Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Neuroimmunomodulation ; 22(3): 203-12, 2015.
Article in English | MEDLINE | ID: mdl-25301381

ABSTRACT

OBJECTIVES: Vasoactive intestinal peptide (VIP) is an immunomodulatory neuropeptide with therapeutic properties in multiple murine models of inflammatory disease including the trinitrobenzene-sulfonic acid (TNBS)-colitis model of Crohn's disease. Understanding the spectrum of biological actions of endogenously produced VIP may help us dissect the complex and multifactorial pathogenesis of such inflammatory diseases. Our goal was to determine the contribution of endogenously produced VIP to TNBS-colitis by using VIP knockout (KO) mice. METHODS: TNBS was intracolonically administered to wild-type (WT) and VIP KO mice, and weight loss and colitis were assessed over time. Colon histopathological changes and myeloperoxidase activities were analyzed and the levels of tumor necrosis factor (TNF)-α and interleukin (IL)-6 in colon and serum quantified. The proliferative response in vitro of splenocytes from TNBS WT and VIP KO administered mice to anti-CD3 and anti-CD28 was determined. RESULTS: VIP KO mice did not exhibit the predicted exacerbated response to TNBS. Instead, they developed a milder clinical profile than WT mice, with lower TNF-α and IL-6 levels. Such potential defects seem selective, because other parameters such as the histopathological scores and the cytokine levels in the colon did not differ between the two strains of mice. Moreover, splenocytes from TNBS-treated VIP KO mice exhibited an enhanced proliferative response to anti-CD3/CD28 stimulation in vitro. CONCLUSION: Chronic loss of VIP in mice leads to a disruption of certain but not all immunological compartments, corroborating recent findings that VIP KO mice exhibit reduced mortality in the lipopolysaccharide-induced endotoxemia model and attenuated clinical development of experimental autoimmune encephalomyelitis while developing robust T-cell responses.


Subject(s)
Colitis/chemically induced , Colitis/pathology , Colon/pathology , Trinitrobenzenesulfonic Acid/toxicity , Vasoactive Intestinal Peptide/deficiency , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colon/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxidase/metabolism , RNA, Messenger/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Time Factors , Vasoactive Intestinal Peptide/genetics
2.
J Immunother ; 43(9): 273-282, 2020.
Article in English | MEDLINE | ID: mdl-32925563

ABSTRACT

Expression of carbonic-anhydrase IX (CAIX) in clear cell renal cell carcinoma (RCC) makes it an attractive vaccine target. We developed a fusion-gene construct, granulocyte-macrophage (GM) colony-stimulating factor+CAIX, delivered by an adenoviral vector (Ad) into autologous dendritic cells (DCs) in this phase 1 study. The injected immature DCs were expected to stimulate an antigen-specific immune response against CAIX expressing RCC. Three dose-escalation cohorts (5, 15, and 50×10 cells/administration) were injected intradermally q2wk×3 doses based on a 3+3 design. The primary objective was the safety of the injections. Secondary objectives were immune responses using enzyme-linked immunosorbent spot, a serum biomarker panel, and clinical response. Fifteen patients with metastatic RCC were enrolled, and 9 patients received all 3 doses. No serious adverse events were seen. There were 3 (33%) patients with grade 1 fatigue, 1 of whom subsequently experienced grade 2 fatigue. One patient (11%) experienced grade 1-2 leukopenia. Only 1 patient (11%) experienced grade 2 flu-like symptoms. Of the 9 patients who received treatment, 1 expired of progressive disease, 2 patients were lost to follow-up and 6 patients are alive. Of the 6 patients, 5 have progressive disease, and 1 has completed treatment with stable disease at 27 months follow-up. Immune response measurements appeared more robust in higher dose cohorts, which appeared to be related to patients with stable disease at 3 months. These early data show that autologous immature DC-AdGMCAIX can be safely given to metastatic RCC patients without any serious adverse events with CAIX-specific immune response elicited by the treatment. These preliminary data support further study of Ad-GMCAIX, particularly with combination therapies that may enhance clinical activity.


Subject(s)
Antigens, Neoplasm/genetics , Cancer Vaccines/administration & dosage , Carbonic Anhydrase IX/genetics , Carcinoma, Renal Cell/therapy , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Kidney Neoplasms/therapy , Antigens, Neoplasm/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/genetics , Cancer Vaccines/metabolism , Carbonic Anhydrase IX/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Dendritic Cells/metabolism , Disease Management , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Treatment Outcome
3.
Nat Cancer ; 1(1): 46-58, 2020.
Article in English | MEDLINE | ID: mdl-34368780

ABSTRACT

Lack of tumor infiltration by immune cells is the main mechanism of primary resistance to programmed cell death protein 1 (PD-1) blockade therapies for cancer. It has been postulated that cancer cell-intrinsic mechanisms may actively exclude T cells from tumors, suggesting that the finding of actionable molecules that could be inhibited to increase T cell infiltration may synergize with checkpoint inhibitor immunotherapy. Here, we show that p21-activated kinase 4 (PAK4) is enriched in non-responding tumor biopsies with low T cell and dendritic cell infiltration. In mouse models, genetic deletion of PAK4 increased T cell infiltration and reversed resistance to PD-1 blockade in a CD8 T cell-dependent manner. Furthermore, combination of anti-PD-1 with the PAK4 inhibitor KPT-9274 improved anti-tumor response compared with anti-PD-1 alone. Therefore, high PAK4 expression is correlated with low T cell and dendritic cell infiltration and a lack of response to PD-1 blockade, which could be reversed with PAK4 inhibition.


Subject(s)
Immune Checkpoint Inhibitors , Immunotherapy , Neoplasms , Programmed Cell Death 1 Receptor , p21-Activated Kinases , Animals , CD8-Positive T-Lymphocytes , Mice , Neoplasms/drug therapy , p21-Activated Kinases/genetics
4.
Cancer Discov ; 10(8): 1140-1157, 2020 08.
Article in English | MEDLINE | ID: mdl-32467343

ABSTRACT

Mechanism-based strategies to overcome resistance to PD-1 blockade therapy are urgently needed. We developed genetic acquired resistant models of JAK1, JAK2, and B2M loss-of-function mutations by gene knockout in human and murine cell lines. Human melanoma cell lines with JAK1/2 knockout became insensitive to IFN-induced antitumor effects, while B2M knockout was no longer recognized by antigen-specific T cells and hence was resistant to cytotoxicity. All of these mutations led to resistance to anti-PD-1 therapy in vivo. JAK1/2-knockout resistance could be overcome with the activation of innate and adaptive immunity by intratumoral Toll-like receptor 9 agonist administration together with anti-PD-1, mediated by natural killer (NK) and CD8 T cells. B2M-knockout resistance could be overcome by NK-cell and CD4 T-cell activation using the CD122 preferential IL2 agonist bempegaldesleukin. Therefore, mechanistically designed combination therapies can overcome genetic resistance to PD-1 blockade therapy. SIGNIFICANCE: The activation of IFN signaling through pattern recognition receptors and the stimulation of NK cells overcome genetic mechanisms of resistance to PD-1 blockade therapy mediated through deficient IFN receptor and antigen presentation pathways. These approaches are being tested in the clinic to improve the antitumor activity of PD-1 blockade therapy.This article is highlighted in the In This Issue feature, p. 1079.


Subject(s)
Drug Resistance, Neoplasm/genetics , Janus Kinase 1/genetics , Janus Kinase 2/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , beta 2-Microglobulin/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Interferons/pharmacology , Interleukin-2/analogs & derivatives , Interleukin-2/immunology , Interleukin-2/pharmacology , Interleukin-2/therapeutic use , Killer Cells, Natural/immunology , Loss of Function Mutation , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Toll-Like Receptor 9/immunology
5.
Nat Commun ; 11(1): 660, 2020 01 31.
Article in English | MEDLINE | ID: mdl-32005809

ABSTRACT

Interleukin-2 (IL-2) is a component of most protocols of adoptive cell transfer (ACT) therapy for cancer, but is limited by short exposure and high toxicities. NKTR-214 is a kinetically-engineered IL-2 receptor ßγ (IL-2Rßγ)-biased agonist consisting of IL-2 conjugated to multiple releasable polyethylene glycol chains resulting in sustained signaling through IL-2Rßγ. We report that ACT supported by NKTR-214 increases the proliferation, homing and persistence of anti-tumor T cells compared to ACT with IL-2, resulting in superior antitumor activity in a B16-F10 murine melanoma model. The use of NKTR-214 increases the number of polyfunctional T cells in murine spleens and tumors compared to IL-2, and enhances the polyfunctionality of T and NK cells in the peripheral blood of patients receiving NKTR-214 in a phase 1 trial. In conclusion, NKTR-214 may have the potential to improve the antitumor activity of ACT in humans through increased in vivo expansion and polyfunctionality of the adoptively transferred T cells.


Subject(s)
Adoptive Transfer , Interleukin-2/analogs & derivatives , Interleukin-2/agonists , Melanoma/drug therapy , Polyethylene Glycols/administration & dosage , Receptors, Interleukin-2/immunology , T-Lymphocytes/immunology , Animals , Humans , Interleukin-2/administration & dosage , Interleukin-2/immunology , Lymphocyte Activation/drug effects , Melanoma/genetics , Melanoma/immunology , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Receptors, Interleukin-2/genetics
6.
J Steroid Biochem Mol Biol ; 193: 105415, 2019 10.
Article in English | MEDLINE | ID: mdl-31226312

ABSTRACT

Breast cancers (BCs) with expression of estrogen receptor-alpha (ERα) occur in more than 70% of newly-diagnosed patients in the U.S. Endocrine therapy with antiestrogens or aromatase inhibitors is an important intervention for BCs that express ERα, and it remains one of the most effective targeted treatment strategies. However, a substantial proportion of patients with localized disease, and essentially all patients with metastatic BC, become resistant to current endocrine therapies. ERα is present in most resistant BCs, and in many of these its activity continues to regulate BC growth. Fulvestrant represents one class of ERα antagonists termed selective ER downregulators (SERDs). Treatment with fulvestrant causes ERα down-regulation, an event that helps overcome several resistance mechanisms. Unfortunately, full antitumor efficacy of fulvestrant is limited by its poor bioavailability in clinic. We have designed and tested a new generation of steroid-like SERDs. Using ERα-positive BC cells in vitro, we find that these compounds suppress ERα protein levels with efficacy similar to fulvestrant. Moreover, these new SERDs markedly inhibit ERα-positive BC cell transcription and proliferation in vitro even in the presence of estradiol-17ß. In vivo, the SERD termed JD128 significantly inhibited tumor growth in MCF-7 xenograft models in a dose-dependent manner (P < 0.001). Further, our findings indicate that these SERDs also interact with ER-positive immune cells in the tumor microenvironment such as myeloid-derived suppressor cells (MDSC), tumor infiltrating lymphocytes and other selected immune cell subpopulations. SERD-induced inhibition of MDSCs and concurrent actions on CD8+ and CD4 + T-cells promotes interaction of immune checkpoint inhibitors with BC cells in preclinical models, thereby leading to enhanced tumor killing even among highly aggressive BCs such as triple-negative BC that lack ERα expression. Since monotherapy with immune checkpoint inhibitors has not been effective for most BCs, combination therapies with SERDs that enhance immune recognition may increase immunotherapy responses in BC and improve patient survival. Hence, ERα antagonists that also promote ER downregulation may potentially benefit patients who are unresponsive to current endocrine therapies.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Estrogen Antagonists/therapeutic use , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/immunology , Estrogen Antagonists/pharmacology , Female , Fulvestrant/pharmacology , Fulvestrant/therapeutic use , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred BALB C , Mice, Nude , Receptors, Estrogen/metabolism
7.
Clin Cancer Res ; 25(7): 2096-2108, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30573690

ABSTRACT

PURPOSE: Transgenic adoptive cell therapy (ACT) targeting the tumor antigen NY-ESO-1 can be effective for the treatment of sarcoma and melanoma. Preclinical models have shown that this therapy can be improved with the addition of dendritic cell (DC) vaccination and immune checkpoint blockade. We studied the safety, feasibility, and antitumor efficacy of transgenic ACT with DC vaccination, with and without CTLA-4 blockade with ipilimumab. PATIENTS AND METHODS: Freshly prepared autologous NY-ESO-1-specific T-cell receptor (TCR) transgenic lymphocytes were adoptively transferred together with NY-ESO-1 peptide-pulsed DC vaccination in HLA-A2.1-positive subjects alone (ESO, NCT02070406) or with ipilimumab (INY, NCT01697527) in patients with advanced sarcoma or melanoma. RESULTS: Six patients were enrolled in the ESO cohort, and four were enrolled in the INY cohort. Four out of six patients treated per ESO (66%), and two out of four patients treated per INY (50%) displayed evidence of tumor regression. Peripheral blood reconstitution with NY-ESO-1-specific T cells peaked within 2 weeks of ACT, indicating rapid in vivo expansion. Tracking of transgenic T cells to the tumor sites was demonstrated in on-treatment biopsies via TCR sequencing. Multiparametric mass cytometry of transgenic cells demonstrated shifting of transgenic cells from memory phenotypes to more terminally differentiated effector phenotypes over time. CONCLUSIONS: ACT of fresh NY-ESO-1 transgenic T cells prepared via a short ex vivo protocol and given with DC vaccination, with or without ipilimumab, is feasible and results in transient antitumor activity, with no apparent clinical benefit of the addition of ipilimumab. Improvements are needed to maintain tumor responses.


Subject(s)
Adoptive Transfer , Antineoplastic Agents, Immunological/pharmacology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Ipilimumab/pharmacology , Neoplasms/immunology , Neoplasms/therapy , Adoptive Transfer/methods , Adult , Animals , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Combined Modality Therapy , Dendritic Cells/metabolism , Female , Gene Knock-In Techniques , Humans , Immunotherapy , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Mice , Middle Aged , Molecular Targeted Therapy , Neoplasms/pathology , Phenotype , Pilot Projects , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Young Adult
8.
Clin Cancer Res ; 25(3): 1000-1011, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30409823

ABSTRACT

PURPOSE: To improve persistence of adoptively transferred T-cell receptor (TCR)-engineered T cells and durable clinical responses, we designed a clinical trial to transplant genetically-modified hematopoietic stem cells (HSCs) together with adoptive cell transfer of T cells both engineered to express an NY-ESO-1 TCR. Here, we report the preclinical studies performed to enable an investigational new drug (IND) application. EXPERIMENTAL DESIGN: HSCs transduced with a lentiviral vector expressing NY-ESO-1 TCR and the PET reporter/suicide gene HSV1-sr39TK and T cells transduced with a retroviral vector expressing NY-ESO-1 TCR were coadministered to myelodepleted HLA-A2/Kb mice within a formal Good Laboratory Practice (GLP)-compliant study to demonstrate safety, persistence, and HSC differentiation into all blood lineages. Non-GLP experiments included assessment of transgene immunogenicity and in vitro viral insertion safety studies. Furthermore, Good Manufacturing Practice (GMP)-compliant cell production qualification runs were performed to establish the manufacturing protocols for clinical use. RESULTS: TCR genetically modified and ex vivo-cultured HSCs differentiated into all blood subsets in vivo after HSC transplantation, and coadministration of TCR-transduced T cells did not result in increased toxicity. The expression of NY-ESO-1 TCR and sr39TK transgenes did not have a detrimental effect on gene-modified HSC's differentiation to all blood cell lineages. There was no evidence of genotoxicity induced by the lentiviral vector. GMP batches of clinical-grade transgenic cells produced during qualification runs had adequate stability and functionality. CONCLUSIONS: Coadministration of HSCs and T cells expressing an NY-ESO-1 TCR is safe in preclinical models. The results presented in this article led to the FDA approval of IND 17471.


Subject(s)
Genetic Therapy/methods , Hematopoietic Stem Cells/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/genetics , Cells, Cultured , Clinical Trials as Topic , Drugs, Investigational/therapeutic use , HLA-A2 Antigen/genetics , Hematopoietic Stem Cells/metabolism , Humans , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
9.
Neuroreport ; 17(3): 309-12, 2006 Feb 27.
Article in English | MEDLINE | ID: mdl-16462603

ABSTRACT

CD4+ (T helper) lymphocytes appear to play important roles in neuron survival and regeneration after injury, although their functions in regulating gene expression in injured neurons are unknown. Mice with targeted mutations in the STAT4 and STAT6 genes are deficient in T helper (Th)1 and Th2 responses, respectively, and have been used to determine the relative importance of T helper subsets in a variety of inflammatory processes. As pituitary adenylyl cyclase-activating peptide mRNA is normally strongly induced in facial motor neurons after axotomy, we examined this induction in Th1 and Th2 lymphocyte-deficient and control Balb/C wild-type mice. As previously reported, pituitary adenylyl cyclase-activating peptide gene expression was strongly induced in ipsilateral but not contralateral motor neurons in the facial motor nucleus of wild-type mice. The mean number of hybridizing motor neurons in STAT4-deficient mice did not differ from that in wild-type mice, whereas the number in STAT6 mice was reduced by more than 50%. The results indicate that STAT6 plays a key role in the upregulation of pituitary adenylyl cyclase-activating peptide gene expression in facial motor neurons after injury, possibly through its role in regulating T helper cell differentiation to the type 2 phenotype.


Subject(s)
Facial Nerve Injuries/metabolism , Facial Nerve Injuries/pathology , Gene Expression Regulation/genetics , Motor Neurons/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , T-Lymphocytes/physiology , Animals , Axotomy/methods , Cell Count/methods , Facial Nerve Injuries/etiology , Functional Laterality/physiology , In Situ Hybridization/methods , Mice , Mice, Inbred BALB C , Mice, Knockout , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , RNA, Messenger/metabolism , STAT4 Transcription Factor/deficiency , STAT6 Transcription Factor/deficiency
11.
Neuroreport ; 15(17): 2647-50, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15570170

ABSTRACT

PACAP is a neuropeptide with putative neuroprotective, regenerative, and immunomodulatory actions. PACAP mRNA is up-regulated in motor neurons following facial nerve axotomy in wild type, but not immunodeficient SCID mice. Because CD4+ lymphocytes appear to be neuroprotective in facial nerve and other injury models, we studied PACAP gene expression in SCID mice preinfused with CD4+ enriched splenocytes. Whereas the mean number of PACAP hybridizing neurons after axotomy was reduced by 75% in uninfused SCID mice, infusion of CD4+ enriched splenocytes restored the number to a value not significantly different than controls. The CD4+ cell-dependent induction of PACAP in motor neurons may thus be a factor in the cascade of events triggered by immune cells that ultimately lead to nerve regeneration.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Gene Expression Regulation/physiology , Nerve Growth Factors/metabolism , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Analysis of Variance , Animals , Axotomy/methods , Facial Nerve/cytology , Facial Nerve/physiology , Flow Cytometry/methods , Functional Laterality/physiology , In Situ Hybridization/methods , Mice , Mice, Inbred BALB C , Mice, SCID , Motor Neurons/metabolism , Nerve Growth Factors/genetics , Neuropeptides/genetics , Neurotransmitter Agents/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide , RNA, Messenger/metabolism , Transcriptional Activation
12.
PLoS One ; 7(5): e36922, 2012.
Article in English | MEDLINE | ID: mdl-22615845

ABSTRACT

Vasoactive intestinal peptide (VIP) is a pleiotropic neuropeptide with immunomodulatory properties. The administration of this peptide has been shown to have beneficial effects in murine models of inflammatory diseases including septic shock, rheumatoid arthritis, multiple sclerosis (MS) and Crohn's disease. However, the role of the endogenous peptide in inflammatory disease remains obscure because VIP-deficient mice were recently found to exhibit profound resistance in a model of MS. In the present study, we analyzed the response of female VIP deficient (KO) mice to intraperitoneal lipopolysaccharide (LPS) administration. We observed significant resistance to LPS in VIP KO mice, as evidenced by lower mortality and reduced tissue damage. The increased survival was associated with decreased levels of proinflammatory cytokines (TNFα, IL-6 and IL-12) in sera and peritoneal suspensions of these mice. Moreover, the expression of TNFα and IL-6 mRNA was reduced in peritoneal cells, spleens and lungs from LPS-treated VIP KO vs. WT mice, suggesting that the resistance might be mediated by an intrinsic defect in the responsiveness of immune cells to endotoxin. In agreement with this hypothesis, peritoneal cells isolated from VIP KO naive mice produced lower levels of proinflammatory cytokines in response to LPS in vitro. Finally, decreased NF-κB pathway activity in peritoneal cells was observed both in vivo and in vitro, as determined by assay of phosphorylated I-κB. The results demonstrate that female VIP KO mice exhibit resistance to LPS-induced shock, explainable in part by the presence of an intrinsic defect in the responsiveness of inflammatory cells to endotoxin.


Subject(s)
Endotoxemia/immunology , Inflammation Mediators/immunology , Inflammation/immunology , Lipopolysaccharides/pharmacology , Neuropeptides/immunology , Vasoactive Intestinal Peptide/deficiency , Vasoactive Intestinal Peptide/immunology , Animals , Endotoxemia/chemically induced , Endotoxins/administration & dosage , Endotoxins/immunology , Female , Inflammation/blood , Inflammation/genetics , Inflammation/metabolism , Interleukin-12/blood , Interleukin-12/immunology , Interleukin-6/blood , Interleukin-6/immunology , Lipopolysaccharides/immunology , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/immunology , NF-kappa B/metabolism , Neuropeptides/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology , Spleen/immunology , Spleen/metabolism , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology , Vasoactive Intestinal Peptide/genetics
13.
J Clin Oncol ; 28(8): 1358-65, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20142597

ABSTRACT

PURPOSE Differences in cellular levels of histone modifications have predicted clinical outcome in certain cancers. Here, we studied the prognostic and predictive value of three histone modifications in pancreatic adenocarcinoma. METHODS Tissue microarrays (TMAs) from two pancreatic adenocarcinoma cohorts were examined, including those from a 195-patient cohort from Radiation Therapy Oncology Group trial RTOG 9704, a multicenter, phase III, randomized treatment trial comparing adjuvant gemcitabine with fluorouracil and a 140-patient cohort of patients with stage I or II cancer from University of California, Los Angeles Medical Center. Immunohistochemistry was performed for histone H3 lysine 4 dimethylation (H3K4me2), histone H3 lysine 9 dimethylation (H3K9me2), and histone H3 lysine 18 acetylation (H3K18ac). Positive tumor cell staining for each histone modification was used to classify patients into low- and high-staining groups, which were related to clinicopathologic parameters and clinical outcome measures. Results Low cellular levels of H3K4me2, H3K9me2, or H3K18ac were each significant and independent predictors of poor survival in univariate and multivariate models, and combined low levels of H3K4me2 and/or H3K18ac were the most significant predictor of overall survival (hazard ratio, 2.93; 95% CI, 1.78 to 4.82) in the University of California, Los Angeles cohort. In subgroup analyses, histone levels were predictive of survival specifically for those patients with node-negative cancer or for those patients receiving adjuvant fluorouracil, but not gemcitabine, in RTOG 9704. CONCLUSION Cellular levels of histone modifications define previously unrecognized subsets of patients with pancreatic adenocarcinoma with distinct epigenetic phenotypes and clinical outcomes and represent prognostic and predictive biomarkers that could inform clinical decisions, including the use of fluorouracil chemotherapy.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/genetics , Epigenesis, Genetic , Histones/metabolism , Pancreatic Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Antineoplastic Agents/pharmacology , Chemotherapy, Adjuvant , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/genetics , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Humans , Multivariate Analysis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Prognosis , Randomized Controlled Trials as Topic , Survival Analysis , Tissue Array Analysis , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL