Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Aug 30.
Published in final edited form as: Best Pract Res Clin Haematol. 2018 Apr 11;31(2):135–146. doi: 10.1016/j.beha.2018.04.001

CAR T cell therapy for B-cell lymphomas

Julio C Chavez a, Frederick L Locke b,*
PMCID: PMC6716161  NIHMSID: NIHMS1046245  PMID: 29909914

Abstract

B-cell non-Hodgkin’s lymphoma (NHLs)is a very heterogonous malignancy with diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL) as the most common subtypes. Standard treatment with anti-CD20 based chemoimmunotherapy is usually very effective for disease control. However a significant proportion of patients with high-risk features (double hit lymphoma, transformed lymphomas or early relapses) will become refractory to standard therapies and will have limited alternatives for cure. Adoptive therapy with chimeric antigen receptor (CAR) T-cells is a new paradigm for effective treatment of poor prognosis lymphomas. Here we review the biology of poor risk DLBCL and FL, the rationale for CAR T-cell therapy in malignant lymphoma and the efficacy/toxicity profile of CD19 directed CAR T cell therapy for DLBCL and FL from early single center studies to multicenter/global clinical trial with different CAR T cell constructs.

Keywords: CART, Aggressive lymphomas, Refractory, Immunotherapy, Diffuse large B cell lymphoma

1. Introduction

The use of chimeric antigen receptor (CAR) modified T-cells targeting CD19 expressing tumor cells have revolutionized the treatment of refractory B-cell lymphoid malignancies. Non-Hodgkin’s lymphomas comprise a heterogeneous group of lymphoid malignancies that originates from B-cell lymphocytes, T-cell lymphocytes and natural killer (NK) cells. The revised World Health Organization (WHO) 2016 have re-classified many B-cell lymphoid malignancies entities based on a deeper understanding of genomics and molecular pathways [1]. The majority of lymphoid malignancies are of B-cell origin and almost invariably express CD19 protein on their cell surface. Thus, the use of CD19 directed CAR therapy has been subject of intense research for B-cell Non-Hodgkin’s lymphomas (NHL).

2. Epidemiology of lymphomas

It is estimated that in 2015, there were 71,000 cases of NHL with approximately 19,700 deaths related to this disease. NHLs is the 7th leading cause of new cancer cases and accounts for approximately 3% of cancer-related deaths in the United States [2]. Of all NHLs, diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma subtype with 32.5% of all newly diagnosed cases, followed by chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) with 18.6% and follicular lymphoma (FL) with 17.1% [3].

3. Rationale for the use of CAR T cell therapy for B-cell non-Hodgkin’s lymphomas

The basic anatomy of a CAR structure consists in an antigen-recognition domain, usually a single-chain variable fragment (scFv) derived from a monoclonal antibody targeting the selected antigen (ie: CD19); a hinge (usually derived from CD8 or Ig4 molecules) that links the recognition site to the transmembrane domain which bridges the membrane; and finally, the intracellular domain that typically contains CD3z chain critical for T cell receptor 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 [48].

CD19 is a transmembrane glycoprotein that is involved in the regulation of the 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 [9]. 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 non-malignant B-cells, it is well established that cancer patients can survive without normal B-cells following chemotherapy or B-cell directed monoclonal antibodies; thus, it appears that there does not seem to be deleterious complications with long-term B-cell aplasia [10]. All these factors make CD19 an attractive target for immunotherapeutic approaches with several companies and academic institutions developing pivotal trials with anti-CD19 CAR T-cells [4,9].

4. Aggressive B-cell lymphomas

Aggressive B-cell lymphomas encompass a heterogeneous group of clinical and molecular diseases with DLBCL being the most common subtype. The WHO 2016 classification differentiated additional important subtypes such as high-grade B-cell lymphomas (HGBL) NOS and high-grade B-cell lymphomas with MYC and BCL2 and/or BCL6 rearrangements, known as double (DHL) or triple hit lymphomas (THL). The prognosis of aggressive B-cell lymphomas is based on clinical, molecular and genetic factors as well as responsiveness to induction chemotherapy.

4.1. Clinical factors as predictive of poor prognosis DLBCL

Prognostic indices have been developed based on clinical factors that predict outcomes in DLBCL. They are commonly utilized in routine practice and in clinical trials. The commonly utilized clinically based scoring system, the International Prognostic Index (IPI), was updated with the Revised International Prognostic Index (R-IPI) which has since been validated in the rituximab era [11,12]. An enhanced scoring system, the NCCN-IPI score stratifies newly diagnosed DLBCL patients in 4 risk groups based on the same prognostic factors used in the IPI score. The NCCN-IPI score was able to better discriminate the low and high-risk subgroups than the IPI score [13,14]. Patients in the high-risk subgroup category per the R-IPI and NCCN-IPI scores have a 5-years overall survival (OS) of 54% and 33%, respectively; representing an unmet need in DLBCL. All scoring systems the IPI, R-IPI and NCCN-IPI can predict clinical outcomes accurately, and are used to different degrees in clinical practice.

4.2. Molecular and genetic factors as predictive of poor prognosis DLBCL

Molecular studies play an important role in identifying DLBCL with high-risk features. Based on the cell of origin (COO) using gene expression profiling (GEP) studies, DLBCL can be classified into activated B-cell (ABC) subtype and germinal center B-cell (GCB) subtype (with 10% being molecularly unclassifiable). Each of these COO subtypes have distinct phenotypic, clinical characteristics and prognosis with ABC –DLBCL having worse outcomes, as compared to GCB – DLBCL, with standard upfront R-CHOP chemotherapy [15,16]. Similarly, next generation sequencing (NGS) studies showed that for GCB lymphomas, specific mutations such as BCL2, EZH2, KMT2D, S1PR2, and GAN13 have independent prognostic significance [17,18]. Widespread use of GEP or NGS remains limited due to cost and complexity, thus immunohistochemistry (IHC) methods continue to be more practical and feasible. The Hans classifier uses 3 markers CD10, BCL6 and MUM1/IRF4) that classifies DLBCL as GCB or non-GCB subtypes [18,19] by IHC. Other IHC based algorithms were developed with good concordance to GEP [20,21]. COO classification based on IHC algorithm is popular and less expensive then molecular techniques, however the predictive value of IHC methods has been questioned [22,23]. Alternative methodology includes RNA deep sequencing and limited set gene expression profile on formalin-fixed-paraffin-embedded (FFPE) tissue (NanoString Lymph2Cx) [2426].

Another high-risk subgroup of DLBCL patients is characterized by the overexpression of the proto-oncogene MYC that promotes uncontrolled cell growth, associated with unregulated cell division and extranodal dissemination of lymphoma [27]. MYC aberrations and/or overexpression can be detected by band cytogenetics, interphase fluorescence in situ hybridization (FISH), genomic comparative hybridization (GCH) studies and IHC. MYC rearrangements are the most common partner IGH detected by FISH or cytogenetics, present in 3–17% of DLBCL, and can be associated with BCL2 and/or BCL6 rearrangements. When MYC rearrangements are present in conjunction with one or both BCL2 and BCL6 rearrangements it is termed double-hit lymphoma (DHL) or triple hit lymphoma (THL) [28]. About 60% of high grade B-cell lymphomas have MYC and BCL2 rearrangements, 20% have MYC and BCL6 and 20% are THLs with all three [29]. An aggressive course of disease with poor outcomes characterizes DHL and THL when treated with standard front-line chemotherapy [30,31]. Unfortunately, cytogenetics, FISH, or molecular exploration of MYC, BCL-2, and BCL-6 from diagnostic tissue is often not ordered or available. Alternatively IHC can be utilized for analysis of MYC protein overexpression in DLBCL and when found in conjunction with BCL-2 or BCL-6 protein overexpression it is called double-expressor lymphoma (DEL). While DEL represents a more heterogeneous disease than DHL, it has also been shown to be associated with poor outcomes across studies [3234]. Clinically, DHL and THL are characterized by advanced disease stage, very elevated LDH and CNS involvement(32, 35). The optimal treatment of DHL has not been well defined but it appears that front-line intensive regimens (such as dose adjusted-EPOCH-R with CNS prophylaxis) are associated with better outcomes than R-CHOP [3537]. Similarly, the role of autologous stem cell transplantation (ASCT) is debatable, as DHL or DEL patients have inferior OS and PFS after ASCT as compared to non-DHL patients [3841]. Thus, the best approach for MYC rearranged DLBCL remains unclear and represents an unmet need given its poor prognosis.

4.3. Response to therapy as prognostic indicator

The addition of rituximab to CHOP improved the OS of patients with DLBCL, however, approximately 30–40% of patients will relapse and become refractory to this regimen [42]. In the majority of cases, lymphoma recurrence occurs within the first 3 years from diagnosis with very few patients relapsing beyond 5 years [43]. In general patients with refractory disease (less than PR to initial treatment) or early relapse (relapse within a year from diagnosis or 6 months after the end of treatment) or progression within 2 years (EFS24) have poorer prognosis [44,45]. Salvage chemotherapy followed by ASCT is the current standard approach [44,46]. About 30–40% of patients with primary refractory disease or early DLBCL relapses will respond to salvage chemotherapy and will be able to undergo ASCT, however about 50% of these patients will ultimately relapse again and further approaches have limited efficacy [44,46,47]. The prognosis of relapsed DLBCL post ASCT is poor with a median OS of 10 months, and particularly worse for relapses within 6 months with a median OS of 5.7 months [48,49].

A retrospective international multicenter study (SCHOLAR-1) was carried out using data from 2 phase III randomized clinical trials (CORAL and CCTG LY.12 trials) and 2 observational studies (MD Anderson Cancer Center and University of Iowa/Mayo Clinic) that evaluated patients that at best achieved stable disease and progressive disease as best response at any point during chemotherapy or relapsed within 12 months of ASCT [50]. The results of this pooled analysis showed an overall response rate of 26% with a median OS of 6.3 months [50]. Another large retrospective study of 15 U.S academic institutions assessed risk factors of refractory DLBCL defined three groups: primary progression (PD with or within 6 weeks of CIT), residual disease (PR or stable disease [SD] after completion of CIT) and early relapse (relapse within 6 month of CIT and having achieved CR) with a 2-year OS of 18.5%, 30.6% and 45.5%, respectively [51]. Multivariate analysis demonstrated that ultra high-risk features (UHR) in refractory DLBCL patients were primary progression, MYC rearrangement present and intermediate-high or high NCCN-IPI score at the time of relapse [51]. The findings of these 2 studies will serve as benchmarking for clinical trials in poor risk refractory DLBCL.

4.4. Efficacy of CART therapy in aggressive B-cell lymphomas

4.4.1. Early studies in DLBCL

The initial studies of anti-CD19 CART cells for lymphoma were carried out in single institutions and included a diverse population of refractory B-cell NHLs, including DLBCL, FL, primary mediastinal B-cell lymphoma (PBMCL), marginal zone lymphomas (MZL) and transformed follicular lymphomas (TFL). Two early reports of anti-CD19 CART cells were in patients with indolent NHL (will be discussed below) [52,53]. The first study to include DLBCL patients utilized a first generation anti CD19z CART (without costimulatory domain) and second generation anti CD19–28z CART. While this study showed significant expansion of CART cells with CD28 costimulation (the nadir was reached at 4–6 weeks), there were no objective responses reported in DLBCL patients. It should be noted that patients received CART cells after at least 6 weeks from last chemotherapy and that there was no lymphodepleting regimen given [54].

The first CART cell study to demonstrate activity in DLBCL was conducted at the NCI using the CD3z-CD28 CART construct (later licensed by Kite pharma for development as axicabtagene ciloleucel). Nine patients with refractory aggressive B-cells NHLs were included: DLBCL [4], PMBCL [4] and DLBCL transformed from CLL [1] in whom CART cells were successfully manufactured at the first attempt. The conditioning regimen consisted of cyclophosphamide (total dose of 60 mg/kg) followed by fludarabine 25 mg/m2 daily for 5 days [55]. There were five CRs and two PRs out of the seven evaluable patients. Three patients who achieved CR had ongoing remissions at the time of publication of the study [55]. In a long-term follow up of responding patients, there were still ongoing remissions and the duration of response ranged from 38 to 56 months [56], highlighting the durability of the therapy for large B cell lymphoma.

Another follow-up report from the NCI included 22 aggressive B cell lymphoma patients (DLBCL: 13, TFL: 2, PMBCL: 1, FL: 2, mantle cell lymphoma: 1 and RT: 1) and demonstrated that low dose conditioning chemotherapy (cyclophosphamide 300–500 mg/m2 and fludarabine 30 mg for 3 days) had effective lymphodepleting activity and was associated with less hematologic and non-hematologic toxicity [57]. In this study, the ORR and CR rates among patients with DLBCL (majority with refractory disease) were 68% and 47%, respectively. The median duration of remission was 12.5 months and the 12-month PFS was 63.3% [57].

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 was conducted based upon strong pre-clinical 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 to those manufactured from effector memory (EM) T-cells in mouse models [58]. 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 [59]. Patients with relapse post-autologous HSCT and post-allogeneic HSCT were also included. This therapy had anti-lymphoma activity with an ORR and CR rates for the whole group of 63 and 33%, respectively; in the subgroup of aggressive lymphomas (DLBCL and TFL) the ORR and CR were 67 and 38%, respectively, and the CAR construct is now licensed by JUNO for development as JCAR017 [59].

Researchers at the University of Pennsylvania developed an alternate anti-CD19 CART also using 4–1BB as a costimulatory domain (called CTL019) with significant anti-lymphoma activity. Preliminary results presented at ASH 2015 confirmed its efficacy in patients with a variety of B-cell NHL, including DLBCL, FL and MCL [60,61]. In the updated analysis there were 38 patients enrolled but 10 patients did not receive treatment for a variety of reasons (rapid disease progression: 4, inability to manufacture CAR T cells due to low T-cell counts: 5, and 1 consent withdrawal) [62]. In the 28 evaluable patients at data cut-off (May 2017) the ORR was 64% and 57% of patients remained free of progression at a median follow up of 28.6 months [60,62]. In the 14 DLBCL patients, the ORR at 3 months and 6 months were 50% and 43%, respectively with no significant differences between GCB/non-GC, double hit status or transformation from FL subtypes [61,62]. The median duration of response was not reached with a 86% of responding DLBCL patients maintaining an ongoing response at last follow up [61,62].

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

4.4.2. Multicenter studies in aggressive lymphomas

Given the preliminary efficacy of CART therapy in single-institution studies, several CAR T cell constructs were licensed for development by pharmaceutical companies for development as therapy for lymphoma. Multicenter phase 1–2 pivotal studies were designed primarily targeting aggressive B-cell lymphomas. With some minor differences, all studies included poor prognosis DLBCL patients that had received, and remained refractory to, standard chemotherapy (Table 1). The ZUMA-1 study included patients with primary refractory DLBCL to front-line anthracycline based chemotherapy, with inclusion of patients relapsing after autologous HSCT.

Table 1.

Patient characteristics in the three largest anti-CD19 multicenter studies CAR T-cells in aggressive B-cell NHLs.

Patients Characteristics ZUMA-1 (Neelapu, 2017) JULIET (Schuster, 2017) TRANSCEND (Abramson, 2017)
No of patients enrolled 111 (101) 141 (85) 91 (67)
Median age, range 58 (23–76) 56 (24–75) 61 (29–82)
Age ≥ 65 24% 21% 17%
Lymphoma subtypes DLBCL, TFL, PMBCL DLBCL, TFL DLBCL, TFL (CORE)a
Double hit lymphomas NR 27% 27%
≥ 3 lines of therapy 69% 50% 50%
Primary refractoriness 26% NR NR
Refractory to > 2nd line 77% NR 76%
Relapse post ASCT 21% 51% 44%

DLBCL: Diffuse Large B-cell Lymphoma, TFL: Transformed lymphoma, FL: Follicular lymphoma, ASCT: Autologous Stem Cell Transplantation, NR: Not Reported.

a

The FULL cohort included: DLBCL transformed from CLL (Richter transformation) and marginal zone lymphoma (MZL), PMBCL and follicular lymphoma 3B

The first multicenter trial to evaluate CAR T for DLBCL was the Phase I-II trial utilizing the NIH CD3 zeta/CD28 CAR construct with a streamlined closed manufacture process. The cell dose and conditioning chemotherapy previously tested at the NCI were confirmed safe in 7 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. The conditioning regimen consistent of cyclophosphamide 500 mg/m2 and fludarabine 30mg/m2 × 3 days followed by infusion of KTE-C19, now called axicabtagene ciloleucel (axi-cel) at a dose of 1–2× 106 CART cells/kg [50,63] was deemed safe. The objective response was 71% with 4 patient achieving CR (57%) at 1 month evaluation. Three patients had ongoing CR at 12 months post axi-cel infusion. Reversible grade 3 neurotoxicity and cytokine release syndrome (CRS) were reported among this cohort. One patient experienced grade 4 CRS and grade 4 encephalopathy and died of intracranial bleeding, which was considered unrelated to axi-cel. This patient had rapidly progressive disease associated with fevers and preplanned retrospective review of cytokine levels before conditioning chemotherapy demonstrated that the patient was already in an extremely inflammatory state prior to CAR T cell infusion. Prior to initiation of the phase 2 portion of the trial, safety criteria were included to check the CRP and delay CAR T cell infusion in patients with fever until appropriate work-up was completed.

The pivotal phase II portion of the ZUMA-1 trial [64] included similar eligibility criteria as the phase I, with two cohorts: cohort 1 for DLBCL and cohort 2 for PMBCL and TFL. The primary end point was objective response rate (ORR) in patients with more than 6 months follow-up post axi-cel infusion, as compared to historical controls. Secondary end points were duration of response (DoR), OS, safety and levels of CAR-T cells and cytokines. The CAR manufacturing success was 99%.

A total of 111 patients were enrolled, with a CAR manufacturing success rate of 99%. Ten patients could not receive axi-cel for various reasons (SAE prior to conditioning regimen: 5, non measurable disease: 2, no product available: 1 and SAE post conditioning regimen: 2).

The 101 patients that received the therapy were the pre-specified intent-to-treat (ITT) analysis cohort. The study met the primary endpoint compared to historical cohort (SCHOLAR-1) with an objective response of 83% and CR of 54% (in comparison to the a pre-specified ORR of 20%, p < 0.0001) representing a 8-fold higher CR rates in comparison to SCHOLAR-1. With a data cut-off of August 11th, 2017, the median follow-up time was 15.4 months (Table 2). The ongoing responses and CR were 42 and 40%, respectively for all patients treated on the phase 1–2 trial. Overall objective response rate and ongoing responses were consistent across key covariates, including advanced stage, age, presence of bulky disease, high IPI score or refractory subgroups (R/R post auto HSCT or > 2nd line of therapy). The median duration of response was 11.1 months in all responders and was not reached in those achieving CR. The 18-month PFS and OS were 41 and 52%.

Table 2.

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

Study ZUMA-1 (Neelapu, 2017) JULIET (Schuster, 2017) TRANSCEND (Abramson, 2017)
No of patients enrolled (treated) 111 (101) 141 (99) NR (91)
67 in CORE
Median age, range 58 (23–76) 56 (24–75) 61 (29–82)
Median follow-up 15.4 months 5.6 months 6.3 months
Costimulatory domain CD28 4–1BB 4–1BB
Bridging chemotherapy Not allowed Allowed Allowed
CART dose 2.0 × 106 cells/kg Median, 3.1 × 108 DL1 5.0 × 107 cellsa
DL2 1.0 × 108 cells
Conditioning regimen Flu 30 mg/m2 ×3d Flu 25/m2× 3d Flu 30 mg/m2 ×3d
Cy 500 mg/m2 ×3d Cy 250 mg/m2 ×3d or B 90 mg/m2 × 2d Cy 300 mg/m2 ×3d
Efficacy
 %ORR (%CR) 82 (54) 59 (43) 84 (61)
 3-mo %ORR (%CR) 44 (39) 45 (37) 65 (53)
 mDOR 11.1 months NR 9.2 months

NR: Not reported, mDOR: median duration of response, Flu: Fludarabine, Cy: cyclophosphamide, B: Bendamustine, ORR: Overall Response Rate, CR: Complete Response.

a

Six patients received double dose of DL1.

The JULIET trial is a phase II multicenter study in patients with refractory DLBCL utilizing CTL019, the anti CD19 CAR construct with a 4–1BB costimulatory domain created at the University of Pennsylvania. Interim results were presented at the American Society of Hematology 59th Annual meeting in 2017 [65,66]. CAR T cells were manufactured centrally, however in contrast to ZUMA-1 cryopreserved apheresis products was utilized and bridging chemotherapy was allowed in order to prevent rapid disease progression. The 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 HCST, or refractory after 2 lines of therapy. Similar to the ZUMA-1 trial, the primary endpoint was ORR.

141 patients were enrolled, however 42 patients were unable to receive CTL019 infusion due to: change in patient status (rapid disease progression in 28 patients, AE in 1 patient, investigator decision in 2, withdrawal in 1 and 1 case of protocol deviation) and inability to manufacture CART cells in 9 cases. Thus, 99 patients received CTL019 infusion with 81 patients having completed more than 3 months follow up at the time of the analysis with a data cutoff of March 8th, 2017. With a median follow up time of 5.6 months, eighty-nine out of 99 infused patients received various bridging chemotherapy regimens. The median dose of CTL019 was 3.1 × 108 (0.1–6.0 × 108) cells. With an objective response of 53% (CR 40%), the JULIET study met its primary endpoint (p < 0.0001 [95% CI, 44–72]). The 3- and 6-months ORR (CR) rates were 38% (32%) and 37% (30%). The median OS and DoR were not reached in responders. The relapse-free survival at 6 months was 74%. If patients were in CR at 3 months, they continued with ongoing CR at data cutoff. Outpatient infusion of CTL019 was feasible and was given to 26 patients and 20 (77%) of those remained as outpatient more than 3 days. No deaths were attributed to CLT019, but 3 patients died within 30 days of infusion (all due to disease progression).

The TRANSCEND-001 study is testing JCAR017 construct, with 4–1BB costimulatory molecule, using a defined CD4:CD8 T-cell ratio [67]. In this trial the inclusion criteria was broader than ZUMA-1 or JULIET: refractory/relapsed DLBCL, TFL, FL grade 3B, mantle cell lymphoma and PMBCL. Patients with CNS involvement, Richter transformation, transformation from marginal zone lymphoma (MZL) and relapsed post-allogeneic HSCT were also included in the study. There were three dose-levels (DL) in this study: DL-1S was 5 × 107, and DL-2S was 1 × 108. A small cohort of patients received double dose of JCAR017 at 5 × 107 that was administered 14 days apart (currently suspended). 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. With a data cutoff of July 17th, 2017, the median follow-up was 6.3 months, The investigators labeled the “FULL” study cohort to include 91 patients and the pivotal population of 67 patients, the “CORE” portion of DLBCL, TFL, and high grade B-cell lymphoma. Although the number of enrolled patients (and those who did not receive the product for various reasons) was reported in the preliminary analysis presented at the ICML-2017, but not reported at the recent ASH 2017 meeting [68,69]. The full cohort analysis data reported an ORR and CR rates of 75 and 56%, respectively. In the CORE data set the best ORR (CR) rates, 3-month and 6 month were 75% (56%), 65% (53%) and 57%(52%) with 55 patients that had more than 3 months follow up. There was a trend towards a dose-dependent improved activity of JCAR017 with a 3-month ORR (CR) rates of 63% (58%) and 40% (27%) in DL2 and DL1, respectively (CORE). The activity of JCAR017 was not different across different high-risk DLBCL groups including double and triple hit lymphomas. In patients achieving CR the median DoR was 9.2 months in the FULL and not reached in the CORE datasets, respectively and the median OS was 13.7 in the FULL and NR in the CORE subgroups [69]. This trial also showed the feasibility and safety of outpatient administration in a small cohort of patients, with CRS and neurotoxicity rates not significant different from inpatient treatment and reducing the duration of hospitalization by 40% [70].

5. Indolent B-cell lymphomas

Indolent NHLs comprise a heterogeneous group of B-cell lymphoproliferative disorders characterized by a slow disease progression and a pattern of relapses over the course of the disease. Diseases that are considered in this category are: follicular lymphoma (FL), marginal zone lymphoma (MZL), lymphoplasmacytic lymphoma (LPL) and small lymphocytic lymphoma (SLL). Although patients with indolent lymphomas can live for many years, they are considered incurable with few exceptions (i.e., limited stage disease or allogeneic stem cell transplantation).

Follicular lymphoma is the most common indolent lymphoma and represents about 35% of all NHLs. There has been an important improvement in the outcomes in FL with median survivals approaching 2 decades, mainly due to the introduction of anti-CD20 monoclonal antibodies (rituximab) in combination with standard chemotherapy [7174]. Given the favorable outcomes of majority of patients with iNHL and FL and durable remissions with available chemoimmunotherapy, PFS and OS may not be adequate to estimate efficacy of new interventions due to long follow up needed, particularly in the front-line setting. Thus, models/prognostic markers that predict outcomes have been developed using clinical and/or molecular factors and/or responsiveness to initial therapy.

5.1. Clinical factors as predictors of outcomes in FL

The Follicular Lymphoma-IPI or FLIPI (age > 60, stage III/IV, elevated LDH, > 4 nodal sites and hemoglobin < 12 g/dl) and FLIPI-2 (age > 60, hemoglobin < 12 g/dl, increased B2 microglobulin, bone marrow involvement, largest lymph node > 6 cm in size) scores was able stratify FL patients into three risk categories: low risk (10-year OS of 71%) intermediate risk (10-year OS of 51%) and high risk (10-year OS of 36%) [75,76].

5.2. Response to therapy as a high-risk indicator for indolent NHL

There have been several studies seeking surrogates of FL survival other than OS and PFS. A large pooled analysis of 13 randomized clinical trials in FL showed that CR rate at month 30 from front-line therapy for FL was predictive of PFS, regardless of the inclusion of rituximab in the regimen [77]. Another strong predictor of survival is the duration of response. The National Lymphcare Study (NLCS) studied the outcomes of FL patients treated with R-CHOP as a front-line regimen. The study identified that event-free survival at 24 months (EFS24) was associated with poor outcomes in comparison to those who did not experience early relapse (5-year OS of 50% and 90%, respectively) [78], and the surrogate endpoint was since validated in other analysis [7981]. Patients not achieving EFS24 remain as an unmet need in FL and constitute as a primary endpoint in clinical trials for FL (and other iNHL).

5.3. Biological and molecular predictors

Follicular lymphoma is graded based on the number of centroblasts per high-power field from grade 1 to grade 3a/3b. Whilst there are no differences in clinical outcomes between grades 1 – 3a and treatment decisions are not affected, grade 3b FL is considered a distinct disease and usually approached as an aggressive lymphoma (similar to DLBCL) and treated with anthracycline based chemotherapy, with a probability of cure of about 40% [82].

The FL tumor microenvironment (TME) has been extensively studied with different methodologies, most commonly using gene expression profiling techniques of non-neoplastic cells. Immune interactions between TME and neoplastic FL cells seem to be prognostically relevant [83]. CD4+ T-cells including FOXP3+ regulatory T-cells (Treg) and follicular helper cells (Tfh), and their interaction with TME, may influence response to therapy and antitumor immunity [84,85].

The mutational landscape of follicular lymphoma is characterized by the presence of several gene alterations, with the majority involving epigenetic regulators and chromatin modifiers but also involving the B-cell receptor pathway genes [8689]. These mutations are believed to promote FL lymphomagenesis by interacting with p53, BCL2 and BCL6 but also resistance and short response to therapy [88].

The m7-FLIPI score was developed incorporating 7 gene alterations known to be recurrently mutated in FL: CREBBP, ARID1A, EZH2, EP300, FOXO1, MEF2B and CARD11. The m7-FLIPI score was better to discriminate high-risk subgroups than traditional FLIPI with a 5-year failure free survival (FFS) and OS of 25% and 42% in the high-risk category [90]. Additionally, the m7-FLIPI score was able to identify better patients in high risk of not achieving EFS24, however was able to qualify only 50% of FL patients with early progression [91].

5.4. CAR-T cell studies in indolent non-Hodgkin lymphomas

To date the majority of NHL patients that have received anti-CD19 CAR T-cells were aggressive B-cell NHLs (mainly DLBCL), some of the first NHL patients to receive this therapy were indolent NHLs. The first FL cases reported no response to first generation anti-CD19 CAR T-cells without costimulation [52]. The first lymphoma patient successfully treated with anti-CD19 CAR T-cells with CD28 as co-stimulatory domain was reported by the NCI and this patient achieved long-term remission [53]. A subsequent report included 5 patients with indolent NHL (4 FL and 1 MZL) that were treated with CAR T-cell single infusion followed by several doses of IL-2 [92]. Patients received a conditioning regimen consisting of cyclophosphamide 60 mg/kg × 2 days and fludarabine 25 mg/m2 × 5 days. Although all patients achieved a response (PR) there were no patients who achieved CR with 75% of FL patients having ongoing response at the time the study was reported [92].

In the 32 patients included in a FHCRC study (with various NHL histologies) that used a 1:1 ratio CD4/CD8 to manufacture CAR T-cells, there were 6 FL patients (there were no other indolent NHL subtypes). In evaluable FL patients the ORR and CR rates were 80% [4/5] and 40% [2/5] [59].

Results of the Phase IIa trial using the CTL019 CAR construct at the University of Pennsylvania included 14 patients with refractory FL that had relapsed within 24 months of initial diagnosis and/or remained refractory to least 2 lines of therapy [60,93]. 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 5), relapsed post-autologous HCT (21%) and allogeneic HCT (1 patient). The updated analysis showed a 3-month ORR and CR of 79% [11/14] and 50% [7/14], respectively. The median PFS was not reached and at the median follow u of 28.6 months 70% of patients were disease free [62,93].

Anti-CD19 CAR therapy for indolent lymphomas seems well tolerated with expected toxicity and appears to provide long-term remission in some cases, thus it may have a curative potential. Although a majority of trials include also aggressive lymphoma, however there are studies dedicated to indolent B-cell NHLs with axi-cel (). To date we do not have enough data to predict which indolent lymphoma patients might benefit from the therapy and characterization of the clinical and molecular prognostic factors is warranted on trial.

6. Toxicities of CAR T-cell therapy in lymphomas

The toxicities related to CAR-T cell therapy were well described in early studies in B-cell lymphomas and acute lymphoblastic leukemia (ALL) [55,59,94,95]. There are two main categories of toxicity: cytokine release syndrome (CRS) and neurotoxicity. which can be accompanied by organ damage (renal failure, cardiac dysfunction, liver dysfunction, etc) [96,97]. A more specifically defined criteria for quantification of neurotoxicity, known as CAR-T cell-related encephalopathy syndrome (CRES), was recently developed [96]. The vast majority of CAR T-cell related toxicities resolve within few weeks, and are reported in both single-center and pivotal multicenter studies [55,59,92]. While, the symptoms and signs of each type of toxicities 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 manuscripts [9698].

There are clinical factors that correlate with the development of CRS such as disease burden (specifically in ALL) and levels of CAR T-cells administered [55,59,94,95]. Increased levels of tumor necrosis factor (TNF-alpha), IL-2R, IL-6, interferon (IFN)-gamma, IL-10, IL-15, and ferritin have been shown to be associated with severity of CRS [55,59]. Neurotoxicity seems to be cytokine driven as well [55]. Peak C-reactive protein (CRP) has been shown to be related to CRS severity and can be used as surrogate for early treatment/supportive care [94].

These toxicities have been also reported in the multicenter studies ZUMA-1, JULIET and TRANSCEND trials, with variable frequency and severity, apparently due to differences in patient population, disease subtypes (and clinical presentation), use of different toxicity grading systems and differences in CAR-T cell constructs. The Lee criteria was used to grade CRS in ZUMA-1 and TRANSCEND while the U Penn Criteria was used for severity stratification of CRS in the JULIET trial. Table 3 describes the frequency and features of CART related toxicities in the 3 multicenter studies in NHL.

Table 3.

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

Study ZUMA-1 (Neelapu, 2017) JULIET (Schuster, 2017) TRANSCENDa (Abramson, 2017)
No patients enrolled (treated) 111 (101) 141 (85) NR (91)
Cytokine release syndrome
 Time to onset, median, range 2 days (1–12) 3 days (1–9) 5 days (1–14)
 Duration, median, range 8 days (NR) 7 days (3–34) 5 days (NR)
  Grade (All) 93% 58% 36%
  Grade 3° 4 13% 23% 1%
  Use of tocilizumab 43% 15% 12%
  Use of vasopressors 17% 6% 24%
  Use of steroids 27% 11% 16%
  Admission to ICU NR 24% NR
Infections
 All Grades 35%1 27% NR
 Grade 3 or 4 31%1 13% NR
Neurotoxicity
 Time to onset, median, range 5 days (1–17) NR 10 days (3–23)
 Duration, median, range 17 days (NR) NR 11 days (NR)
 All Grades 64% 21% 21%
 Grade 3 or 4 28% 12% 15%

NR: Not reported, ICU: Intensive care unit.

1

Febrile Neutropenia.

a

Reported from the FULL cohort data.

7. Expansion/persistance of CAR T cells

In vivo expansion and persistence of CAR T cells has been shown to be associated with clinical outcomes in hematological malignancies and particularly in lymphomas [57,59,95]. The ZUMA-1 study CAR T cells peaked within 14 days of the infusion and were still detectable in majority of patients by day 180. CAR T cells levels by day 28 (area under the curve [AUC 0–28] was 5.4 times higher in those in CR versus non responders (p < 0.001) [99]. In the TRANSCEND study higher concentrations and AUC 0–28 levels of CAR T cells expansion were also associated with clinical and durable responses, as well as with higher tumor burden, peak cytokines, CRS and NT; however, there were no differences in detectable CAR T cells (persistence) in responding and non responding patients in this study a fact that is supported by the long term NCI data that showed non clonal CD19+ B-cell recovery in patients with durable remissions [56,100]. The UPenn data showed that CTL019 CAR T cells were detected at a median of 24 months in 14 out 16 patients who had CR. Recovery of CD19+ B-cells occurred in 50% of responding patients and was not associated with disease relapse [62].

A critical component is the conditioning regimen that leads to lymphodepletion. The most commonly used agents are low dose cyclophosphamide and fludarabine [55]. Others such as bendamustine and low radiation have been utilized. The rationale of inducing lymphodepletion prior to adoptive cell therapy include enhancement of the antitumor activity by decreasing regulatory T-cells (T regs), creating a peritumoral inflammatory milieu by tissue damage and increased activation of antigen-presenting cells [101,102]. A key homeostatic cytokine that promotes T-cell proliferation and activation, IL-15, is increased in the setting of lymphodepletion and has been associated with peak CAR T cells and clinical response. All these factors create a favorable environment for CAR T cell survival and persistence after appropriate lymphodepletion [57,103].

Tumor microenvironment and CAR T cells trafficking into the tumor seem to be associated with clinical activity. In the TRANSCEND-001 study patients who achieved CR had an increased CD8+ CAR T cells concentration in the tumor area [104].

8. Other targets/approaches for cellular therapy in lymphomas

Despite the success of anti-CD19 CARs in B-cell NHLs, relapses do occur. One of the hypothesized mechanisms is CD19 immunological antigen escape; especially since CD19 negative B-cell malignancies relapses have been reported [103]. Failure to express CD19 in some B-cell NHLs and/or epitope/antigen loss of the CD19 through splicing mechanism have been described as resistance mechanism or relapse in patients receiving CD19 directed therapy (such as CARs or bispecific antibodies) [105]. Thus, developing CAR T-cells against alternative lymphoma targets is actively being pursued.

Given the success of anti-CD20 therapy for B-cell NHL in the last decade with the use of monoclonal antibodies, a logical CAR target is the CD20 antigen. Earlier studies showed the feasibility of this approach with modest activity [52,106]. When costimulation with 4–1BB was added to the anti-CD20 CAR T cells, the ORR and CR rates were 80–83% and 17–50% in 2 different trials, respectively [107,108]. Anti-CD20 CAR T-cells with CD28 and 4–1BB as costimulation (“third generation CAR”) was feasible with clinical efficacy and tolerable toxicity profile [108].

Studies with CD22-speficic CAR showed potent activity in CD22+ B-cell leukemias but it also has anti-lymphoma activity in pre-clinical studies [109]. Most of studies reported are focusing on CD22+ B-ALL but there ongoing trials for refractory CD22+ B-cell NHLs ( and ) [110].

One of the most common complications of anti-CD19 CAR T-cell therapy is B-cell aplasia with associated hypogammaglobulinemia that is associated increased frequency of infections. One way to prevent this complication is by targeting on light chain type of the immunoglobulin. One study has showed promising activity of anti-kappa light chain CAR in NHL [111].

Another attractive target is the tyrosine kinase like-orphan receptor 1(ROR-1) that is present in neoplastic cells but no in B-cells, thus preventing B-cell aplasia. ROR-1 specific CAR-T cells are currently being studied mostly in CLL [112,113].

Immune escape is a postulated mechanism of CAR resistance, thus targeting more than one antigen is a proposed approach in order to prevent this immunological event. Bi-specific CARs targeting CD19 and CD20 antigens for B-cell malignancies has been developed (CD19-OR-CD20 CAR) with significant pre-clinical activity, even in CD19 negative tumor cells [114]. Bi-specific CARs targeting CD19 and CD22 are also in development and feasible in preclinical studies, and although majority of the trials focus on B-cell ALL CD22+, some include B-cell NHLs ( and ) [115].

Development of immunogenicity of CARs, especially against the epitopes of the murine derived scFvs portion, has been linked to decreased persistence and activity of CAR T-cells once infused [116]. In order to reduce scFv immunogenicity, fully human CAR T-cells with human derived scFv has been developed and showed interesting preclinical profile with potential for clinical use [117].

9. Conclusions and future directions

CAR T cell therapy has emerged as a treatment option for difficult to treat refractory B-cell non-Hodgkin lymphoma. From early reports to multicenter and global studies the clinical efficacy has been consistent in refractory patients including poor risk B-cell lymphomas. With the FDA approval of axicabtagene ciloleucel, the imminent approval of other CAR T-cell constructs for lymphoma is expected.

Further efforts will be directed towards improving efficacy of CAR T cells by understanding mechanism of CAR T failure and improving safety. As up-regulation o PD-1/PD-L1 pathway is a well-known mechanism it is expected that combination therapy with PD-1 inhibitors may enhance CAR T cell activity [118]. Targeted agents such as ibrutinib and idelalisib could also have a role in improving the CAR T cell efficacy by increasing its proliferative capacity and reversing T-cell dysfunction [119,120]. Utilization of next generation CAR T-cell products such as third generation CAR T cells or bi-specific CAR T cells represent another approach of improving CAR T cell therapy efficacy.

Finally, as the role of CAR T cells in refractory B-cell NHL is becoming standard, it will be important to evaluate the potential role in different settings. Clinical trials testing CAR T cell therapy in primary induction failure for DLBCL, comparing CAR T cell therapy to salvage chemotherapy and autologous HCT, or even testing the therapy for high risk patients in the frontline setting.

Practice points.

  • CAR T cell therapy fills a previously unmet need to for refractory non-Hodgkin’s lymphoma

  • Several anti-CD19 CAR T cell constructs have been studied in multicenter pivotal studies without enough data to compare efficacy

  • CAR T cell related toxicity remains a clinical challenge. This therapy should be limited to specialized center with experience in cell therapies and high-risk immunotherapy.

Research agenda.

  • New CAR T cell products in order to improve efficacy are currently being tested

  • The addition of drugs that target inhibitory immune mechanisms (PD-1/PD-L1), restore immune dysfunction or target the tumor microenvironment will be investigated

  • The role CAR T cell therapy as a second line agent and as up-front therapy for high risk disease may challenge current standard of care approaches

Footnotes

Conflicts of interest

Julio C. Chavez: Kite Pharma (Advisory Board, Speakers Bureau), Novartis (Advisory Board), Genentech (Speakers Bureau), Frederick F. Locke: Kite Pharma (Scientific Advisor), Cellular Biomedicine Group (Consultant).

References

  • [1].Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016;127(20):2375–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Siegel RL, Miller KD, Jemal A. Cancer statistics. CA Cancer J Clin. 2015;65(1):5–29. [DOI] [PubMed] [Google Scholar]
  • [3].Al-Hamadani M, Habermann TM, Cerhan JR, Macon WR, Maurer MJ, Go RS. 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(9):790–5. [DOI] [PubMed] [Google Scholar]
  • [4].Brentjens RJ, Latouche JB, Santos E, Marti F, Gong MC, Lyddane C, et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat Med 2003;9(3):279–86. [DOI] [PubMed] [Google Scholar]
  • [5].Geldres C, Savoldo B, Dotti G. Chimeric antigen receptor-redirected T cells return to the bench. Semin Immunol 2016;28(1):3–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Brudno JN, Kochenderfer JN. Chimeric antigen receptor T-cell therapies for lymphoma. Nat Rev Clin Oncol 2018;15:31–46. [DOI] [PubMed] [Google Scholar]
  • [7].Kochenderfer JN, Feldman SA, Zhao Y, Xu H, Black MA, Morgan RA, et al. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J Immunother 2009;32(7):689–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Sadelain M. CAR therapy: the CD19 paradigm. J Clin Invest 2015;125(9):3392–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Scheuermann RH, Racila E. CD19 antigen in leukemia and lymphoma diagnosis and immunotherapy. Leuk Lymphoma 1995;18(5–6):385–97. [DOI] [PubMed] [Google Scholar]
  • [10].Lim SH, Esler WV, Zhang Y, Zhang J, Periman PO, Burris C, et al. B-cell depletion for 2 years after autologous stem cell transplant for NHL induces prolonged hypogammaglobulinemia beyond the rituximab maintenance period. Leuk Lymphoma 2008;49(1):152–3. [DOI] [PubMed] [Google Scholar]
  • [11].International Non-Hodgkin’s Lymphoma Prognostic Factors P. A predictive model for aggressive non-Hodgkin’s lymphoma. N Engl J Med 1993;329(14):987–94. [DOI] [PubMed] [Google Scholar]
  • [12].Sehn LH, Berry B, Chhanabhai M, Fitzgerald C, Gill K, Hoskins P, et al. The revised International Prognostic Index (R-IPI) is a better predictor of outcome than the standard IPI for patients with diffuse large B-cell lymphoma treated with R-CHOP. Blood 2007;109(5):1857–61. [DOI] [PubMed] [Google Scholar]
  • [13].Zhou Z, Sehn LH, Rademaker AW, Gordon LI, Lacasce AS, Crosby-Thompson A, et al. An enhanced International Prognostic Index (NCCN-IPI) for patients with diffuse large B-cell lymphoma treated in the rituximab era. Blood 2014;123(6):837–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].El-Galaly TC, Villa D, Alzahrani M, Hansen JW, Sehn LH, Wilson D, et al. Outcome prediction by extranodal involvement, IPI, R-IPI, and NCCN-IPI in the PET/CT and rituximab era: a Danish-Canadian study of 443 patients with diffuse-large B-cell lymphoma. Am J Hematol 2015;90(11):1041–6. [DOI] [PubMed] [Google Scholar]
  • [15].Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 2000;403(6769):503–11. [DOI] [PubMed] [Google Scholar]
  • [16].Lenz G, Wright G, Dave SS, Xiao W, Powell J, Zhao H, et al. Stromal gene signatures in large-B-cell lymphomas. N Engl J Med 2008;359(22):2313–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Young RM, Wu T, Schmitz R, Dawood M, Xiao W, Phelan JD, et al. Survival of human lymphoma cells requires B-cell receptor engagement by self-antigens. Proc Natl Acad Sci U S A 2015;112(44):13447–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Pasqualucci L, Trifonov V, Fabbri G, Ma J, Rossi D, Chiarenza A, et al. Analysis of the coding genome of diffuse large B-cell lymphoma. Nat Genet 2011;43(9):830–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Hans CP, Weisenburger DD, Greiner TC, Gascoyne RD, Delabie J, Ott G, et al. Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood 2004;103(1):275–82. [DOI] [PubMed] [Google Scholar]
  • [20].Choi WW, Weisenburger DD, Greiner TC, Piris MA, Banham AH, Delabie J, et al. A new immunostain algorithm classifies diffuse large B-cell lymphoma into molecular subtypes with high accuracy. Clin Cancer Res. 2009;15(17):5494–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Visco C, Li Y, Xu-Monette ZY, Miranda RN, Green TM, Li Y, et al. Comprehensive gene expression profiling and immunohistochemical studies support application of immunophenotypic algorithm for molecular subtype classification in diffuse large B-cell lymphoma: a report from the International DLBCL Rituximab-CHOP Consortium Program Study. Leukemia 2012;26(9):2103–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Gutierrez-Garcia G, Cardesa-Salzmann T, Climent F, Gonzalez-Barca E, Mercadal S, Mate JL, et al. Gene-expression profiling and not immunophenotypic algorithms predicts prognosis in patients with diffuse large B-cell lymphoma treated with immunochemotherapy. Blood 2011;117(18):4836–43. [DOI] [PubMed] [Google Scholar]
  • [23].Coutinho R, Clear AJ, Owen A, Wilson A, Matthews J, Lee A, et al. Poor concordance among nine immunohistochemistry classifiers of cell-of-origin for diffuse large B-cell lymphoma: implications for therapeutic strategies. Clin Cancer Res. 2013;19(24):6686–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Barrans SL, Crouch S, Care MA, Worrillow L, Smith A, Patmore R, et al. Whole genome expression profiling based on paraffin embedded tissue can be used to classify diffuse large B-cell lymphoma and predict clinical outcome. Br J Haematol 2012;159(4):441–53. [DOI] [PubMed] [Google Scholar]
  • [25].Scott DW, Wright GW, Williams PM, Lih CJ, Walsh W, Jaffe ES, et al. Determining cell-of-origin subtypes of diffuse large B-cell lymphoma using gene expression in formalin-fixed paraffin-embedded tissue. Blood 2014;123(8):1214–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Scott DW, Mottok A, Ennishi D, Wright GW, Farinha P, Ben-Neriah S, et al. Prognostic significance of diffuse large B-Cell lymphoma cell of origin determined by digital gene expression in formalin-fixed paraffin-embedded tissue biopsies. J Clin Oncol 2015;33(26):2848–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Adhikary S, Eilers M. Transcriptional regulation and transformation by Myc proteins. Nat Rev Mol Cell Biol 2005;6(8):635–45. [DOI] [PubMed] [Google Scholar]
  • [28].Savage KJ, Johnson NA, Ben-Neriah S, Connors JM, Sehn LH, Farinha P, et al. MYC gene rearrangements are associated with a poor prognosis in diffuse large B-cell lymphoma patients treated with R-CHOP chemotherapy. Blood 2009;114(17):3533–7. [DOI] [PubMed] [Google Scholar]
  • [29].Aukema SM, Siebert R, Schuuring E, van Imhoff GW, Kluin-Nelemans HC, Boerma EJ, et al. Double-hit B-cell lymphomas. Blood 2011;117(8):2319–31. [DOI] [PubMed] [Google Scholar]
  • [30].Barrans S, Crouch S, Smith A, Turner K, Owen R, Patmore R, 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(20):3360–5. [DOI] [PubMed] [Google Scholar]
  • [31].Horn H, Ziepert M, Becher C, Barth TF, Bernd HW, Feller AC, et al. MYC status in concert with BCL2 and BCL6 expression predicts outcome in diffuse large B-cell lymphoma. Blood 2013;121(12):2253–63. [DOI] [PubMed] [Google Scholar]
  • [32].Johnson NA, Slack GW, Savage KJ, Connors JM, Ben-Neriah S, Rogic S, 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(28):3452–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Green TM, Young KH, Visco C, Xu-Monette ZY, Orazi A, Go RS, et al. Immunohistochemical double-hit score is a strong predictor of outcome in patients with diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J Clin Oncol 2012;30(28):3460–7. [DOI] [PubMed] [Google Scholar]
  • [34].Ye Q, Xu-Monette ZY, Tzankov A, Deng L, Wang X, Manyam GC, et al. Prognostic impact of concurrent MYC and BCL6 rearrangements and expression in de novo diffuse large B-cell lymphoma. Oncotarget 2016;7(3):2401–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Petrich AM, Gandhi M, Jovanovic B, Castillo JJ, Rajguru S, Yang DT, et al. Impact of induction regimen and stem cell transplantation on outcomes in double-hit lymphoma: a multicenter retrospective analysis. Blood 2014;124(15):2354–61. [DOI] [PubMed] [Google Scholar]
  • [36].Oki Y, Noorani M, Lin P, Davis RE, Neelapu SS, Ma L, et al. Double hit lymphoma: the MD Anderson Cancer Center clinical experience. Br J Haematol 2014;166(6):891–901. [DOI] [PubMed] [Google Scholar]
  • [37].Howlett C, Snedecor SJ, Landsburg DJ, Svoboda J, Chong EA, Schuster SJ, et al. Front-line, dose-escalated immunochemotherapy is associated with a significant progression-free survival advantage in patients with double-hit lymphomas: a systematic review and meta-analysis. Br J Haematol 2015;170(4):504–14. [DOI] [PubMed] [Google Scholar]
  • [38].Cuccuini W, Briere J, Mounier N, Voelker HU, Rosenwald A, Sundstrom C, et al. MYC+ diffuse large B-cell lymphoma is not salvaged by classical R-ICE or RDHAP followed by BEAM plus autologous stem cell transplantation. Blood 2012;119(20):4619–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Puvvada SD, Stiff PJ, Leblanc M, Cook JR, Couban S, Leonard JP, et al. Outcomes of MYC-associated lymphomas after R-CHOP with and without consolidative autologous stem cell transplant: subset analysis of randomized trial intergroup SWOG S9704. Br J Haematol 2016;174(5):686–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Herrera AF, Mei M, Low L, Kim HT, Griffin GK, Song JY, et al. Relapsed or refractory double-expressor and double-hit lymphomas have inferior progression-free survival after autologous stem-cell transplantation. J Clin Oncol 2017;35(1):24–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Landsburg DJ, Falkiewicz MK, Maly J, Blum KA, Howlett C, Feldman T, et al. Outcomes of patients with double-hit lymphoma who achieve first Complete remission. J Clin Oncol 2017;35(20):2260–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Coiffier B, Thieblemont C, Van Den Neste E, Lepeu G, Plantier I, Castaigne S, 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(12):2040–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Crump M. Management of relapsed diffuse large B-cell lymphoma. Hematol Oncol Clin North Am. 2016;30(6):1195–213. [DOI] [PubMed] [Google Scholar]
  • [44].Gisselbrecht C, Glass B, Mounier N, Singh Gill D, Linch DC, Trneny M, et al. Salvage regimens with autologous transplantation for relapsed large B-cell lymphoma in the rituximab era. J Clin Oncol 2010;28(27):4184–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Maurer MJ, Ghesquieres H, Jais JP, Witzig TE, Haioun C, Thompson CA, et al. Event-free survival at 24 months is a robust end point for disease-related outcome in diffuse large B-cell lymphoma treated with immunochemotherapy. J Clin Oncol 2014;32(10):1066–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Crump M, Kuruvilla J, Couban S, MacDonald DA, Kukreti V, Kouroukis CT, et al. Randomized comparison of gemcitabine, dexamethasone, and cisplatin versus dexamethasone, cytarabine, and cisplatin chemotherapy before autologous stem-cell transplantation for relapsed and refractory aggressive lymphomas: NCICCTG LY.12. J Clin Oncol 2014;32(31):3490–6. [DOI] [PubMed] [Google Scholar]
  • [47].Van Den Neste E, Schmitz N, Mounier N, Gill D, Linch D, Trneny M, et al. Outcome of patients with relapsed diffuse large B-cell lymphoma who fail second-line salvage regimens in the International CORAL study. Bone Marrow Transplant 2016;51(1):51–7. [DOI] [PubMed] [Google Scholar]
  • [48].Van Den Neste E, Schmitz N, Mounier N, Gill D, Linch D, Trneny M, et al. Outcomes of diffuse large B-cell lymphoma patients relapsing after autologous stem cell transplantation: an analysis of patients included in the CORAL study. Bone Marrow Transplant 2017;52(2):216–21. [DOI] [PubMed] [Google Scholar]
  • [49].Nagle SJ, Woo K, Schuster SJ, Nasta SD, Stadtmauer E, Mick R, et al. Outcomes of patients with relapsed/refractory diffuse large B-cell lymphoma with progression of lymphoma after autologous stem cell transplantation in the rituximab era. Am J Hematol 2013;88(10):890–4. [DOI] [PubMed] [Google Scholar]
  • [50].Crump M, Neelapu SS, Farooq U, Van Den Neste E, Kuruvilla J, Westin J, et al. Outcomes in refractory diffuse large B-cell lymphoma: results from the international SCHOLAR-1 study. Blood 2017;130(16):1800–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Costa LJ, Maddocks K, Epperla N, Reddy NM, Karmali R, Umyarova E, 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(2):161–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Jensen MC, Popplewell L, Cooper LJ, DiGiusto D, Kalos M, Ostberg JR, 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(9):1245–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA, 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(20):4099–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Savoldo B, Ramos CA, Liu E, Mims MP, Keating MJ, Carrum G, et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest 2011;121(5):1822–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Kochenderfer JN, Dudley ME, Kassim SH, Somerville RP, Carpenter RO, Stetler-Stevenson M, 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(6):540–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Kochenderfer JN, Somerville RPT, Lu T, Yang JC, Sherry RM, Feldman SA, 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(10):2245–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Kochenderfer JN, Somerville RPT, Lu T, Shi V, Bot A, Rossi J, 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(16):1803–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Sommermeyer D, Hudecek M, Kosasih PL, Gogishvili T, Maloney DG, Turtle CJ, et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia 2016;30(2):492–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Turtle CJ, Hanafi LA, Berger C, Hudecek M, Pender B, Robinson E, 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(355). 355ra116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Schuster SJ, Svoboda J, Dwivedy Nasta S, Porter DL, Chong EA, Landsburg DJ, 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(23). 183-. [Google Scholar]
  • [61].Schuster SJ, Svoboda J, Nasta SD, Chong EA, Winchell N, Landsburg DJ, 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(22). 3026-.28034869 [Google Scholar]
  • [62].Schuster SJ, Svoboda J, Chong EA, Nasta SD, Mato AR, Anak O, et al. Chimeric antigen receptor T cells in refractory B-Cell lymphomas. N Engl J Med 2017;377(26):2545–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Locke FL, Neelapu SS, Bartlett NL, Siddiqi T, Chavez JC, Hosing CM, 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(1):285–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Locke FL, Neelapu SS, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, 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(15_suppl). 7512-. [Google Scholar]
  • [65].Schuster S, Bishop MR, Tam C, Waller EK, Borchmann P, McGuirk J, 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. Hematol Oncol 2017;35(S2). 27-. [Google Scholar]
  • [66].Schuster SJ, Bishop MR, Tam CS, Waller EK, Borchmann P, McGuirk JP, 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]
  • [67].Abramson JS, Palomba ML, Gordon LI, Lunning MA, Arnason JE, Forero-Torres A, 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). J Clin Oncol 2017;15:7513. [Google Scholar]
  • [68].Abramson J, Palomba ML, Gordon L, Lunning M, Arnason J, Wang M, 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-.26177633 [Google Scholar]
  • [69].Abramson JS, Palomba ML, Gordon LI, Lunning MA, Arnason JE, Wang M, 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-.28584136 [Google Scholar]
  • [70].Maloney DG, Abramson JS, Palomba ML, Gordon LI, Lunning MA, Arnason JE, 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]
  • [71].Tan D, Horning SJ, Hoppe RT, Levy R, Rosenberg SA, Sigal BM, et al. Improvements in observed and relative survival in follicular grade 1–2 lymphoma during 4 decades: the Stanford University experience. Blood 2013;122(6):981–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Martin P, Byrtek M, Dawson K, Ziemiecki R, Friedberg JW, Cerhan JR, et al. Patterns of delivery of chemoimmunotherapy to patients with follicular lymphoma in the United States: results of the National LymphoCare Study. Cancer 2013;119(23):4129–36. [DOI] [PubMed] [Google Scholar]
  • [73].Rummel MJ, Niederle N, Maschmeyer G, Banat GA, von Grunhagen U, Losem C, 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(9873):1203–10. [DOI] [PubMed] [Google Scholar]
  • [74].Flinn IW, van der Jagt R, Kahl BS, Wood P, Hawkins TE, Macdonald D, 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(19):2944–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Solal-Celigny P, Roy P, Colombat P, White J, Armitage JO, Arranz-Saez R, et al. Follicular lymphoma international prognostic index. Blood 2004;104(5):1258–65. [DOI] [PubMed] [Google Scholar]
  • [76].Federico M, Bellei M, Marcheselli L, Luminari S, Lopez-Guillermo A, Vitolo U, 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(27):4555–62. [DOI] [PubMed] [Google Scholar]
  • [77].Shi Q, Flowers CR, Hiddemann W, Marcus R, Herold M, Hagenbeek A, et al. Thirty-month Complete response as a surrogate end point in first-line follicular lymphoma therapy: an individual patient-level analysis of multiple randomized trials. J Clin Oncol 2017;35(5):552–60. JCO2016708651. [DOI] [PubMed] [Google Scholar]
  • [78].Casulo C, Byrtek M, Dawson KL, Zhou X, Farber CM, Flowers CR, 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(23):2516–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Maurer MJ, Bachy E, Ghesquieres H, Ansell SM, Nowakowski GS, Thompson CA, et al. Early event status informs subsequent outcome in newly diagnosed follicular lymphoma. Am J Hematol 2016;91(11):1096–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Press OW, Unger JM, Rimsza LM, Friedberg JW, LeBlanc M, Czuczman MS, et al. Phase III randomized intergroup trial of CHOP plus rituximab compared with CHOP chemotherapy plus (131)iodine-tositumomab for previously untreated follicular non-Hodgkin lymphoma: SWOG S0016. J Clin Oncol 2013;31(3):314–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Provencio M, Royuela A, Torrente M, Pollan M, Gomez-Codina J, Sabin P, et al. Prognostic value of event-free survival at 12 and 24 months and long-term mortality for non-Hodgkin follicular lymphoma patients: a study report from the Spanish Lymphoma Oncology Group. Cancer 2017;123(19):3709–16. [DOI] [PubMed] [Google Scholar]
  • [82].Horn H, Schmelter C, Leich E, Salaverria I, Katzenberger T, Ott MM, et al. Follicular lymphoma grade 3B is a distinct neoplasm according to cytogenetic and immunohistochemical profiles. Haematologica 2011;96(9):1327–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Dave SS, Wright G, Tan B, Rosenwald A, Gascoyne RD, Chan WC, et al. Prediction of survival in follicular lymphoma based on molecular features of tumor-infiltrating immune cells. N Engl J Med 2004;351(21):2159–69. [DOI] [PubMed] [Google Scholar]
  • [84].Carreras J, Lopez-Guillermo A, Fox BC, Colomo L, Martinez A, Roncador G, et al. High numbers of tumor-infiltrating FOXP3-positive regulatory T cells are associated with improved overall survival in follicular lymphoma. Blood 2006;108(9):2957–64. [DOI] [PubMed] [Google Scholar]
  • [85].Glas AM, Knoops L, Delahaye L, Kersten MJ, Kibbelaar RE, Wessels LA, et al. Gene-expression and immunohistochemical study of specific T-cell subsets and accessory cell types in the transformation and prognosis of follicular lymphoma. J Clin Oncol 2007;25(4):390–8. [DOI] [PubMed] [Google Scholar]
  • [86].Morin RD, Johnson NA, Severson TM, Mungall AJ, An J, Goya R, et al. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat Genet 2010;42(2):181–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Morin RD, Mendez-Lago M, Mungall AJ, Goya R, Mungall KL, Corbett RD, et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 2011;476(7360):298–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Pasqualucci L, Dominguez-Sola D, Chiarenza A, Fabbri G, Grunn A, Trifonov V, et al. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature 2011;471(7337):189–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Krysiak K, Gomez F, White BS, Matlock M, Miller CA, Trani L, et al. Recurrent somatic mutations affecting B-cell receptor signaling pathway genes in follicular lymphoma. Blood 2017;129(4):473–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Pastore A, Jurinovic V, Kridel R, Hoster E, Staiger AM, Szczepanowski M, 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(9):1111–22. [DOI] [PubMed] [Google Scholar]
  • [91].Jurinovic V, Kridel R, Staiger AM, Szczepanowski M, Horn H, Dreyling MH, et al. Clinicogenetic risk models predict early progression of follicular lymphoma after first-line immunochemotherapy. Blood 2016;128(8):1112–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, 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(12):2709–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Chong EA, Svoboda J, Nasta SD, Porter DL, Winchell N, Landsburg DJ, 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(22). 1100-. [Google Scholar]
  • [94].Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 2014;6(224). 224ra25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014;371(16):1507–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, et al. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol 2018. January;15(1):47–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 2016;127(26):3321–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014;124(2):188–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-Cell lymphoma. N Engl J Med 2017;377(26):2531–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Heipel M, Smith J, Brown W, Karimi M, Xie B, Li D, et al. Pharmacokinetic, pharmacodynamic and blood analytes associated with clinical response and safety in relapsed/refractory aggressive B-NHL patients treated with JCAR017. Blood 2017;130(Suppl 1). 2835-. [Google Scholar]
  • [101].Klebanoff CA, Khong HT, Antony PA, Palmer DC, Restifo NP. Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol 2005;26(2):111–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Yao X, Ahmadzadeh M, Lu YC, Liewehr DJ, Dudley ME, Liu F, et al. Levels of peripheral CD4(+)FoxP3(+) regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer. Blood 2012;119(24):5688–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, Hudecek M, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest 2016;126(6):2123–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Swanson C, Do T, Merrigan S, Lonning S, Merriam K, Prentiss J, et al. Predicting clinical response and safety of JCAR017 in B-NHL patients: potential importance of tumor microenvironment biomarkers and CAR T-cell tumor infiltration. Blood 2017;130(Suppl 1). 194-. [Google Scholar]
  • [105].Sotillo E, Barrett DM, Black KL, Bagashev A, Oldridge D, Wu G, et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 2015;5(12):1282–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Till BG, Jensen MC, Wang J, Chen EY, Wood BL, Greisman HA, et al. Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood 2008;112(6):2261–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Wang Y, Zhang WY, Han QW, Liu Y, Dai HR, Guo YL, 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(2):160–75. [DOI] [PubMed] [Google Scholar]
  • [108].Till BG, Jensen MC, Wang J, Qian X, Gopal AK, Maloney DG, 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(17):3940–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].James SE, Greenberg PD, Jensen MC, Lin Y, Wang J, Till BG, et al. Antigen sensitivity of CD22-specific chimeric TCR is modulated by target epitope distance from the cell membrane. J Immunol 2008;180(10):7028–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Shah NN, Stetler-Stevenson M, Yuan CM, Shalabi H, Yates B, Delbrook C, 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(22). 650-.27281794 [Google Scholar]
  • [111].Ramos CA, Savoldo B, Torrano V, Ballard B, Zhang H, Dakhova O, et al. Clinical responses with T lymphocytes targeting malignancy-associated kappa light chains. J Clin Invest 2016;126(7):2588–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Balakrishnan A, Goodpaster T, Randolph-Habecker J, Hoffstrom BG, Jalikis FG, Koch LK, et al. Analysis of ROR1 protein expression in human cancer and normal tissues. Clin Cancer Res. 2017;23(12):3061–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Berger C, Sommermeyer D, Hudecek M, Berger M, Balakrishnan A, Paszkiewicz PJ, et al. Safety of targeting ROR1 in primates with chimeric antigen receptor-modified T cells. Cancer Immunol Res. 2015;3(2):206–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].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. 2016;4(6):498–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Qin H, Haso W, Nguyen SM, Fry TJ. Preclinical development of bispecific chimeric antigen receptor targeting both CD19 and CD22. Blood 2015;126(23). 4427-. [Google Scholar]
  • [116].Lamers CH, Willemsen R, van Elzakker P, van Steenbergen-Langeveld S, Broertjes M, Oosterwijk-Wakka J, et al. Immune responses to transgene and retroviral vector in patients treated with ex vivo-engineered T cells. Blood 2011;117(1):72–82. [DOI] [PubMed] [Google Scholar]
  • [117].Sommermeyer D, Hill T, Shamah SM, Salter AI, Chen Y, Mohler KM, et al. Fully human CD19-specific chimeric antigen receptors for T-cell therapy. Leukemia 2017;31(10):2191–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Cherkassky L, Morello A, Villena-Vargas J, Feng Y, Dimitrov DS, Jones DR, et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J Clin Invest 2016;126(8):3130–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Fraietta JA, Beckwith KA, Patel PR, Ruella M, Zheng Z, Barrett DM, et al. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood 2016;127(9):1117–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Bowers JS, Majchrzak K, Nelson MH, Aksoy BA, Wyatt MM, Smith AS, et al. PI3Kdelta inhibition enhances the antitumor fitness of adoptively transferred CD8(+) T cells. Front Immunol 2017;8:1221. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES