Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
1.
Immunol Cell Biol ; 101(6): 514-524, 2023 Jul.
Article En | MEDLINE | ID: mdl-36401824

Influenza A viruses (IAVs) exist as distinct serological subtypes, with limited antibody cross reactivity compared with T-cell responses, leading to universal vaccines that elicit robust T-cell responses entering clinical trials to combat pandemic and zoonotic outbreaks. Previously we have extensively characterized the viral-vectored universal vaccine, Wyeth/IL-15/5flu, a group 1 hemagglutinin, H5N1-based vaccine using a vaccinia backbone with interleukin (IL)-15. The vaccine elicits robust T-cell responses to provide heterosubtypic protection from lethal infection; however, we have also observed short-term morbidity of vaccinated mice with a disparity between the effects of sublethal infection with group 1 and 2 IAV strains. At day 3 of H3N2 (group 2 IAV) infection, there was a heavily skewed T helper type 1 response in vaccinated infected mice with overproduction of cytokines and reduced chemokines, whereas H1N1 (group 1 IAV) infection had increased innate cellular responses. These findings suggest that increased and early immune activation by T-cell activating vaccines may induce mild immunopathology when there is a mismatch between non-neutralizing antibody and cross-reactive memory T-cell responses leading to exuberant cytokine production. Therefore, to avoid overstimulating proinflammatory immune responses upon infection, universal influenza vaccines that elicit strong T-cell immunity will need a robust cross-reactive antibody response.


Influenza A Virus, H1N1 Subtype , Influenza A Virus, H5N1 Subtype , Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Animals , Mice , Humans , Cytokines , Influenza A Virus, H3N2 Subtype , Antibodies, Viral
2.
Blood ; 140(18): 1951-1963, 2022 11 03.
Article En | MEDLINE | ID: mdl-35921533

Adult T-cell leukemia/lymphoma (ATLL) is one of the aggressive peripheral T-cell neoplasms with a poor prognosis. Accumulating evidence demonstrates that escape from adaptive immunity is a hallmark of ATLL pathogenesis. However, the mechanisms by which ATLL cells evade natural killer (NK)-cell-mediated immunity have been poorly understood. Here we show that CD48 expression in ATLL cells determines the sensitivity for NK-cell-mediated cytotoxicity against ATLL cells. We performed unbiased genome-wide clustered regularly interspaced short palindromic repeat (CRISPR) screening using 2 ATLL-derived cell lines and discovered CD48 as one of the best-enriched genes whose knockout conferred resistance to YT1-NK cell line-mediated cytotoxicity. The ability of CD48-knockout ATLL cells to evade NK-cell effector function was confirmed using human primary NK cells with reduced interferon-γ (IFNγ) induction and degranulation. We found that primary ATLL cells had reduced CD48 expression along with disease progression. Furthermore, other subgroups among aggressive peripheral T-cell lymphomas (PTCLs) also expressed lower concentrations of CD48 than normal T cells, suggesting that CD48 is a key molecule in malignant T-cell evasion of NK-cell surveillance. Thus, this study demonstrates that CD48 expression is likely critical for malignant T-cell lymphoma cell regulation of NK-cell-mediated immunity and provides a rationale for future evaluation of CD48 as a molecular biomarker in NK-cell-associated immunotherapies.


Leukemia-Lymphoma, Adult T-Cell , Lymphoma, T-Cell, Peripheral , Adult , Humans , CD48 Antigen/genetics , CD48 Antigen/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Leukemia-Lymphoma, Adult T-Cell/genetics , Lymphoma, T-Cell, Peripheral/genetics , Killer Cells, Natural
3.
Sci Adv ; 8(14): eabl5209, 2022 04 08.
Article En | MEDLINE | ID: mdl-35385318

To determine the potential for viral adaptation to T cell responses, we probed the full influenza virus genome by next-generation sequencing directly ex vivo from infected mice, in the context of an experimental T cell-based vaccine, an H5N1-based viral vectored vaccinia vaccine Wyeth/IL-15/5Flu, versus the current standard-of-care, seasonal inactivated influenza vaccine (IIV) and unvaccinated conditions. Wyeth/IL-15/5Flu vaccination was coincident with increased mutation incidence and frequency across the influenza genome; however, mutations were not enriched within T cell epitope regions, but high allele frequency mutations within conserved hemagglutinin stem regions and PB2 mammalian adaptive mutations arose. Depletion of CD4+ and CD8+ T cell subsets led to reduced frequency of mutants in vaccinated mice; therefore, vaccine-mediated T cell responses were important drivers of virus diversification. Our findings suggest that Wyeth/IL-15/5Flu does not generate T cell escape mutants but increases stochastic events for virus adaptation by stringent bottlenecks.


Influenza A Virus, H5N1 Subtype , Influenza Vaccines , Influenza, Human , Animals , Genome, Viral , Humans , Influenza A Virus, H5N1 Subtype/genetics , Influenza, Human/prevention & control , Interleukin-15/genetics , Mice , Mutation , Orthomyxoviridae Infections , Prevalence , Vaccination
4.
Microbes Infect ; 23(8): 104841, 2021.
Article En | MEDLINE | ID: mdl-34022375

SARS-CoV-2 has infected more than 167 million individuals globally. Highly effective and safe vaccines are required to accelerate the development of herd immunity to end the pandemic. This review focuses on vaccines that are being developed at unprecedented speed globally and are completing late phase clinical trials to meet this urgent need.


COVID-19 Vaccines/immunology , COVID-19/prevention & control , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , SARS-CoV-2/immunology
5.
J Immunother Cancer ; 9(4)2021 04.
Article En | MEDLINE | ID: mdl-33883258

BACKGROUND: Full application of cytokines as oncoimmunotherapeutics requires identification of optimal regimens. Our initial effort with intravenous bolus recombinant human interleukin-15 (rhIL-15) was limited by postinfusional reactions. Subcutaneous injection and continuous intravenous infusion for 10 days (CIV-10) provided rhIL-15 with less toxicity with CIV-10 giving the best increases in CD8+ lymphocytes and natural killer (NK) cells. To ease rhIL-15 administration, we shortened time of infusion. Treatment with rhIL-15 at a dose of 3-5 µg/kg as a 5-day continuous intravenous infusion (CIV-5) had no dose-limiting toxicities while effector cell stimulation was comparable to the CIV-10 regimen. METHODS: Eleven patients with metastatic cancers were treated with rhIL-15 CIV-5, 3 µg (n=4), 4 µg (n=3), and 5 µg/kg/day (n=4) in a phase I dose-escalation study (April 6, 2012). RESULTS: Impressive expansions of NK cells were seen at all dose levels (mean 34-fold), including CD56bright NK cells (mean 144-fold for 4 µg/kg), as well as an increase in CD8+ T cells (mean 3.38-fold). At 5 µg/kg/day, there were no dose-limiting toxicities but pulmonary capillary leak and slower patient recovery. This led to our choice of the 4 µg/kg as CIV-5 dose for further testing. Cytolytic capacity of CD56bright and CD56dim NK cells was increased by interleukin-15 assayed by antibody-dependent cellular cytotoxicity (ADCC), natural cytotoxicity and natural killer group 2D-mediated cytotoxicity. The best response was stable disease. CONCLUSIONS: IL-15 administered as CIV-5 substantially expanded NK cells with increased cytotoxic functions. Tumor-targeting monoclonal antibodies dependent on ADCC as their mechanism of action including alemtuzumab, obinutuzumab, avelumab, and mogamulizumab could benefit from those NK cell expansions and provide a promising therapeutic strategy. TRIAL REGISTRATION NUMBERS: NCT01572493, NCT03759184, NCT03905135, NCT04185220 and NCT02689453.


Antineoplastic Agents/administration & dosage , Cell Proliferation/drug effects , Interleukin-15/administration & dosage , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Neoplasms/drug therapy , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Coculture Techniques , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Interleukin-15/adverse effects , K562 Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Count , Male , Maryland , Middle Aged , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/metabolism , Time Factors , Treatment Outcome
6.
Clin Sci (Lond) ; 134(10): 1151-1166, 2020 05 29.
Article En | MEDLINE | ID: mdl-32420596

A percentage of celiac disease (CD) patients develop refractory type-2 disease (RCD2), a condition associated with increased risk of enteropathy-associated T-cell-lymphoma (EATL) and without therapeutic option. Therefore, we profiled the miRNome in series of peripheral T-cell lymphomas (PTCLs), CD, RCD1 or 2 and in the murine interleukin-15 (IL15)-transgenic (TG) model of RCD. The transcriptome was analyzed in 18 intestinal T-cell lymphomas (ITLs). Bioinformatics pipelines provided significant microRNA (miRNA) lists and predicted targets that were confirmed in a second set of patients. Our data show that ITLs have a unique miRNA profile with respect to other PTCLs. The c-MYC regulated miR-17/92 cluster distinguishes monomorphic epitheliotropic ITL (MEITL) from EATL and prognosticates EATL outcome. These miRNAs are decreased in IL15-TG mice upon Janus kinase (JAK) inhibition. The random forest algorithm identified a signature of 38 classifier miRNAs, among which, the miR-200 and miR-192/215 families were progressively lost in RCD2 and ITL-CD, whereas miR-17/92 and C19MC miRNAs were up-regulated. Accordingly, SMAD3, MDM2, c-Myc and activated-STAT3 were increased in RCD2 and EATL tissues while JAK inhibition in IL15-TG mice restored their levels to baseline. Our data suggest that miRNAs circuit supports activation of STAT3 and c-Myc oncogenic signaling in RCD2, thus contributing to lymphomagenesis. This novel understanding might pave the way to personalized medicine approaches for RCD and EATL.


Carcinogenesis/genetics , Celiac Disease/genetics , Gene Expression Regulation, Neoplastic , Lymphoma/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins c-myc/metabolism , Algorithms , Animals , Biomarkers, Tumor/metabolism , Female , Intestines/pathology , Lymphoma/pathology , Male , Mice, Transgenic , MicroRNAs/metabolism , Models, Biological , Prognosis , Proto-Oncogene Proteins c-mdm2/metabolism , Smad3 Protein/metabolism , Up-Regulation/genetics
7.
World J Oncol ; 11(1): 1-8, 2020 Feb.
Article En | MEDLINE | ID: mdl-32095184

BACKGROUND: The rise in human papillomavirus (HPV) infection rates over the last few decades in the USA has contributed to a significant increase in the overall incidence of patients diagnosed with squamous cell carcinoma of the head and neck. These head and neck carcinomas develop in the oropharynx, with more than 90% of them caused by infection with high-risk HPV type 16. Patients diagnosed with HPV-induced oropharyngeal squamous cell carcinomas (OPSCCs) have a better prognosis and treatment response than those diagnosed with head and neck cancers caused by alcohol consumption and tobacco use. To identify patients with HPV-positive OPSCC, new guidelines recommend positive staining of oropharyngeal tissues for p16 INK4a (p16) by immunohistochemistry (IHC). Herein we discuss the testing algorithm that was adopted to address discrepant results between p16 IHC and a DNA in situ hybridization (ISH) test used routinely to diagnose HPV-positive OPSCC patients. METHODS: A DNA polymerase chain reaction (PCR) test that amplifies HPV16 and HPV18 E7 was developed to aid in the diagnosis of HPV-positive OPSCC in a subset of patients. Specimens from these patients stained positive for p16 by an IHC test, but negative for high-risk HPV by a commercial DNA ISH test. Moreover, these results did not match the histopathological characteristics of the specimens, nor the clinical presentations of the patients. RESULTS: Of 21 patients' specimens that were tested for p16 by IHC, 11 specimens showed concordant results with the high-risk HPV 16/18 DNA ISH test. Whereas, in eight p16 IHC positive specimens, HPV viral DNA was not detected by HPV16/18 DNA ISH, and two specimens were not tested by DNA ISH. When these eight p16 IHC positive specimens with discrepant p16 IHC and DNA ISH results were further tested by DNA PCR, six specimens showed concordance with p16 IHC with positive results for HPV16 E7, while two specimens were negative for HPV16 E7 by DNA PCR. All tested specimens were negative for HPV18 E7 by DNA PCR. Thus, the addition of the HPV16 and HPV18 E7 DNA PCR test identified a significant number of false negative test results by the HPV16/18 DNA ISH test and likely several false positive results by p16 IHC. CONCLUSIONS: Inclusion of an HPV16 E7 DNA PCR test improved the robustness of HPV-associated OPSCC diagnosis in patients with discrepant results from p16 IHC staining and a DNA ISH test, and identified patients for proper management with less misclassification.

8.
Clin Cancer Res ; 25(16): 4945-4954, 2019 08 15.
Article En | MEDLINE | ID: mdl-31142503

PURPOSE: The first-in-human clinical trial with human bolus intravenous infusion IL15 (rhIL15) was limited by treatment-associated toxicity. Here, we report toxicity, immunomodulation, and clinical activity of rhIL15 administered as a 10-day continuous intravenous infusion (CIV) to patients with cancers in a phase I trial. PATIENTS AND METHODS: Patients received treatment for 10 days with CIV rhIL15 in doses of 0.125, 0.25, 0.5, 1, 2, or 4 µg/kg/day. Correlative laboratory tests included IL15 pharmacokinetic (PK) analyses, and assessment of changes in lymphocyte subset numbers. RESULTS: Twenty-seven patients were treated with rhIL15; 2 µg/kg/day was identified as the MTD. There were eight serious adverse events including two bleeding events, papilledema, uveitis, pneumonitis, duodenal erosions, and two deaths (one due to likely drug-related gastrointestinal ischemia). Evidence of antitumor effects was observed in several patients, but stable disease was the best response noted. Patients in the 2 µg/kg/day group had a 5.8-fold increase in number of circulating CD8+ T cells, 38-fold increase in total NK cells, and 358-fold increase in CD56bright NK cells. Serum IL15 concentrations were markedly lower during the last 3 days of infusion. CONCLUSIONS: This phase I trial identified the MTD for CIV rhIL15 and defined a treatment regimen that produced significant expansions of CD8+ T and NK effector cells in circulation and tumor deposits. This regimen has identified several biological features, including dramatic increases in numbers of NK cells, supporting trials of IL15 with anticancer mAbs to increase antibody-dependent cell-mediated cytotoxicity and anticancer efficacy.


Interleukin-15/administration & dosage , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Cytokines/metabolism , Female , Humans , Immunohistochemistry , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Immunologic Factors/pharmacokinetics , Immunomodulation/drug effects , Inflammation Mediators/metabolism , Infusions, Intravenous , Interleukin-15/adverse effects , Interleukin-15/pharmacokinetics , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphocyte Subsets/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/diagnosis , Neoplasms/metabolism , Treatment Outcome
9.
Vaccine ; 36(29): 4198-4206, 2018 07 05.
Article En | MEDLINE | ID: mdl-29887326

There is a diverse array of influenza viruses which circulate between different species, reassort and drift over time. Current seasonal influenza vaccines are ineffective in controlling these viruses. We have developed a novel universal vaccine which elicits robust T cell responses and protection against diverse influenza viruses in mouse and human models. Vaccine mediated protection was dependent on influenza-specific CD4+ T cells, whereby depletion of CD4+ T cells at either vaccination or challenge time points significantly reduced survival in mice. Vaccine memory CD4+ T cells were needed for early antibody production and CD8+ T cell recall responses. Furthermore, influenza-specific CD4+ T cells from vaccination manifested primarily Tfh and Th1 profiles with anti-viral cytokine production. The vaccine boosted H5-specific T cells from human PBMCs, specifically CD4+ and CD8+ T effector memory type, ensuring the vaccine was truly universal for its future application. These findings have implications for the development and optimization of T cell activating vaccines for universal immunity against influenza.


CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Influenza Vaccines/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Influenza Vaccines/administration & dosage , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Survival Analysis
10.
Am J Hematol ; 92(9): 892-901, 2017 Sep.
Article En | MEDLINE | ID: mdl-28543380

With the emerging success of treating CD19 expressing B cell malignancies with ex vivo modified, autologous T cells that express CD19-directed chimeric antigen receptors (CAR), there is intense interest in expanding this evolving technology to develop effective modalities to treat other malignancies including solid tumors. Exploiting this approach to develop a therapeutic modality for T cell malignancies for which the available regimens are neither curative, nor confer long term survival we generated a lentivirus-based CAR gene transfer system to target the chemokine receptor CCR4 that is over-expressed in a spectrum of T cell malignancies as well as in CD4+ CD25+ Foxp3+ T regulatory cells that accumulate in the tumor microenvironment constituting a barrier against anti-tumor immunity. Ex vivo modified, donor-derived T cells that expressed CCR4 directed CAR displayed antigen-dependent potent cytotoxicity against patient-derived cell lines representing ATL, CTCL, ALCL and a subset of HDL. Furthermore, these CAR T cells also eradicated leukemia in a mouse xenograft model of ATL illustrating the potential utility of this modality in the treatment of a wide spectrum of T cell malignancies.


Hematologic Neoplasms , Receptors, Antigen, T-Cell , Receptors, CCR4/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Humans , Leukemia, T-Cell/genetics , Leukemia, T-Cell/immunology , Leukemia, T-Cell/pathology , Leukemia, T-Cell/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, CCR4/genetics , Receptors, CCR4/immunology , T-Lymphocytes, Regulatory/pathology , Xenograft Model Antitumor Assays
11.
PLoS One ; 10(10): e0141141, 2015.
Article En | MEDLINE | ID: mdl-26505634

Malaria remains a major global public health problem with an estimated 200 million cases detected in 2012. Although the most advanced candidate malaria vaccine (RTS,S) has shown promise in clinical trials, its modest efficacy and durability have created uncertainty about the impact of RTS,S immunization (when used alone) on global malaria transmission. Here we describe the development and characterization of a novel modified vaccinia virus Ankara (MVA)-based malaria vaccine which co-expresses the Plasmodium yoelii circumsporozoite protein (CSP) and IL-15. Vaccination/challenge studies showed that C57BL/6 mice immunized with the MVA-CSP/IL15 vaccine were protected significantly better against a P. yoelii 17XNL sporozoite challenge than either mice immunized with an MVA vaccine expressing only CSP or naïve controls. Importantly, the levels of total anti-CSP IgG were elevated about 100-fold for the MVA-CSP/IL15 immunized group compared to mice immunized with the MVA-CSP construct that does not express IL-15. Among the IgG subtypes, the IL-15 expressing MVA-CSP vaccine induced levels of IgG1 (8 fold) and IgG2b (80 fold) higher than the MVA-CSP construct. The significantly enhanced humoral responses and protection detected after immunization with the MVA-CSP/IL15 vaccine suggest that this IL-15 expressing MVA construct could be considered in the development of future malaria immunization strategies.


Immunity, Innate/genetics , Interleukin-15/biosynthesis , Malaria Vaccines/immunology , Malaria/immunology , Animals , Female , Humans , Interleukin-15/genetics , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Mice , Plasmodium yoelii/drug effects , Plasmodium yoelii/immunology , Plasmodium yoelii/pathogenicity , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Vaccination , Vaccinia virus/genetics , Vaccinia virus/immunology
12.
J Clin Immunol ; 35(7): 661-7, 2015 Oct.
Article En | MEDLINE | ID: mdl-26453583

PURPOSE: Autoimmune lymphoproliferative syndrome (ALPS) is a non-malignant genetic disorder of lymphocyte homeostasis with defective Fas-mediated apoptosis. Current therapies for ALPS primarily target autoimmune manifestations with non-specific immune suppressants with variable success thus highlighting the need for better therapeutics for this disorder. METHODS: The spectrum of clinical manifestations of ALPS is mirrored by MRL/lpr mice that carry a loss of function mutation in the Fas gene and have proven to be a valuable model in predicting the efficacy of several therapeutics that are front-line modalities for the treatment of ALPS. We evaluated the potential efficacy of tofacitinib, an orally active, pan-JAK inhibitor currently approved for rheumatoid arthritis as a single agent modality against ALPS using MRL/lpr mice. RESULTS: We demonstrate that a 42-day course of tofacitinib therapy leads to a lasting reversal of lymphadenopathy and autoimmune manifestations in the treated MRL/lpr mice, Specifically, in treated mice the peripheral blood white blood cell counts were reversed to near normal levels with almost a 50 % reduction in the TCRαß(+)CD4(-)CD8(-)T lymphocyte numbers that coincided with a parallel increase in CD8(+) T cells without a demonstrable effect on CD4(+) lymphocytes including FoxP3(+) regulatory T cells. The elevated plasma IgG and IgA levels were also drastically lowered along with a significant reduction in plasmablasts and plasmacytes in the spleen. CONCLUSION: On the basis of these results, it is likely that tofacitinib would prove to be a potent single agent therapeutic modality capable of ameliorating both offending lymphadenopathy as well as autoimmunity in ALPS patients.


Autoimmune Lymphoproliferative Syndrome/drug therapy , Piperidines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Autoimmune Lymphoproliferative Syndrome/immunology , Disease Models, Animal , Humans , Immunoglobulins/blood , Janus Kinase 3/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mutation/genetics , Piperidines/adverse effects , Protein Kinase Inhibitors/adverse effects , Pyrimidines/adverse effects , Pyrroles/adverse effects , T-Lymphocyte Subsets/physiology , T-Lymphocytes, Regulatory/physiology , fas Receptor/genetics
13.
J Clin Oncol ; 33(1): 74-82, 2015 Jan 01.
Article En | MEDLINE | ID: mdl-25403209

PURPOSE: Interleukin-15 (IL-15) has significant potential in cancer immunotherapy as an activator of antitumor CD8 T and natural killer (NK) cells. The primary objectives of this trial were to determine safety, adverse event profile, dose-limiting toxicity, and maximum-tolerated dose of recombinant human IL-15 (rhIL-15) administered as a daily intravenous bolus infusion for 12 consecutive days in patients with metastatic malignancy. PATIENTS AND METHODS: We performed a first in-human trial of Escherichia coli-produced rhIL-15. Bolus infusions of 3.0, 1.0, and 0.3 µg/kg per day of IL-15 were administered for 12 consecutive days to patients with metastatic malignant melanoma or metastatic renal cell cancer. RESULTS: Flow cytometry of peripheral blood lymphocytes revealed dramatic efflux of NK and memory CD8 T cells from the circulating blood within minutes of IL-15 administration, followed by influx and hyperproliferation yielding 10-fold expansions of NK cells that ultimately returned to baseline. Up to 50-fold increases of serum levels of multiple inflammatory cytokines were observed. Dose-limiting toxicities observed in patients receiving 3.0 and 1.0 µg/kg per day were grade 3 hypotension, thrombocytopenia, and elevations of ALT and AST, resulting in 0.3 µg/kg per day being determined the maximum-tolerated dose. Indications of activity included clearance of lung lesions in two patients. CONCLUSION: IL-15 could be safely administered to patients with metastatic malignancy. IL-15 administration markedly altered homeostasis of lymphocyte subsets in blood, with NK cells and γδ cells most dramatically affected, followed by CD8 memory T cells. To reduce toxicity and increase efficacy, alternative dosing strategies have been initiated, including continuous intravenous infusions and subcutaneous IL-15 administration.


CD4-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Interleukin-15/therapeutic use , Killer Cells, Natural/drug effects , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Area Under Curve , CD4-Positive T-Lymphocytes/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Fever/chemically induced , Humans , Infusions, Intravenous , Interleukin-15/adverse effects , Interleukin-15/genetics , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Male , Metabolic Clearance Rate , Middle Aged , Nausea/chemically induced , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/metabolism , Neutropenia/chemically induced , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Treatment Outcome , Young Adult
14.
Proc Natl Acad Sci U S A ; 111(15): 5676-81, 2014 Apr 15.
Article En | MEDLINE | ID: mdl-24706798

Current influenza vaccines are ineffective against novel viruses and the source or the strain of the next outbreak of influenza is unpredictable; therefore, establishing universal immunity by vaccination to limit the impact of influenza remains a high priority. To meet this challenge, a novel vaccine has been developed using the immunogenic live vaccinia virus as a vaccine vector, expressing multiple H5N1 viral proteins (HA, NA, M1, M2, and NP) together with IL-15 as a molecular adjuvant. Previously, this vaccine demonstrated robust sterile cross-clade protection in mice against H5 influenza viruses, and herein its use has been extended to mediate heterosubtypic immunity toward viruses from both group 1 and 2 HA lineages. The vaccine protected mice against lethal challenge by increasing survival and significantly reducing lung viral loads against the most recent human H7N9, seasonal H3N2, pandemic-2009 H1N1, and highly pathogenic H7N7 influenza A viruses. Influenza-specific antibodies elicited by the vaccine failed to neutralize heterologous viruses and were unable to confer protection by passive transfer. Importantly, heterologous influenza-specific CD4(+) and CD8(+) T-cell responses that were elicited by the vaccine were effectively recalled and amplified following viral challenge in the lungs and periphery. Selective depletion of T-cell subsets in the immunized mice revealed an important role for CD4(+) T cells in heterosubtypic protection, despite low sequence conservation among known MHC-II restricted epitopes across different influenza viruses. This study illustrates the potential utility of our multivalent Wyeth/IL-15/5Flu as a universal influenza vaccine with a correlate of protective immunity that is independent of neutralizing antibodies.


CD4-Positive T-Lymphocytes/immunology , Influenza Vaccines/immunology , Interleukin-15/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Vaccinia virus/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Female , Flow Cytometry , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H7N7 Subtype/immunology , Influenza A Virus, H7N9 Subtype/immunology , Interleukin-15/administration & dosage , Mice , Mice, Inbred BALB C
15.
Hum Vaccin Immunother ; 9(9): 1841-8, 2013 Sep.
Article En | MEDLINE | ID: mdl-23787486

An intense effort has been launched to develop improved anthrax vaccines that confer rapid, long lasting protection preferably with an extended stability profile amenable for stockpiling. Protective antigen (PA)-based vaccines are most favored as immune responses directed against PA are singularly protective, although the actual protective mechanism remains to be unraveled. Herein we show that contrary to the prevailing view, an efficacious PA-based vaccine confers protection against inhalation anthrax by preventing the establishment of a toxin-releasing systemic infection. Equally importantly, antibodies measured by the in vitro lethal toxin neutralization activity assay (TNA) that is considered as a reliable correlate of protection, especially for PA protein-based vaccines adjuvanted with aluminum salts appear to be not absolutely essential for this protective immune response.


Anthrax Vaccines/immunology , Anthrax/immunology , Anthrax/prevention & control , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/prevention & control , Animals , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Female , Humans , Rabbits
16.
J Clin Immunol ; 33(3): 586-94, 2013 Apr.
Article En | MEDLINE | ID: mdl-23269601

Celiac disease (CD) is an immune-mediated, inflammatory disorder of the small intestines with a defined genetic etiological component associated with the expression of HLA-DQ2 and/or HLA-DQ8 haplotypes. The dietary consumption of gluten-rich cereals triggers a gluten-specific immune response in genetically susceptible individuals leading to a spectrum of clinical manifestations ranging from an inapparent subclinical disease, to overt enteropathy that can in some individuals progress to enteropathy-associated T cell lymphoma (EATL). The tissue-destructive pathologic process of CD is driven by activated NK-like intraepithelial CD8(+) lymphocytes and the proinflammatory cytokine IL-15 has emerged to be pivotal in orchestrating this perpetual tissue destruction and inflammation. Moreover, transgenic mice that over-express human IL-15 from an enterocyte-specific promoter (T3(b)-hIL-15 Tg) recapitulate many of the disease-defining T and B cell-mediated pathologic features of CD, further supporting the evolving consensus that IL-15 represents a valuable target in devising therapeutic interventions against the form of the disease that is especially refractory to gluten-free diet. In the present study, we evaluated the potential efficacy of tofacitinib, a pan-JAK inhibitor that abrogates IL-15 signaling, as a therapeutic modality against CD using T3(b)-hIL-15 Tg mice. We demonstrate that tofacitinib therapy leads to a lasting reversal of pathologic manifestations in the treated mice, thereby highlighting the potential value of tofacitininb as a therapeutic modality against refractory CD for which no effective therapy exists currently. Additionally, the visceral adiposity observed in the tofacitinib-treated mice underscores the importance of continued evaluation of the drug's impact on the lipid metabolism.


Celiac Disease/enzymology , Celiac Disease/genetics , Interleukin-15/genetics , Janus Kinases/antagonists & inhibitors , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Animals , Celiac Disease/drug therapy , Disease Models, Animal , Female , Humans , Immunophenotyping , Interleukin-15/immunology , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/pathology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Mice , Mice, Transgenic , Piperidines/administration & dosage , Piperidines/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrroles/administration & dosage , Pyrroles/adverse effects , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Time Factors
17.
Microbes Infect ; 14(3): 247-61, 2012 Mar.
Article En | MEDLINE | ID: mdl-22064066

Interleukin-15 (IL-15) is a pleiotropic cytokine with a broad range of biological functions in many diverse cell types. It plays a major role in the development of inflammatory and protective immune responses to microbial invaders and parasites by modulating immune cells of both the innate and adaptive immune systems. This review provides an overview of the mechanisms by which IL-15 modulates the host response to infectious agents and its utility as a cytokine adjuvant in vaccines against infectious pathogens.


Immunity , Inflammation/immunology , Interleukin-15/immunology , Interleukin-15/therapeutic use , Adjuvants, Immunologic/therapeutic use , Animals , Dendritic Cells/immunology , Host-Pathogen Interactions , Humans , Killer Cells, Natural/immunology , Macrophages/immunology , Mast Cells/immunology , Monocytes/immunology , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Neutrophils/immunology , Signal Transduction , T-Lymphocytes/immunology , Tuberculosis/immunology , Tuberculosis/microbiology , Virus Diseases/immunology , Virus Diseases/virology , Viruses/immunology , Viruses/pathogenicity
18.
PLoS One ; 6(12): e28164, 2011.
Article En | MEDLINE | ID: mdl-22205939

BACKGROUND: Given the considerable geographic overlap in the endemic regions for malaria and tuberculosis, it is probable that co-infections with Mycobacterium tuberculosis and Plasmodium species are prevalent. Thus, it is quite likely that both malaria and TB vaccines may be used in the same populations in endemic areas. While novel vaccines are currently being developed and tested individually against each of these pathogens, the efficacy of these vaccines has not been evaluated in co-infection models. To further assess the effectiveness of these new immunization strategies, we investigated whether co-infection with malaria would impact the anti-tuberculosis protection induced by four different types of TB vaccines in a mouse model of pulmonary tuberculosis. PRINCIPAL FINDINGS: Here we show that the anti-tuberculosis protective immunity induced by four different tuberculosis vaccines was not impacted by a concurrent infection with Plasmodium yoelii NL, a nonlethal form of murine malaria. After an aerogenic challenge with virulent M. tuberculosis, the lung bacterial burdens of vaccinated animals were not statistically different in malaria infected and malaria naïve mice. Multi-parameter flow cytometric analysis showed that the frequency and the median fluorescence intensities (MFI) for specific multifunctional T (MFT) cells expressing IFN-γ, TNF-α, and/or IL-2 were suppressed by the presence of malaria parasites at 2 weeks following the malaria infection but was not affected after parasite clearance at 7 and 10 weeks post-challenge with P. yoelii NL. CONCLUSIONS: Our data indicate that the effectiveness of novel TB vaccines in protecting against tuberculosis was unaffected by a primary malaria co-infection in a mouse model of pulmonary tuberculosis. While the activities of specific MFT cell subsets were reduced at elevated levels of malaria parasitemia, the T cell suppression was short-lived. Our findings have important relevance in developing strategies for the deployment of new TB vaccines in malaria endemic areas.


BCG Vaccine/immunology , Malaria/complications , Tuberculosis, Pulmonary/complications , Tuberculosis, Pulmonary/immunology , Animals , Female , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Plasmodium yoelii/pathogenicity , Tuberculosis, Pulmonary/parasitology
19.
J Clin Immunol ; 31(6): 1038-44, 2011 Dec.
Article En | MEDLINE | ID: mdl-21938511

Celiac disease (CD) is a chronic immune-mediated intestinal inflammatory disorder afflicting genetically susceptible individuals triggered by the consumption of dietary cereals with high gluten content. As with many other organ-specific autoimmune diseases, the dominant tissue-destructive inflammation in CD is T cell-mediated. The proinflammatory cytokine IL-15 that is overexpressed in the intestinal epithelium of CD patients has emerged as a pivotal element that orchestrates intestinal inflammation and T cell-mediated autoimmune tissue destruction. Although no animal model exists that recapitulates the full spectrum of CD pathophysiology, we have previously reported that transgenic mice that overexpress human IL-15 in enterocytes (T3(b)-hlL-15 Tg) display many of the T cell-mediated pathologic features seen in CD. Extending these observations, we now report that T3(b)-hlL-15 Tg mice in addition to recapitulating T cell-mediated effects also display autoantibodies including those against tissue transglutaminase 2 and extensive lamina propria plasmacytosis, all of which are characteristic of CD, thereby reflecting the possibility that locally expressed IL-15 drives both T and B cell pathologic effects seen in CD. More importantly, these findings support the validity and utility of T3(b)-hlL-15 Tg mice as a reasonable model to investigate not only tissue-destructive pathologic processes in CD, but also to explore novel therapeutic modalities for the treatment of this disease.


Celiac Disease/immunology , Disease Models, Animal , Enterocytes/metabolism , Interleukin-15/metabolism , Plasma Cells/pathology , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , B-Lymphocytes/immunology , Celiac Disease/genetics , Cell Movement , Enterocytes/immunology , Enterocytes/pathology , GTP-Binding Proteins/immunology , Humans , Inflammation , Interleukin-15/genetics , Interleukin-15/immunology , Intestinal Mucosa/pathology , Lymphocyte Activation , Mice , Mice, Transgenic , Protein Glutamine gamma Glutamyltransferase 2 , T-Lymphocytes/immunology , Transgenes/genetics , Transglutaminases/immunology
20.
Blood ; 117(18): 4787-95, 2011 May 05.
Article En | MEDLINE | ID: mdl-21385847

IL-15 uses the heterotrimeric receptor IL-2/IL-15Rß and the γ chain shared with IL-2 and the cytokine-specific IL-15Rα. Although IL-15 shares actions with IL-2 that include activation of natural killer (NK) and CD8 T cells, IL-15 is not associated with capillary leak syndrome, activation-induced cell death, or with a major effect on the number of functional regulatory T cells. To prepare for human trials to determine whether IL-15 is superior to IL-2 in cancer therapy, recombinant human IL-15 (rhIL-15) was produced under current good manufacturing practices. A safety study in rhesus macaques was performed in 4 groups of 6 animals each that received vehicle diluent control or rhIL-15 at 10, 20, or 50 µg/kg/d IV for 12 days. The major toxicity was grade 3/4 transient neutropenia. Bone marrow examinations demonstrated increased marrow cellularity, including cells of the neutrophil series. Furthermore, neutrophils were observed in sinusoids of enlarged livers and spleens, suggesting that IL-15 mediated neutrophil redistribution from the circulation to tissues. The observation that IL-15 administration was associated with increased numbers of circulating NK and CD8 central and effector-memory T cells, in conjunction with efficacy studies in murine tumor models, supports the use of multiple daily infusions of rhIL-15 in patients with metastatic malignancies.


Interleukin-15/toxicity , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Blood Coagulation/drug effects , Bone Marrow/drug effects , Bone Marrow/pathology , Humans , Immunotherapy , Infusions, Intravenous , Interleukin-15/administration & dosage , Interleukin-15/immunology , Interleukin-15/pharmacokinetics , Liver/drug effects , Liver/pathology , Macaca mulatta , Neoplasms/immunology , Neoplasms/therapy , Neutropenia/blood , Neutropenia/chemically induced , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/toxicity
...