cell product JCAR017 (TRANSCEND NHL 001). Hematol Oncol 2017; 35: 138. [Abstract] [Google Scholar]
86. Maloney DG, Abramson JS, Palomba ML, et al. Preliminary safety profile of the CD19-directed defined composition CAR T cell product JCAR017 in relapsed/refractory aggressive B-NHL patients: potential for outpatient administration. Blood 2017; 130(Suppl. 1): 1552. [Google Scholar]
87. Abramson JS, Gordon LI, Palomba ML, et al. Updated safety and long term clinical outcomes in TRANSCEND NHL 001, pivotal trial of lisocabtagene maraleucel (JCAR017) in R/R aggressive NHL. J Clin Oncol, abstract 7505. [Google Scholar]
88. Nastoupil LJ, Jain MD, Spiegel JY, et al. Axicabtagene ciloleucel (axi-cel) CD19 chimeric antigen receptor (CAR) T-cell therapy for relapsed/refractory large B-cell lymphoma: real world experience. Blood 2018; 132: 91. [Google Scholar]
89. Jacobson CA, Hunter B, Armand P, et al. Axicabtagene ciloleucel in the real world: outcomes and predictors of response, resistance and toxicity. Blood 2018; 132: 92. [Google Scholar]
90. Kochenderfer JN, Dudley ME, Feldman SA, et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 2012; 119: 2709–2720. [Europe PMC free article] [Abstract] [Google Scholar]
91. Chong EA, Svoboda J, Nasta SD, et al. Chimeric antigen receptor modified T cells directed against CD19 (CTL019) in patients with poor prognosis, relapsed or refractory CD19+ follicular lymphoma: prolonged remissions relative to antecedent therapy. Blood 2016; 128: 1100. [Google Scholar]
92. 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 2014; 6: 224ra25. [Europe PMC free article] [Abstract] [Google Scholar]
93. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014; 371: 1507–1517. [Europe PMC free article] [Abstract] [Google Scholar]
94. Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol 2017. [Europe PMC free article] [Abstract] [Google Scholar]
95. Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 2016; 127: 3321–3330. [Europe PMC free article] [Abstract] [Google Scholar]
96. Lee DW, Gardner R, Porter DL, et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014; 124: 188–195. [Europe PMC free article] [Abstract] [Google Scholar]
97. Giavridis T, Van der Stegen SJC, Eyquem J, et al. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med 2018; 24: 731–738. [Europe PMC free article] [Abstract] [Google Scholar]
98. Norelli M, Camisa B, Barbiera G, et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med 2018; 24: 739–748. [Abstract] [Google Scholar]
99. Locke FL, Rossi J, Xue X, et al. Abstract CT020: immune signatures of cytokine release syndrome and neurologic events in a multicenter registrational trial (ZUMA-1) in subjects with refractory diffuse large B cell lymphoma treated with axicabtagene ciloleucel (KTE-C19). In: Proceedings of the AACR Annual Meeting, 1–5 April 2017 Washington, DC. [Google Scholar]
100. Siddiqi T, Abramson JS, Li D, Brown W, et al. Patient characteristics and pre-infusion biomarkers of inflammation correlate with clinical outcomes after treatment with the defined composition, CD19-targeted CAR T cell product, JCAR017. Blood 2017; 130: 193. [Google Scholar]
101. Gust J, Hay KA, Hanafi LA, et al. Endothelial activation and blood-brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov 2017; 7: 1404–1419. [Europe PMC free article] [Abstract] [Google Scholar]
102. Porter D, Frey N, Wood PA, Weng Y, Grupp SA. Grading of cytokine release syndrome associated with the CAR T cell therapy tisagenlecleucel. J Hematol Oncol 2018; 11: 35. [Europe PMC free article] [Abstract] [Google Scholar]
103. 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 2016; 126: 2123–2138. [Europe PMC free article] [Abstract] [Google Scholar]
104. Sotillo E, Barrett DM, Black KL, et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov 2015; 5: 1282–1295. [Europe PMC free article] [Abstract] [Google Scholar]
105. Orlando EJ, Han X, Tribouley C, et al. Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia. Nat Med 2018; 24: 1504–1506. [Abstract] [Google Scholar]
106. Till BG, Jensen MC, Wang J, et al. Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood 2008; 112: 2261–2271. [Europe PMC free article] [Abstract] [Google Scholar]
107. Wang Y, Zhang WY, Han QW, et al. Effective response and delayed toxicities of refractory advanced diffuse large B-cell lymphoma treated by CD20-directed chimeric antigen receptor-modified T cells. Clin Immunol 2014; 155: 160–175. [Abstract] [Google Scholar]
108. 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 2012; 119: 3940–3950. [Europe PMC free article] [Abstract] [Google Scholar]
109. James SE, Greenberg PD, Jensen MC, et al. Antigen sensitivity of CD22-specific chimeric TCR is modulated by target epitope distance from the cell membrane. J Immunol 2008; 180: 7028–7038. [Europe PMC free article] [Abstract] [Google Scholar]
110. Shah NN, Stetler-Stevenson M, Yuan CM, et al. Minimal residual disease negative complete remissions following anti-CD22 chimeric antigen receptor (CAR) in children and young adults with relapsed/refractory acute lymphoblastic leukemia (ALL). Blood 2016; 128: 650. [Abstract] [Google Scholar]
111. Zah E, Lin MY, Silva-Benedict A, et al. T cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B cells. Cancer Immunol Res 2016; 4: 498–508. [Europe PMC free article] [Abstract] [Google Scholar]
112. Hossain N, Sahaf B, Abramian M, et al. Phase I experience with a bi-specific CAR targeting CD19 and CD22 in adults with B-cell malignancies. Blood 2018; 132(Suppl. 1): 490. [Google Scholar]
113. Hill BT, Roberts ZJ, Rossi JM, et al. Marked re-expansion of chimeric antigen receptor (CAR) T cells and tumor regression following nivolumab treatment in a patient treated with axicabtagene ciloleucel (axi-cel; KTE-C19) for refractory diffuse large B cell lymphoma (DLBCL). Blood 2017; 130: 2825. [Google Scholar]
114. Chong EA, Melenhorst JJ, Svoboda J, et al. Phase I/II study of pembrolizumab for progressive diffuse large B cell lymphoma after anti-CD19 directed chimeric antigen receptor modified T cell therapy. Blood 2017; 130: 4121. [Google Scholar]
115. Locke FL, Westin JR, Miklos DB, et al. Phase 1 results from ZUMA-6: axicabtagene ciloleucel (axi-cel; KTE-C19) in combination with atezolizumab for the treatment of patients with refractory diffuse large B cell lymphoma (DLBCL). Blood 2017; 130: 2826. [Google Scholar]
116. Jacobson CA, Locke FL, Miklos DB, et al. End of phase 1 results from Zuma-6: axicabtagene ciloleucel (Axi-Cel) in combination with atezolizumab for the treatment of patients with refractory diffuse large B cell lymphoma. Blood 2018; 132(Suppl. 1): 4192. [Google Scholar]
117. Chow VA, Shadman M, Gopal AK. Translating anti-CD19 CAR T-cell therapy into clinical practice for relapsed/refractory diffuse large B-cell lymphoma. Blood 2018; 132: 777–781. [Europe PMC free article] [Abstract] [Google Scholar]
118. 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 2013; 122: 4129–4139. [Europe PMC free article] [Abstract] [Google Scholar]
119. Smith M, Zakrzewski J, James S, et al. Posttransplant chimeric antigen receptor therapy. Blood 2018; 131: 1045–1052. [Europe PMC free article] [Abstract] [Google Scholar]
120. 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 2016; 34: 1112–1121. [Europe PMC free article] [Abstract] [Google Scholar]
121. Joung JK, Sander JD. TALENs: a widely applicable technology for targeted genome editing. Nat Rev Mol Cell Biol 2013; 14: 49–55. [Europe PMC free article] [Abstract] [Google Scholar]
122. Hernandez I, Prasad V, Gellad WF. Accounting for all costs in the total cost of chimeric antigen receptor T-cell immunotherapy-reply. JAMA Oncol 2018; 4: 1785–1786. [Abstract] [Google Scholar]

Articles from Therapeutic Advances in Hematology are provided here courtesy of SAGE Publications

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/59552796
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/59552796

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1177/2040620719841581

Supporting
Mentioning
Contrasting
0
140
0

Article citations


Go to all (113) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.