CAR T Cells in Chronic Lymphocytic Leukemia

Ugo Testa1, Elvira Pelosi1, Germana Castelli1, Alberto Fresa2,3, and Luca Laurenti2,3.

1 Istituto Superiore di Sanità, Roma, Italy.
2 Dipartimento di Diagnostica per  Immagini,  Radioterapia  Oncologica  ed  Ematologia,  Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy. Sezione Di Ematologia. Roma, Italy.
3 Dipartimento di Scienze Radiologiche Ed Ematologiche, Università Cattolica Del Sacro Cuore, Roma, Italy.

Correspondence to: Ugo Testa. E-mail: ugo.testa@iss.it

Published: May 01, 2024
Received: April 02, 2024
Accepted: April 18, 2024
Mediterr J Hematol Infect Dis 2024, 16(1): e2024045 DOI 10.4084/MJHID.2024.045

This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(
https://creativecommons.org/licenses/by-nc/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

The treatment outcomes of patients with chronic lymphocytic leukemia (CLL) have considerably improved with the introduction of targeted therapies based on Bruton kinase inhibitors (BTKIs), venetoclax, and anti-CD20 monoclonal antibodies. However, despite these consistent improvements, patients who become resistant to these agents have poor outcomes and need new and more efficacious therapeutic strategies.
Among these new treatments, a potentially curative approach consists of the use of chimeric antigen receptor T (CAR-T) cell therapy, which achieved remarkable success in various B-cell malignancies, including B-cell Non-Hodgkin Lymphomas (NHLs) and B-acute lymphoblastic Leukemia (ALL). However, although CAR-T cells were initially used for the treatment of CLL, their efficacy in CLL patients was lower than in other B-cell malignancies. This review analyses possible mechanisms of these failures, highlighting some recent developments that could offer the perspective of the incorporation of CAR-T cells in treatment protocols for relapsed/refractory CLL patients.



Introduction

Chronic lymphocytic leukemia (CLL) is the most frequent leukemia in adult subjects in Western countries, with a mean age at diagnosis around 69 years.[1] At the cellular level, this leukemia is characterized by the progressive accumulation of mature CD5-positive B-lymphocytes in the peripheral blood, bone marrow, and secondary lymphoid organs.[2] The clinical stratification of CLL patients currently relies on the Rai and Binet classification: low-risk (Rai 0/Binet A), intermediate-risk (Rai I and II or Binet B), and high-risk (Rai III and IV or Binet C). The molecular stratification relies on molecular prognostic markers that include cytogenetic abnormalities and the assessment of the mutational status of immunoglobulin heavy-chain variable (IGVH) and TP53 genes. High-risk markers are represented by the presence of TP53 mutations, del(17p), a complex karyotype, or unmutated IGVH.[2]
The treatment algorithm of CLL radically changed over the past few years, becoming almost completely chemo-free. Therefore, according to the current treatment recommendations, patients with the symptomatic disease receive a covalent Bruton tyrosine kinase inhibitor (i.e., ibrutinib, acalabrutinib, zanubrutinib), the BCL-2 inhibitor venetoclax +/- anti-CD20 monoclonal antibody, or a combination of the classes.[2,3] In relapsed/refractory (R/R) patients, exposure to one class of drug does not prevent the patient from being treated with the other one, with effective results.[1] However, the outcomes of double-resistant patients are poor, with an overall survival of only a few months.[4-7]
Therefore, there is an absolute need to develop novel therapies for these double-resistant patients who have a dismal prognosis. Concerning cellular therapy, slightly better results were obtained from allogeneic stem cell transplantation.[7,8] Studies carried out in the last years have suggested that chimeric antigen receptors (CAR) designed to target T cells to antigens expressed on CLL cells may offer a therapeutic opportunity for these patients. In this review, we explored and analyzed the current available evidence on the use of CAR-T cell therapy in CLL patients. The most significant results of trials with CAR-T in CLL and Richter's transformation are summarized in Table 1.


Table 1
Table 1. CAR-T cell therapy in chronic lymphocytic leukemia. The most relevant clinical studies are reported.


CD19-targeted CAR-T Cells in CLL

CD19-targeted CAR-T cell therapy was successfully experimented with for the treatment of several B-cell malignancies, including B-cell lymphomas and B-acute lymphoblastic leukemia.[9-19] Four CAR-T cell products are commercially available and approved by the FDA for clinical use: Axicabtagene ciloleucel (Axi-Cel), Brexucabtagene-autoleucel (Brexu-Cel), Tisagenlecleucel (Tisa-Cel) and Lisocabtagene maraleucel (Liso-Cel). These CAR-T products were also evaluated in CLL patients.
Liso-Cel. Liso-Cel is a second-generation anti-CD19 CAR-T cell that utilizes the 4-1BB costimulatory domain and is produced from separated subsets of CD4- and CD8-positive cells to make a 1:1 CD4/CD8 ratio in CAR-T.[20]
The first study involving the investigation of Liso-Cel in CLL patients was performed on 24 R/R CLL patients, in large part with high-risk features (such as complex karyotype and/or 17p deletion).[21] The overall response rate (ORR) was 74%, with 21% complete response (CR) and 53% partial response (PR).[21] Recently, long-term follow-up of this study was reported, including the analysis of phase I and II of this study; the study included a total of 49 patients with R/R CLL and 10 patients with Richter’s transformation.[22] Patients received CAR-T cell therapy without (30 patients) or with concurrent ibrutinib treatment (19 patients); patients were evaluated on day +28 for response by standard criteria, and their measurable residual disease (MRD) status was assessed by multiparameter flow cytometry (MFC) and IGH next-generation sequencing.[22] Of the enrolled patients, 96% had complex karyotype and/or 17p deletion, and 94% were ibrutinib-refractory. ORR and CRR were 70% and 17%, respectively, and the median duration of response (mDoR) was 21.3 months; 5-year overall survival (OS) and progression-free survival (PFS) were 35% and 21%, respectively. MRD by MFC was observed in 75% of patients, and 72% of these patients were negative for NGS; in uMRD patients by NGS, the mDoR was 55.9 months.[22]
In 2018, Siddiqi et al. reported the first results of the monotherapy section of the TRASCEND CLL 004 phase I/II clinical trial, which initially involved 10 R/R CLL patients.[23] The patients with standard risk received 3 and those with high risk 2 prior lines of therapy, including a Bruton’s tyrosine kinase inhibitor (BTKi); after lymphodepletion, the patients received a single-infusion of Liso-Cel at either dosage 1 (DL1, 5x107CAR-T cells) or dosage 2 (DL2, 1x108CAR-T cells).[23] At 30 days post-CAR-T cell infusion, 75% of patients displayed an OR, with 50% of CR; all patients with CR remained with a negative MRD status at 3 months post-dose.[4] In 2022, the same authors reported the results of phase I of the TRASCEND CLL 004 study, involving a total of 23 R/R CLL patients, including 100% of patients who had prior ibrutinib and 65% venetoclax treatment; 83% of these patients had high-risk features, including TP53 mutations and del (17p).[24] In this study, 74% of patients had cytokine release syndrome (CRS, 9% of grade 3) and 39% of neurological events (22% of grade 3). ORR was 42.9%, and CR was 11%; the patients who achieved MRD negativity were 75% and 60% in peripheral blood and bone marrow, respectively.[24] The results of this study have represented the basis for a phase II study at 1x108CAR-T cell dosage. A more recent report of this phase I/II study reported the results of 117 CLL patients.[25] In the patients treated with dosage level 2 (DL2, 1x10
8CAR-T cells), 88% (95/108) were BTKi refractory, 78% (89/108) were venetoclax refractory, 61% (66/108) were double-refractory. The efficacy evaluation at DL2 showed an ORR of 42.9% with 18.4% CR; the median DoR was 35.3 months, with a median follow-up of 19.7 months.[25] The same efficacy was reported in the evaluable double-refractory patients [ORR was 42% (21/49), with 18% (9/49) of CR, median DoR 35.25 months]. Undetectable MRD was obtained in 64% of double refractory patients in peripheral blood and 59% in bone marrow. The median duration of response among patients with CR was not reached. In exploratory analyses of undetectable MRD and progression-free survival, PFS was 26.2 months in patients with uMRD and 2.8 months in those with detectable MRD in blood. Grade 3 CRS occurred in 8.5% of cases, and grade 3 neurological events in 17.9% of cases.[25] With a longer follow-up (median follow-up of 23.5 months), Lico-Cel continues to demonstrate durable CR, high MRD rates, and a manageable safety profile in this population of heavily pretreated CLL patients.[26]
Tisa-Cel.  Tisa-Cel is  a  second-generation  anti-CD19 CAR-T cell with 4-1BB costimulatory domain and CD8a hinge region.[27]
The initial study carried out using autologous T cell transduced with a CD19-directed CAR (CTL-109, Tisa-Cel) reported the results observed in 14 R/R CLL patients: the ORR was 57%, with 4 CR and 4PR.[28] A final evaluation of this study involved the treatment of 38 R/R CLL patients with CTL109, showing an ORR of 44%, with 28% of CR.[29] Patients achieving a CR showed an OS longer than those who did not, with a mOS not reached vs 64 months.[8] The median PFS was 40.2 months in patients with CR, compared to 1 month in those without CR.[29]
Interestingly, Melenhorst et al. reported the cellular and molecular analysis of two CLL patients exhibiting long-term remission after a CTL 109 infusion performed in 2010.[9] The anti-leukemia activity of anti-CD19 CTL 109 cells displayed two distinct phases: an initial phase characterized by CD8+or CD4-CD8-Helioshi γδ CAR-T cells, followed by the predominance of proliferative CD4
+CAR-T cells in the ensuing years.[30] Surprisingly, CD4+T cells seem responsible for mediating anti-CD19 cytotoxic activity; functional characterization of these cells, as well as upregulation of antigen-mediated signaling pathways and upregulation of G 2MK, G2MA, and PRF1, support this view.[30]
Brexu-Cel. Brexu-Cel is a second-generation anti-CD19 CAR-T cell that utilizes the CD28 costimulatory domain and CD28 hinge region; an additional step is introduced in the manufacturing of these CAR-T cells to remove malignant cells from the leukapheresis product.[31]
The ZUMA-8 phase I/II clinical trial explored the safety and efficacy of Brexu-Cel (KTE-19), anti-CD19 CAR-T cells. In phase I, the enrolment of 12-18 patients was planned to assess dose-limiting toxicities, while in phase II, 60 patients would be enrolled to assess safety and efficacy.[32] Phase I results of the ZUMA-8 trial involving 15 R/R CLL patients have involved four cohorts of patients: cohorts 1 and 2 were treated with 1x106 and 2x10
6 KTE-19 cells/Kg, cohort 3 involved the enrolment of low-tumor burden patients and cohort 4 any patients.[33] The enrolled patients were heavily pretreated. Seven out of fifteen patients exhibited an objective response, with 3 CR; two of these patients with a low tumor burden had a CR.[33] Significant CAR-T cell expansion occurred in 4 patients, mostly in those with low tumor burden.[33]
Axi-Cel. Axi-Cel is a second-generation anti-CD19 CAR-T cell utilizing CD28 costimulatory domain and CD28 hinge region.[34] Axi-Cel, autologous anti-CD19 CAR-T cells have been explored in a few R/R CLL patients. An initial study by Kochenderfer et al. also included 4 R/R CLL patients.[35] A later study included 8 R/R CLL patients and explored the long-term effects of Axi-Cel CAR-T cell therapy. The ORR was 88%, with 63% of CR and 25% of PR; 3/5 of the patients who achieved a CR maintained a response ≥3 years.[36] The median duration of ongoing responses was 82 months; median EFS was 40.5 months.[36]
CAR-T GLPG 5201. A recent study reported the first results obtained using GLPC 5201, a second-generation anti-CD19/4-1BB CAR-T product, administered as a fresh product in a single dose, without the need for cryopreservation within 7 days of apheresis.[36] Seven patients were enrolled in the ongoing phase I study; all these 7 patients were diagnosed as R/R CLL, and 4/7 had Richter's transformation (RT); no grade 3-4 CRS or neurologic events were observed; 7/7 patients responded to the treatment, with 6/7 CRs; responding patients remained in CR at a 7-month follow-up.[37] These results, although still preliminary, are considered very promising.
A more extensive report of this study was presented at the ASH Meeting 2023 and involved a total of 12 patients: 5 with R/R CLL and 7 with RT.[38] ORR and CRR were 86% and 7!% for RT patients.[38] Robust expansion of CAR-T cells was observed, with detectable CAR-T cells up to 9 months after CAR-T cell infusion.[38] Infused CAR-T cells exhibited a preserved early memory phenotype for CD4
+ and CD8+ CAR-T cells.[38]
Varnicambtagene autoleucel. Varnicambtagene autoleucel (Var-Cel), an academic anti-CD19 CAR product manufactured by the Hospital Clinic of Barcelona, Spain, was used primarily for the treatment of non-Hodgkin lymphoma in the CART19-BE-01 trial. Recently, the safety profile and the efficacy of Var-Cel were explored in 20 R/R CLL patients, including 63% of patients with RT.[39] Var-Cel manufacturing was performed in a median of 8 days; with a median follow-up of 8.4 months, the ORR was 83%, with 83% of patients achieving an MRD negativity; CR in BM and PB was 67%, achieving CR at the level of extramedullary sites.[39] PFS was shorter in patients with RT and with higher BM infiltration.[39] These preliminary observations support further development of Var-Cel in these high-risk populations of CLL patients.

CD19-targeted CAR-T Cell Therapy in Richter’s Transformation

Richter’s transformation is a relatively rare event involving the development of aggressive B-cell lymphoma in patients with CLL or SLL; the prognosis is dismal, with a short OS.[40] Table 2 reports the results of the trials involving CAR-T use in Richter's transformation.

Table 2
Table 2. CAR-T cell therapy in Richter’s transformation. The most relevant clinical studies are reported.

Benjamini and coworkers reported the analysis of the safety and efficacy of CD19 CAR-T cells in 8 patients with Richter’s transformation generated using a retroviral vector encoding a CAR comprising FMC63 anti-CD19 ScFv, linked to a CD28 costimulatory domain and CD3-zeta intracellular signaling domain.[38] All these patients had a prior history of CLL, and 83% of them had del 17p/TP53 mutation.[41] Patients had at least 3 previous lines of therapy, including ibrutinib and venetoclax treatment.[41] Of these patients, 71% had a CR following treatment with CAR-t cells, and 2 patients proceeded to allo-HSCT.[38] Three patients had grade 3-4 CRS.[41]
Kittai et al. reported a retrospective analysis of 9 patients with Richter's transformation treated with Axi-Cel in the context of an institutional experience of off-label use of these CAR-T cells.[39] Eight of these 9 patients were evaluated for response, and most of them continued treatment with BTK inhibitor.[42] All of these patients displayed a response, with 5 CR and 3 PR.[42] The median follow-up was 6 months: one patient relapsed during these 6 months, while the remaining 7 patients remained in remission.[42]
Bensader et al. have retrospectively analyzed 12 patients with Richter’s transformation treated with either Axi-Cel (5 patients) or Tisa-Cel (7 patients), with 50% ORR and 42 CRs.[43] Of the treated patients, 25% had grade 3 CRS. All the patients included in this analysis were heavily pretreated.
Blackmon reported 7 patients who have been treated with Liso-Cel and relapsed after CAR-T cell therapy with Richter’s syndrome.[41] All these patients had CLL with high-risk factors prior to CAR-T cell therapy (including TP53 mutations, del17p, NOTCH1 mutations, and SF3B1 mutations.[44]

CD19-targeted CAR-T Cell Therapy in Combination with Ibrutinib

Several clinical observations have strongly supported the rationale of associating CAR-T cells with ibrutinib administration: (i) ibrutinib treatment of CLL patients increases in vivo persistence of activated T cells, decreases Treg/CD4+T cell ratio, and decreases the immunosuppressive properties of CLL cells;[45] (ii) previous cycles of ibrutinib therapy improve the in vivo expansion of CD19-directed CAR-T cells, in association with decreased expression of the immunosuppressive molecules PD-1 and CD200 on the membrane of T-lymphocytes;[46] (iii) Ibrutinib administration after CAR-T cell infusion decreases cytokine release syndrome;[47] assays demonstrated enhanced anti-CLL T-cell killing function during ibrutinib-rituximab treatment, including a switch from predominantly CD4+ T-cell: CLL immune synapses at baseline to increased CD8+ lytic synapses on-therapy.[48]
Gauthier et al. have conducted a pilot study to evaluate the safety and feasibility of administering ibrutinib concurrently with CD19 CAR-T cell therapy.[49] Thus, 19 R/R CLL patients, with a median number of 5 prior therapies, have been treated with Liso-Cel and ibrutinib; 13 of these patients received the scheduled ibrutinib treatment and were evaluated for safety and response to the treatment. The 4-week ORR was 83%, with 61% of patients achieving an MRD-negativity in bone marrow, as assessed by NGS.[46] In these patients, the 1-yr OS and PFS were 86% and 59% respectively. Patients treated with ibrutinib and CAR-T cells displayed lower CRS severity and lower serum concentrations of CRS-associated cytokines.[49]
Gill et al. reported the results of a phase II clinical study in which anti-CD19 humanized binding domain CAR-T cells (hu-CART 19) were used in combination with ibrutinib in 19R/R CLL patients not in CR despite ≥6 months of ibrutinib.[50] CRR evaluated at 3 months was 44%; at 12 months, 72% of patients had no measurable MRD.[47] Of 15 patients with undetectable MRD at 6 months, 13 remained in CR at the last follow-up.[50]

Mechanisms of Resistance to CD19 CAR-T Cell Therapy in CLL

As mentioned above, clinical trials of CD19-targeted CAR-T cell therapy have shown a durable antitumor response in a limited proportion of R/R CLL patients, around 20-25%, while most patients are resistant to this treatment.
Several studies have explored the characteristics of CAR-T cells, as well as the genomic abnormalities and other molecular features of CLL that could correlate with response or resistance to CAR-T cells.[51]
Frazetta and coworkers have investigated the transcriptomic profile of CAR-T cells of R/R CLL patients undergoing CAR-T cell therapy: responding patients were enriched in memory-related genes, whereas T cells from non-responders upregulated the expression of genes involved in effector differentiation, glycolysis, exhaustion, and apoptosis.[52] Furthermore, patients with sustained mission showed an elevated frequency of CD27+CD45RO-CD8
+T cells with memory-like characteristics before CAR-T cell generation.[52] An additional feature of functionally active CAR-T cells consisted of the production of STAT3-related cytokines and serum IL6 levels correlated with CAR-T cell expansion.[52]
Resting CD8
+lymphocytes in CLL have an altered mitochondrial profile (increased mitochondrial respiration, membrane potential, and level of reactive oxygen species), which impairs the development of efficacious CAR-T cells in these patients.[53] In line with these observations, CD8+CAR-T cells of CLL patients responding to CAR-T cell therapy have increased mitochondrial mass with respect to non-responders, and this property is correlated with CAR-T cell expansion in vivo.[53]
CAR-T cells harbor an engineered receptor that is delivered through lentiviral vector integration and may modify the cellular genome by insertional mutagenesis. In some patients, retroviral integration within the host genes, such as the TET2 gene[50] or genes involved in cell signaling and chromatin modification pathways,[54] may promote therapeutic T cell proliferation.
As discussed above, non-responding or partially responding CLL patients exhibited marginal or no expansion of their transformed CAR-T cells; in contrast, patients with full response possessed CAR-T cells with pronounced proliferative capacity and sustained persistence.[55] In addition to these observations, the level and the duration of CAR expression on the surface of T cells are a key determinant of clinical efficacy since patients exhibiting complete remission have a permanent CAR expression. In contrast, non-responding patients lost their cell-surface CAR detection at the time of relapse, thus suggesting that CAR extinction at the cell surface is an important mechanism of resistance to therapy.[56] Epigenetic mechanisms seem to be responsible for the extinction of CAR expression; particularly, the bromodomain and extra-terminal (BET) family of chromatin adaptors seem to be involved in this epigenetic silencing of CAR expression.[56] BET protein inhibition decreased TET2 levels and improved the proliferative capacity of exhausted CAR-T cells and their antitumor activity.[56]
The reduced CAR-T cell expansion and persistence may also be attributed to the activation of naturally occurring negative immune checkpoint molecules (such as PD-1, TM-3, LAG-3, and CTLA-4). Agarwal et al. have explored whether the disruption of the co-inhibitory receptors CTLA-4 or PD-1 could restore CAR-T function.[57] CRISPR-Cas9-mediated deletion of CTLA-4 in preclinical models of leukemia and in T-cells from patients with CLL who previously failed CAR-T cell treatment reinvigorates dysfunctional T cells, thus suggesting a strategy for increasing patient responses to CAR-T cell therapy.[57]

CD20-targeted CAR-T Cells in CLL

Only an ongoing phase I clinical trial is evaluating the safety and efficacy of CD20-targeted CAR-T cells in CLL patients.[54] MB-106 is a fully human third-generation CD20-targeted CAR-T product with both CD28 and 4-1BB costimulatory domains; an ongoing phase I/II clinical trial is evaluating the safety and the efficacy of MB-106 in R/R CD20+ B-cell malignancies.[58] The results on the first 16 patients included 1 patient with R/R CLL achieving CR, with MRD-negative status following treatment with MB-106.[58]
Other clinical trials have started the evaluation of bispecific CAR-T cells targeting both CD19 and CD20 antigens. In phase I clinical trial enrolling patients with B-cell lymphomas and CC, it was evaluated the safety and the efficacy of LV20.19, a CAR construct targeting both CD20 and CD19: 3 patients with R/R CLL were treated, with two patients achieving CR and one patient achieving PR.[59]
Another study of tandem CAR-T targeting CD19 and CD20 also included one R/R CLL patient, who achieved a CR to this therapy.[60]
Park et al. reported the initial evaluation of a third-generation CAR-T targeting CD19 and containing two costimulatory domains, CD28 and 4-1BB.[61] In the context of a phase I evaluation of CAR-T cells developed using this CAR construct, 9 patients with R/R CLLL and 2 with Richter’s transformation were evaluated: 2/9 R/R CLL and 1 / 2 patients with Richter’s transformation achieved a CR.[61] In the same phase I study, 7/8 and 2/2 patients with large B cell lymphoma and follicular lymphoma, respectively, achieved a CR.[61]

Allogeneic CAR-T Cells

Most of the studies performed with CAR-T cells in CLL, as well as in other tumors, involve the use of autologous, patient-derived CAR-T cells. However, allogeneic CAR-T (alloCAR-T) cells may represent a treatment alternative to autoCAR-T cells. Two types of allogeneic CAR-T cells can be utilized: donor-compatible donors and off-the-shelf CAR-T cells. Donor-derived CAR-T cells are CAR-T cells obtained from HLA-compatible donors or donors in patients with a history of allo-HSCT. HLA compatibility is required for donor-relative CAR-T, while "off-the-shelf" CAR-T cells require techniques to reduce the rejection risk of non-HLA-matched products. An example of the technology used for off-the-shelf CAR-T cells is given by UCART 19 cells, recently used for the treatment of B-ALL patients.[62] UCART 19 is a first-in-class “off-the-shelf” alloCAR-T cell based on genetic engineering of T cells from a normal donor to express an anti-CD19 (murine 4G7 scFv)/4-1BB/CD3ζ CAR together with RQR8 safety switch (a suicide gene); T cells were further genome-edited through disruption of T cell receptor alpha chain (to prevent GVHD) and CD52 gene knockout (to protect donor cells from early rejection).[63]
Studies with "off-the-shelf" CAR-T cells have not yet been reported in CLL. Only one study with donor-derived alloCAR-T cells reported the treatment of a few R/R CLL patients.[64] In fact, Brudno et al. reported the results of a phase I study involving the treatment of 20 patients with B-cell malignancies, including 5 R/R CLL patients.[60] In this study, CAR-T cells were generated from the patient's prior allo-HSCT donor. They were based on a CAR composed of a murine scFv anti-CD19, a CD28 costimulatory domain, and a CD3ζ T-cell activation domain.[64] None of the treated patients developed new onset GVHD. Among the 5 CLL patients included in the study, 1 achieved a CR, 1 a PR, and 3 did not respond to the treatment.[64] Interestingly, the CLL patient achieving a CR showed rapid elimination of leukemic cells after alloCAR-T at the level of bone marrow, peripheral blood, and lymph nodes.[64]

Conclusions

CLL was the first hematopoietic malignancy for which CAR-T cells were administered for therapeutic purposes. However, subsequent development was challenged by the finding of inferior responses observed in CLL patients compared to those observed in other B-cell malignancies. These more limited responses are related to the presence of patient comorbidities, immunodeficiency, and immunosubversion of the CAR-T cell product.
Thus, none of the four approved CAR-T cell products, Axi-Cel, Brexu-Cel, Lisi-Cell, and Tisa-Cel for NHL and B-ALL, are currently approved for CLL. The recent data reported for Liso-Cel could provide sufficient support for its approval for the treatment of R/R CLL patients.
Although preliminary and based on only 12 R/R CLL patients, the results observed using fresh, unfrozen CAR-T cells GLPG 5201 were particularly promising, manufactured, and reinfused to the patients within 7 days of blood draw. Frozen peripheral blood mononucleated cells as starting material and frozen CAR-T infusion products maintain high antitumor activity, but fresh CAR-T infusion product exhibits higher antitumor reactivity.[65] Future studies will assess whether freshly manufactured CAR-T cells have a more potent antitumor activity compared to CAR-T cells manufactured using frozen cell preparations for the treatment of R/R CLL patients.
The development of allogeneic CAR-T cells and natural killer cells from healthy donors may represent a promising solution to address the reduced fitness of T cells observed in CLL patients.
Combined treatment with ibrutinib and CAR-T cells appears to be a therapeutic strategy associated with increased efficacy due to synergistic effects between BTKi and cell therapy and provides some safety benefits. Future studies will clarify whether next-generation BTK inhibitors in association with CAR-T cells could further improve the therapeutic benefit observed with ibrutinib.
At the moment, the role of CAR-T cell therapy is limited to CLL patients with relapsed/refractory disease. Additional improvements in the safety and efficacy of CAR-T cells are required to integrate CAR-T cell therapy in earlier lines of treatment and patients with Richter's transformation. It is of interest to note that some RT patients treated with CAR-T cell therapy achieved long-term remission with prolonged survival.[66]
Interestingly, a very recent study supports the targeting of CLL cells expressing a tumor-specific antigen, a B-cell receptor light chain neoepitope defined by a characteristic point mutation (IGLV3-21R110), for selective targeting of a poor-risk subset of CLL with CAR-T cells.[67] CAR-T cells targeting this tumor neoantigen exert a significant anti-leukemia effect, sparing normal B cells.

References   

  1. SEER Cancer Stat Facts: Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma. National Cancer Institute. Bethesda, MD, https://seer.cancer.gov/statfacts/html/cllsll.html
  2. Eichhorst B, Robak T, Montserrat E, et al. Chronic lymphocytic leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2021;32(1):23-33. doi:10.1016/j.annonc.2020.09.019 https://doi.org/10.1016/j.annonc.2020.09.019 PMid:33091559
  3. NCCN Guidelines Update: Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma. J Natl Compr Canc Netw. 2023;21(5.5):563-566. doi:10.6004/jnccn.2023.5007 https://doi.org/10.6004/jnccn.2023.5007
  4. Lew TE, Lin VS, Cliff ER, Blombery P, Thompson ER, Handunnetti SM, Westerman DA, Kuss BJ, Tam CS, Huang D, et al. Outcomes of patients with CLL sequentially resistant to both BCL2 and BTK inhibition. Blood Adv 2021; 5: 4054-4063. https://doi.org/10.1182/bloodadvances.2021005083 PMid:34478505 PMCid:PMC8945613
  5. Mato, A.R.; Hess, L.M.; Chen, Y.; Abada, P.B.; Konig, H.; Pagel, J.M.; Walgren, R. Outcomes for Patients with Chronic Lymphocytic Leukemia (CLL) Previously Treated With Both a Covalent BTK and BCL2 Inhibitor in the United States: A Real-World Database Study. Clin. Lymphoma Myeloma Leuk. 2023, 23, 57-67. https://doi.org/10.1016/j.clml.2022.09.007 PMid:36335022
  6. Eyre, T.A.; Hess, L.M.; Sugihara, T.; He, D.; Khanal, M.; Pagel, J.M.; Walgren, R.; Abada, P.B.; Konig, H.; Roeker, L.; Mato, A. Clinical outcomes among patients with chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL) who received treatment with a covalent BTK and BCL2 inhibitor in the United States: A real-world database study. Leuk. Lymphoma 2023, 64, 1005-1016. https://doi.org/10.1080/10428194.2023.2190436 PMid:36987650
  7. Innocenti I, Fresa A, Tomasso A, Tarnani M, De Padua L, Benintende G, Pasquale R, Galli E, Morelli F, Giannarelli D, Autore F, Laurenti L. Treatment Sequencing and Outcome of Chronic Lymphocytic Leukemia Patients Treated at Fondazione Policlinico Universitario Agostino Gemelli IRCCS: A Thirty-Year Single-Center Experience. Cancers (Basel). 2023 Nov 26;15(23):5592. doi: 10.3390/cancers15235592. https://doi.org/10.3390/cancers15235592 PMid:38067296 PMCid:PMC10705134
  8. Roeker LE, Dreger P, Brown JR, Lahoud OB, Eyre TA, Brander DM, et al. Allogeneic stem cell transplantation for chronic lymphocytic leukemia in the era of novel agents. Blood Adv [Internet]. 2020 Aug 25;4(16):3977-89. https://doi.org/10.1182/bloodadvances.2020001956 PMid:32841336 PMCid:PMC7448605
  9. 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 Dec 28;377(26):2531-2544. doi: 10.1056/NEJMoa1707447. Epub 2017 Dec 10. https://doi.org/10.1056/NEJMoa1707447 PMid:29226797 PMCid:PMC5882485
  10. Jacobson CA, Chavez JC, Sehgal AR, William BM, Munoz J, Salles G, et al. Axicabtagene ciloleucel in relapsed or refractory indolent non-Hodgkin lymphoma (ZUMA-5): a single-arm, multicentre, phase 2 trial. Lancet Oncol. 2022 Jan;23(1):91-103. doi: 10.1016/S1470-2045(21)00591-X. Epub 2021 Dec 8. https://doi.org/10.1016/S1470-2045(21)00591-X PMid:34895487
  11. Locke FL, Miklos DB, Jacobson CA, Perales MA, Kersten MJ, Oluwole OO, et al; All ZUMA-7 Investigators and Contributing Kite Members. Axicabtagene Ciloleucel as Second-Line Therapy for Large B-Cell Lymphoma. N Engl J Med. 2022 Feb 17;386(7):640-654. doi: 10.1056/NEJMoa2116133. Epub 2021 Dec 11. https://doi.org/10.1056/NEJMoa2116133 PMid:34891224
  12. Wang M, Munoz J, Goy A, Locke FL, Jacobson CA, Hill BT, et al. KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. N Engl J Med. 2020 Apr 2;382(14):1331-1342. doi: 10.1056/NEJMoa1914347. https://doi.org/10.1056/NEJMoa1914347 PMid:32242358 PMCid:PMC7731441
  13. Shah BD, Ghobadi A, Oluwole OO, Logan AC, Boissel N, Cassaday RD, et al. KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet. 2021 Aug 7;398(10299):491-502. doi: 10.1016/S0140-6736(21)01222-8. Epub 2021 Jun 4. https://doi.org/10.1016/S0140-6736(21)01222-8 PMid:34097852
  14. Abramson JS, Solomon SR, Arnason J, Johnston PB, Glass B, Bachanova V, et al. Lisocabtagene maraleucel as second-line therapy for large B-cell lymphoma: primary analysis of the phase 3 TRANSFORM study. Blood. 2023 Apr 6;141(14):1675-1684. doi: 10.1182/blood.2022018730. https://doi.org/10.1182/blood.2022018730 PMid:36542826 PMCid:PMC10646768  
  15. Abramson JS, Palomba ML, Gordon LI, Lunning MA, Wang M, Arnason J, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020 Sep 19;396(10254):839-852. doi: 10.1016/S0140-6736(20)31366-0. Epub 2020 Sep 1. https://doi.org/10.1016/S0140-6736(20)31366-0 PMid:32888407  
  16. Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med. 2018 Feb 1;378(5):439-448. doi: 10.1056/NEJMoa1709866.  https://doi.org/10.1056/NEJMoa1709866 PMid:29385370 PMCid:PMC5996391  
  17. Mueller KT, Waldron E, Grupp SA, Levine JE, Laetsch TW, Pulsipher MA, et al. Clinical Pharmacology of Tisagenlecleucel in B-cell Acute Lymphoblastic Leukemia. Clin Cancer Res. 2018 Dec 15;24(24):6175-6184. doi: 10.1158/1078-0432.CCR-18-0758. Epub 2018 Sep 6.. https://doi.org/10.1158/1078-0432.CCR-18-0758 PMid:30190371 PMCid:PMC7433345  
  18. Schuster SJ, Bishop MR, Tam CS, Waller EK, Borchmann P, McGuirk JP, et al; JULIET Investigators. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N Engl J Med. 2019 Jan 3;380(1):45-56. doi: 10.1056/NEJMoa1804980. Epub 2018 Dec 1. https://doi.org/10.1056/NEJMoa1804980 PMid:30501490
  19. Fowler NH, Dickinson M, Dreyling M, Martinez-Lopez J, Kolstad A, Butler J, et al. Tisagenlecleucel in adult relapsed or refractory follicular lymphoma: the phase 2 ELARA trial. Nat Med. 2022 Feb;28(2):325-332. doi: 10.1038/s41591-021-01622-0. Epub 2021 Dec 17. https://doi.org/10.1038/s41591-021-01622-0 PMid:34921238  
  20. Sommermeyer D, Hudecek M, Kosasih PL, Gogishvili T, Maloney DG, Turtle CJ, Riddell SR. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia. 2016 Feb;30(2):492-500. doi: 10.1038/leu.2015.247. Epub 2015 Sep 15. https://doi.org/10.1038/leu.2015.247 PMid:26369987 PMCid:PMC4746098  
  21. Turtle CJ, Hay KA, Hanafi LA, Li D, Cherian S, Chen X, Wood B, Lozanski A, Byrd JC, Heimfeld S, et al. Durable molecular remissions in chronic lymphocytic leukemia treated with CD19-specific chimeric antigen receptor-modified T cells after failure of ibrutinib. J Clin Oncol 2017; 35: 3010-3020. https://doi.org/10.1200/JCO.2017.72.8519 PMid:28715249 PMCid:PMC5590803  
  22. Liang E, Hirayama A, Kimble E, Portuguese, Albittar A, Chapouis A, Shadman M, Till B, Cassaday R, Milano F, et al. Long-term follow-up update and multivariable analyses of factors associated with duration of response after CD19 CAR T-cell therapy for relapsed/refractory CLL. Hemasphere 2023; 7 (suppl.1): e472395b. https://doi.org/10.1097/01.HS9.0000969368.47239.5b PMCid:PMC10429589
  23. Siddiqi T, Soumarai JD, Wierda WG, D ubovsky JA, Gillenwater HH, Pharm LG, Mitchell A, Thorpe J, Yang LDorritie KA, et al. Rapid MRD-negative responses in patients with relapsed/refractory CLL treated with Liso-Cel, a CD19-directed CAR T-cell product: preliminary results from transcend CLL 004, a phase 1 / 2 study including patients with high-risk disease previously treated with ibrutinib. Blood 2018; 132 (suppl. 1):300. https://doi.org/10.1182/blood-2018-99-110462  
  24. Siddiqi T, Soumerai JD, Dorritie KA, Stephens DM, Riedell PA, Arnason JA, Kipps TJ, Gillenwater HH, Gong L, Yang L, et al. Phase 1 TRASCEND CELL 004 study of lisocabtagene maraleucel in patients with relapsed/refractory CLL or SLL. Blood 2022; 139: 1794-1806. https://doi.org/10.1182/blood.2021011895 PMid:34699592 PMCid:PMC10652916
  25. Siddiqi T, Maloney D, Kenderian SS, Brander DM, Dorritie K, Soumerai J. Lisocabtagene maraleucel in chronic lymphocytic leukemia and small lymphocytic lymphoma (TRASCEND CLL 004): a multicentre, open-label, single-arm, phase 1-2 study. Lancet 2023; 402: 641-654. https://doi.org/10.1016/S0140-6736(23)01052-8 PMid:37295445
  26. Siddiqi T, Maloney DG, Kenderian SS, Brander DM, Dorritie K, Soumnerai J, Riedell PA, Shah NV, Nath R, Fakhri B, et al. Lisocabtagene Maraleucel (liso-cel) in R/R CLL/SLL: 24-median follow-up of TRASCEND CLL-004. Blood 2023; 142 (suppl.1): 330. https://doi.org/10.1182/blood-2023-179529  
  27. Milone MC, Fish JD, Carpenito C, Carroll RG, Binder GK, Teachey D, Samanta M, Lakhal M, Gloss B, Danet-Desnoyers G, Campana D, Riley JL, Grupp SA, June CH. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther. 2009 Aug;17(8):1453-64. doi: 10.1038/mt.2009.83. Epub 2009 Apr 21. Erratum in: Mol Ther. 2015 Jul;23(7):1278. PMID: 19384291; PMCID: PMC2805264. https://doi.org/10.1038/mt.2009.83 PMid:19384291 PMCid:PMC2805264
  28. Porter DL, Hwang WT, Frey NV, Lacey SF, Shaw PA, Loren AW, Bagg A, Marcucci KT, Shen A, Gonzalez V, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl 2015; 7: 303ra139. https://doi.org/10.1126/scitranslmed.aac5415  
  29. Frey NV, Gill S, Hexner EO, Schuster S, Nasta S, Loren A, Svoboda J, Stadtmauer E, Landsburg DJ, Mato A, et al. Long-term outcomes from a randomized dose optimization study of chimeric antigen receptor modified T cells in relapsed chronic lymphocytic leukemia. J Clin Oncol 2020; 38: 2862-2871. https://doi.org/10.1200/JCO.19.03237 PMid:32298202 PMCid:PMC8265376  
  30. Melenhorst JJ, Chen GM, Wang M, Porter DL, Chen C, Collins MA, Gao P, Brabyopadhyay S, n H, Zhao Z, et al. Decade-long leukemia remissions with persistence of CD4+ CAR T cells. Nature 2022; 602: 503-509. https://doi.org/10.1038/s41586-021-04390-6 PMid:35110735 PMCid:PMC9166916
  31. Wang M, Munoz J, Goy A, Locke FL, Jacobson CA, Hill BT, Timmerman JM, Holmes H, Jaglowski S, Flinn IW, McSweeney PA, Miklos DB, Pagel JM, Kersten MJ, Milpied N, Fung H, Topp MS, Houot R, Beitinjaneh A, Peng W, Zheng L, Rossi JM, Jain RK, Rao AV, Reagan PM. KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. N Engl J Med. 2020 Apr 2;382(14):1331-1342. doi: 10.1056/NEJMoa1914347.  https://doi.org/10.1056/NEJMoa1914347 PMid:32242358 PMCid:PMC7731441
  32. Flinn I, Marris M, Wierda WG, Coutre S, Pagel JM, Byrd JC, Goyal L, Goodman K, Zheng Y, Milletti F, et al. ZUMA-8: a phase 1-2 multicenter study evaluating KTE-X19 in patients (pts) with relapsed/refractory (R/R) chronic lymphocytic leukemia (CLL). J Clin Oncol 2019; 37(suppl. 16): TPS7566. https://doi.org/10.1200/JCO.2019.37.15_suppl.TPS7566
  33. Davids MS, Kenderian SS, Flinn IW, Hill BT, Maris M, Ghia P, Byrne M, Barlett NL, Pagel JM, Zheng Y, et al. ZUMA-8: a phase 1 study of KTE-X19, an anti-CD19 chimeric antigen receptor (CAR) T-cell therapy, in patients with relapsed/refractory chronic lymphocytic leukemia. Blood 2022; 140 (suppl. 1): 7454-7456. https://doi.org/10.1182/blood-2022-167868  
  34. Kochenderfer JN, Feldman SA, Zhao Y, Xu H, Black MA, Morgan RA, Wilson WH, Rosenberg SA. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J Immunother. 2009 Sep;32(7):689-702. doi: 10.1097/CJI.0b013e3181ac6138. PMID: 19561539; PMCID: PMC2747302. https://doi.org/10.1097/CJI.0b013e3181ac6138 PMid:19561539 PMCid:PMC2747302
  35. Kochenderfer JN, Dudley ME, Kassim SH, Somerville R, Carpenter RO, Setler-Stevenson M, Yang JC, Phan GQ, Hughes MS, Sherry RM, 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. https://doi.org/10.1200/JCO.2014.56.2025 PMid:25154820 PMCid:PMC4322257  
  36. Cappell KM, Sherry RM, Yang JC, Goff SL, Vanasse DA, McInntyre L, Rosenberg SA, Kochenderfer JN. Long-term follow-up of anti-CD19 chimeric antigen receptor T-cell therapy. J Clin Oncol 2020; 38: 3805-3815. https://doi.org/10.1200/JCO.20.01467 PMid:33021872 PMCid:PMC7655016
  37. Martinez-Cibrian N, Betriu S, Ortiz-Maldonado V, Esteban D, Tovar N, Moreno AT, Alserawan L; Montoro M, Van Muyden A, Pont M, et al. Initial clinical results of EUPLAGIA-1, a phase 1-2 trial of point-of-care manufactured GLPG5201 in R/R CLL/SLL with or without Richter's transformation. Hemasphere 2023; 7(S3): 2714-2715. https://doi.org/10.1097/01.HS9.0000972484.02679.4e PMCid:PMC10430544
  38. Tovar N, Ortiz-Maldonado V, Martinez-Cibrian N, Betriu S, Esteban D, Triguero A, Verbruggen N, Spoon M, Liefaard MC, Pont M, van Muyden A. Seven-day vein-tovein point-of-care manufactured CD19 CAR T cells (GLPG5201) in relapsed/refractory CLL/SLL including Richter's transformation: results from the phase 1 Euplagia-1 trial. Blood 2023; 142 (suppl.1): 2112. https://doi.org/10.1182/blood-2023-189321
  39. Ortiz-Maldonado V, Martinez-Cibrian N, De Campo Balguerias G, Espanol-Rego M, Navarro S, Lopez-Oreja I, Nadeu F, Cobo A, Brillembourg H, Alserawan L, et al. Varnicambtagene autoleucel (ARI-0001) for relapsed or refractory chronic lymphocytic leukemia (CLL) and Richter transformation (RT). Blood 2023 142 (suppl.1): 3483. https://doi.org/10.1182/blood-2023-182896
  40. Douglas M. Richter transformation: clinical manifestations, evaluation, and management. J Adv Pract Oncol 2022; 13: 525-534. https://doi.org/10.6004/jadpro.2022.13.5.6 PMid:35910504 PMCid:PMC9328451
  41. Benjamini O, Shimoni A, Besser M, Shem-Tov N, Danylesko I, Yerushalmi R, Merkel DG, Tadmor T, Lavie D, et al. Safety and efficacy of CD19-CAR T cells in Richter's transformation after targeted therapy for chronic lymphocytic leukemia. Blood 2020; 136(suppl.1): 40. https://doi.org/10.1182/blood-2020-138904  
  42. Kittai AS, Bond DA, William B, Saad A, Penza S, Efebera Y, Larkin K, Wall SA, Choe HK, Bhatnagar B, et al. Clinical activity of axicabtagene ciloleucel in adult patients with Richter syndrome. Blood Adv 2020; 4: 4648-4652. https://doi.org/10.1182/bloodadvances.2020002783 PMid:33002129 PMCid:PMC7556158
  43. Bensaber H, Bachy E, Beauvais D, Dulery R, Gastinne T, Villemagne B, Roulin L, Paubelle E, Castilla-Llorente C, Longval T, et al. Anti-CD19 CAR T-cell therapy for patients with Richter syndrome: a Lysa study from the Descar-T registry. Blood 2022; 140 (suppl.1): 3803-3804. https://doi.org/10.1182/blood-2022-158807
  44. Blackmon A, Danilov AV, Wang L, Pillai R, rshkarlo HB, Rosen ST, Siddiqi T. Richter's transformation after CD-19 directed CAR-T cells for relapsed/refractory chronic lymphocytic leukemia (CLL). Blood 2021; 138 (suppl. 1): 1430. https://doi.org/10.1182/blood-2021-149815  
  45. Long M, Beckwith K, Do P, Mundy BL, Gordon A, Lehman AM, Maddocks KJ, Cheney C, Jones JA, et al. Ibrutinib treatment improves T cell number and function in CLL patients. J Clin Invest 2017; 127: 3052-3064. https://doi.org/10.1172/JCI89756 PMid:28714866 PMCid:PMC5531425  
  46. Fraietta JA, Beckwith KA, Patel PR, Ruella M, Zheng Z, Barrett DM, Lacey SF, Melenhorst J, McGettigan SE, et al. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood 2016; 127: 1117-1127. https://doi.org/10.1182/blood-2015-11-679134 PMid:26813675 PMCid:PMC4778162  
  47. Ruella M, Kenderian SS, Shestova O, ichinsky M, Melenhorst JJ, Wasik MA, Lacey SF, June CH, Gill S, et al. Kinase inhibitor ibrutinib to prevent cytokine-release syndrome after anti-CD19 chimeric antigen receptor T cells for B-cell neoplasms. Leukemia 2017; 31: 246-248. https://doi.org/10.1038/leu.2016.262 PMid:27677739  
  48. Papazoglou D, Wang XV, Shanafelt TD, Lesnick CE, Ioannou N, De Rossi G, Herter S, Bacac M, Klein C, Tallman MS, Kay NE, Ramsay AG. Ibrutinib-based therapy reinvigorates CD8+ T cells compared to chemoimmunotherapy: immune monitoring from the E1912 trial. Blood. 2024 Jan 4;143(1):57-63. doi: 10.1182/blood.2023020554.. https://doi.org/10.1182/blood.2023020554 PMid:37824808 PMCid:PMC10797553
  49. Gauthier J, Hirayama AV, Purushe J, Hay KA, Lymp J, Li DH, Yeung C, Sheih A, Pender BS, Hawkins RM, et al. Feasibility and efficacy of CD19-targeted CAR T cells with concurrent ibrutinib for CLL after ibrutinib failure. Blood 2020; 135: 1650-1660. https://doi.org/10.1182/blood.2019002936 PMid:32076701 PMCid:PMC7205814
  50. Gill S, Vides V, Frey NV, Hexner EO, Metzeger S, O'Brien M, Hwang WT, Brogdon JL, Davis MM, Fraietta JA, et al. Anti-CD10 CAR T cells in combination with ibrutinib for the treatment of chronic lymphocytic leukemia. Blood Adv 2022; 6: 5774-5781. https://doi.org/10.1182/bloodadvances.2022007317 PMid:35349631 PMCid:PMC9647791
  51. Vitale C, Griggio V, Perutelli F, Coscia M. CAR-modified cellular therapies in chronic lymphocytic leukemia: is the uphili road getting less steep? HemaSphere 2023; 7: 12(e988). https://doi.org/10.1097/HS9.0000000000000988 PMid:38044959 PMCid:PMC10691795
  52. Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinci I, Gohil M, Lundh S, Boesteanu AC, Wang Y, O'Connor RS, Hwang WT, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med 2018; 24: 563-571. https://doi.org/10.1038/s41591-018-0010-1 PMid:29713085 PMCid:PMC6117613
  53. van Bruggen JAC, Martens AWJ, Fraietta JA, Hofland T, Tonino SH, Eldering E, Levin MD, Siska PJ, Endstra S, Rathmell JC, June CH, Porter DL, Melenhorst JJ, Kater AP, van der Windt GJW. Chronic lymphocytic leukemia cells impair mitochondrial fitness in CD8+ T cells and impede CAR T-cell efficacy. Blood. 2019 Jul 4;134(1):44-58. https://doi.org/10.1182/blood.2018885863 PMID: 31076448; PMCID: PMC7022375. 
  54. Fraietta JA, Nobles CL, Sammons MA, Lundh S, Carty SA, Reich TJ, Cogdill AP, Morrissette JJD, DeNizio JE, Reddy S, Hwang Y, Gohil M, Kulikovskaya I, Nazimuddin F, Gupta M, Chen F, Everett JK, Alexander KA, Lin-Shiao E, Gee MH, Liu X, Young RM, Ambrose D, Wang Y, Xu J, Jordan MS, Marcucci KT, Levine BL, Garcia KC, Zhao Y, Kalos M, Porter DL, Kohli RM, Lacey SF, Berger SL, Bushman FD, June CH, Melenhorst JJ. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature. 2018 Jun;558(7709):307-312. https://doi.org/10.1038/s41586-018-0178-z PMID: 29849141; PMCID: PMC6320248.
  55. Nobles CL, Sherill-Mix S, Everett JK, Reddy S, Fraietta JA, Porter DL, Frey N, Gill S, Grupp SA, Maude SL, et al. CD19-targeting CAR T cell immunotherapy outcomes correlate with genomic modification by vector integration. J Clin Invest 2020; 130: 673-685. https://doi.org/10.1172/JCI130144 PMid:31845905 PMCid:PMC6994131  
  56. Kong W, Dimitri A, Wang W, Jung IY, Ott CJ, Fasolino M, Wang Y, Kulikovskaya I, Gupta M, Yoder T, et al. BET bromodomain protein inhibition reverses chimeric antigen receptor chimeric antigen receptor extinction and reinvigorates exhausted T cells in chronic lymphocytic leukemia. J Clin Invest 2021; 13: e145459. https://doi.org/10.1172/JCI145459 PMid:34396987 PMCid:PMC8363276  
  57. Agarwal S, Aznar MA, Rech AJ, Good CR, Kuramitsu S, Da T, Gahil M, Chen L, Hong SJ, Ravikumur S, et al. Deletion of the co-inhibitory co-receptor CTLA4 enhances and invigorates chimer antigen receptor T cells. Immunity 2023; 56: 1-20. https://doi.org/10.1016/j.immuni.2023.09.001 PMid:37776850  
  58. Shadman M,Yeung CC, Redman M, Lee SY, Lee DH, Ra S, Ujjani CS, Dezube BJ, Poh C, Warren EH, et al. High efficacy and low toxicity of MB-106, a third generation CD20 targeted CAR-T for treatment of relapsed/refractory b-NHL and CLL. Transplant Cell Ther 2022; 28: S182-S183. https://doi.org/10.1016/S2666-6367(22)00386-4  
  59. Shah NN, Johnson BD, Schneider D, Zhu F, Szabo A, Keever-Taylor CA, Krueger W, Worden AA, Kadan MJ, Yim S.; et al. Bispecific anti-CD20, anti-CD19 CAR T cells for relapsed B cell malignancies: a phase 1 dose-escalation and expansion trial. Nat Med 2020; 26: 1569-1575. https://doi.org/10.1038/s41591-020-1081-3 PMid:33020647
  60. Tong C, Zhang Y, Liu Y, Ji X, Zhang WY, Guo Y, Han X, Ti D, Dai H, Wang C., et al. Optimized tandem CD19/CD20 CAR-engineered T cells in refractory/relapsed B cell lymphoma. Blood 2020; 136: 1632-1644. https://doi.org/10.1182/blood.2020005278 PMid:32556247 PMCid:PMC7596761
  61. Park JH, Palomba ML, Bellevi CL, Riviere I, Wang X, Senechal B, Furman RR, Bernal Y, Hall M, Pineda J, et al. A phase I first-in-human clinical trial of CD19-targeted 19-28z/4-1BBL "Armored" CAR T cells in aptients with relapsed or refractory NHL and CLL including Richter's transformation. Blood 2018; 132(suppl.1): 224. https://doi.org/10.1182/blood-2018-99-117737  
  62. Benjamin R, Jain N, Maus MV, Boissel N, Graham C, Jorwik A, Yallop D, Konopleva M, Frigault MJ, Teshima T, et al. UCART19, a first-in-class allogeneic anti-CD19 chimeric antigen receptor T-cell therapy for adults with relapsed or refractory B-cell acute lymphoblastic leukemia (CALM): a phase 1, dose-escalation trial. Lancet Haematol 2022; 9: e833-e843. https://doi.org/10.1016/S2352-3026(22)00245-9 PMid:36228643  
  63. Dupouy S, Marciq I, Derippe T, Almena-Carrasco M, Jozwik A, Fouliard S, Adimy Y, Geronimi J, Graham C, Jain N, et al. Clinical pharmacology and determinants of response to UCART19, an allogeneic anti-CD19 CAR-T cell product, in adult B-cell acute lymphoblastic leukemia. Cancer Res commun 2022; 2: 1520-1530. https://doi.org/10.1158/2767-9764.CRC-22-0175 PMid:36970059 PMCid:PMC10035397  
  64. Brudno JN, Sommerville R, Shi V, Rose J, Halverson DC, Fowler DH, Gea-Banacloche JC, Pavletic SZ, Hickstein DD, Lu TL, 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; 344: 1112-1121. https://doi.org/10.1200/JCO.2015.64.5929 PMid:26811520 PMCid:PMC4872017
  65. Brezinger-Dayan K, Itzhaki O, Melnichenko J, Kubi A, Zelter L, Jacoby E, Avigdot A, Shapira Frommer R, Besser MJ. Impact of cryopreservation on CAR T production and clinical response. Front Oncol 2022; 12: 1024362. https://doi.org/10.3389/fonc.2022.1024362 PMid:36276077 PMCid:PMC9582437
  66. Kutsch N, Godel P, Voltin CA, Hallek M, Scheid C, Borchmann P, Holtick U. Long-term remission in a patient with relapsed Richter's transformation treated with CD19-directed chimeric antigen-receptor T-cells after allogeneic stem cell transplantation. Eur J Haematol 2024; in press. https://doi.org/10.1111/ejh.14182 PMid:38316549
  67. Markl F, Schultheib C, Ali M, Chen SS, Zintchenko M, Egli L, Mietz J, Chijioke D, Paschold L, Spajic S, et al. Mutation-specific CAR T cells as precision therapy for IGLV3-21R110 expressing high-risk chronic lymphocytic leukemia. Nat Commun 2024; 15: 993. https://doi.org/10.1038/s41467-024-45378-w PMid:38307904 PMCid:PMC10837166