Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3933, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730243

ABSTRACT

As a strategy to improve the therapeutic success of chimeric antigen receptor T cells (CART) directed against solid tumors, we here test the combinatorial use of CART and IMSA101, a newly developed stimulator of interferon genes (STING) agonist. In two syngeneic tumor models, improved overall survival is observed when mice are treated with intratumorally administered IMSA101 in addition to intravenous CART infusion. Transcriptomic analyses of CART isolated from tumors show elevated T cell activation, as well as upregulated cytokine pathway signatures, in particular IL-18, in the combination treatment group. Also, higher levels of IL-18 in serum and tumor are detected with IMSA101 treatment. Consistent with this, the use of IL-18 receptor negative CART impair anti-tumor responses in mice receiving combination treatment. In summary, we find that IMSA101 enhances CART function which is facilitated through STING agonist-induced IL-18 secretion.


Subject(s)
Interleukin-18 , Membrane Proteins , Receptors, Chimeric Antigen , Animals , Interleukin-18/metabolism , Membrane Proteins/agonists , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Humans , Cell Line, Tumor , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Lymphocyte Activation/drug effects , Immunotherapy, Adoptive/methods , Female , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy
2.
Proc Natl Acad Sci U S A ; 121(10): e2317735121, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38408246

ABSTRACT

Chimeric antigen receptor (CAR) T cell dysfunction is a major barrier to achieving lasting remission in hematologic cancers, especially in chronic lymphocytic leukemia (CLL). We have shown previously that Δ133p53α, an endogenous isoform of the human TP53 gene, decreases in expression with age in human T cells, and that reconstitution of Δ133p53α in poorly functional T cells can rescue proliferation [A. M. Mondal et al., J. Clin. Invest. 123, 5247-5257 (2013)]. Although Δ133p53α lacks a transactivation domain, it can form heterooligomers with full-length p53 and modulate the p53-mediated stress response [I. Horikawa et al., Cell Death Differ. 24, 1017-1028 (2017)]. Here, we show that constitutive expression of Δ133p53α potentiates the anti-tumor activity of CD19-directed CAR T cells and limits dysfunction under conditions of high tumor burden and metabolic stress. We demonstrate that Δ133p53α-expressing CAR T cells exhibit a robust metabolic phenotype, maintaining the ability to execute effector functions and continue proliferating under nutrient-limiting conditions, in part due to upregulation of critical biosynthetic processes and improved mitochondrial function. Importantly, we show that our strategy to constitutively express Δ133p53α improves the anti-tumor efficacy of CAR T cells generated from CLL patients that previously failed CAR T cell therapy. More broadly, our results point to the potential role of the p53-mediated stress response in limiting the prolonged antitumor functions required for complete tumor clearance in patients with high disease burden, suggesting that modulation of the p53 signaling network with Δ133p53α may represent a translationally viable strategy for improving CAR T cell therapy.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive/methods , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Antigens, CD19 , Cell- and Tissue-Based Therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
3.
Immunity ; 56(10): 2388-2407.e9, 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37776850

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy targeting CD19 has achieved tremendous success treating B cell malignancies; however, some patients fail to respond due to poor autologous T cell fitness. To improve response rates, we investigated whether disruption of the co-inhibitory receptors CTLA4 or PD-1 could restore CART function. CRISPR-Cas9-mediated deletion of CTLA4 in preclinical models of leukemia and myeloma improved CAR T cell proliferation and anti-tumor efficacy. Importantly, this effect was specific to CTLA4 and not seen upon deletion of CTLA4 and/or PDCD1 in CAR T cells. Mechanistically, CTLA4 deficiency permitted unopposed CD28 signaling and maintenance of CAR expression on the T cell surface under conditions of high antigen load. In clinical studies, deletion of CTLA4 rescued the function of T cells from patients with leukemia that previously failed CAR T cell treatment. Thus, selective deletion of CTLA4 reinvigorates dysfunctional chronic lymphocytic leukemia (CLL) patient T cells, providing a strategy for increasing patient responses to CAR T cell therapy.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Receptors, Chimeric Antigen , Humans , Receptors, Antigen, T-Cell/metabolism , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , T-Lymphocytes , Immunotherapy, Adoptive , Antigens, CD19
4.
Sci Transl Med ; 15(714): eadi1145, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37651540

ABSTRACT

In the absence of cell surface cancer-specific antigens, immunotherapies such as chimeric antigen receptor (CAR) T cells, monoclonal antibodies, or bispecific T cell engagers typically target lineage antigens. Currently, such immunotherapies are individually designed and tested for each disease. This approach is inefficient and limited to a few lineage antigens for which the on-target/off-tumor toxicities are clinically tolerated. Here, we sought to develop a universal CAR T cell therapy for blood cancers directed against the pan-leukocyte marker CD45. To protect healthy hematopoietic cells, including CAR T cells, from CD45-directed on-target/off-tumor toxicity while preserving the essential functions of CD45, we mapped the epitope on CD45 that is targeted by the CAR and used CRISPR adenine base editing to install a function-preserving mutation sufficient to evade CAR T cell recognition. Epitope-edited CD45 CAR T cells were fratricide resistant and effective against patient-derived acute myeloid leukemia, B cell lymphoma, and acute T cell leukemia. Epitope-edited hematopoietic stem cells (HSCs) were protected from CAR T cells and, unlike CD45 knockout cells, could engraft, persist, and differentiate in vivo. Ex vivo epitope editing in HSCs and T cells enables the safe and effective use of CD45-directed CAR T cells and bispecific T cell engagers for the universal treatment of hematologic malignancies and might be exploited for other diseases requiring intensive hematopoietic ablation.


Subject(s)
Hematologic Neoplasms , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Epitopes , Gene Editing , Hematologic Neoplasms/therapy , Immunotherapy
5.
Mol Ther ; 31(8): 2309-2325, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37312454

ABSTRACT

Multiple clinical studies have treated mesothelin (MSLN)-positive solid tumors by administering MSLN-directed chimeric antigen receptor (CAR) T cells. Although these products are generally safe, efficacy is limited. Therefore, we generated and characterized a potent, fully human anti-MSLN CAR. In a phase 1 dose-escalation study of patients with solid tumors, we observed two cases of severe pulmonary toxicity following intravenous infusion of this product in the high-dose cohort (1-3 × 108 T cells per m2). Both patients demonstrated progressive hypoxemia within 48 h of infusion with clinical and laboratory findings consistent with cytokine release syndrome. One patient ultimately progressed to grade 5 respiratory failure. An autopsy revealed acute lung injury, extensive T cell infiltration, and accumulation of CAR T cells in the lungs. RNA and protein detection techniques confirmed low levels of MSLN expression by benign pulmonary epithelial cells in affected lung and lung samples obtained from other inflammatory or fibrotic conditions, indicating that pulmonary pneumocyte and not pleural expression of mesothelin may lead to dose-limiting toxicity. We suggest patient enrollment criteria and dosing regimens of MSLN-directed therapies consider the possibility of dynamic expression of mesothelin in benign lung with a special concern for patients with underlying inflammatory or fibrotic conditions.


Subject(s)
Mesothelin , Neoplasms , Humans , GPI-Linked Proteins/genetics , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Neoplasms/therapy , T-Lymphocytes
6.
Blood Adv ; 7(14): 3416-3430, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37058474

ABSTRACT

A challenge when targeting T-cell lymphoma with chimeric antigen receptor (CAR) T-cell therapy is that target antigens are often shared between T cells and tumor cells, resulting in fratricide between CAR T cells and on-target cytotoxicity on normal T cells. CC chemokine receptor 4 (CCR4) is highly expressed in many mature T-cell malignancies, such as adult T-cell leukemia/lymphoma (ATLL) and cutaneous T-cell lymphoma (CTCL), and has a unique expression profile in normal T cells. CCR4 is predominantly expressed by type-2 and type-17 helper T cells (Th2 and Th17) and regulatory T cells (Treg), but it is rarely expressed by other T helper (Th) subsets and CD8+ cells. Although fratricide in CAR T cells is generally thought to be detrimental to anticancer functions, in this study, we demonstrated that anti-CCR4 CAR T cells specifically depleted Th2 and Tregs, while sparing CD8+ and Th1 T cells. Moreover, fratricide increased the percentage of CAR+ T cells in the final product. CCR4-CAR T cells were characterized by high transduction efficiency, robust T-cell expansion, and rapid fratricidal depletion of CCR4-positive T cells during CAR transduction and expansion. Furthermore, mogamulizumab-based CCR4-CAR T cells induced superior antitumor efficacy and long-term remission in mice engrafted with human T-cell lymphoma cells. In summary, CCR4-depleted anti-CCR4 CAR T cells are enriched in Th1 and CD8+ T cells and exhibit high antitumor efficacy against CCR4-expressing T-cell malignancies.


Subject(s)
Lymphoma, T-Cell, Cutaneous , Lymphoma, T-Cell, Peripheral , Lymphoma, T-Cell , Skin Neoplasms , Adult , Humans , Animals , Mice , Receptors, CCR4/metabolism , T-Lymphocytes, Regulatory
7.
Sci Adv ; 9(2): eade2526, 2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36630514

ABSTRACT

Incomplete surgery of solid tumors is a risk factor for primary treatment failure. Here, we have investigated whether chimeric antigen receptor T cells (CARTs) could be used as an adjuvant therapy to clear residual cancer cells. We tested the feasibility of this approach in two partial resection xenograft models using mesothelin-specific CARTs. In addition, we developed a previously unexplored in vivo toxicity model to evaluate safety and effects on wound healing in immunocompetent C57BL/6 mice. We found that the local delivery of CARTs in a fibrin glue-based carrier was effective in clearing residual cancer cells following incomplete surgery. This resulted in significantly longer overall survival when compared to mice treated with surgery and CARTs without fibrin glue. On-target off-tumor toxicity was diminished, and wound healing complications were not seen in any of the mice. On the basis of these observations, a clinical trial in patients with locally advanced breast cancer is planned.

8.
Blood Cancer Discov ; 4(2): 118-133, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36413381

ABSTRACT

We conducted a phase I clinical trial of anti-BCMA chimeric antigen receptor T cells (CART-BCMA) with or without anti-CD19 CAR T cells (huCART19) in multiple myeloma (MM) patients responding to third- or later-line therapy (phase A, N = 10) or high-risk patients responding to first-line therapy (phase B, N = 20), followed by early lenalidomide or pomalidomide maintenance. We observed no high-grade cytokine release syndrome (CRS) and only one instance of low-grade neurologic toxicity. Among 15 subjects with measurable disease, 10 exhibited partial response (PR) or better; among 26 subjects responding to prior therapy, 9 improved their response category and 4 converted to minimal residual disease (MRD)-negative complete response/stringent complete response. Early maintenance therapy was safe, feasible, and coincided in some patients with CAR T-cell reexpansion and late-onset, durable clinical response. Outcomes with CART-BCMA + huCART19 were similar to CART-BCMA alone. Collectively, our results demonstrate favorable safety, pharmacokinetics, and antimyeloma activity of dual-target CAR T-cell therapy in early lines of MM treatment. SIGNIFICANCE: CAR T cells in early lines of MM therapy could be safer and more effective than in the advanced setting, where prior studies have focused. We evaluated the safety, pharmacokinetics, and efficacy of CAR T cells in patients with low disease burden, responding to current therapy, combined with standard maintenance therapy. This article is highlighted in the In This Issue feature, p. 101.


Subject(s)
Multiple Myeloma , Receptors, Chimeric Antigen , Humans , Multiple Myeloma/therapy , Receptors, Chimeric Antigen/therapeutic use , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Lenalidomide/therapeutic use , Antigens, CD19/therapeutic use , T-Lymphocytes
12.
Nature ; 607(7918): 360-365, 2022 07.
Article in English | MEDLINE | ID: mdl-35676488

ABSTRACT

Synthetic receptor signalling has the potential to endow adoptively transferred T cells with new functions that overcome major barriers in the treatment of solid tumours, including the need for conditioning chemotherapy1,2. Here we designed chimeric receptors that have an orthogonal IL-2 receptor extracellular domain (ECD) fused with the intracellular domain (ICD) of receptors for common γ-chain (γc) cytokines IL-4, IL-7, IL-9 and IL-21 such that the orthogonal IL-2 cytokine elicits the corresponding γc cytokine signal. Of these, T cells that signal through the chimeric orthogonal IL-2Rß-ECD-IL-9R-ICD (o9R) are distinguished by the concomitant activation of STAT1, STAT3 and STAT5 and assume characteristics of stem cell memory and effector T cells. Compared to o2R T cells, o9R T cells have superior anti-tumour efficacy in two recalcitrant syngeneic mouse solid tumour models of melanoma and pancreatic cancer and are effective even in the absence of conditioning lymphodepletion. Therefore, by repurposing IL-9R signalling using a chimeric orthogonal cytokine receptor, T cells gain new functions, and this results in improved anti-tumour activity for hard-to-treat solid tumours.


Subject(s)
Cell- and Tissue-Based Therapy , Immunotherapy, Adoptive , Interleukin Receptor Common gamma Subunit , Neoplasms , Receptors, Interleukin-9 , Recombinant Fusion Proteins , T-Lymphocytes , Animals , Cell- and Tissue-Based Therapy/methods , Immunotherapy, Adoptive/methods , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/immunology , Interleukins/genetics , Interleukins/immunology , Melanoma/immunology , Mice , Neoplasms/genetics , Neoplasms/immunology , Pancreatic Neoplasms/immunology , Receptors, Interleukin-9/genetics , Receptors, Interleukin-9/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , STAT Transcription Factors/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
13.
Cancer Discov ; 12(7): 1625-1633, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35417527

ABSTRACT

CD19- and B-cell maturation antigen (BCMA)-directed chimeric antigen receptor (CAR) T cells have enabled unprecedented responses in a subset of refractory patients with B-cell and plasma cell malignancies, leading to their approval by the FDA for the treatment of leukemia, lymphoma, and myeloma. These "living drugs" can become part of a synthetic immune system, persisting at least a decade in some patients. However, despite this tremendous impact, significant unmet treatment needs remain for patients with hematologic malignancies and solid cancers. In this perspective, we highlight recent innovations that advance the field toward production of a more potent and universal cellular immunotherapy of the future. Next-generation CAR T cells will incorporate advances in gene engineering and synthetic biology to enhance functionality and persistence, and reduce treatment-associated toxicities. The combination of autologous CAR T cells with various allogeneic cell treatment strategies designed to target the immunosuppressive tumor microenvironment will broaden the impact of future CAR T-cell therapies.


Subject(s)
Immunotherapy, Adoptive , Multiple Myeloma , Antigens, CD19 , B-Cell Maturation Antigen , Humans , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes , Tumor Microenvironment
14.
Immunity ; 55(3): 557-574.e7, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35263570

ABSTRACT

The clinical benefit of T cell immunotherapies remains limited by incomplete understanding of T cell differentiation and dysfunction. We generated an epigenetic and transcriptional atlas of T cell differentiation from healthy humans that included exhausted CD8 T cells and applied this resource in three ways. First, we identified modules of gene expression and chromatin accessibility, revealing molecular coordination of differentiation after activation and between central memory and effector memory. Second, we applied this healthy molecular framework to three settings-a neoadjuvant anti-PD1 melanoma trial, a basal cell carcinoma scATAC-seq dataset, and autoimmune disease-associated SNPs-yielding insights into disease-specific biology. Third, we predicted genome-wide cis-regulatory elements and validated this approach for key effector genes using CRISPR interference, providing functional annotation and demonstrating the ability to identify targets for non-coding cellular engineering. These studies define epigenetic and transcriptional regulation of human T cells and illustrate the utility of interrogating disease in the context of a healthy T cell atlas.


Subject(s)
Epigenomics , Lymphocyte Activation , CD8-Positive T-Lymphocytes , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , Epigenesis, Genetic , Humans , Lymphocyte Activation/genetics
15.
Nature ; 602(7897): 503-509, 2022 02.
Article in English | MEDLINE | ID: mdl-35110735

ABSTRACT

The adoptive transfer of T lymphocytes reprogrammed to target tumour cells has demonstrated potential for treatment of various cancers1-7. However, little is known about the long-term potential and clonal stability of the infused cells. Here we studied long-lasting CD19-redirected chimeric antigen receptor (CAR) T cells in two patients with chronic lymphocytic leukaemia1-4 who achieved a complete remission in 2010. CAR T cells remained detectable more than ten years after infusion, with sustained remission in both patients. Notably, a highly activated CD4+ population emerged in both patients, dominating the CAR T cell population at the later time points. This transition was reflected in the stabilization of the clonal make-up of CAR T cells with a repertoire dominated by a small number of clones. Single-cell profiling demonstrated that these long-persisting CD4+ CAR T cells exhibited cytotoxic characteristics along with ongoing functional activation and proliferation. In addition, longitudinal profiling revealed a population of gamma delta CAR T cells that prominently expanded in one patient concomitant with CD8+ CAR T cells during the initial response phase. Our identification and characterization of these unexpected CAR T cell populations provide novel insight into the CAR T cell characteristics associated with anti-cancer response and long-term remission in leukaemia.


Subject(s)
CD4-Positive T-Lymphocytes , Immunotherapy, Adoptive , Leukemia , Receptors, Chimeric Antigen , Antigens, CD19/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Separation , Humans , Leukemia/immunology , Leukemia/therapy , Receptors, Chimeric Antigen/immunology , Time Factors
16.
Cell ; 184(25): 6081-6100.e26, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34861191

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has achieved remarkable success in hematological malignancies but remains ineffective in solid tumors, due in part to CAR T cell exhaustion in the solid tumor microenvironment. To study dysfunction of mesothelin-redirected CAR T cells in pancreatic cancer, we establish a robust model of continuous antigen exposure that recapitulates hallmark features of T cell exhaustion and discover, both in vitro and in CAR T cell patients, that CAR dysregulation is associated with a CD8+ T-to-NK-like T cell transition. Furthermore, we identify a gene signature defining CAR and TCR dysregulation and transcription factors, including SOX4 and ID3 as key regulators of CAR T cell exhaustion. Our findings shed light on the plasticity of human CAR T cells and demonstrate that genetic downmodulation of ID3 and SOX4 expression can improve the efficacy of CAR T cell therapy in solid tumors by preventing or delaying CAR T cell dysfunction.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Pancreatic Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , HEK293 Cells , Humans , Inhibitor of Differentiation Proteins/immunology , Male , Mice , Mice, Knockout , Mice, Nude , Mice, SCID , Neoplasm Proteins/immunology , SOXC Transcription Factors/immunology
17.
J Clin Invest ; 131(16)2021 08 16.
Article in English | MEDLINE | ID: mdl-34396987

ABSTRACT

Chimeric antigen receptor (CAR) T cells have induced remarkable antitumor responses in B cell malignancies. Some patients do not respond because of T cell deficiencies that hamper the expansion, persistence, and effector function of these cells. We used longitudinal immune profiling to identify phenotypic and pharmacodynamic changes in CD19-directed CAR T cells in patients with chronic lymphocytic leukemia (CLL). CAR expression maintenance was also investigated because this can affect response durability. CAR T cell failure was accompanied by preexisting T cell-intrinsic defects or dysfunction acquired after infusion. In a small subset of patients, CAR silencing was observed coincident with leukemia relapse. Using a small molecule inhibitor, we demonstrated that the bromodomain and extra-terminal (BET) family of chromatin adapters plays a role in downregulating CAR expression. BET protein blockade also ameliorated CAR T cell exhaustion as manifested by inhibitory receptor reduction, enhanced metabolic fitness, increased proliferative capacity, and enriched transcriptomic signatures of T cell reinvigoration. BET inhibition decreased levels of the TET2 methylcytosine dioxygenase, and forced expression of the TET2 catalytic domain eliminated the potency-enhancing effects of BET protein targeting in CAR T cells, providing a mechanism linking BET proteins and T cell dysfunction. Thus, modulating BET epigenetic readers may improve the efficacy of cell-based immunotherapies.


Subject(s)
Immunotherapy, Adoptive , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Proteins/antagonists & inhibitors , Proteins/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Antigens, CD19/immunology , Azepines/pharmacology , Epigenesis, Genetic , Glycolysis/drug effects , Humans , Immune Tolerance , Immunologic Memory , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Oxidative Phosphorylation/drug effects , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Triazoles/pharmacology
18.
Nat Med ; 27(5): 842-850, 2021 05.
Article in English | MEDLINE | ID: mdl-33888899

ABSTRACT

While CD19-directed chimeric antigen receptor (CAR) T cells can induce remission in patients with B cell acute lymphoblastic leukemia (ALL), a large subset relapse with CD19- disease. Like CD19, CD22 is broadly expressed by B-lineage cells and thus serves as an alternative immunotherapy target in ALL. Here we present the composite outcomes of two pilot clinical trials ( NCT02588456 and NCT02650414 ) of T cells bearing a 4-1BB-based, CD22-targeting CAR in patients with relapsed or refractory ALL. The primary end point of these studies was to assess safety, and the secondary end point was antileukemic efficacy. We observed unexpectedly low response rates, prompting us to perform detailed interrogation of the responsible CAR biology. We found that shortening of the amino acid linker connecting the variable heavy and light chains of the CAR antigen-binding domain drove receptor homodimerization and antigen-independent signaling. In contrast to CD28-based CARs, autonomously signaling 4-1BB-based CARs demonstrated enhanced immune synapse formation, activation of pro-inflammatory genes and superior effector function. We validated this association between autonomous signaling and enhanced function in several CAR constructs and, on the basis of these observations, designed a new short-linker CD22 single-chain variable fragment for clinical evaluation. Our findings both suggest that tonic 4-1BB-based signaling is beneficial to CAR function and demonstrate the utility of bedside-to-bench-to-bedside translation in the design and implementation of CAR T cell therapies.


Subject(s)
4-1BB Ligand/metabolism , Immunotherapy, Adoptive/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Chimeric Antigen/metabolism , Sialic Acid Binding Ig-like Lectin 2/metabolism , T-Lymphocytes/transplantation , Adult , Animals , Antigens, CD19/metabolism , B-Lymphocytes/immunology , CD28 Antigens/genetics , Cells, Cultured , Child , Child, Preschool , Female , Humans , Male , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...