Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
4.
Front Immunol ; 12: 757804, 2021.
Article in English | MEDLINE | ID: mdl-34630437

ABSTRACT

Defective DNA mismatch repair (dMMR) is associated with many cancer types including colon, gastric, endometrial, ovarian, hepatobiliary tract, urinary tract, brain and skin cancers. Lynch syndrome - a hereditary cause of dMMR - confers increased lifetime risk of malignancy in different organs and tissues. These Lynch syndrome pathogenic alleles are widely present in humans at a 1:320 population frequency of a single allele and associated with an up to 80% risk of developing microsatellite unstable cancer (microsatellite instability - high, or MSI-H). Advanced MSI-H tumors can be effectively treated with checkpoint inhibitors (CPI), however, that has led to response rates of only 30-60% despite their high tumor mutational burden and favorable immune gene signatures in the tumor microenvironment (TME). We and others have characterized a subset of MSI-H associated highly recurrent frameshift mutations that yield shared immunogenic neoantigens. These frameshifts might serve as targets for off-the-shelf cancer vaccine designs. In this review we discuss the current state of research around MSI-H cancer vaccine development, its application to MSI-H and Lynch syndrome cancer patients and the utility of MSI-H as a biomarker for CPI therapy. We also summarize the tumor intrinsic mechanisms underlying the high occurrence rates of certain frameshifts in MSI-H. Finally, we provide an overview of pivotal clinical trials investigating MSI-H as a biomarker for CPI therapy and MSI-H vaccines. Overall, this review aims to inform the development of novel research paradigms and therapeutics.


Subject(s)
Cancer Vaccines , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mismatch Repair/genetics , Immune Checkpoint Inhibitors/therapeutic use , Microsatellite Instability , Translational Science, Biomedical/trends , Biomarkers, Tumor , Cancer Vaccines/therapeutic use , Clinical Trials as Topic , Colorectal Neoplasms, Hereditary Nonpolyposis/immunology , Colorectal Neoplasms, Hereditary Nonpolyposis/prevention & control , Colorectal Neoplasms, Hereditary Nonpolyposis/therapy , Disease Management , Drug Repositioning , Drug Resistance/genetics , Frameshift Mutation , Humans , INDEL Mutation , Models, Genetic , Models, Immunological
5.
Blood Adv ; 5(23): 5086-5097, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34581778

ABSTRACT

Myelofibrosis (MF) is a clonal stem cell neoplasm characterized by abnormal JAK-STAT signaling, chronic inflammation, cytopenias, and risk of transformation to acute leukemia. Despite improvements in the therapeutic options for patients with MF, allogeneic hematopoietic stem cell transplantation remains the only curative treatment. We previously demonstrated multiple immunosuppressive mechanisms in patients with MF, including increased expression of programmed cell death protein 1 (PD-1) on T cells compared with healthy controls. Therefore, we conducted a multicenter, open-label, phase 2, single-arm study of pembrolizumab in patients with Dynamic International Prognostic Scoring System category of intermediate-2 or greater primary, post-essential thrombocythemia or post-polycythemia vera myelofibrosis that were ineligible for or were previously treated with ruxolitinib. The study followed a Simon 2-stage design and enrolled a total of 10 patients, 5 of whom had JAK2V617mutation, 2 had CALR mutation, and 6 had additional mutations. Most patients were previously treated with ruxolitinib. Pembrolizumab treatment was well tolerated, but there were no objective clinical responses, so the study closed after the first stage was completed. However, immune profiling by flow cytometry, T-cell receptor sequencing, and plasma proteomics demonstrated changes in the immune milieu of patients, which suggested improved T-cell responses that can potentially favor antitumor immunity. The fact that these changes were not reflected in a clinical response strongly suggests that combination immunotherapeutic approaches rather than monotherapy may be necessary to reverse the multifactorial mechanisms of immune suppression in myeloproliferative neoplasms. This trial was registered at www.clinicaltrials.gov as #NCT03065400.


Subject(s)
Myeloproliferative Disorders , Polycythemia Vera , Primary Myelofibrosis , Thrombocythemia, Essential , Humans , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/genetics , Programmed Cell Death 1 Receptor , Thrombocythemia, Essential/drug therapy , Thrombocythemia, Essential/genetics
6.
Cancer Cell ; 39(9): 1187-1189, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34478641

ABSTRACT

Despite the demonstrated efficacy and broad applicability of checkpoint blockade, the mechanisms by which it exerts its antitumor effects are incompletely understood. A recent article in Nature Medicine describes an ex vivo platform for assessing early responses to checkpoint blockade and the properties of tumor immune contexture in correlation to clinical responses.


Subject(s)
Neoplasms , Programmed Cell Death 1 Receptor , Biomarkers, Tumor , Humans , Immunity , Neoplasms/drug therapy , Organoids
7.
STAR Protoc ; 2(3): 100758, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34458873

ABSTRACT

Determining the antigen specificities of the endogenous T-cell repertoire is important for screening naturally occurring or therapy-induced T-cell immunity and may help identify novel targets for T-cell-based therapies. Here, we describe a rapid, sensitive, and high-throughput protocol for expanding antigen-specific T cells from human peripheral blood mononuclear cells in vitro following peptide stimulation and detecting antigen-specific effector cytokine formation by flow cytometry. Our approach can be applied to examining specific T-cell subsets from various tissues. For complete details on the use and execution of this protocol, please refer to Roudko et al. (2020) and Cimen Bozkus et al. (2019).


Subject(s)
Cytokines/metabolism , Immunologic Techniques/methods , T-Lymphocyte Subsets/immunology , Cell Culture Techniques/methods , Cryopreservation , Cytokines/pharmacology , Flow Cytometry , Humans , Immunologic Techniques/instrumentation , Leukocytes, Mononuclear/immunology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/metabolism
8.
Nat Rev Immunol ; 20(10): 593, 2020 10.
Article in English | MEDLINE | ID: mdl-32873888
9.
Cancer Discov ; 9(9): 1192-1207, 2019 09.
Article in English | MEDLINE | ID: mdl-31266769

ABSTRACT

Somatic frameshift mutations in the calreticulin (CALR) gene are key drivers of cellular transformation in myeloproliferative neoplasms (MPN). All patients carrying these mutations (CALR + MPN) share an identical sequence in the C-terminus of the mutated CALR protein (mut-CALR), with the potential for utility as a shared neoantigen. Here, we demonstrate that although a subset of patients with CALR + MPN develop specific T-cell responses against the mut-CALR C-terminus, PD-1 or CTLA4 expression abrogates the full complement of responses. Significantly, blockade of PD-1 and CLTA4 ex vivo by mAbs and of PD-1 in vivo by pembrolizumab administration restores mut-CALR-specific T-cell immunity in some patients with CALR + MPN. Moreover, mut-CALR elicits antigen-specific responses from both CD4+ and CD8+ T cells, confirming its broad applicability as an immunogen. Collectively, these results establish mut-CALR as a shared, MPN-specific neoantigen and inform the design of novel immunotherapies targeting mut-CALR. SIGNIFICANCE: Current treatment modalities for MPN are not effective in eliminating malignant cells. Here, we show that mutations in the CALR gene, which drive transformation in MPN, elicit T-cell responses that can be further enhanced by checkpoint blockade, suggesting immunotherapies could be employed to eliminate CALR + malignant cells in MPN.This article is highlighted in the In This Issue feature, p. 1143.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Calreticulin/genetics , Myeloproliferative Disorders/drug therapy , T-Lymphocytes/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/pharmacology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Calreticulin/chemistry , Calreticulin/immunology , Case-Control Studies , Cell Line, Tumor , Frameshift Mutation , Humans , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/immunology , Peptides/immunology
10.
J Immunol ; 195(11): 5237-50, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26491198

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature cells that expand during benign and cancer-associated inflammation and are characterized by their ability to inhibit T cell immunity. Increased metabolism of l-Arginine (l-Arg), through the enzymes arginase 1 and NO synthase 2 (NOS2), is well documented as a major MDSC suppressive mechanism. Therefore, we hypothesized that restricting MDSC uptake of l-Arg is a critical control point to modulate their suppressor activity. Using murine models of prostate-specific inflammation and cancer, we have identified the mechanisms by which extracellular l-Arg is transported into MDSCs. We have shown that MDSCs recruited to localized inflammation and tumor sites upregulate cationic amino acid transporter 2 (Cat2), coordinately with Arg1 and Nos2. Cat2 expression is not induced in MDSCs in peripheral organs. CAT2 contributes to the transport of l-Arg in MDSCs and is an important regulator of MDSC suppressive function. MDSCs that lack CAT2 have significantly reduced suppressive ability ex vivo and display impaired capacity for regulating T cell responses in vivo as evidenced by increased T cell expansion and decreased tumor growth in Cat2(-/-) mice. The abrogation of suppressive function is due to low intracellular l-Arg levels, which leads to the impaired ability of NOS2 to catalyze l-Arg-dependent metabolic processes. Together, these findings demonstrate that CAT2 modulates MDSC function. In the absence of CAT2, MDSCs display diminished capacity for controlling T cell immunity in prostate inflammation and cancer models, where the loss of CAT2 results in enhanced antitumor activity.


Subject(s)
Amino Acid Transport Systems, Basic/genetics , Cationic Amino Acid Transporter 2/biosynthesis , Myeloid Cells/immunology , Prostatic Neoplasms/immunology , T-Lymphocytes/immunology , Amino Acid Transport Systems, Basic/biosynthesis , Animals , Arginase/biosynthesis , Arginine/metabolism , Biological Transport , Cationic Amino Acid Transporter 2/genetics , Cell Line, Tumor , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Prostatic Neoplasms/pathology , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL