Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Sci Adv ; 9(1): eade8272, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36598976

ABSTRACT

Spleen tyrosine kinase (SYK) is a previously unidentified therapeutic target that inhibits neutrophil and macrophage activation in coronavirus disease 2019 (COVID-19). Fostamatinib, a SYK inhibitor, was studied in a phase 2 placebo-controlled randomized clinical trial and was associated with improvements in many secondary end points related to efficacy. Here, we used a multiomic approach to evaluate cellular and soluble immune mediator responses of patients enrolled in this trial. We demonstrated that SYK inhibition was associated with reduced neutrophil activation, increased circulation of mature neutrophils (CD10+CD33-), and decreased circulation of low-density granulocytes and polymorphonuclear myeloid-derived suppressor cells (HLA-DR-CD33+CD11b-). SYK inhibition was also associated with normalization of transcriptional activity in circulating monocytes relative to healthy controls, an increase in frequency of circulating nonclassical and HLA-DRhi classical monocyte populations, and restoration of interferon responses. Together, these data suggest that SYK inhibition may mitigate proinflammatory myeloid cellular and soluble mediator responses thought to contribute to immunopathogenesis of severe COVID-19.


Subject(s)
COVID-19 , Humans , Syk Kinase , Oxazines/pharmacology , Oxazines/therapeutic use , HLA-DR Antigens , Homeostasis
2.
Mol Ther Oncolytics ; 28: 74-87, 2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36699615

ABSTRACT

Multiple clinical trials exploring the potential of adoptive natural killer (NK) cell therapy for cancer have employed ex vivo expansion using feeder cells to obtain large numbers of NK cells. We have previously utilized the rhesus macaque model to clonally track the NK cell progeny of barcode-transduced CD34+ stem and progenitor cells after transplant. In this study, NK cells from barcoded rhesus macaques were used to study the changes in NK cell clonal patterns that occurred during ex vivo expansion using culture protocols similar to those employed in clinical preparation of human NK cells including irradiated lymphoblastoid cell line (LCL) feeder cells or K562 cells expressing 4-1BBL and membrane-bound interleukin-21 (IL-21). NK expansion cultures resulted in the proliferation of clonally diverse NK cells, which, at day 14 harvest, contained greater than 50% of the starting barcode repertoire. Diversity as measured by Shannon index was maintained after culture. With both LCL and K562 feeders, proliferation of long-lived putative memory-like NK cell clones was observed, with these clones continuing to constitute a mean of 31% of the total repertoire of expanded cells. These experiments provide insight into the clonal makeup of expanded NK cell clinical products.

3.
J Immunother Cancer ; 10(2)2022 02.
Article in English | MEDLINE | ID: mdl-35135865

ABSTRACT

BACKGROUND: Adoptive transfer of natural killer (NK) cells with augmented antibody-dependent cellular cytotoxicity (ADCC) capabilities and resistance to CD38 targeting has the potential to enhance the clinical anti-myeloma activity of daratumumab (DARA). Therefore, we sought to develop an efficient CRISPR/Cas9-based gene editing platform to disrupt CD38 expression (CD38 knockout (KO)) in ex vivo expanded NK cells and simultaneously arm CD38KO NK cells with a high-affinity CD16 (CD16-158V) receptor. METHODS: CD38KO human NK cells were generated using Cas9 ribonucleoprotein complexes. The platform was expanded by incorporating messenger RNA (mRNA) transfection of CD38KO NK cells and targeted gene insertion at the CD38 locus to mediate gene knockin (KI). The capacity of these gene-edited NK cells to persist and mediate ADCC in the presence of DARA was tested in vitro and in a MM.1S xenograft mouse model. RESULTS: Highly efficient CD38 gene disruption was achieved in ex vivo expanded NK cells without affecting their proliferative or functional capacity. CD38 KO conferred resistance to DARA-induced NK cell fratricide, enabling persistence and augmented ADCC against myeloma cell lines in the presence of DARA in vitro and in a MM.1S xenograft mouse model. CD38KO NK cells could be further modified by transfection with mRNA encoding a CD16-158V receptor, resulting in augmented DARA-mediated ADCC. Finally, we observed that a homology-directed repair template targeted to the CD38 locus facilitated an efficient 2-in-1 CD38 KO coupled with KI of a truncated CD34 reporter and CD16-158V receptor, with CD38KO/CD16KI NK cells demonstrating a further enhancement of DARA-mediated ADCC both in vitro and in vivo. CONCLUSIONS: Adoptive immunotherapy using ex vivo expanded CD38KO/CD16KI NK cells has the potential to boost the clinical efficacy of DARA. By incorporating complementary genetic engineering strategies into a CD38 KO manufacturing platform, we generated NK cells with substantially augmented CD38-directed antitumor activity, establishing a strong rationale for exploring this immunotherapy strategy in the clinic.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , CRISPR-Cas Systems/immunology , Gene Editing/methods , Immunotherapy/methods , Killer Cells, Natural/metabolism , Animals , Cell Line, Tumor , Humans , Luciferases, Firefly , Mice , Mice, Inbred NOD , Transfection
4.
Clin Infect Dis ; 75(1): e491-e498, 2022 08 24.
Article in English | MEDLINE | ID: mdl-34467402

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19) requiring hospitalization is characterized by robust antibody production, dysregulated immune response, and immunothrombosis. Fostamatinib is a novel spleen tyrosine kinase inhibitor that we hypothesize will ameliorate Fc activation and attenuate harmful effects of the anti-COVID-19 immune response. METHODS: We conducted a double-blind, randomized, placebo-controlled trial in hospitalized adults requiring oxygen with COVID-19 where patients receiving standard of care were randomized to receive fostamatinib or placebo. The primary outcome was serious adverse events by day 29. RESULTS: A total of 59 patients underwent randomization (30 to fostamatinib and 29 to placebo). Serious adverse events occurred in 10.5% of patients in the fostamatinib group compared with 22% in placebo (P = .2). Three deaths occurred by day 29, all receiving placebo. The mean change in ordinal score at day 15 was greater in the fostamatinib group (-3.6 ±â€…0.3 vs -2.6 ±â€…0.4, P = .035) and the median length in the intensive care unit was 3 days in the fostamatinib group vs 7 days in placebo (P = .07). Differences in clinical improvement were most evident in patients with severe or critical disease (median days on oxygen, 10 vs 28, P = .027). There were trends toward more rapid reductions in C-reactive protein, D-dimer, fibrinogen, and ferritin levels in the fostamatinib group. CONCLUSION: For COVID-19 requiring hospitalization, the addition of fostamatinib to standard of care was safe and patients were observed to have improved clinical outcomes compared with placebo. These results warrant further validation in larger confirmatory trials. CLINICAL TRIALS REGISTRATION: Clinicaltrials.gov, NCT04579393.


Subject(s)
COVID-19 Drug Treatment , Adult , Aminopyridines , Double-Blind Method , Hospitalization , Humans , Morpholines , Oxazines/therapeutic use , Oxygen , Pyridines/therapeutic use , Pyrimidines , SARS-CoV-2 , Treatment Outcome
5.
Mol Ther Methods Clin Dev ; 20: 559-571, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33665226

ABSTRACT

Transduction of primary human natural killer (NK) cells with lentiviral vectors has historically been challenging. We sought to evaluate multiple parameters to optimize lentiviral transduction of human peripheral blood NK cells being expanded to large numbers using a good manufacturing practice (GMP)-compliant protocol that utilizes irradiated lymphoblastoid (LCL) feeder cells. Although prestimulation of NK cells with interleukin (IL)-2 for 2 or more days facilitated transduction with vesicular stomatitis virus glycoprotein (VSVG)-pseudotyped lentivirus, there was a subsequent impairment in the capacity of transduced NK cells to proliferate when stimulated with LCL feeder cells. In contrast, incubation of human NK cells with LCL feeder cells plus IL-2 before transduction in the presence of the TBK1 inhibitor BX795 resulted in efficient lentiviral integration (mean of 23% transgene+ NK cells) and successful subsequent proliferation of the transduced cells. Investigation of multiple internal promoter sequences within the same lentiviral vector revealed differences in percentage and level of transgene expression per NK cell. Bicistronic lentiviral vectors encoding both GFP and proteins suitable for the isolation of transduced cells with magnetic beads led to efficient transgene expression in NK cells. The optimized approaches described herein provide a template for protocols that generate large numbers of fully functional and highly purified lentivirus-transduced NK cells for clinical trials.

6.
Mol Ther ; 29(1): 47-59, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33010232

ABSTRACT

Many investigational adoptive immunotherapy regimens utilizing natural killer (NK) cells require the administration of interleukin-2 (IL-2) or IL-15, but these cytokines cause serious dose-dependent toxicities. To reduce or preclude the necessity for IL-2 use, we investigated whether genetic engineering of NK cells to express the erythropoietin (EPO) receptor (EPOR) or thrombopoietin (TPO) receptor (c-MPL) could be used as a method to improve NK cell survival and function. Viral transduction of NK-92 cells to express EPOR or c-MPL receptors conveyed signaling via appropriate pathways, protected cells from apoptosis, augmented cellular proliferation, and increased cell cytotoxic function in response to EPO or TPO ligands in vitro. In the presence of TPO, viral transduction of primary human NK cells to express c-MPL enhanced cellular proliferation and increased degranulation and cytokine production toward target cells in vitro. In contrast, transgenic expression of EPOR did not augment the proliferation of primary NK cells. In immunodeficient mice receiving TPO, in vivo persistence of primary human NK cells genetically modified to express c-MPL was higher compared with control NK cells. These data support the concept that genetic manipulation of NK cells to express hematopoietic growth factor receptors could be used as a strategy to augment NK cell proliferation and antitumor immunity.


Subject(s)
Gene Expression , Immunomodulation/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Neoplasms/immunology , Neoplasms/therapy , Receptors, Erythropoietin/genetics , Receptors, Thrombopoietin/genetics , Animals , Disease Models, Animal , Genetic Engineering , Humans , Immunotherapy/methods , Mice , Transgenes
7.
Hepatology ; 62(2): 546-57, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25712247

ABSTRACT

UNLABELLED: Clinical evidence suggests that many cases of serious idiosyncratic drug-induced liver injury are mediated by the adaptive immune system in response to hepatic drug-protein adducts, also referred to as "drug-induced allergic hepatitis"; but detailed mechanistic proof has remained elusive due to the lack of animal models. We have hypothesized that drug-induced allergic hepatitis is as rare in animals as it is in humans due at least in part to the tolerogenic nature of the liver. We provide evidence that immune tolerance can be overcome in a murine model of halothane-induced liver injury initiated by trifluoroacetylated protein adducts of halothane formed in the liver. Twenty-four hours after female Balb/cJ mice were initially treated with halothane, perivenous necrosis and an infiltration of CD11b(+) Gr-1(high) cells were observed in the liver. Further study revealed a subpopulation of myeloid-derived suppressor cells within the CD11b(+) Gr-1(high) cell fraction that inhibited the proliferation of both CD4(+) and CD8(+) T cells. When CD11b(+) Gr-1(high) cells were depleted from the liver with Gr-1 antibody treatment, enhanced liver injury was observed at 9 days after halothane rechallenge. Toxicity was associated with increased serum levels of interleukin-4 and immunoglobulins G1 and E directed against hepatic trifluoroacetylated protein adducts, as well as increased hepatic infiltration of eosinophils and CD4(+) T cells, all features of an allergic reaction. When hepatic CD4(+) T cells were depleted 5 days after halothane rechallenge, trifluoroacetylated protein adduct-specific serum immunoglobulin and hepatotoxicity were reduced. CONCLUSION: Our data provide a rational approach for developing animal models of drug-induced allergic hepatitis mediated by the adaptive immune system and suggest that impaired liver tolerance may predispose patients to this disease.


Subject(s)
CD11b Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Chemical and Drug Induced Liver Injury/immunology , Halothane/toxicity , Hepatitis/immunology , Myeloid Cells/metabolism , Alanine Transaminase/metabolism , Analysis of Variance , Animals , CD11b Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Female , Flow Cytometry , Hepatitis/pathology , Immunohistochemistry , Mice , Mice, Inbred BALB C , Myeloid Cells/drug effects , Nitric Oxide/metabolism , Random Allocation
8.
Hepatology ; 60(5): 1741-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24723460

ABSTRACT

UNLABELLED: Liver eosinophilia has been associated with incidences of drug-induced liver injury (DILI) for more than 50 years, although its role in this disease has remained largely unknown. In this regard, it was recently shown that eosinophils played a pathogenic role in a mouse model of halothane-induced liver injury (HILI). However, the signaling events that drove hepatic expression of eosinophil-associated chemokines, eotaxins, eosinophil infiltration, and subsequent HILI were unclear. We now provide evidence implicating hepatic epithelial-derived cytokine thymic stromal lymphopoietin (TSLP) and type 2 immunity, in particular, interleukin-4 (IL-4) production, in mediating hepatic eosinophilia and injury during HILI. TSLP was constitutively expressed by mouse hepatocytes and increased during HILI. Moreover, the severity of HILI was reduced in mice deficient in either the TSLP receptor (TSLPR) or IL-4 and was accompanied by decreases in serum levels of eotaxins and hepatic eosinophilia. Similarly, concanavalin A-induced liver injury, where type 2 cytokines and eosinophils play a significant role in its pathogenesis, was also reduced in TSLPR-deficient mice. Studies in vitro revealed that mouse and human hepatocytes produce TSLP and eotaxins in response to treatment with combinations of IL-4 and proinflammatory cytokines IL-1ß and tumor necrosis factor alpha. CONCLUSION: This report provides the first evidence implicating roles for hepatic TSLP signaling, type 2 immunity, and eosinophilia in mediating liver injury caused by a drug.


Subject(s)
Anesthetics, Inhalation/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Cytokines/metabolism , Halothane/adverse effects , Interleukin-4/metabolism , Animals , Concanavalin A , Female , Hepatitis, Animal/metabolism , Hepatocytes/metabolism , Humans , Mice, Inbred BALB C , Thymic Stromal Lymphopoietin
9.
Hepatology ; 57(5): 2026-36, 2013 May.
Article in English | MEDLINE | ID: mdl-23238640

ABSTRACT

UNLABELLED: Drug-induced liver injury (DILI) is a major health issue, as it remains difficult to predict which new drugs will cause injury and who will be susceptible to this disease. This is due in part to the lack of animal models and knowledge of susceptibility factors that predispose individuals to DILI. In this regard, liver eosinophilia has often been associated with DILI, although its role remains unclear. We decided to investigate this problem in a murine model of halothane-induced liver injury (HILI). When female Balb/cJ mice were administered halothane, eosinophils were detected by flow cytometry in the liver within 12 hours and increased thereafter proportionally to liver damage. Chemokines, eotaxin-1 (CCL11) and eotaxin-2 (CCL24), which are known to attract eosinophils, increased in response to halothane treatment. The severity of HILI was decreased significantly when the study was repeated in wildtype mice made deficient in eosinophils with a depleting antibody and in eosinophil lineage-ablated ΔdblGata(-/-) mice. Moreover, depletion of neutrophils by pretreating animals with Gr-1 antibody prior to halothane administration failed to reduce the severity of HILI at antibody concentrations that did not affect hepatic eosinophils. Immunohistochemical staining for the granule protein, major basic protein, revealed that eosinophils accumulated exclusively around areas of hepatocellular necrosis. CONCLUSION: Our findings indicate that eosinophils have a pathologic role in HILI in mice and suggest that they may contribute similarly in many clinical cases of DILI.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/physiopathology , Eosinophils/physiology , Halothane/adverse effects , Animals , Cell Movement , Chemical and Drug Induced Liver Injury/pathology , Chemokine CCL11/metabolism , Chemokine CCL24/metabolism , Comorbidity , Disease Models, Animal , Eosinophilia/epidemiology , Eosinophils/pathology , Female , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Prevalence
10.
Chem Res Toxicol ; 25(1): 83-93, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22107450

ABSTRACT

In a recent study, we reported that interleukin (IL)-4 had a protective role against acetaminophen (APAP)-induced liver injury (AILI), although the mechanism of protection was unclear. Here, we carried out more detailed investigations and have shown that one way IL-4 may control the severity of AILI is by regulating glutathione (GSH) synthesis. In the present studies, the protective role of IL-4 in AILI was established definitively by showing that C57BL/6J mice made deficient in IL-4 genetically (IL-4(-/-)) or by depletion with an antibody, were more susceptible to AILI than mice not depleted of IL-4. The increased susceptibility of IL-4(-/-) mice was not due to elevated levels of hepatic APAP-protein adducts but was associated with a prolonged reduction in hepatic GSH that was attributed to decreased gene expression of γ-glutamylcysteine ligase (γ-GCL). Moreover, administration of recombinant IL-4 to IL-4(-/-) mice postacetaminophen treatment diminished the severity of liver injury and increased γ-GCL and GSH levels. We also report that the prolonged reduction of GSH in APAP-treated IL-4(-/-) mice appeared to contribute toward increased liver injury by causing a sustained activation of c-Jun-N-terminal kinase (JNK) since levels of phosphorylated JNK remained significantly higher in the IL-4(-/-) mice up to 24 h after APAP treatment. Overall, these results show for the first time that IL-4 has a role in regulating the synthesis of GSH in the liver under conditions of cellular stress. This mechanism appears to be responsible at least in part for the protective role of IL-4 against AILI in mice and may have a similar role not only in AILI in humans but also in pathologies of the liver caused by other drugs and etiologies.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Glutathione/metabolism , Interleukin-4/metabolism , Animals , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 CYP2E1/metabolism , Glutamate-Cysteine Ligase/metabolism , Interleukin-4/deficiency , Interleukin-4/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/metabolism
11.
Clin Cancer Res ; 14(13): 4241-9, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18594006

ABSTRACT

PURPOSE: Exposing human tumor cells to sublethal doses of external beam radiation up-regulates expression of tumor antigen and accessory molecules, rendering tumor cells more susceptible to killing by antigen-specific CTLs. This study explored the possibility that exposure to palliative doses of a radiopharmaceutical agent could alter the phenotype of tumor cells to render them more susceptible to T cell-mediated killing. EXPERIMENTAL DESIGN: Here, 10 human tumor cell lines (4 prostate, 2 breast, and 4 lung) were exposed to increasing doses of the radiopharmaceutical samarium-153-ethylenediaminetetramethylenephosphonate ((153)Sm-EDTMP) used in cancer patients to treat pain due to bone metastasis. Fluorescence-activated cell sorting analysis and quantitative real-time PCR analysis for expression of five surface molecules and several tumor-associated antigens involved in prostate cancer were done. LNCaP human prostate cancer cells were exposed to (153)Sm-EDTMP and incubated with tumor-associated antigen-specific CTL in a CTL killing assay to determine whether exposure to (153)Sm-EDTMP rendered LNCaP cells more susceptible to T cell-mediated killing. RESULTS: Tumor cells up-regulated the surface molecules Fas (100% of cell lines up-regulated Fas), carcinoembryonic antigen (90%), mucin-1 (60%), MHC class I (50%), and intercellular adhesion molecule-1 (40%) in response to (153)Sm-EDTMP. Quantitative real-time PCR analysis revealed additional up-regulated tumor antigens. Exposure to (153)Sm-EDTMP rendered LNCaP cells more susceptible to killing by CTLs specific for prostate-specific antigen, carcinoembryonic antigen, and mucin-1. CONCLUSIONS: Doses of (153)Sm-EDTMP equivalent to palliative doses delivered to bone alter the phenotype of tumor cells, suggesting that (153)Sm-EDTMP may work synergistically with immunotherapy to increase the susceptibility of tumor cells to CTL killing.


Subject(s)
Breast Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Lung Neoplasms/therapy , Organometallic Compounds/therapeutic use , Organophosphorus Compounds/therapeutic use , Prostatic Neoplasms/therapy , Radioisotopes/pharmacology , Samarium/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Cell Line, Tumor , Cell Separation , Female , Humans , Male , Phenotype , T-Lymphocytes, Cytotoxic/metabolism , Treatment Outcome
12.
Clin Cancer Res ; 14(13): 4316-25, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18594015

ABSTRACT

PURPOSE: Saccharomyces cerevisiae, a nonpathogenic yeast, has been used previously as a vehicle to elicit immune responses to foreign antigens, and tumor-associated antigens, and has been shown to reduce tumor burden in mice. Studies were designed to determine if vaccination of human carcinoembryonic antigen (CEA)-transgenic (CEA-Tg) mice (where CEA is a self-antigen) with a recombinant S. cerevisiae construct expressing human CEA (yeast-CEA) elicits CEA-specific T-cell responses and antitumor activity. EXPERIMENTAL DESIGN: CEA-Tg mice were vaccinated with yeast-CEA, and CD4(+) and CD8(+) T-cell responses were assessed after one and multiple administrations or vaccinations at multiple sites per administration. Antitumor activity was determined by tumor growth and overall survival in both pulmonary metastasis and s.c. pancreatic tumor models. RESULTS: These studies demonstrate that recombinant yeast can break tolerance and that (a) yeast-CEA constructs elicit both CEA-specific CD4(+) and CD8(+) T-cell responses; (b) repeated yeast-CEA administration causes increased antigen-specific T-cell responses after each vaccination; (c) vaccination with yeast-CEA at multiple sites induces a greater T-cell response than the same dose given at a single site; and (d) tumor-bearing mice vaccinated with yeast-CEA show a reduction in tumor burden and increased overall survival compared to mock-treated or control yeast-vaccinated mice in both pulmonary metastasis and s.c. pancreatic tumor models. CONCLUSIONS: Vaccination with a heat-killed recombinant yeast expressing the tumor-associated antigen CEA induces CEA-specific immune responses, reduces tumor burden, and extends overall survival in CEA-Tg mice. These studies thus form the rationale for the incorporation of recombinant yeast-CEA and other recombinant yeast constructs in cancer immunotherapy protocols.


Subject(s)
Antigens, Neoplasm/chemistry , Antineoplastic Agents/pharmacology , Carcinoembryonic Antigen/chemistry , Gene Expression Regulation , Immunotherapy/methods , Saccharomyces cerevisiae/metabolism , Vaccines, DNA/chemistry , Animals , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines , Carcinoembryonic Antigen/metabolism , Cell Proliferation , Female , Humans , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Recombinant Proteins/chemistry , Vaccines, DNA/metabolism
13.
Biochem Biophys Res Commun ; 374(1): 6-10, 2008 Sep 12.
Article in English | MEDLINE | ID: mdl-18586006

ABSTRACT

Recent studies in mice suggest that stress-activated c-Jun N-terminal protein kinase 2 (JNK2) plays a pathologic role in acetaminophen (APAP)-induced liver injury (AILI), a major cause of acute liver failure (ALF). In contrast, we present evidence that JNK2 can have a protective role against AILI. When male C57BL/6J wild type (WT) and JNK2(-/-) mice were treated with 300mg APAP/kg, 90% of JNK2(-/-) mice died of ALF compared to 20% of WT mice within 48h. The high susceptibility of JNK2(-/-) mice to AILI appears to be due in part to deficiencies in hepatocyte proliferation and repair. Therefore, our findings are consistent with JNK2 signaling playing a protective role in AILI and further suggest that the use of JNK inhibitors as a potential treatment for AILI, as has been recommended by other investigators, should be reconsidered.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Liver Failure, Acute/chemically induced , Liver Failure, Acute/enzymology , Liver/drug effects , Mitogen-Activated Protein Kinase 9/physiology , Animals , Cyclin D , Cyclins/metabolism , Liver/enzymology , Liver/pathology , Liver Failure, Acute/genetics , Liver Regeneration/genetics , Male , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 9/genetics
14.
Cancer Immunol Immunother ; 57(8): 1173-83, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18256832

ABSTRACT

Radiolabeled monoclonal antibodies (mAb) have demonstrated measurable antitumor effects in hematologic malignancies. This outcome has been more difficult to achieve for solid tumors due, for the most part, to difficulties in delivering sufficient quantities of mAb to the tumor mass. Previous studies have shown that nonlytic levels of external beam radiation can render tumor cells more susceptible to T cell-mediated killing. The goal of these studies was to determine if the selective delivery of a radiolabeled mAb to tumors would modulate tumor cell phenotype so as to enhance vaccine-mediated T-cell killing. Here, mice transgenic for human carcinoembryonic antigen (CEA) were transplanted with a CEA expressing murine carcinoma cell line. Radioimmunotherapy consisted of yttrium-90 (Y-90)-labeled anti-CEA mAb, used either alone or in combination with vaccine therapy. A single dose of Y-90-labeled anti-CEA mAb, in combination with vaccine therapy, resulted in a statistically significant increase in survival in tumor-bearing mice over vaccine or mAb alone; this was shown to be mediated by engagement of the Fas/Fas ligand pathway. Mice receiving the combination therapy also showed a significant increase in the percentage of viable tumor-infiltrating CEA-specific CD8(+) T cells compared to vaccine alone. Mice cured of tumors demonstrated an antigen cascade resulting in CD4(+) and CD8(+) T-cell responses not only for CEA, but for p53 and gp70. These results show that systemic radiotherapy in the form of radiolabeled mAb, in combination with vaccine, promotes effective antitumor response, which may have implications in the design of future clinical trials.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cancer Vaccines/therapeutic use , Hematologic Neoplasms/radiotherapy , Radioimmunotherapy/methods , Animals , Antibodies, Monoclonal/pharmacokinetics , Antigens, Neoplasm/immunology , Cancer Vaccines/pharmacokinetics , Cell Line, Tumor , Mice , Mice, Transgenic , Sensitivity and Specificity , Tissue Distribution , Treatment Outcome , Up-Regulation/immunology , Yttrium Radioisotopes/pharmacokinetics , Yttrium Radioisotopes/therapeutic use , fas Receptor/immunology
15.
Vaccine ; 26(4): 509-21, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-18155327

ABSTRACT

Recombinant Saccharomyces cerevisiae (yeast) represents a unique and attractive vehicle to deliver antigens in vaccine immunotherapy protocols for cancer or infectious disease, in that it has been shown to be extremely safe and can be administered multiple times to hosts. In the studies reported here, we describe the effects of treatment with recombinant yeast on murine immature dendritic cells (DCs). Yeast expressing human carcinoembryonic antigen (CEA) as a model antigen was studied. Injection of mice subcutaneously with yeast-CEA resulted in rapid increases in MHC class II(+) cells and total antigen-presenting cells in draining lymph nodes. Post-treatment with yeast-CEA, DCs rapidly elevated both MHC class I and class II, numerous costimulatory molecules and other DC maturation markers, and secreted a range of Type I inflammatory cytokines. Gene expression arrays also revealed the rapid up-regulation of numerous cytokine and chemokine mRNAs, as well as genes involved in signal transduction and antigen uptake. Functional studies demonstrated enhanced allospecific reactivity of DCs following treatment with yeast-CEA or control yeast. Additionally, treatment of DCs with yeast-CEA resulted in specific activation of CEA-specific CD8(+) T cells in an MHC-restricted manner in vitro. Lastly, vaccination of CEA-transgenic mice with yeast-CEA elicited antigen-specific CD4(+) and CD8(+) immune responses in vivo. Thus, these studies taken together form a scientific rationale for the use of recombinant yeast in vaccination protocols for cancer or infectious diseases.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Genetic Vectors/immunology , Saccharomyces cerevisiae/immunology , Vaccination , Animals , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Carcinoembryonic Antigen/genetics , Cytokines/biosynthesis , Female , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Injections, Subcutaneous , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Neoplasms/therapy , Saccharomyces cerevisiae/genetics
16.
Front Biosci ; 12: 4900-10, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17569618

ABSTRACT

Since its discovery more than a hundred years ago, radiation has been used to treat cancer. In recent decades, advances in radiation technology have expanded the role and value of radiation in imaging and treating many forms of cancer. Currently, there is a growing interest in combining radiation with other modalities, such as immunotherapy, to treat a broad range of malignancies. This article reviews the use of standard and novel combinations of radiation therapy and immunotherapy to eradicate tumor cells. The combination of radiation therapy and immunotherapy holds particular promise as a strategy for cancer therapeutics for a variety of reasons. First, there is evidence that immunotherapy is most beneficial when employed early in the disease process and in combination with standard therapies. In addition, radiation may act synergistically with immunotherapy to enhance immune responses, inhibit immunosuppression, and/or alter the phenotype of tumor cells, thus rendering them more susceptible to immune-mediated killing. Finally, as monotherapies, both immunotherapy and radiation may be insufficient to eliminate tumor masses. However, following immunization with a cancer vaccine, the destruction of even a small percentage of tumor cells by radiation could result in cross-priming and presentation of tumor antigens to the immune system, thereby potentiating antitumor responses.


Subject(s)
Immunotherapy , Neoplasms/therapy , Brachytherapy , Humans , Neoplasm Metastasis , Neoplasms/radiotherapy , Radioimmunodetection , Radioimmunotherapy , Radiotherapy, Adjuvant
17.
Cancer Immunol Immunother ; 56(9): 1471-84, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17318654

ABSTRACT

Blockade of CTLA-4 by monoclonal antibodies (mAb) can mediate regression of tumors and increase the efficacy of tumor antigen specific vaccines. Blockade of CTLA-4 has also been shown to significantly increase the avidity of antigen-specific T cells after immunization with live recombinant viral vector based vaccine. Here, we demonstrate a biological synergy between CTLA-4 blockade and active vaccine therapy consisting of recombinant vaccinia and avipox viruses expressing carcinoembryonic antigen (CEA) and three T cell costimulatory molecules to enhance antitumor effects. However, this synergy was very much dependent on the temporal relationship of scheduling of the two agents. We evaluated the strategies in both a foreign antigen model using beta-galactosidase as immunogen, and in a "self" antigen model using CEA as immunogen. For antitumor activity the model used consisted of mice transgenic for human CEA and a murine carcinoma cell line transfected with CEA. The enhanced antitumor activity after vaccine and CTLA-4 blockade did not result in any signs of autoimmunity. These studies form a rational basis for the use of vector-based vaccines with anti-CTLA-4 and demonstrate that both enhancement of positive costimulatory signals and inhibition of negative costimulatory signals can be simultaneously exploited. These studies also underscore the importance of "drug" scheduling in vaccine combination therapies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cancer Vaccines/pharmacology , Drug Synergism , Immunization/methods , Lymphocyte Activation , Neoplasms/therapy , T-Lymphocytes/immunology , Vaccinia virus/genetics , Animals , Antibody Affinity , Antigens, CD/drug effects , Antigens, Differentiation/drug effects , CTLA-4 Antigen , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Recombinant Proteins/genetics
18.
J Exp Med ; 203(5): 1259-71, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16636135

ABSTRACT

Radiotherapy is one of the most successful cancer therapies. Here the effect of irradiation on antigen presentation by MHC class I molecules was studied. Cell surface expression of MHC class I molecules was increased for many days in a radiation dose-dependent manner as a consequence of three responses. Initially, enhanced degradation of existing proteins occurred which resulted in an increased intracellular peptide pool. Subsequently, enhanced translation due to activation of the mammalian target of rapamycin pathway resulted in increased peptide production, antigen presentation, as well as cytotoxic T lymphocyte recognition of irradiated cells. In addition, novel proteins were made in response to gamma-irradiation, resulting in new peptides presented by MHC class I molecules, which were recognized by cytotoxic T cells. We show that immunotherapy is successful in eradicating a murine colon adenocarcinoma only when preceded by radiotherapy of the tumor tissue. Our findings indicate that directed radiotherapy can improve the efficacy of tumor immunotherapy.


Subject(s)
Adenocarcinoma/immunology , Antigen Presentation/radiation effects , Colonic Neoplasms/immunology , Gamma Rays , HLA-A2 Antigen/immunology , Immunotherapy , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Animals , Antigen Presentation/immunology , Colonic Neoplasms/genetics , Colonic Neoplasms/therapy , Dose-Response Relationship, Radiation , Gene Expression Regulation, Neoplastic/immunology , Gene Expression Regulation, Neoplastic/radiation effects , HLA-A2 Antigen/genetics , Humans , Mice , Mice, Transgenic , Peptides/immunology , Protein Biosynthesis/immunology , Protein Biosynthesis/radiation effects , Protein Kinases/immunology , Radiotherapy , T-Lymphocytes, Cytotoxic/immunology , TOR Serine-Threonine Kinases
19.
Curr Pharm Des ; 12(3): 351-61, 2006.
Article in English | MEDLINE | ID: mdl-16454749

ABSTRACT

For the immune system to mount an effective antitumor T-cell response, an adequate number of T-cells specific for the antigens expressed by the malignancy must be activated [1]. Since most antigens expressed by tumors are "self"-antigens, tumor antigens often lack endogenous immunogenicity and thus do not sufficiently activate T-cells to levels that can mediate tumor eradication. In addition, virtually all solid tumor cells lack the costimulatory molecules necessary to activate tumor-specific T-cells. Approaches that stimulate immune responses to these tumor antigens have the potential to alter this poor responsiveness. This theory has promoted the use of active immunotherapy to generate immune responses against tumor-associated antigens (TAAs) for the treatment of cancer. As one such vaccine strategy, we have utilized poxviruses as delivery vehicles for TAAs in combination with T-cell costimulatory molecules. Initial studies have demonstrated that the insertion of costimulatory molecule trangenes into viral vectors, along with a TAA transgene, greatly enhances the immune response to the antigen. Using this approach, a TRIad of COstimulatory Molecules (TRICOM; B7-1, ICAM-1 and LFA-3) has been shown to enhance T-cell responses to TAAs to levels far greater than any one or two of the costimulatory molecules in combination. In this article, preclinical findings and recent clinical applications of TRICOM-based vaccines as a cancer immunotherapy are reviewed.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Animals , Clinical Trials as Topic , Combined Modality Therapy , Humans , Neoplasms/physiopathology , Neoplasms/prevention & control , Neoplasms/radiotherapy , Signal Transduction/drug effects , T-Lymphocytes/immunology , Vaccination , Vaccines, Synthetic/therapeutic use
20.
J Immunol ; 174(10): 5994-6004, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15879092

ABSTRACT

Recent studies in both animal models and clinical trials have demonstrated that the avidity of T cells is a major determinant of antitumor and antiviral immunity. In this study, we evaluated several different vaccine strategies for their ability to enhance both the quantity and avidity of CTL responses. CD8(+) T cell quantity was measured by tetramer binding precursor frequency, and avidity was measured by both tetramer dissociation and quantitative cytolytic function. We have evaluated a peptide, a viral vector expressing the Ag transgene alone, with one costimulatory molecule (B7-1), and with three costimulatory molecules (B7-1, ICAM-1, and LFA-3), with anti-CTLA-4 mAb, with GM-CSF, and combinations of the above. We have evaluated these strategies in both a foreign Ag model using beta-galactosidase as immunogen, and in a "self" Ag model, using carcinoembryonic Ag as immunogen in carcinoembryonic Ag transgenic mice. The combined use of several of these strategies was shown to enhance not only the quantity, but, to a greater magnitude, the avidity of T cells generated; a combination strategy is also shown to enhance antitumor effects. The results reported in this study thus demonstrate multiple strategies that can be used in both antitumor and antiviral vaccine settings to generate higher avidity host T cell responses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cytotoxicity Tests, Immunologic/methods , Lymphocyte Activation/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Adjuvants, Immunologic/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antigens, CD , Antigens, Differentiation/immunology , CTLA-4 Antigen , Cancer Vaccines/genetics , Carcinoembryonic Antigen/administration & dosage , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , Combined Modality Therapy , Female , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Binding/immunology , beta-Galactosidase/administration & dosage , beta-Galactosidase/genetics , beta-Galactosidase/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...