Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 12(4)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38604813

ABSTRACT

BACKGROUND: Despite recent advances in immunotherapy, a substantial population of late-stage melanoma patients still fail to achieve sustained clinical benefit. Lack of translational preclinical models continues to be a major challenge in the field of immunotherapy; thus, more optimized translational models could strongly influence clinical trial development. To address this unmet need, we designed a preclinical model reflecting the heterogeneity in melanoma patients' clinical responses that can be used to evaluate novel immunotherapies and synergistic combinatorial treatment strategies. Using our all-autologous humanized melanoma mouse model, we examined the efficacy of a novel engineered interleukin 2 (IL-2)-based cytokine variant immunotherapy. METHODS: To study immune responses and antitumor efficacy for human melanoma tumors, we developed an all-autologous humanized melanoma mouse model using clinically annotated, matched patient tumor cells and peripheral blood mononuclear cells (PBMCs). After inoculating immunodeficient NSG mice with patient tumors and an adoptive cell transfer of autologous PBMCs, mice were treated with anti-PD-1, a novel investigational engineered IL-2-based cytokine (nemvaleukin), or recombinant human IL-2 (rhIL-2). The pharmacodynamic effects and antitumor efficacy of these treatments were then evaluated. We used tumor cells and autologous PBMCs from patients with varying immunotherapy responses to both model the diversity of immunotherapy efficacy observed in the clinical setting and to recapitulate the heterogeneous nature of melanoma. RESULTS: Our model exhibited long-term survival of engrafted human PBMCs without developing graft-versus-host disease. Administration of an anti-PD-1 or nemvaleukin elicited antitumor responses in our model that were patient-specific and were found to parallel clinical responsiveness to checkpoint inhibitors. An evaluation of nemvaleukin-treated mice demonstrated increased tumor-infiltrating CD4+ and CD8+ T cells, preferential expansion of non-regulatory T cell subsets in the spleen, and significant delays in tumor growth compared with vehicle-treated controls or mice treated with rhIL-2. CONCLUSIONS: Our model reproduces differential effects of immunotherapy in melanoma patients, capturing the inherent heterogeneity in clinical responses. Taken together, these data demonstrate our model's translatability for novel immunotherapies in melanoma patients. The data are also supportive for the continued clinical investigation of nemvaleukin as a novel immunotherapeutic for the treatment of melanoma.


Subject(s)
Melanoma , Humans , Animals , Mice , CD8-Positive T-Lymphocytes , Interleukin-2/pharmacology , Interleukin-2/therapeutic use , Leukocytes, Mononuclear/pathology , Cytokines , Immunotherapy
2.
J Immunother Cancer ; 10(9)2022 09.
Article in English | MEDLINE | ID: mdl-36472839

ABSTRACT

BACKGROUND: Small cell lung cancer (SCLC) is a deadly disease with a 5-year survival of less than 7%. The addition of immunotherapy to chemotherapy was recently approved as first-line treatment; however, the improved clinical benefit is modest, highlighting an urgent need for new treatment strategies. Nemvaleukin alfa, a novel engineered interleukin-2 fusion protein currently in phase I-III studies, is designed to selectively expand cytotoxic natural killer (NK) cells and CD8+ T cells. Here, using a novel SCLC murine model, we investigated the effects of a mouse version of nemvaleukin (mNemvaleukin) on tumor growth and antitumor immunity. METHODS: A novel Rb1 -/- p53 -/- p130 -/- SCLC model that mimics human disease was generated. After confirming tumor burden by MRI, mice were randomized into four treatment groups: vehicle, mNemvaleukin alone, chemotherapy (cisplatin+etoposide) alone, or the combination of mNemvaleukin and chemotherapy. Tumor growth was measured by MRI and survival was recorded. Tumor-infiltrating lymphocytes and peripheral blood immune cells were analyzed by flow cytometry. Cytokine and chemokine secretion were quantified and transcriptomic analysis was performed to characterize the immune gene signatures. RESULTS: mNemvaleukin significantly inhibited SCLC tumor growth, which was further enhanced by the addition of chemotherapy. Combining mNemvaleukin with chemotherapy provided the most significant survival benefit. Profiling of tumor-infiltrating lymphocytes revealed mNemvaleukin expanded the total number of tumor-infiltrating NK and CD8+ T cells. Furthermore, mNemvaleukin increased the frequencies of activated and proliferating NK and CD8+ T cells in tumors. Similar immune alterations were observed in the peripheral blood of mNemvaleukin-treated mice. Of note, combining mNemvaleukin with chemotherapy had the strongest effects in activating effector and cytotoxic CD8+ T cells. mNemvaleukin alone, and in combination with chemotherapy, promoted proinflammatory cytokine and chemokine production, which was further confirmed by transcriptomic analysis. CONCLUSIONS: mNemvaleukin, a novel cytokine-based immunotherapy, significantly inhibited murine SCLC tumor growth and prolonged survival, which was further enhanced by the addition of chemotherapy. mNemvaleukin alone, and in combination with chemotherapy, drove a strong antitumor immune program elicited by cytotoxic immune cells. Our findings support the evaluation of nemvaleukin alone or in combination with chemotherapy in clinical trials for the treatment of SCLC.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Mice , Animals , Interleukin-2 , Small Cell Lung Carcinoma/drug therapy , CD8-Positive T-Lymphocytes , Lung Neoplasms/drug therapy , Chemokines
3.
J Pharmacol Exp Ther ; 379(2): 203-210, 2021 11.
Article in English | MEDLINE | ID: mdl-34362793

ABSTRACT

Nemvaleukin alfa (nemvaleukin, ALKS 4230) is a novel cytokine created by the fusion of circularly permuted interleukin-2 (IL-2) to the IL-2Rα subunit of the IL-2 receptor (IL-2R) complex that confers selectivity for the intermediate-affinity IL-2R expressed on CD8+ T cells and natural killer (NK) cells. The pharmacokinetics and selective pharmacodynamic properties of nemvaleukin have been demonstrated using in vitro and in vivo mouse models. The pharmacokinetic/pharmacodynamic effects of nemvaleukin on immune cell subtypes were evaluated in cynomolgus monkeys after intravenous and subcutaneous administration to inform dose selection and predict pharmacodynamic effects in humans. Male drug-naïve cynomolgus monkeys (N = 15) were administered either single-dose (0.3 mg/kg i.v.; 0.3 mg/kg or 1.0 mg/kg s.c.) or repeated doses (0.1 mg/kg i.v. on days 1-5 or 0.5 mg/kg s.c. on days 1 and 4) of nemvaleukin. Serial blood samples were collected for pharmacokinetic assessment, immunophenotyping by flow cytometry, and profiling of serum cytokines. Repeat-dose subcutaneous administration of nemvaleukin with less frequent dosing resulted in total systemic exposure and trough serum concentrations comparable to those seen with intravenous administration, with lower peak serum concentrations. Transient elevation of interferon-γ and IL-6 peaked at 2 and 8 hours after intravenous and subcutaneous administration, respectively. Selective expansion of immunoprotective central memory, effector memory, terminal effector CD8+ T cells, and CD56+ NK cells, and minimal expansion of immunosuppressive CD4+CD25+FoxP3+ regulatory T cells was observed after both intravenous and subcutaneous administration. These data support the ongoing clinical evaluation of intravenous and subcutaneous nemvaleukin. SIGNIFICANCE STATEMENT: Administration of the novel interleukin-2 receptor agonist nemvaleukin alfa to cynomolgus monkeys resulted in selective expansion of immune effector cells, including CD8+ T and natural killer cells with minimal effects on immunosuppressive CD4+ regulatory T cells, confirming the design of nemvaleukin and highlighting its potential as a cancer immunotherapy. Subcutaneous administration of nemvaleukin achieved systemic exposure and immunostimulatory effects similar to those observed after more frequent intravenous dosing and may represent a practical alternative in a clinical setting.


Subject(s)
Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2/pharmacokinetics , Receptors, Interleukin-2/agonists , Receptors, Interleukin-2/metabolism , Administration, Intravenous , Animals , Dose-Response Relationship, Drug , Humans , Injections, Subcutaneous , Interleukin-2/administration & dosage , Lymphocytes/drug effects , Lymphocytes/metabolism , Macaca fascicularis , Male
4.
J Immunother Cancer ; 8(1)2020 04.
Article in English | MEDLINE | ID: mdl-32317293

ABSTRACT

BACKGROUND: Interleukin-2 (IL-2) plays a pivotal role in immune homeostasis due to its ability to stimulate numerous lymphocyte subsets including natural killer (NK) cells, effector CD4+ and CD8+ T cells, and regulatory T cells (Tregs). Low concentrations of IL-2 induce signaling through the high-affinity IL-2 receptor (IL-2R) comprised of IL-2Rα, IL-2Rß, and common γ chain (γc), preferentially expressed on Tregs. Higher concentrations of IL-2 are necessary to induce signaling through the intermediate-affinity IL-2R, composed of IL-2Rß and γc, expressed on memory CD8+ T cells and NK cells. Recombinant human IL-2 (rhIL-2) is approved for treatment of metastatic melanoma and renal cell carcinoma (RCC), but adverse events including capillary leak syndrome, potentially mediated through interaction with the high-affinity IL-2R, limit its therapeutic use. Furthermore, antitumor efficacy of IL-2 may also be limited by preferential expansion of immunosuppressive Tregs. ALKS 4230 is an engineered fusion protein comprised of a circularly-permuted IL-2 with the extracellular domain of IL-2Rα, designed to selectively activate effector lymphocytes bearing the intermediate-affinity IL-2R. RESULTS: ALKS 4230 was equipotent to rhIL-2 in activating human cells bearing the intermediate-affinity IL-2R, and less potent than rhIL-2 on cells bearing the high-affinity IL-2R. As observed in vitro with primary human cells from healthy donors and advanced cancer patients, ALKS 4230 induced greater activation and expansion of NK cells with reduced expansion of Tregs relative to rhIL-2. Similarly, in mice, ALKS 4230 treatment stimulated greater expansion of NK cells and memory-phenotype CD8+ T cells at doses that did not expand or activate Tregs. ALKS 4230 treatment induced significantly lower levels of proinflammatory cytokines, including tumor necrosis factor alpha, interleukin-6, and interferon gamma relative to rhIL-2. Furthermore, ALKS 4230 exhibited superior antitumor efficacy in the mouse B16F10 lung tumor model, where ALKS 4230 could be administered via multiple routes of administration and dosing schedules while achieving equivalent antitumor efficacy. CONCLUSIONS: ALKS 4230 exhibited enhanced pharmacokinetic and selective pharmacodynamic properties resulting in both improved antitumor efficacy and lower indices of toxicity relative to rhIL-2 in mice. These data highlight the potential of ALKS 4230 as a novel cancer immunotherapy, and as such, the molecule is being evaluated clinically.


Subject(s)
Antineoplastic Agents/administration & dosage , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2/metabolism , Lung Neoplasms/therapy , Melanoma, Experimental/therapy , Recombinant Fusion Proteins/administration & dosage , T-Lymphocytes, Regulatory/immunology , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis , Cell Proliferation , Female , Genetic Engineering , Humans , Immunotherapy , Interleukin-2/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lymphocyte Activation/immunology , Lymphocytes/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured
5.
Nature ; 453(7192): 236-40, 2008 May 08.
Article in English | MEDLINE | ID: mdl-18368049

ABSTRACT

T helper cells that produce IL-17 (T(H)17 cells) promote autoimmunity in mice and have been implicated in the pathogenesis of human inflammatory diseases. At mucosal surfaces, T(H)17 cells are thought to protect the host from infection, whereas regulatory T (T(reg)) cells control immune responses and inflammation triggered by the resident microflora. Differentiation of both cell types requires transforming growth factor-beta (TGF-beta), but depends on distinct transcription factors: RORgammat (encoded by Rorc(gammat)) for T(H)17 cells and Foxp3 for T(reg) cells. How TGF-beta regulates the differentiation of T cells with opposing activities has been perplexing. Here we demonstrate that, together with pro-inflammatory cytokines, TGF-beta orchestrates T(H)17 cell differentiation in a concentration-dependent manner. At low concentrations, TGF-beta synergizes with interleukin (IL)-6 and IL-21 (refs 9-11) to promote IL-23 receptor (Il23r) expression, favouring T(H)17 cell differentiation. High concentrations of TGF-beta repress IL23r expression and favour Foxp3+ T(reg) cells. RORgammat and Foxp3 are co-expressed in naive CD4+ T cells exposed to TGF-beta and in a subset of T cells in the small intestinal lamina propria of the mouse. In vitro, TGF-beta-induced Foxp3 inhibits RORgammat function, at least in part through their interaction. Accordingly, lamina propria T cells that co-express both transcription factors produce less IL-17 (also known as IL-17a) than those that express RORgammat alone. IL-6, IL-21 and IL-23 relieve Foxp3-mediated inhibition of RORgammat, thereby promoting T(H)17 cell differentiation. Therefore, the decision of antigen-stimulated cells to differentiate into either T(H)17 or T(reg) cells depends on the cytokine-regulated balance of RORgammat and Foxp3.


Subject(s)
Forkhead Transcription Factors/metabolism , Interleukin-17/metabolism , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Thyroid Hormone/antagonists & inhibitors , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects , Transforming Growth Factor beta/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Forkhead Transcription Factors/genetics , Gene Expression Regulation/drug effects , Humans , Interleukin-17/biosynthesis , Interleukin-17/genetics , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , T-Lymphocytes, Helper-Inducer/metabolism
6.
Sci STKE ; 2007(393): pe36, 2007 Jul 03.
Article in English | MEDLINE | ID: mdl-17609478

ABSTRACT

The forkhead family transcription factor Foxp3 is critical for the development and function of CD4(+)CD25(+) regulatory T cells (Tregs). A series of reports have begun to shed light on the precise role of Foxp3 in the regulation of the Treg transcriptome. Foxp3 can bind to specific gene elements, thereby altering transcription of target genes directly, and Foxp3 can alter the expression of genes encoding other transcription factors, thereby having an indirect effect on the transcription of target genes. Cells retaining aspects of Treg differentiation persist in the absence of Foxp3, which is suggestive of a Foxp3-independent aspect of Treg biology.


Subject(s)
Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Forkhead Transcription Factors/genetics , Gene Regulatory Networks , Humans , Immune Tolerance/immunology , Mice
7.
J Immunol ; 177(5): 3133-42, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16920951

ABSTRACT

Foxp3 has been shown to be both necessary and sufficient for the development and function of naturally arising CD4+ CD25+ regulatory T cells in mice. Mutation of Foxp3 in Scurfy mice and FOXP3 in humans with IPEX results in fatal, early onset autoimmune disease and demonstrates the critical role of FOXP3 in maintaining immune homeostasis. The FOXP3 protein encodes several functional domains, including a C2H2 zinc finger, a leucine zipper, and a winged-helix/forkhead (FKH) domain. We have shown previously that FOXP3 functions as a transcriptional repressor and inhibits activation-induced IL-2 gene transcription. To characterize the role of each predicted functional domain on the in vivo activity of FOXP3, we have evaluated the location of point mutations identified in a large cohort of patients with the immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) and found them to cluster primarily within the FKH domain and the leucine zipper, but also present within the poorly defined N-terminal portion of the protein. The molecular functions of each of the IPEX-targeted domains were investigated. We show that FOXP3 is constitutively localized to the nucleus and this localization requires sequences at both the amino and C-terminal ends of its FKH domain. Moreover, FOXP3 was found to homodimerize through its leucine zipper. We also identify a novel functional domain within the N-terminal half of FOXP3, which is required for FOXP3-mediated repression of transcription from both a constitutively active and a NF-AT-inducible promoter. Furthermore, we demonstrate that IPEX mutations in these domains correlate with deficiencies in FOXP3 repressor function, corroborating their in vivo relevance.


Subject(s)
Forkhead Transcription Factors/metabolism , Transcription, Genetic/genetics , Active Transport, Cell Nucleus , Cell Line , Dimerization , Forkhead Transcription Factors/genetics , Humans , Leucine Zippers , Mutation/genetics , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/metabolism , Promoter Regions, Genetic/genetics , Transcriptional Activation
8.
Front Biosci ; 11: 1607-19, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16368541

ABSTRACT

FoxP3 recently entered the spotlight as a critical component of regulatory T cell development and function. Several groups are presently engaged in an effort to uncover the mechanistic details of its contribution to this critical T cell subset. Despite this, the mechanism of FoxP3-mediated transcriptional repression and the affected target genes are still largely unknown. First, we discuss insights from work on other Fox family members with an emphasis on those with known roles in the immune system. Second, we review recent data concerning the molecular mechanism of FoxP3 function and its role in human disease. Finally, we consider what is known about FoxP3 target genes and their effect on T cell physiology.


Subject(s)
Forkhead Transcription Factors/physiology , Gene Expression Regulation , Transcription, Genetic , Amino Acid Sequence , Animals , Antigens, CD , Antigens, Differentiation/metabolism , Apoptosis , Autoimmunity , CTLA-4 Antigen , Cell Proliferation , Forkhead Transcription Factors/metabolism , Humans , Immune System/metabolism , Models, Genetic , Molecular Sequence Data , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Protein Structure, Tertiary , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Structure-Activity Relationship , T-Lymphocytes/metabolism , Zinc Fingers
9.
Cell Microbiol ; 6(3): 235-42, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14764107

ABSTRACT

Flagellin is the structural component of flagella produced by many pathogenic bacteria and is a potent proinflammatory molecule that mediates these effects through Toll-like receptor (TLR) 5. In Listeria monocytogenes (LM), flagellin expression is regulated by temperature and has been described as being shut off at 37 degrees C. In this study, we demonstrate that TLR5-mediated cell activation and flagellin expression is maintained at 37 degrees C in some laboratory-adapted strains and in approximately 20% of LM clinical isolates. To determine the role of flagellin in LM infection, a targeted mutation in the structural gene for flagellin (flaA) was generated in a parental LM strain that expressed flagellin under all conditions examined. In vitro studies demonstrated that this deltaflaA mutant was (i). non-motile; (ii). not able to activate TLR5-transfected HeLa cells; and (iii). induced tumour necrosis factor (TNF)-alpha production in approximately 50% fewer CD11b+ cells in splenocytes from normal mice compared with the parental strain. However, there was no significant alteration in virulence of the deltaflaA mutant after either intravenous or oral murine infection. Similarly, there was no difference in the generation of LM-specific CD8 or CD4 T cells after intravenous or oral infection. These data indicate that flagellin is not essential for LM pathogenesis or for the induction of LM-specific adaptive immune responses in normal mice.


Subject(s)
Flagellin/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Animals , Disease Models, Animal , Flagellin/genetics , Flagellin/metabolism , Listeria monocytogenes/metabolism , Listeria monocytogenes/physiology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...