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.
Neuro Oncol ; 26(2): 348-361, 2024 02 02.
Article in English | MEDLINE | ID: mdl-37715730

ABSTRACT

BACKGROUND: Recurrent brain tumors are the leading cause of cancer death in children. Indoleamine 2,3-dioxygenase (IDO) is a targetable metabolic checkpoint that, in preclinical models, inhibits anti-tumor immunity following chemotherapy. METHODS: We conducted a phase I trial (NCT02502708) of the oral IDO-pathway inhibitor indoximod in children with recurrent brain tumors or newly diagnosed diffuse intrinsic pontine glioma (DIPG). Separate dose-finding arms were performed for indoximod in combination with oral temozolomide (200 mg/m2/day x 5 days in 28-day cycles), or with palliative conformal radiation. Blood samples were collected at baseline and monthly for single-cell RNA-sequencing with paired single-cell T cell receptor sequencing. RESULTS: Eighty-one patients were treated with indoximod-based combination therapy. Median follow-up was 52 months (range 39-77 months). Maximum tolerated dose was not reached, and the pediatric dose of indoximod was determined as 19.2 mg/kg/dose, twice daily. Median overall survival was 13.3 months (n = 68, range 0.2-62.7) for all patients with recurrent disease and 14.4 months (n = 13, range 4.7-29.7) for DIPG. The subset of n = 26 patients who showed evidence of objective response (even a partial or mixed response) had over 3-fold longer median OS (25.2 months, range 5.4-61.9, p = 0.006) compared to n = 37 nonresponders (7.3 months, range 0.2-62.7). Four patients remain free of active disease longer than 36 months. Single-cell sequencing confirmed emergence of new circulating CD8 T cell clonotypes with late effector phenotype. CONCLUSIONS: Indoximod was well tolerated and could be safely combined with chemotherapy and radiation. Encouraging preliminary evidence of efficacy supports advancing to Phase II/III trials for pediatric brain tumors.


Subject(s)
Brain Neoplasms , Brain Stem Neoplasms , Humans , Child , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Temozolomide , Tryptophan , Immunologic Factors , Immunotherapy , Brain Stem Neoplasms/pathology
2.
Immunity ; 54(10): 2354-2371.e8, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34614413

ABSTRACT

Monocytic-lineage inflammatory Ly6c+CD103+ dendritic cells (DCs) promote antitumor immunity, but these DCs are infrequent in tumors, even upon chemotherapy. Here, we examined how targeting pathways that inhibit the differentiation of inflammatory myeloid cells affect antitumor immunity. Pharmacologic inhibition of Bruton's tyrosine kinase (BTK) and the tryptophan-degrading enzyme indoleamine 2,3-dioxygenase (IDO) or deletion of Btk or Ido1 allowed robust differentiation of inflammatory Ly6c+CD103+ DCs during chemotherapy, promoting antitumor T cell responses and inhibiting tumor growth. Immature Ly6c+c-kit+ precursor cells had epigenetic profiles similar to conventional DC precursors; deletion of Btk or Ido1 promoted differentiation of these cells. Mechanistically, a BTK-IDO axis inhibited a tryptophan-sensitive differentiation pathway driven by GATOR2 and mTORC1, and disruption of the GATOR2 in monocyte-lineage precursors prevented differentiation into inflammatory DCs in vivo. IDO-expressing DCs and monocytic cells were present across a range of human tumors. Thus, a BTK-IDO axis represses differentiation of inflammatory DCs during chemotherapy, with implications for targeted therapies.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Agammaglobulinaemia Tyrosine Kinase/immunology , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Male , Mice , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Signal Transduction/immunology , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
3.
Cancer Immunol Res ; 6(2): 201-208, 2018 02.
Article in English | MEDLINE | ID: mdl-29305519

ABSTRACT

Although an immune response to tumors may be generated using vaccines, so far, this approach has only shown minimal clinical success. This is attributed to the tendency of cancer to escape immune surveillance via multiple immune suppressive mechanisms. Successful cancer immunotherapy requires targeting these inhibitory mechanisms along with enhancement of antigen-specific immune responses to promote sustained tumor-specific immunity. Here, we evaluated the effect of indoximod, an inhibitor of the immunosuppressive indoleamine-(2,3)-dioxygenase (IDO) pathway, on antitumor efficacy of anti-OX40 agonist in the context of vaccine in the IDO- TC-1 tumor model. We demonstrate that although the addition of anti-OX40 to the vaccine moderately enhances therapeutic efficacy, incorporation of indoximod into this treatment leads to enhanced tumor regression and cure of established tumors in 60% of treated mice. We show that the mechanisms by which the IDO inhibitor leads to this therapeutic potency include (i) an increment of vaccine-induced tumor-infiltrating effector T cells that is facilitated by anti-OX40 and (ii) a decrease of IDO enzyme activity produced by nontumor cells within the tumor microenvironment that results in enhancement of the specificity and the functionality of vaccine-induced effector T cells. Our findings suggest a translatable strategy to enhance the overall efficacy of cancer immunotherapy. Cancer Immunol Res; 6(2); 201-8. ©2018 AACR.


Subject(s)
Antigens, Differentiation/pharmacology , Lung Neoplasms/drug therapy , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan/analogs & derivatives , Animals , Antigens, Differentiation/immunology , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Epitopes, T-Lymphocyte , Female , Humans , Immunotherapy/methods , Lung Neoplasms/immunology , Mice , Mice, Inbred C57BL , Tryptophan/pharmacology , Tryptophan Oxygenase/immunology , Xenograft Model Antitumor Assays
4.
J Immunother Cancer ; 5(1): 64, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28807056

ABSTRACT

BACKGROUND: We previously demonstrated that in addition to generating an antigen-specific immune response, Listeria monocytogenes (Lm)-based immunotherapy significantly reduces the ratio of regulatory T cells (Tregs)/CD4+ and myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment. Since Lm-based immunotherapy is able to inhibit the immune suppressive environment, we hypothesized that combining this treatment with agonist antibody to a co-stimulatory receptor that would further boost the effector arm of immunity will result in significant improvement of anti-tumor efficacy of treatment. METHODS: Here we tested the immune and therapeutic efficacy of Listeria-based immunotherapy combination with agonist antibody to glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) in TC-1 mouse tumor model. We evaluated the potency of combination on tumor growth and survival of treated animals and profiled tumor microenvironment for effector and suppressor cell populations. RESULTS: We demonstrate that combination of Listeria-based immunotherapy with agonist antibody to GITR synergizes to improve immune and therapeutic efficacy of treatment in a mouse tumor model. We show that this combinational treatment leads to significant inhibition of tumor-growth, prolongs survival and leads to complete regression of established tumors in 60% of treated animals. We determined that this therapeutic benefit of combinational treatment is due to a significant increase in tumor infiltrating effector CD4+ and CD8+ T cells along with a decrease of inhibitory cells. CONCLUSION: To our knowledge, this is the first study that exploits Lm-based immunotherapy combined with agonist anti-GITR antibody as a potent treatment strategy that simultaneously targets both the effector and suppressor arms of the immune system, leading to significantly improved anti-tumor efficacy. We believe that our findings depicted in this manuscript provide a promising and translatable strategy that can enhance the overall efficacy of cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Glucocorticoid-Induced TNFR-Related Protein/agonists , Immunotherapy/methods , Listeria monocytogenes/immunology , Lung Neoplasms/therapy , Myeloid-Derived Suppressor Cells/drug effects , Animals , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Combined Modality Therapy , Humans , Lung Neoplasms/immunology , Mice , T-Lymphocytes, Regulatory/drug effects , Treatment Outcome , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
5.
Cancer Res ; 77(15): 4135-4145, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28615225

ABSTRACT

Inhibition of specific Akt isoforms in CD8+ T cells promotes favored differentiation into memory versus effector cells, the former of which are superior in mediating antitumor immunity. In this study, we investigated the role of upstream PI3K isoforms in CD8+ T-cell differentiation and assessed the potential use of PI3K isoform-specific inhibitors to favorably condition CD8+ T cells for adoptive cell therapy. The phenotype and proliferative ability of tumor antigen-specific CD8+ T cells was assessed in the presence of PI3K-α, -ß, or -δ inhibitors. Inhibition of PI3K-δ, but not PI3K-α or PI3K-ß, delayed terminal differentiation of CD8+ T cells and maintained the memory phenotype, thus enhancing their proliferative ability and survival while maintaining their cytokine and granzyme B production ability. This effect was preserved in vivo after ex vivo PI3K-δ inhibition in CD8+ T cells destined for adoptive transfer, enhancing their survival and also the antitumor therapeutic activity of a tumor-specific peptide vaccine. Our results outline a mechanism by which inhibitions of a single PI3K isoform can enhance the proliferative potential, function, and survival of CD8+ T cells, with potential clinical implications for adoptive cell transfer and vaccine-based immunotherapies. Cancer Res; 77(15); 4135-45. ©2017 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Melanoma, Experimental/pathology , Phosphoinositide-3 Kinase Inhibitors , Animals , CD8-Positive T-Lymphocytes/enzymology , Cell Differentiation/drug effects , Class I Phosphatidylinositol 3-Kinases , Enzyme Inhibitors/pharmacology , Female , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
6.
Cancer Res ; 77(8): 1892-1904, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28108509

ABSTRACT

To modulate T-cell function for cancer therapy, one challenge is to selectively attenuate regulatory but not conventional CD4+ T-cell subsets [regulatory T cell (Treg) and conventional T cell (Tconv)]. In this study, we show how a functional dichotomy in Class IA PI3K isoforms in these two subsets of CD4+ T cells can be exploited to target Treg while leaving Tconv intact. Studies employing isoform-specific PI3K inhibitors and a PI3Kδ-deficient mouse strain revealed that PI3Kα and PI3Kß were functionally redundant with PI3Kδ in Tconv. Conversely, PI3Kδ was functionally critical in Treg, acting there to control T-cell receptor signaling, cell proliferation, and survival. Notably, in a murine model of lung cancer, coadministration of a PI3Kδ-specific inhibitor with a tumor-specific vaccine decreased numbers of suppressive Treg and increased numbers of vaccine-induced CD8 T cells within the tumor microenvironment, eliciting potent antitumor efficacy. Overall, our results offer a mechanistic rationale to employ PI3Kδ inhibitors to selectively target Treg and improve cancer immunotherapy. Cancer Res; 77(8); 1892-904. ©2017 AACR.


Subject(s)
CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/immunology , Neoplasms, Experimental/therapy , Phosphatidylinositol 3-Kinases/immunology , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Drug Synergism , Enzyme Inhibitors/pharmacology , Female , Immunotherapy/methods , Isoenzymes , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/immunology , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Purines/pharmacology , Quinazolinones/pharmacology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology
7.
Mol Cancer Res ; 12(7): 1029-41, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24743655

ABSTRACT

UNLABELLED: African Americans are disproportionately affected by early-onset, high-grade malignancies. A fraction of this cancer health disparity can be explained by genetic differences between individuals of African or European descent. Here the wild-type Pro/Pro genotype at the TP53Pro72Arg (P72R) polymorphism (SNP: rs1042522) is more frequent in African Americans with cancer than in African Americans without cancer (51% vs. 37%), and is associated with a significant increase in the rates of cancer diagnosis in African Americans. To test the hypothesis that Tp53 allele-specific gene expression may contribute to African American cancer disparities, TP53 hemizygous knockout variants were generated and characterized in the RKO colon carcinoma cell line, which is wild type for TP53 and heterozygous at the TP53Pro72Arg locus. Transcriptome profiling, using RNAseq, in response to the DNA-damaging agent etoposide revealed a large number of Tp53-regulated transcripts, but also a subset of transcripts that were TP53Pro72Arg allele specific. In addition, a shRNA-library suppressor screen for Tp53 allele-specific escape from Tp53-induced arrest was performed. Several novel RNAi suppressors of Tp53 were identified, one of which, PRDM1ß (BLIMP-1), was confirmed to be an Arg-specific transcript. Prdm1ß silences target genes by recruiting H3K9 trimethyl (H3K9me3) repressive chromatin marks, and is necessary for stem cell differentiation. These results reveal a novel model for African American cancer disparity, in which the TP53 codon 72 allele influences lifetime cancer risk by driving damaged cells to differentiation through an epigenetic mechanism involving gene silencing. IMPLICATIONS: TP53 P72R polymorphism significantly contributes to increased African American cancer disparity.


Subject(s)
Black or African American/genetics , Colonic Neoplasms/ethnology , Colonic Neoplasms/genetics , Genes, p53 , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Alleles , Genetic Predisposition to Disease , Humans , Polymorphism, Single Nucleotide , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...