Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Clin Invest ; 134(8)2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38441967

ABSTRACT

Antitumor responses of CD8+ T cells are tightly regulated by distinct metabolic fitness. High levels of glutathione (GSH) are observed in the majority of tumors, contributing to cancer progression and treatment resistance in part by preventing glutathione peroxidase 4-dependent (GPX4-dependent) ferroptosis. Here, we show the necessity of adenosine A2A receptor (A2AR) signaling and the GSH/GPX4 axis in orchestrating metabolic fitness and survival of functionally competent CD8+ T cells. Activated CD8+ T cells treated ex vivo with simultaneous inhibition of A2AR and lipid peroxidation acquire a superior capacity to proliferate and persist in vivo, demonstrating a translatable means to prevent ferroptosis in adoptive cell therapy. Additionally, we identify a particular cluster of intratumoral CD8+ T cells expressing a putative gene signature of GSH metabolism (GMGS) in association with clinical response and survival across several human cancers. Our study addresses a key role of GSH/GPX4 and adenosinergic pathways in fine-tuning the metabolic fitness of antitumor CD8+ T cells.


Subject(s)
Neoplasms , Receptor, Adenosine A2A , Humans , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Neoplasms/genetics , Neoplasms/therapy , Neoplasms/metabolism , CD8-Positive T-Lymphocytes/metabolism , Glutathione/metabolism
2.
GEN Biotechnol ; 2(3): 228-246, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37363412

ABSTRACT

Off-the shelf immune cell therapies are potentially curative and may offer cost and manufacturing advantages over autologous products, but further development is needed. The NK92 cell line has a natural killer-like phenotype, has efficacy in cancer clinical trials, and is safe after irradiation. However, NK92 cells lose activity post-injection, limiting efficacy. This may be addressed by engineering NK92 cells to express stimulatory factors, and comparative analysis is needed. Thus, we systematically explored the expression of synthetic cytokines for enhancing NK92 cell production and performance. All synthetic cytokines evaluated (membrane-bound IL2 and IL15, and engineered versions of Neoleukin-2/15, IL15, IL12, and decoy resistant IL18) enhanced NK92 cell cytotoxicity. Engineered cells were preferentially expanded by expressing membrane-bound but not soluble synthetic cytokines, without compromising the radiosensitivity required for safety. Some membrane-bound cytokines conferred cell-contact independent paracrine activity, partly attributable to extracellular vesicles. Finally, we characterized interactions within consortia of differently engineered NK92 cells.

3.
Front Immunol ; 11: 1333, 2020.
Article in English | MEDLINE | ID: mdl-32733447

ABSTRACT

Although the strategy of therapeutic vaccination for the treatment of prostate cancer has advanced to and is available in the clinic (Sipuleucel-T), the efficacy of such therapy remains limited. Here, we develop Immunostimulatory Spherical Nucleic Acid (IS-SNA) nanostructures comprised of CpG oligonucleotides as adjuvant and prostate cancer peptide antigens, and evaluate their antitumor efficacy in syngeneic mouse models of prostate cancer. IS-SNAs with the specific structural feature of presenting both antigen and adjuvant CpG on the surface (hybridized model (HM) SNAs) induce stronger cytotoxic T lymphocyte (CTL) mediated antigen-specific killing of target cells than that for IS-SNAs with CpG on the surface and antigen encapsulated within the core (encapsulated model (EM) SNAs). Mechanistically, HM SNAs increase the co-delivery of CpG and antigen to dendritic cells over that for EM SNAs or admixtures of linear CpG and peptide, thereby improving cross-priming of antitumor CD8+ T cells. As a result, vaccination with HM SNAs leads to more effective antitumor immune responses in two prostate cancer models. These data demonstrate the importance of the structural positioning of peptide antigens together with adjuvants within IS-SNAs to the efficacy of IS-SNA-based cancer immunotherapy.


Subject(s)
Cancer Vaccines/pharmacology , Immunotherapy/methods , Nanostructures , Oligodeoxyribonucleotides/pharmacology , Prostatic Neoplasms , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/pharmacology , Cancer Vaccines/immunology , Cross-Priming/drug effects , Cross-Priming/immunology , Cytotoxicity, Immunologic/drug effects , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/immunology , Prostatic Neoplasms/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
5.
Blood ; 132(12): 1265-1278, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30064973

ABSTRACT

Genetic studies have identified recurrent somatic mutations in acute myeloid leukemia (AML) patients, including in the Wilms' tumor 1 (WT1) gene. The molecular mechanisms by which WT1 mutations contribute to leukemogenesis have not yet been fully elucidated. We investigated the role of Wt1 gene dosage in steady-state and pathologic hematopoiesis. Wt1 heterozygous loss enhanced stem cell self-renewal in an age-dependent manner, which increased stem cell function over time and resulted in age-dependent leukemic transformation. Wt1-haploinsufficient leukemias were characterized by progressive genetic and epigenetic alterations, including those in known leukemia-associated alleles, demonstrating a requirement for additional events to promote hematopoietic transformation. Consistent with this observation, we found that Wt1 depletion cooperates with Flt3-ITD mutation to induce fully penetrant AML. Our studies provide insight into mechanisms of Wt1-loss leukemogenesis and into the evolutionary events required to induce transformation of Wt1-haploinsufficient stem/progenitor cells.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Mutation , Repressor Proteins/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Self Renewal , Gene Deletion , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Leukemia, Myeloid, Acute/pathology , Leukopoiesis , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Myeloid Cells/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , WT1 Proteins , fms-Like Tyrosine Kinase 3/genetics
6.
J Clin Invest ; 128(2): 789-804, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29355841

ABSTRACT

Patients with myeloproliferative neoplasms (MPNs) frequently progress to bone marrow failure or acute myeloid leukemia (AML), and mutations in epigenetic regulators such as the metabolic enzyme isocitrate dehydrogenase (IDH) are associated with poor outcomes. Here, we showed that combined expression of Jak2V617F and mutant IDH1R132H or Idh2R140Q induces MPN progression, alters stem/progenitor cell function, and impairs differentiation in mice. Jak2V617F Idh2R140Q-mutant MPNs were sensitive to small-molecule inhibition of IDH. Combined inhibition of JAK2 and IDH2 normalized the stem and progenitor cell compartments in the murine model and reduced disease burden to a greater extent than was seen with JAK inhibition alone. In addition, combined JAK2 and IDH2 inhibitor treatment also reversed aberrant gene expression in MPN stem cells and reversed the metabolite perturbations induced by concurrent JAK2 and IDH2 mutations. Combined JAK2 and IDH2 inhibitor therapy also showed cooperative efficacy in cells from MPN patients with both JAK2mut and IDH2mut mutations. Taken together, these data suggest that combined JAK and IDH inhibition may offer a therapeutic advantage in this high-risk MPN subtype.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Isocitrate Dehydrogenase/genetics , Janus Kinase 2/genetics , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Aged , Animals , Disease Progression , Epigenesis, Genetic , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Middle Aged , Mutation , Phenotype , Stem Cells
8.
Leuk Res ; 49: 62-5, 2016 10.
Article in English | MEDLINE | ID: mdl-27568819

ABSTRACT

We examined the genetic implications and clinical impact of telomere length (TL) in 67 patients with acute myeloid leukemia (AML). There was a trend toward improved survival at 6 months in patients with longer TL. We found that patients with activating mutations, such as FLT3-ITD, had shorter TL, while those with mutations in epigenetic modifying enzymes, particularly IDH1 and IDH2, had longer TL. These are intriguing findings that warrant further investigation in larger cohorts. Our data show the potential of TL as a predictive biomarker in AML and identify genetic subsets that may be particularly vulnerable to telomere-targeted therapies.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Mutation , Telomere/ultrastructure , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Female , Humans , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Prognosis , Survival Analysis , Treatment Outcome , fms-Like Tyrosine Kinase 3/genetics
9.
J Exp Med ; 213(9): 1723-40, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27481130

ABSTRACT

Primary myelofibrosis (PMF) is a fatal neoplastic disease characterized by clonal myeloproliferation and progressive bone marrow (BM) fibrosis thought to be induced by mesenchymal stromal cells stimulated by overproduced growth factors. However, tissue fibrosis in other diseases is associated with monocyte-derived fibrocytes. Therefore, we sought to determine whether fibrocytes play a role in the induction of BM fibrosis in PMF. In this study, we show that BM from patients with PMF harbors an abundance of clonal, neoplastic collagen- and fibronectin-producing fibrocytes. Immunodeficient mice transplanted with myelofibrosis patients' BM cells developed a lethal myelofibrosis-like phenotype. Treatment of the xenograft mice with the fibrocyte inhibitor serum amyloid P (SAP; pentraxin-2) significantly prolonged survival and slowed the development of BM fibrosis. Collectively, our data suggest that neoplastic fibrocytes contribute to the induction of BM fibrosis in PMF, and inhibiting fibrocyte differentiation with SAP may interfere with this process.


Subject(s)
Fibroblasts/physiology , Monocytes/cytology , Primary Myelofibrosis/etiology , Animals , Bone Marrow/pathology , Bone Marrow Transplantation , Cells, Cultured , Fibroblasts/drug effects , Fibrosis , Homeodomain Proteins/pharmacology , Humans , Mice , Mice, SCID , Nitriles , Primary Myelofibrosis/pathology , Pyrazoles/pharmacology , Pyrimidines , Recombinant Proteins/pharmacology , Serum Amyloid P-Component/pharmacology
10.
Cancer Cell ; 28(1): 15-28, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-26175413

ABSTRACT

Although clinically tested JAK inhibitors reduce splenomegaly and systemic symptoms, molecular responses are not observed in most myeloproliferative neoplasm (MPN) patients. We previously demonstrated that MPN cells become persistent to type I JAK inhibitors that bind the active conformation of JAK2. We investigated whether CHZ868, a type II JAK inhibitor, would demonstrate activity in JAK inhibitor persistent cells, murine MPN models, and MPN patient samples. JAK2 and MPL mutant cell lines were sensitive to CHZ868, including type I JAK inhibitor persistent cells. CHZ868 showed significant activity in murine MPN models and induced reductions in mutant allele burden not observed with type I JAK inhibitors. These data demonstrate that type II JAK inhibition is a viable therapeutic approach for MPN patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Myeloproliferative Disorders/drug therapy , Protein Kinase Inhibitors/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Benzamides/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/administration & dosage , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism , Sequence Analysis, RNA , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
11.
Cell Rep ; 9(5): 1841-1855, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25482556

ABSTRACT

Somatic mutations in IDH1/IDH2 and TET2 result in impaired TET2-mediated conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). The observation that WT1 inactivating mutations anticorrelate with TET2/IDH1/IDH2 mutations in acute myeloid leukemia (AML) led us to hypothesize that WT1 mutations may impact TET2 function. WT1 mutant AML patients have reduced 5hmC levels similar to TET2/IDH1/IDH2 mutant AML. These mutations are characterized by convergent, site-specific alterations in DNA hydroxymethylation, which drive differential gene expression more than alterations in DNA promoter methylation. WT1 overexpression increases global levels of 5hmC, and WT1 silencing reduced 5hmC levels. WT1 physically interacts with TET2 and TET3, and WT1 loss of function results in a similar hematopoietic differentiation phenotype as observed with TET2 deficiency. These data provide a role for WT1 in regulating DNA hydroxymethylation and suggest that TET2 IDH1/IDH2 and WT1 mutations define an AML subtype defined by dysregulated DNA hydroxymethylation.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/physiology , WT1 Proteins/genetics , 5-Methylcytosine/analogs & derivatives , Animals , Cell Differentiation , Cytosine/analogs & derivatives , Cytosine/physiology , Dioxygenases , Enhancer Elements, Genetic , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/metabolism , Mice, Knockout , Mutation , Promoter Regions, Genetic , Protein Binding , Sequence Analysis, DNA
12.
Proc Natl Acad Sci U S A ; 111(50): E5401-10, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25516983

ABSTRACT

Patients with myeloproliferative neoplasms (MPNs) are at significant, cumulative risk of leukemic transformation to acute myeloid leukemia (AML), which is associated with adverse clinical outcome and resistance to standard AML therapies. We performed genomic profiling of post-MPN AML samples; these studies demonstrate somatic tumor protein 53 (TP53) mutations are common in JAK2V617F-mutant, post-MPN AML but not in chronic-phase MPN and lead to clonal dominance of JAK2V617F/TP53-mutant leukemic cells. Consistent with these data, expression of JAK2V617F combined with Tp53 loss led to fully penetrant AML in vivo. JAK2V617F-mutant, Tp53-deficient AML was characterized by an expanded megakaryocyte erythroid progenitor population that was able to propagate the disease in secondary recipients. In vitro studies revealed that post-MPN AML cells were sensitive to decitabine, the JAK1/2 inhibitor ruxolitinib, or the heat shock protein 90 inhibitor 8-(6-iodobenzo[d][1.3]dioxol-5-ylthio)-9-(3-(isopropylamino)propyl)-9H-purine-6-amine (PU-H71). Treatment with ruxolitinib or PU-H71 improved survival of mice engrafted with JAK2V617F-mutant, Tp53-deficient AML, demonstrating therapeutic efficacy for these targeted therapies and providing a rationale for testing these therapies in post-MPN AML.


Subject(s)
Hematologic Neoplasms/complications , Janus Kinase 2/genetics , Leukemia, Myeloid, Acute/genetics , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/genetics , Tumor Suppressor Protein p53/genetics , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Benzodioxoles/pharmacology , Blotting, Western , Colony-Forming Units Assay , Decitabine , Exome/genetics , Flow Cytometry , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/etiology , Mice , Mutation, Missense/genetics , Nitriles , Purines/pharmacology , Pyrazoles/pharmacology , Pyrimidines
13.
Blood ; 123(22): e123-33, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24740812

ABSTRACT

Genomic studies have identified somatic alterations in the majority of myeloproliferative neoplasms (MPN) patients, including JAK2 mutations in the majority of MPN patients and CALR mutations in JAK2-negative MPN patients. However, the role of JAK-STAT pathway activation in different MPNs, and in patients without JAK2 mutations, has not been definitively delineated. We used expression profiling, single nucleotide polymorphism arrays, and mutational profiling to investigate a well-characterized cohort of MPN patients. MPN patients with homozygous JAK2V617F mutations were characterized by a distinctive transcriptional profile. Notably, a transcriptional signature consistent with activated JAK2 signaling is seen in all MPN patients regardless of clinical phenotype or mutational status. In addition, the activated JAK2 signature was present in patients with somatic CALR mutations. Conversely, we identified a gene expression signature of CALR mutations; this signature was significantly enriched in JAK2-mutant MPN patients consistent with a shared mechanism of transformation by JAK2 and CALR mutations. We also identified a transcriptional signature of TET2 mutations in MPN patent samples. Our data indicate that MPN patients, regardless of diagnosis or JAK2 mutational status, are characterized by a distinct gene expression signature with upregulation of JAK-STAT target genes, demonstrating the central importance of the JAK-STAT pathway in MPN pathogenesis.


Subject(s)
Genomics , Janus Kinases/metabolism , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Calreticulin , Case-Control Studies , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cluster Analysis , Female , Gene Expression Profiling , Homozygote , Humans , Janus Kinase 2/genetics , Janus Kinases/genetics , Male , Mutation , STAT Transcription Factors/genetics , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...