Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Mol Ther Methods Clin Dev ; 32(1): 101171, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38298420

ABSTRACT

Chimeric antigen receptor T cells (CART) have demonstrated curative potential for hematological malignancies, but the optimal manufacturing has not yet been determined and may differ across products. The first step, T cell selection, removes contaminating cell types that can potentially suppress T cell expansion and transduction. While positive selection of CD4/CD8 T cells after leukapheresis is often used in clinical trials, it may modulate signaling cascades downstream of these co-receptors; indeed, the addition of a CD4/CD8-positive selection step altered CD22 CART potency and toxicity in patients. While negative selection may avoid this drawback, it is virtually absent from good manufacturing practices. Here, we performed both CD4/CD8-positive and -negative clinical scale selections of mononuclear cell apheresis products and generated CD22 CARTs per our ongoing clinical trial (NCT02315612NCT02315612). While the selection process did not yield differences in CART expansion or transduction, positively selected CART exhibited a significantly higher in vitro interferon-γ and IL-2 secretion but a lower in vitro tumor killing rate. Notably, though, CD22 CART generated from both selection protocols efficiently eradicated leukemia in NSG mice, with negatively selected cells exhibiting a significant enrichment in γδ CD22 CART. Thus, our study demonstrates the importance of the initial T cell selection process in clinical CART manufacturing.

2.
Haematologica ; 108(2): 457-471, 2023 02 01.
Article in English | MEDLINE | ID: mdl-35950535

ABSTRACT

Chimeric antigen receptor (CAR) T-cell immunotherapies targeting CD19 or CD22 induce remissions in the majority of patients with relapsed/refractory B-cell acute lymphoblastic leukemia (ALL), although relapse due to target antigen loss or downregulation has emerged as a major clinical dilemma. Accordingly, great interest exists in developing CAR T cells directed against alternative leukemia cell surface antigens that may help to overcome immunotherapeutic resistance. The fms-like tyrosine kinase 3 receptor (FLT3) is constitutively activated via FLT3 mutation in acute myeloid leukemia (AML) or wild-type FLT3 overexpression in KMT2A (lysine-specific methyltransferase 2A)-rearranged ALL, which are associated with poor clinical outcomes in children and adults. We developed monovalent FLT3-targeted CAR T cells (FLT3CART) and bispecific CD19xFLT3CART and assessed their anti-leukemia activity in preclinical models of FLT3-mutant AML and KMT2A-rearranged infant ALL. We report robust in vitro FLT3CART-induced cytokine production and cytotoxicity against AML and ALL cell lines with minimal cross-reactivity against normal hematopoietic and non-hematopoietic tissues. We also observed potent in vivo inhibition of leukemia proliferation in xenograft models of both FLT3-mutant AML and KMT2A-rearranged ALL, including a post-tisagenlecleucel ALL-to-AML lineage switch patient-derived xenograft model pairing. We further demonstrate significant in vitro and in vivo activity of bispecific CD19xFLT3CART against KMT2Arearranged ALL and posit that this additional approach might also diminish potential antigen escape in these high-risk leukemias. Our preclinical data credential FLT3CART as a highly effective immunotherapeutic strategy for both FLT3- mutant AML and KMT2A-rearranged ALL which is poised for further investigation and clinical translation.


Subject(s)
Leukemia, Myeloid, Acute , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Receptors, Chimeric Antigen , Adult , Child , Humans , Receptors, Chimeric Antigen/genetics , Leukemia, Myeloid, Acute/genetics , Immunotherapy , T-Lymphocytes/metabolism , fms-Like Tyrosine Kinase 3/genetics
3.
Blood ; 140(5): 451-463, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35605184

ABSTRACT

Remission durability following single-antigen targeted chimeric antigen receptor (CAR) T-cells is limited by antigen modulation, which may be overcome with combinatorial targeting. Building upon our experiences targeting CD19 and CD22 in B-cell acute lymphoblastic leukemia (B-ALL), we report on our phase 1 dose-escalation study of a novel murine stem cell virus (MSCV)-CD19/CD22-4-1BB bivalent CAR T-cell (CD19.22.BBζ) for children and young adults (CAYA) with B-cell malignancies. Primary objectives included toxicity and dose finding. Secondary objectives included response rates and relapse-free survival (RFS). Biologic correlatives included laboratory investigations, CAR T-cell expansion and cytokine profiling. Twenty patients, ages 5.4 to 34.6 years, with B-ALL received CD19.22.BBζ. The complete response (CR) rate was 60% (12 of 20) in the full cohort and 71.4% (10 of 14) in CAR-naïve patients. Ten (50%) developed cytokine release syndrome (CRS), with 3 (15%) having ≥ grade 3 CRS and only 1 experiencing neurotoxicity (grade 3). The 6- and 12-month RFS in those achieving CR was 80.8% (95% confidence interval [CI]: 42.4%-94.9%) and 57.7% (95% CI: 22.1%-81.9%), respectively. Limited CAR T-cell expansion and persistence of MSCV-CD19.22.BBζ compared with EF1α-CD22.BBζ prompted laboratory investigations comparing EF1α vs MSCV promoters, which did not reveal major differences. Limited CD22 targeting with CD19.22.BBζ, as evaluated by ex vivo cytokine secretion and leukemia eradication in humanized mice, led to development of a novel bicistronic CD19.28ζ/CD22.BBζ construct with enhanced cytokine production against CD22. With demonstrated safety and efficacy of CD19.22.BBζ in a heavily pretreated CAYA B-ALL cohort, further optimization of combinatorial antigen targeting serves to overcome identified limitations (www.clinicaltrials.gov #NCT03448393).


Subject(s)
Burkitt Lymphoma , Lymphoma, B-Cell , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Receptors, Chimeric Antigen , Animals , Antigens, CD19 , Cytokine Release Syndrome , Cytokines , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Recurrence , T-Lymphocytes
4.
J Immunother Cancer ; 9(9)2021 09.
Article in English | MEDLINE | ID: mdl-34531250

ABSTRACT

BACKGROUND: Successful development of chimeric antigen receptor (CAR) T cell immunotherapy for children and adults with relapsed/refractory acute myeloid leukemia (AML) is highly desired given their poor clinical prognosis and frequent inability to achieve cure with conventional chemotherapy. Initial experiences with CD19 CAR T cell immunotherapy for patients with B-cell malignancies highlighted the critical impact of intracellular costimulatory domain selection (CD28 vs 4-1BB (CD137)) on CAR T cell expansion and in vivo persistence that may impact clinical outcomes. However, the impact of costimulatory domains on the efficacy of myeloid antigen-directed CAR T cell immunotherapy remains unknown. METHODS: In this preclinical study, we developed six CAR constructs targeting CD33, a highly expressed and validated AML target, comprised of one of three single-chain variable fragments with CD3ζ and either CD28 or 4-1BB costimulatory domains. We systematically compared the preclinical in vitro and in vivo efficacy of T cells lentivirally transduced with CD33 CAR constructs (CD33CARTs) against human AML. RESULTS: We observed potent in vitro cytokine production and cytotoxicity of CD33CARTs incubated with human CD33+ AML cell lines, as well as robust in vivo antileukemia activity in cell line and childhood AML patient-derived xenograft (PDX) models. Gemtuzumab-based CD33CARTs were unexpectedly toxic in vivo in animal models despite observed in vitro anti-leukemia activity. CD28-based CD33CARTs consistently induced more robust inhibition of leukemia proliferation in AML cell line and PDX models than did 4-1BB-based CD33CARTs. A 'best-in-class' lintuzumab-CD28/CD3ζ CAR construct was thus selected for clinical translation. CONCLUSIONS: CD33 is a critical antigen for potential immunotherapeutic targeting in patients with AML. Based on this rigorous preclinical evaluation, our validated clinical grade lintuzumab-CD28/CD3ζ CD33CART immunotherapy is now under evaluation in a first-in-child/first-in-human phase 1 clinical trial for children and adolescents/young adults with relapsed/refractory AML. TRIAL REGISTRATION NUMBER: clinicaltrials.gov; NCT03971799.


Subject(s)
Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/drug therapy , Receptors, Chimeric Antigen/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , T-Lymphocytes/immunology , Animals , Female , Humans , Male , Mice
5.
J Clin Invest ; 130(10): 5425-5443, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32925169

ABSTRACT

Late-onset inflammatory toxicities resembling hemophagocytic lymphohistiocytosis (HLH) or macrophage activation syndrome (MAS) occur after chimeric antigen receptor T cell (CAR T cell) infusion and represent a therapeutic challenge. Given the established link between perforin deficiency and primary HLH, we investigated the role of perforin in anti-CD19 CAR T cell efficacy and HLH-like toxicities in a syngeneic murine model. Perforin contributed to both CD8+ and CD4+ CAR T cell cytotoxicity but was not required for in vitro or in vivo leukemia clearance. Upon CAR-mediated in vitro activation, perforin-deficient CAR T cells produced higher amounts of proinflammatory cytokines compared with WT CAR T cells. Following in vivo clearance of leukemia, perforin-deficient CAR T cells reexpanded, resulting in splenomegaly with disruption of normal splenic architecture and the presence of hemophagocytes, which are findings reminiscent of HLH. Notably, a substantial fraction of patients who received anti-CD22 CAR T cells also experienced biphasic inflammation, with the second phase occurring after the resolution of cytokine release syndrome, resembling clinical manifestations of HLH. Elevated inflammatory cytokines such as IL-1ß and IL-18 and concurrent late CAR T cell expansion characterized the HLH-like syndromes occurring in the murine model and in humans. Thus, a murine model of perforin-deficient CAR T cells recapitulated late-onset inflammatory toxicities occurring in human CAR T cell recipients, providing therapeutically relevant mechanistic insights.


Subject(s)
Immunotherapy, Adoptive/adverse effects , Perforin/deficiency , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Animals , Cytokines/biosynthesis , Disease Models, Animal , Humans , In Vitro Techniques , Inflammation Mediators/metabolism , Lymphohistiocytosis, Hemophagocytic/etiology , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/pathology , Macrophage Activation Syndrome/etiology , Macrophage Activation Syndrome/immunology , Macrophage Activation Syndrome/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Perforin/genetics , T-Lymphocytes/pathology
6.
Sci Transl Med ; 9(417)2017 Nov 22.
Article in English | MEDLINE | ID: mdl-29167392

ABSTRACT

Chimeric antigen receptor (CAR)-expressing T cells induce durable remissions in patients with relapsed/refractory B cell malignancies. CARs are synthetic constructs that, when introduced into mature T cells, confer a second, non-major histocompatibility complex-restricted specificity in addition to the endogenous T cell receptor (TCR). The implications of TCR activation on CAR T cell efficacy has not been well defined. Using an immunocompetent, syngeneic murine model of CD19-targeted CAR T cell therapy for pre-B cell acute lymphoblastic leukemia in which the CAR is introduced into T cells with known TCR specificity, we demonstrate loss of CD8 CAR T cell efficacy associated with T cell exhaustion and apoptosis when TCR antigen is present. CD4 CAR T cells demonstrate equivalent cytotoxicity to CD8 CAR T cells and, in contrast, retain in vivo efficacy despite TCR stimulation. Gene expression profiles confirm increased exhaustion and apoptosis of CD8 CAR T cells upon dual receptor stimulation compared to CD4 CAR T cells and indicate inherent differences between CD4 and CD8 CAR T cells in the use of T cell-associated signaling pathways. These results provide insights into important aspects of CAR T cell immune biology and indicate opportunities to rationally design CAR constructs to optimize clinical efficacy.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Apoptosis/physiology , Cell Line , Female , Flow Cytometry , Lymphocyte Activation/physiology , Mice , Mice, Inbred C57BL , Receptors, Antigen/metabolism
7.
Pharmacol Ther ; 168: 53-60, 2016 12.
Article in English | MEDLINE | ID: mdl-27595927

ABSTRACT

Hematopoietic cells are increasingly recognized as playing key roles in tumor growth and metastatic progression. Although many studies have focused on the functional interaction of hematopoietic cells with tumor cells, few have examined the regulation of hematopoiesis by the hematopoietic stem cell (HSC) niche in the setting of cancer. Hematopoiesis occurs primarily in the bone marrow, and processes including expansion, mobilization, and differentiation of hematopoietic progenitors are tightly regulated by the specialized stem cell niche. Loss of niche components or the ability of stem cells to localize to the stem cell niche relieves HSCs of the restrictions imposed under normal homeostasis. In this review, we discuss how tumor-derived factors and therapeutic interventions disrupt structural and regulatory properties of the stem cell niche, resulting in niche invasion by hematopoietic malignancies, extramedullary hematopoiesis, myeloid skewing by peripheral tissue microenvironments, and lymphopenia. The key regulatory roles played by the bone marrow niche in hematopoiesis has implications for therapy-related toxicity and the successful development of immune-based therapies for cancer.


Subject(s)
Hematopoietic Stem Cells/metabolism , Neoplasms/metabolism , Stem Cell Niche , Animals , Disease Progression , Hematologic Neoplasms/immunology , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/therapy , Hematopoiesis/physiology , Humans , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/therapy , Neoplastic Stem Cells/metabolism , Tumor Microenvironment
8.
Nat Commun ; 7: 12320, 2016 07 27.
Article in English | MEDLINE | ID: mdl-27460500

ABSTRACT

Adoptive immunotherapy using chimeric antigen receptor (CAR) expressing T cells targeting the CD19 B lineage receptor has demonstrated marked success in relapsed pre-B-cell acute lymphoblastic leukaemia (ALL). Persisting CAR-T cells generate sustained pressure against CD19 that may drive unique mechanisms of resistance. Pre-B ALL originates from a committed pre-B cell or an earlier progenitor, with potential to reprogram into other hematopoietic lineages. Here we report changes in lineage markers including myeloid conversion in patients following CD19 CAR therapy. Using murine ALL models we study the long-term effects of CD19 CAR-T cells and demonstrate partial or complete lineage switch as a consistent mechanism of CAR resistance depending on the underlying genetic oncogenic driver. Deletion of Pax5 or Ebf1 recapitulates lineage reprogramming occurring during CD19 CAR pressure. Our findings establish lineage switch as a mechanism of CAR resistance exposing inherent plasticity in genetic subtypes of pre-B-cell ALL.


Subject(s)
Cell Lineage , Cell Plasticity , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Cell Line, Tumor , Chromatin/metabolism , Clone Cells , Epigenesis, Genetic , Gene Editing , Genome , Mice, Knockout , Myeloid Cells/metabolism , Myeloid Cells/pathology , Phenotype , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Recurrence
9.
Blood ; 127(10): 1361-70, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26660684

ABSTRACT

Acute lymphoblastic leukemia (ALL) persisting or relapsing following bone marrow transplantation (BMT) has a dismal prognosis. Success with chimeric antigen receptor (CAR) T cells offers an opportunity to treat these patients with leukemia-redirected donor-derived T cells, which may be more functional than T cells derived from patients with leukemia but have the potential to mediate graft-versus-host disease (GVHD). We, together with others, have previously demonstrated tumor-specific T-cell dysfunction in the allogeneic environment. Here, we studied CAR T-cell function following BMT using an immunocompetent murine model of minor mismatched allogeneic transplantation followed by donor-derived CD19-CAR T cells. Allogeneic donor-derived CD19-CAR T cells eliminated residual ALL with equal potency to those administered after syngeneic BMT. Surprisingly, allogeneic CAR T cells mediated lethal acute GVHD with early mortality, which is atypical for this minor mismatch model. We demonstrated that both allogeneic and syngeneic CAR T cells show initial expansion as effector T cells, with a higher peak but rapid deletion of allogeneic CAR T cells. Interestingly, CAR-mediated acute GVHD was only seen in the presence of leukemia, suggesting CAR-target interactions induced GVHD. Indeed, serum interleukin (IL)-6 was elevated only in the presence of both leukemia and CAR T cells, and IL-6 neutralization ameliorated the severity of GVHD in a delayed donor lymphocyte infusion model. Finally, allogeneic CD4(+) CAR T cells were responsible for GVHD, which correlated with their ability to produce IL-6 upon CAR stimulation. Altogether, we demonstrate that donor-derived allogeneic CAR T cells are active but have the capacity to drive GVHD.


Subject(s)
Antigens, CD19/immunology , Graft vs Host Disease/immunology , Lymphocyte Transfusion , Neoplasms, Experimental/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, G-Protein-Coupled/immunology , T-Lymphocytes/immunology , Allografts , Animals , Antigens, CD19/genetics , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Calcium-Sensing , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/pathology , T-Lymphocytes/transplantation
10.
Blood ; 124(12): 1976-86, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25079358

ABSTRACT

Selective targeting of non-T cells, including antigen-presenting cells (APCs), is a potential strategy to prevent graft-versus-host-disease (GVHD) but to maintain graft-versus-tumor (GVT) effects. Because type I and II interferons signal through signal transducer and activator of transcription-1 (STAT1), and contribute to activation of APCs after allogeneic bone marrow transplant (alloBMT), we examined whether the absence of STAT1 in donor APCs could prevent GVHD while preserving immune competence. Transplantation of STAT1(-/-) bone marrow (BM) prevented GVHD induced by STAT1(+/+) T cells, leading to expansion of B220(+) cells and regulatory T cells. STAT1(-/-) BM also preserved GVT activity and enhanced overall survival of tumor-challenged mice in the setting of GVHD. Furthermore, recipients of allogeneic STAT1(-/-) BM demonstrated increased CD9(-)Siglec H(hi) plasmacytoid dendritic cells (pDCs), and depletion of pDCs after STAT1(-/-) BM transplantation prevented GVHD resistance. STAT1(-/-) pDCs were found to produce decreased free radicals, IFNα, and interleukin (IL)-12, and increased IL-10. Additionally, STAT1(-/-) pDCs that were isolated after alloBMT showed increased gene expression of S100A8 and S100A9, and transplantation of S100A9(-/-) BM reduced GVHD-free survival. Finally, elevated STAT3 was found in STAT1(-/-) pDCs isolated after alloBMT. We conclude that interfering with interferon signaling in APCs such as pDCs provides a novel approach to regulate the GVHD/GVT axis.


Subject(s)
Dendritic Cells/metabolism , Dendritic Cells/transplantation , Graft vs Host Disease/prevention & control , STAT1 Transcription Factor/deficiency , STAT3 Transcription Factor/metabolism , Allografts , Animals , Bone Marrow Transplantation/adverse effects , Calgranulin A/genetics , Calgranulin B/genetics , Calgranulin B/metabolism , Dendritic Cells/immunology , Female , Gene Expression , Graft vs Host Disease/etiology , Graft vs Host Disease/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , STAT1 Transcription Factor/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tissue Donors
11.
Front Oncol ; 4: 95, 2014.
Article in English | MEDLINE | ID: mdl-24847444

ABSTRACT

Leukemia remains the most common diagnosis in pediatric oncology and, despite dramatic progress in upfront therapy, is also the most common cause of cancer-related death in children. Much of the initial improvement in outcomes for acute lymphoblastic leukemia (ALL) was due to identification of cytotoxic agents that are active against leukemia followed by the recognition that combination of these cytotoxic agents and prolonged therapy are essential for cure. Recent data demonstrating lack of progress in patients for whom standard chemotherapy fails suggests that the ability to improve outcome for these children will not be dramatically impacted through more intensive or newer cytotoxic agents. Thus, much of the recent research focus has been in the area of improving our understanding of the genetics and the biology of leukemia. Although in vitro studies remain critical, given the complexity of a living system and the increasing recognition of the contribution of leukemia extrinsic factors such as the bone marrow microenvironment, in vivo models have provided important insights. The murine systems that are used can be broadly categorized into syngeneic models in which a murine leukemia can be studied in immunologically intact hosts and xenograft models where human leukemias are studied in highly immunocompromised murine hosts. Both of these systems have limitations such that neither can be used exclusively to study all aspects of leukemia biology and therapeutics for humans. This review will describe the various ALL model systems that have been developed as well as discuss the advantages and disadvantages inherent to these systems that make each particularly suitable for specific types of studies.

12.
Mol Endocrinol ; 28(6): 846-59, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24678732

ABSTRACT

Transcription of the HER2 oncogene can be repressed by estrogen (E2). We now show that, a splice isoform of the nuclear receptor coactivator AIB1, AIB1-Δ4, is able to reverse E2 repression of HER2 gene expression in breast cancer cells. The first 224 amino acids of AIB1 that are absent in AIB1-Δ4, bind a co-repressor, ANCO1. Using chromatin immunoprecipitation assay approaches in MCF7 and BT474 cell lines, we demonstrate that AIB1 and AIB1-Δ4 can bind to the E2 regulatory site in the first intron of the HER2 gene, after E2 treatment, but only full-length AIB1 recruits ANCO1. Consistent with E2-induced chromatin repression, the AIB1-ANCO1 complex recruits HDAC3 and HDAC4 to the intronic estrogen response element and the proximal promoter acquires the repressive chromatin mark H3K9me3 and loses H3K4me1. In contrast, AIB1-Δ4 does not recruit ANCO 1, HDAC3, or HDAC4 and the proximal promoter retains activation marks of H3K4me1. In cell lines with low levels of ANCO1 (T47D), E2 does not repress HER2 gene transcription but the repressive response can be restored by overexpression of ANCO1. ANCO1 can also repress other E2-responsive genes, indicating that AIB1, AIB1-Δ4 and ANCO1 are important determinants of endocrine and growth factor responsiveness in breast cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Nuclear Receptor Coactivator 3/physiology , Receptor, ErbB-2/genetics , Repressor Proteins/physiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chromatin Assembly and Disassembly , Estradiol/physiology , Female , HEK293 Cells , Histone Deacetylases/metabolism , Humans , MCF-7 Cells , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Oncogenes , Protein Binding , Receptor, ErbB-2/metabolism , Response Elements , Signal Transduction , Transcription, Genetic
13.
Mol Endocrinol ; 27(7): 1113-27, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23660594

ABSTRACT

The oncogene nuclear receptor coactivator amplified in breast cancer 1 (AIB1) is a transcriptional coactivator that is overexpressed in various types of human cancers. However, the molecular mechanisms controlling AIB1 expression in the majority of cancers remain unclear. In this study, we identified a novel interacting protein of AIB1, forkhead-box protein G1 (FoxG1), which is an evolutionarily conserved forkhead-box transcriptional corepressor. We show that FoxG1 expression is low in breast cancer cell lines and that low levels of FoxG1 are correlated with a worse prognosis in breast cancer. We also demonstrate that transient overexpression of FoxG1 can suppress endogenous levels of AIB1 mRNA and protein in MCF-7 breast cancer cells. Exogenously expressed FoxG1 in MCF-7 cells also leads to apoptosis that can be rescued in part by AIB1 overexpression. Using chromatin immunoprecipitation, we determined that FoxG1 is recruited to a region of the AIB1 gene promoter previously characterized to be responsible for AIB1-induced, positive autoregulation of transcription through the recruitment of an activating, multiprotein complex, involving AIB1, E2F transcription factor 1, and specificity protein 1. Increased FoxG1 expression significantly reduces the recruitment of AIB1, E2F transcription factor 1 and E1A-binding protein p300 to this region of the endogenous AIB1 gene promoter. Our data imply that FoxG1 can function as a pro-apoptotic factor in part through suppression of AIB1 coactivator transcription complex formation, thereby reducing the expression of the AIB1 oncogene.


Subject(s)
Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Receptor Coactivator 3/genetics , Repressor Proteins/metabolism , Transcription, Genetic , Apoptosis/genetics , Down-Regulation/genetics , E2F1 Transcription Factor/metabolism , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , MCF-7 Cells , Models, Biological , Nuclear Receptor Coactivator 3/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Protein Stability , Sp1 Transcription Factor/metabolism
14.
Mol Cell ; 46(6): 759-70, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22578841

ABSTRACT

The attachment of the SUMO modifier to proteins controls cellular signaling pathways through noncovalent binding to SUMO-interaction motifs (SIMs). Canonical SIMs contain a core of hydrophobic residues that bind to a hydrophobic pocket on SUMO. Negatively charged residues of SIMs frequently contribute to binding by interacting with a basic surface on SUMO. Here we define acetylation within this basic interface as a central mechanism for the control of SUMO-mediated interactions. The acetyl-mediated neutralization of basic charges on SUMO prevents binding to SIMs in PML, Daxx, and PIAS family members but does not affect the interaction between RanBP2 and SUMO. Acetylation is controlled by HDACs and attenuates SUMO- and PIAS-mediated gene silencing. Moreover, it affects the assembly of PML nuclear bodies and restrains the recruitment of the corepressor Daxx to these structures. This acetyl-dependent switch thus expands the regulatory repertoire of SUMO signaling and determines the selectivity and dynamics of SUMO-SIM interactions.


Subject(s)
Protein Interaction Maps , SUMO-1 Protein/metabolism , Acetylation , Binding Sites , Gene Silencing , HEK293 Cells , HeLa Cells , Histone Deacetylases/metabolism , Humans , SUMO-1 Protein/genetics
15.
J Biol Chem ; 286(30): 26813-27, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21636853

ABSTRACT

The oncogene amplified in breast cancer 1 (AIB1) is a nuclear receptor coactivator that plays a major role in the progression of various cancers. We previously identified a splice variant of AIB1 called AIB1-Δ4 that is overexpressed in breast cancer. Using mass spectrometry, we define the translation initiation of AIB1-Δ4 at Met(224) of the full-length AIB1 sequence and have raised an antibody to a peptide representing the acetylated N terminus. We show that AIB1-Δ4 is predominantly localized in the cytoplasm, although leptomycin B nuclear export inhibition demonstrates that AIB1-Δ4 can enter and traffic through the nucleus. Our data indicate an import mechanism enhanced by other coactivators such as p300/CBP. We report that the endogenously and exogenously expressed AIB1-Δ4 is recruited as efficiently as full-length AIB1 to estrogen-response elements of genes, and it enhances estrogen-dependent transcription more effectively than AIB1. Expression of an N-terminal AIB1 protein fragment, which is lost in the AIB1-Δ4 isoform, potentiates AIB1 as a coactivator. This suggests a model whereby the transcriptional activity of AIB1 is squelched by a repressive mechanism utilizing the N-terminal domain and that the increased coactivator function of AIB1-Δ4 is due to the loss of this inhibitory domain. Finally, we show, using Scorpion primer technology, that AIB1-Δ4 expression is correlated with metastatic capability of human cancer cell lines.


Subject(s)
Cell Nucleus/metabolism , Nuclear Receptor Coactivator 3/metabolism , Transcription, Genetic , Active Transport, Cell Nucleus/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , CHO Cells , COS Cells , Cell Nucleus/genetics , Chlorocebus aethiops , Cricetinae , Cricetulus , Cytoplasm/genetics , Cytoplasm/metabolism , Dogs , Fatty Acids, Unsaturated/pharmacology , HEK293 Cells , Humans , Mice , Nuclear Receptor Coactivator 3/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Response Elements/genetics
16.
Mol Cell Biol ; 28(21): 6580-93, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18765637

ABSTRACT

Overexpression and activation of the steroid receptor coactivator amplified in breast cancer 1 (AIB1)/steroid receptor coactivator-3 (SRC-3) have been shown to have a critical role in oncogenesis and are required for both steroid and growth factor signaling in epithelial tumors. Here, we report a new mechanism for activation of SRC coactivators. We demonstrate regulated tyrosine phosphorylation of AIB1/SRC-3 at a C-terminal tyrosine residue (Y1357) that is phosphorylated after insulin-like growth factor 1, epidermal growth factor, or estrogen treatment of breast cancer cells. Phosphorylated Y1357 is increased in HER2/neu (v-erb-b2 erythroblastic leukemia viral oncogene homolog 2) mammary tumor epithelia and is required to modulate AIB1/SRC-3 coactivation of estrogen receptor alpha (ERalpha), progesterone receptor B, NF-kappaB, and AP-1-dependent promoters. c-Abl (v-Abl Abelson murine leukemia viral oncogene homolog 1) tyrosine kinase directly phosphorylates AIB1/SRC-3 at Y1357 and modulates the association of AIB1 with c-Abl, ERalpha, the transcriptional cofactor p300, and the methyltransferase coactivator-associated arginine methyltransferase 1, CARM1. AIB1/SRC-3-dependent transcription and phenotypic changes, such as cell growth and focus formation, can be reversed by an Abl kinase inhibitor, imatinib. Thus, the phosphorylation state of Y1357 can function as a molecular on/off switch and facilitates the cross talk between hormone, growth factor, and intracellular kinase signaling pathways in cancer.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Histone Acetyltransferases/metabolism , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/metabolism , Trans-Activators/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Epidermal Growth Factor/pharmacology , Female , Fusion Proteins, bcr-abl , Humans , Insulin-Like Growth Factor I/pharmacology , Mice , Nuclear Receptor Coactivator 3 , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Interaction Mapping , Transcription Factors/metabolism
17.
Clin Immunol ; 124(2): 119-30, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17584532

ABSTRACT

We assessed the ability of several factors to increase the size of tumor-antigen-specific CD8(+) T cell responses elicited by vaccines incorporating peptides and CpG-containing oligodeoxynucleotides (CpG). Neither granulocyte-macrophage colony-stimulating factor (GM-CSF) nor an immunogenic MHC class II-presented "helper" peptide increased the size of epitope-specific CD8+ T cell responses elicited by peptide+CpG-containing vaccines. In contrast, low-dose subcutaneous interleukin (IL)-2 dramatically increased the size of splenic and peripheral blood epitope-specific CD8(+) T cell responses generated by peptide+CpG-containing vaccines. Moreover, peptide+CpG-containing vaccines plus low-dose IL-2 mediated anti-tumor immunity. A prime-boost vaccination schedule elicited larger CD8(+) T cell responses than a weekly vaccination schedule. Including larger doses of peptide in vaccines led to larger vaccine-elicited CD8(+) T cell responses. Clinical trials of CpG-containing peptide vaccines are ongoing. These findings suggest strategies to increase the size of CD8(+) T cell responses generated by CpG-containing peptide vaccines that could be tested in future clinical trials.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Oligodeoxyribonucleotides/immunology , Vaccines, Subunit/immunology , Adjuvants, Immunologic/pharmacology , Animals , Cancer Vaccines/immunology , Cell Line, Tumor , Dose-Response Relationship, Immunologic , Epitopes , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunization Schedule , Interleukin-2/immunology , Interleukin-2/pharmacology , Intramolecular Oxidoreductases/immunology , Intramolecular Oxidoreductases/pharmacology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Peptide Fragments/immunology , Peptide Fragments/pharmacology
18.
Blood ; 110(1): 450-60, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17371943

ABSTRACT

Development of CD8(+) T-cell responses targeting tumor-associated antigens after autologous stem cell transplantations (ASCTs) might eradicate residual tumor cells and decrease relapse rates. Because thymic function dramatically decreases with aging, T-cell reconstitution in the first year after ASCT in middle-aged patients occurs primarily by homeostatic peripheral expansion (HPE) of mature T cells. To study antigen-specific T-cell responses during HPE, we performed syngeneic bone marrow transplantations (BMTs) on thymectomized mice and then vaccinated the mice with peptides plus CpG-containing oligodeoxynucleotides (CpGs) in incomplete Freund adjuvant and treated the mice with systemic interleukin-2 (IL-2). When CD8(+) T-cell responses were measured ex vivo, up to 9.1% of CD8(+) T cells were specific for tumor-associated epitopes. These large T-cell responses were generated by synergism between CpG and IL-2. When we injected mice subcutaneously with tumor cells 14 days after BMT and then treated them with peptide + CpG-containing vaccines plus systemic IL-2, survival was increased and tumor growth was inhibited in an epitope-specific manner. Depletion of CD8(+) T cells eliminated epitope-specific antitumor immunity. This is the first report to demonstrate that CD8(+) T-cell responses capable of executing antitumor immunity can be elicited by CpG-containing vaccines during HPE.


Subject(s)
Bone Marrow Transplantation , Cancer Vaccines/chemistry , Cancer Vaccines/pharmacology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , CpG Islands , Immunity , Interleukin-2/therapeutic use , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/therapeutic use
19.
J Immunol ; 177(12): 8860-73, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17142789

ABSTRACT

Novel anticancer vaccination regimens that can elicit large numbers of Ag-specific T cells are needed. When we administered therapeutic vaccines containing the MHC class I-presented self-peptide tyrosinase-related protein (TRP)-2(180-188) and CpG-containing oligodeoxynucleotides (CpG ODN) to mice, growth of the TRP-2-expressing B16F1 melanoma was not inhibited compared with growth in mice that received control vaccinations. When we added systemic IL-2 to the TRP-2(180-188) plus CpG ODN vaccines, growth of B16F1 was inhibited in a CD8-dependent, epitope-specific manner. Vaccines containing TRP-2(180-188) without CpG ODN did not cause epitope-specific tumor growth inhibition when administered with IL-2. The antitumor efficacy of the different regimens correlated with their ability to elicit TRP-2(180-188)-specific CD8+ T cell responses. When we administered TRP-2(180-188) plus CpG ODN-containing vaccines with systemic IL-2, 18.2% of CD8+ T cells were specific for TRP-2(180-188). Identical TRP-2(180-188) plus CpG ODN vaccines given without IL-2 elicited a TRP-2(180-188)-specific CD8+ T cell response of only 1.1% of CD8+ T cells. Vaccines containing TRP-2(180-188) without CpG ODN elicited TRP-2(180-188)-specific responses of 2.8% of CD8+ T cells when administered with IL-2. There was up to a 221-fold increase in the absolute number of TRP-2(180-188)-specific CD8+ T cells when IL-2 was added to TRP-2(180-188) plus CpG ODN-containing vaccines. Peptide plus CpG ODN vaccines administered with IL-2 generated epitope-specific CD8+ T cells by a mechanism that depended on endogenous IL-6. This is the first report of synergism between CpG ODN and IL-2. This synergism caused a striking increase in vaccine-elicited CD8+ T cells and led to epitope-specific antitumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/chemistry , Interleukin-2/therapeutic use , Melanoma, Experimental/therapy , Oligodeoxyribonucleotides/therapeutic use , Animals , Antigens, Neoplasm , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , CpG Islands , Drug Synergism , Epitopes, T-Lymphocyte , Immunity/drug effects , Interleukin-2/pharmacology , Intramolecular Oxidoreductases/immunology , Mice , Oligodeoxyribonucleotides/pharmacology
20.
Exp Hematol ; 34(11): 1490-5, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17046568

ABSTRACT

OBJECTIVE: The physiologic role of platelet FcgammaRIIA, the only Fc receptor for IgG on human platelets, is largely unknown. FcgammaRIIA is also expressed on phagocytes such as monocytes and neutrophils, where it mediates the binding and internalization of both soluble IgG-containing complexes and IgG-coated cells. We previously reported the creation and characterization of a transgenic mouse that expresses human FcgammaRIIA on platelets and macrophages at levels comparable to that seen in humans. Using the transgenic mouse model, we observed that FcgammaRIIA mediates the clearance of IgG-coated cells. With the hypothesis that FcgammaRIIA on platelets may serve to remove IgG complexes from the circulation, we studied the capacity of human platelet FcgammaRIIA to bind and internalize such complexes. METHODS: We demonstrated by flow cytometry and electron microscopy that human platelets at 37 degrees C can bind and endocytose IgG complexes. We also utilized platelets from FcgammaRIIA transgenic mice to study endocytosis of IgG complexes by platelet FcgammaRIIA. RESULTS: Wild-type mouse platelets do not express Fcgamma receptors. While platelets from wild-type mice did not bind or endocytose IgG complexes, the presence of transgenic FcgammaRIIA on mouse platelets allowed the platelets to bind and endocytose IgG complexes. CONCLUSION: Our data indicate that platelet FcgammaRIIA binds and internalizes IgG complexes and suggest that human platelets may function to clear soluble IgG complexes from the circulation.


Subject(s)
Antigen-Antibody Complex/metabolism , Antigens, CD/physiology , Blood Platelets/metabolism , Immunoglobulin G/physiology , Receptors, IgG/physiology , Animals , Antigens, CD/blood , Endocytosis/physiology , Flow Cytometry , Humans , Mice , Mice, Transgenic , Microscopy, Electron , Protein Binding , Receptors, IgG/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...