Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nature ; 615(7952): 507-516, 2023 03.
Article in English | MEDLINE | ID: mdl-36890224

ABSTRACT

Although chimeric antigen receptor (CAR) T cells have altered the treatment landscape for B cell malignancies, the risk of on-target, off-tumour toxicity has hampered their development for solid tumours because most target antigens are shared with normal cells1,2. Researchers have attempted to apply Boolean-logic gating to CAR T cells to prevent toxicity3-5; however, a truly safe and effective logic-gated CAR has remained elusive6. Here we describe an approach to CAR engineering in which we replace traditional CD3ζ domains with intracellular proximal T cell signalling molecules. We show that certain proximal signalling CARs, such as a ZAP-70 CAR, can activate T cells and eradicate tumours in vivo while bypassing upstream signalling proteins, including CD3ζ. The primary role of ZAP-70 is to phosphorylate LAT and SLP-76, which form a scaffold for signal propagation. We exploited the cooperative role of LAT and SLP-76 to engineer logic-gated intracellular network (LINK) CAR, a rapid and reversible Boolean-logic AND-gated CAR T cell platform that outperforms other systems in both efficacy and prevention of on-target, off-tumour toxicity. LINK CAR will expand the range of molecules that can be targeted with CAR T cells, and will enable these powerful therapeutic agents to be used for solid tumours and diverse diseases such as autoimmunity7 and fibrosis8. In addition, this work shows that the internal signalling machinery of cells can be repurposed into surface receptors, which could open new avenues for cellular engineering.


Subject(s)
Cell Engineering , Immunotherapy, Adoptive , Logic , Neoplasms , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen , Signal Transduction , T-Lymphocytes , Humans , Cell Engineering/methods , Immunotherapy, Adoptive/adverse effects , Leukemia, B-Cell , Lymphoma, B-Cell , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
2.
Nature ; 586(7831): 779-784, 2020 10.
Article in English | MEDLINE | ID: mdl-33087934

ABSTRACT

Antibodies that antagonize extracellular receptor-ligand interactions are used as therapeutic agents for many diseases to inhibit signalling by cell-surface receptors1. However, this approach does not directly prevent intracellular signalling, such as through tonic or sustained signalling after ligand engagement. Here we present an alternative approach for attenuating cell-surface receptor signalling, termed receptor inhibition by phosphatase recruitment (RIPR). This approach compels cis-ligation of cell-surface receptors containing ITAM, ITIM or ITSM tyrosine phosphorylation motifs to the promiscuous cell-surface phosphatase CD452,3, which results in the direct intracellular dephosphorylation of tyrosine residues on the receptor target. As an example, we found that tonic signalling by the programmed cell death-1 receptor (PD-1) results in residual suppression of T cell activation, but is not inhibited by ligand-antagonist antibodies. We engineered a PD-1 molecule, which we denote RIPR-PD1, that induces cross-linking of PD-1 to CD45 and inhibits both tonic and ligand-activated signalling. RIPR-PD1 demonstrated enhanced inhibition of checkpoint blockade compared with ligand blocking by anti-PD1 antibodies, and increased therapeutic efficacy over anti-PD1 in mouse tumour models. We also show that the RIPR strategy extends to other immune-receptor targets that contain activating or inhibitory ITIM, ITSM or ITAM motifs; for example, inhibition of the macrophage SIRPα 'don't eat me' signal with a SIRPα-CD45 RIPR molecule potentiates antibody-dependent cellular phagocytosis beyond that of SIRPα blockade alone. RIPR represents a general strategy for direct attenuation of signalling by kinase-activated cell-surface receptors.


Subject(s)
Leukocyte Common Antigens/metabolism , Phosphoric Monoester Hydrolases/metabolism , Receptors, Immunologic/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Carcinoma, Small Cell/drug therapy , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cross-Linking Reagents , Disease Models, Animal , Disease Progression , Female , HEK293 Cells , Humans , Leukocyte Common Antigens/antagonists & inhibitors , Leukocyte Common Antigens/chemistry , Ligands , Lymphocyte Activation/drug effects , Male , Mice , Nivolumab/pharmacology , Phosphorylation , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
3.
Cancer Discov ; 10(5): 702-723, 2020 05.
Article in English | MEDLINE | ID: mdl-32193224

ABSTRACT

Insufficient reactivity against cells with low antigen density has emerged as an important cause of chimeric antigen receptor (CAR) T-cell resistance. Little is known about factors that modulate the threshold for antigen recognition. We demonstrate that CD19 CAR activity is dependent upon antigen density and that the CAR construct in axicabtagene ciloleucel (CD19-CD28ζ) outperforms that in tisagenlecleucel (CD19-4-1BBζ) against antigen-low tumors. Enhancing signal strength by including additional immunoreceptor tyrosine-based activation motifs (ITAM) in the CAR enables recognition of low-antigen-density cells, whereas ITAM deletions blunt signal and increase the antigen density threshold. Furthermore, replacement of the CD8 hinge-transmembrane (H/T) region of a 4-1BBζ CAR with a CD28-H/T lowers the threshold for CAR reactivity despite identical signaling molecules. CARs incorporating a CD28-H/T demonstrate a more stable and efficient immunologic synapse. Precise design of CARs can tune the threshold for antigen recognition and endow 4-1BBζ-CARs with enhanced capacity to recognize antigen-low targets while retaining a superior capacity for persistence. SIGNIFICANCE: Optimal CAR T-cell activity is dependent on antigen density, which is variable in many cancers, including lymphoma and solid tumors. CD28ζ-CARs outperform 4-1BBζ-CARs when antigen density is low. However, 4-1BBζ-CARs can be reengineered to enhance activity against low-antigen-density tumors while maintaining their unique capacity for persistence.This article is highlighted in the In This Issue feature, p. 627.


Subject(s)
Receptors, Chimeric Antigen/metabolism , Animals , Humans , Mice , Signal Transduction
4.
Clin Cancer Res ; 25(8): 2560-2574, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30655315

ABSTRACT

PURPOSE: Patients with relapsed pediatric solid tumors and CNS malignancies have few therapeutic options and frequently die of their disease. Chimeric antigen receptor (CAR) T cells have shown tremendous success in treating relapsed pediatric acute lymphoblastic leukemia, but this has not yet translated to treating solid tumors. This is partially due to a paucity of differentially expressed cell surface molecules on solid tumors that can be safely targeted. Here, we present B7-H3 (CD276) as a putative target for CAR T-cell therapy of pediatric solid tumors, including those arising in the central nervous system. EXPERIMENTAL DESIGN: We developed a novel B7-H3 CAR whose binder is derived from a mAb that has been shown to preferentially bind tumor tissues and has been safely used in humans in early-phase clinical trials. We tested B7-H3 CAR T cells in a variety of pediatric cancer models. RESULTS: B7-H3 CAR T cells mediate significant antitumor activity in vivo, causing regression of established solid tumors in xenograft models including osteosarcoma, medulloblastoma, and Ewing sarcoma. We demonstrate that B7-H3 CAR T-cell efficacy is largely dependent upon high surface target antigen density on tumor tissues and that activity is greatly diminished against target cells that express low levels of antigen, thus providing a possible therapeutic window despite low-level normal tissue expression of B7-H3. CONCLUSIONS: B7-H3 CAR T cells could represent an exciting therapeutic option for patients with certain lethal relapsed or refractory pediatric malignancies, and should be tested in carefully designed clinical trials.


Subject(s)
Antigens, Neoplasm/immunology , B7 Antigens/immunology , Brain Neoplasms/etiology , Brain Neoplasms/metabolism , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , B7 Antigens/antagonists & inhibitors , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Disease Models, Animal , Humans , Immunohistochemistry , Immunotherapy, Adoptive/methods , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
5.
Cytotherapy ; 20(10): 1259-1266, 2018 10.
Article in English | MEDLINE | ID: mdl-30309710

ABSTRACT

Targeted adoptive immunotherapy with engineered T cells is a promising treatment for refractory hematologic malignancies. However, many patients achieving early complete remissions ultimately relapse. Immunosuppressive ligands are expressed on tumor and supportive cells in the tumor microenvironment (TME). When activated, T cells express associated "checkpoint" receptors. Binding of co-inhibitory ligands and receptors may directly contribute to T-cell functional exhaustion. It is not known whether all T cells engineered to express chimeric antigen receptors (CARs) are subject to checkpoint-mediated regulation. It is also unknown whether distinct CAR signaling moieties modulate T-cell responsiveness to these inhibitory pathways. We have, therefore, directly compared functional co-inhibition in engineered T cells identically targeted to the tumor-associated antigen CD123, but distinct in their mode of T-cell activation: via the endogenous T-cell receptor (ENG), or downstream of CD28 or 41BB-containing CARs. In all cases, we have observed antigen-independent T-cell activation associated with upregulation of the co-inhibitory receptors programmed cell death protein 1 (PD-1, CD279), Tim-3 and Lag-3. Notably, CD28.CAR T cells were uniquely susceptible to PD-1/PD-L1 mediated checkpoint inhibition. Together, our data indicate that PD-1/PD-L1 checkpoint blocking agents may be considered clinically when CD28.CAR T cells do not perform optimally in human trials.


Subject(s)
Programmed Cell Death 1 Receptor/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , CD28 Antigens/genetics , CD28 Antigens/immunology , Cell Line, Tumor , Cytotoxicity Tests, Immunologic/methods , Genetic Engineering , Hepatitis A Virus Cellular Receptor 2/immunology , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Immunotherapy, Adoptive/methods , Interleukin-3 Receptor alpha Subunit/genetics , K562 Cells , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Lymphocyte Activation , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/physiology , Tumor Microenvironment
6.
Nat Med ; 24(5): 572-579, 2018 05.
Article in English | MEDLINE | ID: mdl-29662203

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) and other diffuse midline gliomas (DMGs) with mutated histone H3 K27M (H3-K27M)1-5 are aggressive and universally fatal pediatric brain cancers 6 . Chimeric antigen receptor (CAR)-expressing T cells have mediated impressive clinical activity in B cell malignancies7-10, and recent results suggest benefit in central nervous system malignancies11-13. Here, we report that patient-derived H3-K27M-mutant glioma cell cultures exhibit uniform, high expression of the disialoganglioside GD2. Anti-GD2 CAR T cells incorporating a 4-1BBz costimulatory domain 14 demonstrated robust antigen-dependent cytokine generation and killing of DMG cells in vitro. In five independent patient-derived H3-K27M+ DMG orthotopic xenograft models, systemic administration of GD2-targeted CAR T cells cleared engrafted tumors except for a small number of residual GD2lo glioma cells. To date, GD2-targeted CAR T cells have been well tolerated in clinical trials15-17. Although GD2-targeted CAR T cell administration was tolerated in the majority of mice bearing orthotopic xenografts, peritumoral neuroinflammation during the acute phase of antitumor activity resulted in hydrocephalus that was lethal in a fraction of animals. Given the precarious neuroanatomical location of midline gliomas, careful monitoring and aggressive neurointensive care management will be required for human translation. With a cautious multidisciplinary clinical approach, GD2-targeted CAR T cell therapy for H3-K27M+ diffuse gliomas of pons, thalamus and spinal cord could prove transformative for these lethal childhood cancers.


Subject(s)
Brain Neoplasms/immunology , Gangliosides/metabolism , Glioma/immunology , Histones/metabolism , Immunotherapy, Adoptive , Lysine/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Humans , Methylation , Mice , Xenograft Model Antitumor Assays
7.
Am J Physiol Lung Cell Mol Physiol ; 311(5): L903-L912, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27638902

ABSTRACT

Acute respiratory distress syndrome (ARDS) remains a leading cause of morbidity and mortality in both adult and pediatric intensive care units. A key event in the development of ARDS is neutrophil recruitment into the lungs leading to tissue damage and destruction. Interleukin-8 (IL-8) is the major human chemokine responsible for neutrophil recruitment into the lungs. Protein phosphatase 2A (PP2A) has been shown to be a key regulator of the mitogen-activated protein kinase (MAPK) cascades, which control the production of IL-8. Previously, our laboratory employed an in vitro model to show that inhibition of PP2A results in an increase in IL-8 production in human alveolar epithelial cells. The objective of this study was to determine whether PP2A regulated this response in vivo by investigating the impact of pharmacologic activation of PP2A on chemokine production and activation of the MAPK cascade and lung injury using endotoxin- and bacterial-challenge models of ARDS in mice. N6-cyclopentyladenosine (N6-CPA) increased PP2A activity and inhibited endotoxin-induced cytokine production in a murine alveolar macrophage cell line. N6-CPA pretreatment in mice challenged with intratracheal endotoxin decreased chemokine production, reduced neutrophil infiltration, and attenuated lung injury. Following initiation of lung injury with live Pseudomonas aeruginosa, mice that received N6-CPA 4 h following bacterial challenge showed attenuated chemokine production and reduced neutrophil infiltration compared with control mice. Pharmacologic PP2A activation both limited and prevented inflammation and tissue injury in two direct injury models of ARDS. These results suggest modulation of PP2A activity as a therapeutic target in ARDS.


Subject(s)
Acute Lung Injury/enzymology , Inflammation/metabolism , Inflammation/pathology , Protein Phosphatase 2/metabolism , Acute Lung Injury/pathology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Cell Line , Chemokines/biosynthesis , Disease Models, Animal , Endotoxins , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Mice, Inbred C57BL , Phosphorylation/drug effects , Pseudomonas aeruginosa/drug effects , Respiratory Distress Syndrome/enzymology , Respiratory Distress Syndrome/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...