Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 4 de 4
1.
Nature ; 622(7984): 850-862, 2023 Oct.
Article En | MEDLINE | ID: mdl-37794185

Immune checkpoint blockade is effective for some patients with cancer, but most are refractory to current immunotherapies and new approaches are needed to overcome resistance1,2. The protein tyrosine phosphatases PTPN2 and PTPN1 are central regulators of inflammation, and their genetic deletion in either tumour cells or immune cells promotes anti-tumour immunity3-6. However, phosphatases are challenging drug targets; in particular, the active site has been considered undruggable. Here we present the discovery and characterization of ABBV-CLS-484 (AC484), a first-in-class, orally bioavailable, potent PTPN2 and PTPN1 active-site inhibitor. AC484 treatment in vitro amplifies the response to interferon and promotes the activation and function of several immune cell subsets. In mouse models of cancer resistant to PD-1 blockade, AC484 monotherapy generates potent anti-tumour immunity. We show that AC484 inflames the tumour microenvironment and promotes natural killer cell and CD8+ T cell function by enhancing JAK-STAT signalling and reducing T cell dysfunction. Inhibitors of PTPN2 and PTPN1 offer a promising new strategy for cancer immunotherapy and are currently being evaluated in patients with advanced solid tumours (ClinicalTrials.gov identifier NCT04777994 ). More broadly, our study shows that small-molecule inhibitors of key intracellular immune regulators can achieve efficacy comparable to or exceeding that of antibody-based immune checkpoint blockade in preclinical models. Finally, to our knowledge, AC484 represents the first active-site phosphatase inhibitor to enter clinical evaluation for cancer immunotherapy and may pave the way for additional therapeutics that target this important class of enzymes.


Immunotherapy , Neoplasms , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Drug Resistance, Neoplasm , Immune Checkpoint Inhibitors , Immunotherapy/methods , Interferons/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 2/antagonists & inhibitors , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
2.
Nat Immunol ; 23(10): 1495-1506, 2022 10.
Article En | MEDLINE | ID: mdl-36151395

The immune system can eliminate tumors, but checkpoints enable immune escape. Here, we identify immune evasion mechanisms using genome-scale in vivo CRISPR screens across cancer models treated with immune checkpoint blockade (ICB). We identify immune evasion genes and important immune inhibitory checkpoints conserved across cancers, including the non-classical major histocompatibility complex class I (MHC class I) molecule Qa-1b/HLA-E. Surprisingly, loss of tumor interferon-γ (IFNγ) signaling sensitizes many models to immunity. The immune inhibitory effects of tumor IFN sensing are mediated through two mechanisms. First, tumor upregulation of classical MHC class I inhibits natural killer cells. Second, IFN-induced expression of Qa-1b inhibits CD8+ T cells via the NKG2A/CD94 receptor, which is induced by ICB. Finally, we show that strong IFN signatures are associated with poor response to ICB in individuals with renal cell carcinoma or melanoma. This study reveals that IFN-mediated upregulation of classical and non-classical MHC class I inhibitory checkpoints can facilitate immune escape.


CD8-Positive T-Lymphocytes , Neoplasms , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Immune Checkpoint Inhibitors , Immune Evasion , Interferon-gamma/genetics , Interferon-gamma/metabolism , NK Cell Lectin-Like Receptor Subfamily C
3.
Immunity ; 54(3): 571-585.e6, 2021 03 09.
Article En | MEDLINE | ID: mdl-33497609

CRISPR-Cas9 genome engineering has increased the pace of discovery for immunology and cancer biology, revealing potential therapeutic targets and providing insight into mechanisms underlying resistance to immunotherapy. However, endogenous immune recognition of Cas9 has limited the applicability of CRISPR technologies in vivo. Here, we characterized immune responses against Cas9 and other expressed CRISPR vector components that cause antigen-specific tumor rejection in several mouse cancer models. To avoid unwanted immune recognition, we designed a lentiviral vector system that allowed selective CRISPR antigen removal (SCAR) from tumor cells. The SCAR system reversed immune-mediated rejection of CRISPR-modified tumor cells in vivo and enabled high-throughput genetic screens in previously intractable models. A pooled in vivo screen using SCAR in a CRISPR-antigen-sensitive renal cell carcinoma revealed resistance pathways associated with autophagy and major histocompatibility complex class I (MHC class I) expression. Thus, SCAR presents a resource that enables CRISPR-based studies of tumor-immune interactions and prevents unwanted immune recognition of genetically engineered cells, with implications for clinical applications.


Carcinoma, Renal Cell/immunology , Genetic Testing/methods , Genetic Vectors/genetics , Immunotherapy/methods , Kidney Neoplasms/immunology , Killer Cells, Natural/immunology , Lentivirus/genetics , Animals , Antigen Presentation , Autophagy , Carcinoma, Renal Cell/therapy , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats , Genetic Engineering , Histocompatibility Antigens Class I/metabolism , Kidney Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Targeted Therapy
4.
Nature ; 565(7737): 43-48, 2019 01.
Article En | MEDLINE | ID: mdl-30559380

Most patients with cancer either do not respond to immune checkpoint blockade or develop resistance to it, often because of acquired mutations that impair antigen presentation. Here we show that loss of function of the RNA-editing enzyme ADAR1 in tumour cells profoundly sensitizes tumours to immunotherapy and overcomes resistance to checkpoint blockade. In the absence of ADAR1, A-to-I editing of interferon-inducible RNA species is reduced, leading to double-stranded RNA ligand sensing by PKR and MDA5; this results in growth inhibition and tumour inflammation, respectively. Loss of ADAR1 overcomes resistance to PD-1 checkpoint blockade caused by inactivation of antigen presentation by tumour cells. Thus, effective anti-tumour immunity is constrained by inhibitory checkpoints such as ADAR1 that limit the sensing of innate ligands. The induction of sufficient inflammation in tumours that are sensitized to interferon can bypass the therapeutic requirement for CD8+ T cell recognition of cancer cells and may provide a general strategy to overcome immunotherapy resistance.


Adenosine Deaminase/deficiency , Adenosine Deaminase/metabolism , Cell Cycle Checkpoints/drug effects , Drug Resistance, Neoplasm/drug effects , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Adenosine Deaminase/genetics , Animals , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Histocompatibility Antigens Class I/immunology , Immunotherapy , Inflammation/genetics , Inflammation/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Interferons/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/radiotherapy , Mice , Mice, Inbred C57BL , Phenotype , RNA Editing , RNA, Double-Stranded/genetics , RNA-Binding Proteins/genetics , Receptors, G-Protein-Coupled/metabolism
...