Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Clin Oncol ; 39(9): 966-977, 2021 03 20.
Article in English | MEDLINE | ID: mdl-33197226

ABSTRACT

PURPOSE: The role of maintenance therapy for gastric (GC) or gastroesophageal junction cancer (GEJC) is unclear. We investigated avelumab (anti-programmed death ligand-1 [PD-L1]) maintenance after first-line induction chemotherapy for GC/GEJC. PATIENTS AND METHODS: JAVELIN Gastric 100 was a global, open-label, phase III trial. Eligible patients had untreated, unresectable, human epidermal growth factor receptor 2-negative, locally advanced or metastatic GC or GEJC. Patients without progressive disease after 12 weeks of first-line chemotherapy with oxaliplatin plus a fluoropyrimidine were randomly assigned 1:1 to avelumab 10 mg/kg every 2 weeks or continued chemotherapy, stratified by region (Asia v non-Asia). The primary end point was overall survival (OS) after induction chemotherapy in all randomly assigned patients or the PD-L1-positive randomly assigned population (≥ 1% of tumor cells; 73-10 assay). RESULTS: A total of 805 patients received induction; 499 were randomly assigned to avelumab (n = 249) or continued chemotherapy (n = 250). Median OS was 10.4 months (95% CI, 9.1 to 12.0 months) versus 10.9 months (95% CI, 9.6 to 12.4 months) and 24-month OS rate was 22.1% versus 15.5% with avelumab versus chemotherapy, respectively (hazard ratio [HR], 0.91; 95% CI, 0.74 to 1.11; P = .1779). In the PD-L1-positive population (n = 54), the HR for OS was 1.13 (95% CI, 0.57 to 2.23; P = .6352). In an exploratory analysis of the PD-L1-positive population, defined as combined positive score ≥ 1 (22C3 assay; n = 137), median OS was 14.9 months (95% CI, 8.7 to 17.3 months) with avelumab versus 11.6 months (95% CI, 8.4 to 12.6 months) with chemotherapy (unstratified HR, 0.72; 95% CI, 0.49 to 1.05). With avelumab and chemotherapy, treatment-related adverse events (TRAEs) occurred in 149 (61.3%) and 184 (77.3%) patients, including grade ≥ 3 TRAEs in 31 (12.8%) and 78 (32.8%) patients, respectively. CONCLUSION: JAVELIN Gastric 100 did not demonstrate superior OS with avelumab maintenance versus continued chemotherapy in patients with advanced GC or GEJC overall or in a prespecified PD-L1-positive population.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Induction Chemotherapy/mortality , Maintenance Chemotherapy/mortality , Stomach Neoplasms/drug therapy , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Capecitabine/administration & dosage , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Humans , Male , Middle Aged , Oxaliplatin/administration & dosage , Prognosis , Stomach Neoplasms/pathology , Survival Rate
2.
Clin Cancer Res ; 25(1): 99-109, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30131389

ABSTRACT

PURPOSE: The NHS-IL12 immunocytokine is composed of two IL12 heterodimers fused to the NHS76 antibody. Preclinical studies have shown that this antibody targets IL12 to regions of tumor necrosis by binding histones on free DNA fragments in these areas, resulting in enhanced antitumor activity. The objectives of this phase I study were to determine the maximum tolerated dose (MTD) and pharmacokinetics of NHS-IL12 in subjects with advanced solid tumors. PATIENTS AND METHODS: Subjects (n = 59) were treated subcutaneously with NHS-IL12 in a single ascending-dose cohort followed by a multiple ascending-dose cohort (n = 37 with every 4-week dosing). RESULTS: The most frequently observed treatment-related adverse events (TRAE) included decreased circulating lymphocytes, increased liver transaminases, and flu-like symptoms. Of the grade ≥3 TRAEs, all were transient and only one was symptomatic (hyperhidrosis). The MTD is 16.8 µg/kg. A time-dependent rise in IFNγ and an associated rise in IL10 were observed following NHS-IL12. Of peripheral immune cell subsets evaluated, most noticeable were increases in frequencies of activated and mature natural killer (NK) cells and NKT cells. Based on T-cell receptor sequencing analysis, increases in T-cell receptor diversity and tumor-infiltrating lymphocyte density were observed after treatment where both biopsies and peripheral blood mononuclear cells were available. Although no objective tumor responses were observed, 5 subjects had durable stable disease (range, 6-30+ months). CONCLUSIONS: NHS-IL12 was well tolerated up to a dose of 16.8 µg/kg, which is the recommended phase II dose. Early clinical immune-related activity warrants further studies, including combination with immune checkpoint inhibitors.See related commentary by Lyerly et al., p. 9.


Subject(s)
Immunoglobulin G/administration & dosage , Interleukin-12/immunology , Neoplasms, Second Primary/drug therapy , Neoplasms/drug therapy , Recombinant Fusion Proteins/administration & dosage , Adult , Aged , Cell Line, Tumor , DNA Fragmentation/drug effects , Drug-Related Side Effects and Adverse Reactions/immunology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Immunoglobulin G/adverse effects , Influenza, Human/chemically induced , Influenza, Human/pathology , Interleukin-12/administration & dosage , Interleukin-12/adverse effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Male , Maximum Tolerated Dose , Middle Aged , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Neoplasms/immunology , Neoplasms/pathology , Neoplasms, Second Primary/immunology , Neoplasms, Second Primary/pathology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/adverse effects , Transaminases/metabolism
3.
Cancer Immunol Res ; 3(4): 326-32, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25527356

ABSTRACT

Programmed death 1 ligand 1 (PD-L1) is an immune regulatory molecule that limits antitumor immune activity. Targeting of PD-L1 and other immune checkpoint proteins has shown therapeutic activity in various tumor types. The expression of PD-L1 and its correlation with response to neoadjuvant chemotherapy in breast cancer has not been studied extensively. Our goal was to assess PD-L1 expression in a cohort of breast cancer patients treated with neoadjuvant chemotherapy. Pretreatment biopsies from 105 patients with breast cancer from Yale New Haven Hospital that subsequently received neoadjuvant chemotherapy were assessed for PD-L1 protein expression by automated quantitative analysis with a rabbit monoclonal antibody (E1L3N) to the cytoplasmic domain of PD-L1. In addition, tumor-infiltrating lymphocytes (TIL) were assessed on hematoxylin and eosin slides. PD-L1 expression was observed in 30% of patients, and it was positively associated with hormone-receptor-negative and triple-negative status and high levels of TILs. Both TILs and PD-L1 measured in the epithelium or stroma predicted pathologic complete response (pCR) to neoadjuvant chemotherapy in univariate and multivariate analyses. However, because they are strongly associated, TILs and PD-L1 cannot both be included in a significant multivariate model. PD-L1 expression is prevalent in breast cancer, particularly hormone-receptor-negative and triple-negative patients, indicating a subset of patients that may benefit from immune therapy. Furthermore, PD-L1 and TILs correlate with pCR, and high PD-L1 predicts pCR in multivariate analysis.


Subject(s)
B7-H1 Antigen/biosynthesis , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Aged , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Cohort Studies , Female , Humans , Middle Aged , Neoadjuvant Therapy , Neoplasm Proteins/biosynthesis , Prognosis , Treatment Outcome
4.
Clin Cancer Res ; 18(16): 4449-57, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22661537

ABSTRACT

PURPOSE: To deepen our understanding of mutant ROS1 expression, localization, and frequency in non-small cell lung cancer (NSCLC), we developed a highly specific and sensitive immunohistochemistry (IHC)-based assay that is useful for the detection of wild-type and mutant ROS1. EXPERIMENTAL DESIGN: We analyzed 556 tumors with the ROS1 D4D6 rabbit monoclonal antibody IHC assay to assess ROS1 expression levels and localization. A subset of tumors was analyzed by FISH to determine the percentage of these tumors harboring ROS1 translocations. Using specific and sensitive IHC assays, we analyzed the expression of anaplastic lymphoma kinase (ALK), EGFR L858R, and EGFR E746-A750del mutations in a subset of lung tumors, including those expressing ROS1. RESULTS: In our NSCLC cohort of Chinese patients, we identified 9 (1.6%) tumors expressing ROS1 and 22 (4.0%) tumors expressing ALK. FISH identified tumors with ALK or ROS1 rearrangements, and IHC alone was capable of detecting all cases with ALK and ROS1 rearrangements. ROS1 fusion partners were determined by reverse transcriptase PCR identifying CD74-ROS1, SLC34A2-ROS1, and FIG-ROS1 fusions. Some of the ALK and ROS1 rearranged tumors may also harbor coexisting EGFR mutations. CONCLUSIONS: NSCLC tumors with ROS1 rearrangements are uncommon in the Chinese population and represent a distinct entity of carcinomas. The ROS1 IHC assay described here is a valuable tool for identifying patients expressing mutant ROS1 and could be routinely applied in clinical practice to detect lung cancers that may be responsive to targeted therapies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carrier Proteins/metabolism , Lung Neoplasms/metabolism , Membrane Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing , Anaplastic Lymphoma Kinase , Animals , Base Sequence , Carcinoma, Non-Small-Cell Lung/genetics , Carrier Proteins/genetics , Cell Line , Cell Proliferation , Gene Expression , Genes, erbB-1 , Genotype , Golgi Matrix Proteins , Humans , Lung Neoplasms/genetics , Membrane Proteins/genetics , Membrane Transport Proteins , Mice , Mutation , Oncogene Proteins, Fusion/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Transplantation, Heterologous
5.
Neural Netw ; 26: 29-58, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22079270

ABSTRACT

How does working memory store multiple spatial positions to control sequences of eye movements, particularly when the same items repeat at multiple list positions, or ranks, during the sequence? An Item-Order-Rank model of working memory shows how rank-selective representations enable storage and recall of items that repeat at arbitrary list positions. Rank-related activity has been observed in many areas including the posterior parietal cortices (PPC), prefrontal cortices (PFC) and supplementary eye fields (SEF). The model shows how rank information, originating in PPC, may support rank-sensitive PFC working memory representations and how SEF may select saccades stored in working memory. It also proposes how SEF may interact with downstream regions such as the frontal eye fields (FEF) during memory-guided sequential saccade tasks, and how the basal ganglia (BG) may control the flow of information. Model simulations reproduce behavioral, anatomical and electrophysiological data under multiple experimental paradigms, including visually- and memory-guided single and sequential saccade tasks. Simulations reproduce behavioral data during two SEF microstimulation paradigms, showing that their seemingly inconsistent findings about saccade latency can be reconciled.


Subject(s)
Eye Movements/physiology , Memory, Short-Term/physiology , Models, Neurological , Neurons/physiology , Visual Cortex/physiology , Visual Fields/physiology , Attention/physiology , Brain Mapping , Computer Simulation , Humans , Movement , Photic Stimulation , Signal Detection, Psychological , Visual Pathways/physiology
6.
Inflamm Res ; 59(3): 207-18, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19763788

ABSTRACT

OBJECTIVE: Mast cell and basophil activation contributes to inflammation, bronchoconstriction, and airway hyperresponsiveness in asthma. Because IL-33 expression is inflammation inducible, we investigated IL-33-mediated effects in concert with both IgE-mediated and IgE-independent stimulation. METHODS: Because the HMC-1 mast cell line can be activated by GPCR and RTK signaling, we studied the effects of IL-33 on these pathways. The IL-33- and SCF-stimulated HMC-1 cells were co-cultured with human lung fibroblasts and airway smooth muscle cells in a collagen gel contraction assay. IL-33 effects on IgE-mediated activation were studied in primary mast cells and basophils. RESULT: IL-33 synergized with adenosine, C5a, SCF, and NGF receptor activation. IL-33-stimulated and SCF-stimulated HMC-1 cells demonstrated enhanced collagen gel contraction when cultured with fibroblasts or smooth muscle cells. IL-33 also synergized with IgE receptor activation of primary human mast cells and basophils. CONCLUSION: IL-33 amplifies inflammation in both IgE-independent and IgE-dependent responses.


Subject(s)
Basophils/drug effects , Basophils/metabolism , Immunoglobulin E/metabolism , Interleukins/pharmacology , Mast Cells/drug effects , Mast Cells/metabolism , Basophils/cytology , Cell Line , Chemokines/metabolism , Coculture Techniques , Collagen/metabolism , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Histamine/metabolism , Humans , Interleukin-33 , MAP Kinase Kinase 4/metabolism , Mast Cells/cytology , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Receptors, Purinergic P1/metabolism , Stem Cell Factor/pharmacology
7.
Behav Brain Res ; 177(1): 160-4, 2007 Feb 12.
Article in English | MEDLINE | ID: mdl-17145084

ABSTRACT

The activity of single units in the intermediate and deep layers of the superior colliculus was recorded while rats performed an operant conditioning task. On all trials, each animal pressed a bar and then inserted his snout into a food cup; on half of the trials, food reinforcement was available. To test for tactile sensitivity, on half of the trials the rats received a puff of air to the face when the snout entered the food cup. Activity of most cells was correlated with the motor activity of inserting the snout into the food cup, even when reinforcement was not available. For many cells, a larger burst of activity was seen on the reinforced trials than on trials when rats made the same movements without the presence of reward. There was no evidence that an increase in tactile sensitivity occurred when the animal retrieved the reinforcement. These results suggest that cells in the superior colliculus have an increase in activity associated with reward retrieval, which for some neurons is not dependent on simple sensory or motor factors.


Subject(s)
Action Potentials/physiology , Neurons/physiology , Reward , Superior Colliculi/cytology , Animals , Behavior, Animal , Conditioning, Operant/physiology , Male , Physical Stimulation/methods , Rats , Rats, Long-Evans
8.
Endocrinology ; 146(8): 3351-61, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15878963

ABSTRACT

A full-length transcript encoding a functional type II GnRH receptor was cloned from the pituitary of the sea lamprey, Petromyzon marinus. The current study is the first to identify a pituitary GnRH receptor transcript in an agnathan, which is the oldest vertebrate lineage. The cloned receptor retains the conserved structural features and amino acid motifs of other known GnRH receptors and notably includes a C-terminal intracellular tail of approximately 120 amino acids, the longest C-terminal tail of any vertebrate GnRH receptor identified to date. The lamprey GnRH receptor was shown to activate the inositol phosphate (IP) signaling system; stimulation with either lamprey GnRH-I or lamprey GnRH-III led to dose-dependent responses in transiently transfected COS7 cells. Furthermore, analyses of serially truncated lamprey GnRH receptor mutants indicate perturbations of the C-terminal tail disrupts IP accumulation, however, the tailless lamprey GnRH receptor was not only functional but was also capable of stimulating IP levels equal to wild type. Expression of the receptor transcript was demonstrated in the pituitary and testes using RT-PCR, whereas in situ hybridization showed expression and localization of the transcript in the proximal pars distalis of the pituitary. The phylogenetic placement and structural and functional features of this GnRH receptor suggest that it is representative of an ancestral GnRH receptor. In addition to having an important role in lamprey reproductive processes, the extensive C-terminal tail of this lamprey GnRH receptor may have great significance for understanding the evolutionary change of this vital structural feature within the GnRH receptor family.


Subject(s)
Lampreys/genetics , Receptors, LHRH/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Consensus Sequence , DNA Primers , Humans , Lampreys/classification , Molecular Sequence Data , Phylogeny , Receptors, LHRH/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Vertebrates
9.
Gen Comp Endocrinol ; 139(1): 85-94, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15474539

ABSTRACT

The lamprey, which are divided into three families, including the Petromyzonidae, Geotriidae, and Mordaciidae, have been shown to regulate the reproductive axis through a functional hypothalamic-pituitary-gonadal axis. To date, two forms of gonadotropin-releasing hormone (GnRH) have been identified in the sea lamprey (Petromyzon marinus), lamprey GnRH-I (decapeptide and cDNA) and lamprey GnRH-III (decapeptide), both of which have been shown to be expressed in the preoptic-anterior hypothalamic region and both forms have been demonstrated to regulate reproductive function (i.e. steroidogenesis and gametogenesis). The objective of this study was to isolate the cDNA encoding the prepro-lamprey GnRH-III from eight species of lamprey using a PCR based subcloning procedure. A degenerate primer designed to the lamprey GnRH-III decapeptide was used to amplify the 3' end of each transcript, while gene specific primers were used to amplify the 5' ends. Phylogenetic analysis using the prepro-lamprey GnRH-III amino acid sequences was performed, in which the lamprey GnRH-III sequences divided into three groups, supporting the current view of the lamprey lineage at the family level. Finally, a phylogenetic analysis of these newly identified deduced amino acid sequences together with 64 previously described GnRH sequences suggests that the lamprey GnRHs are unique, as they group together separately from the three previously described paralogous lineages of the GnRH family.


Subject(s)
Cloning, Molecular , DNA, Complementary , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Lampreys/genetics , Lampreys/metabolism , Oligopeptides/genetics , Oligopeptides/metabolism , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/metabolism , Amino Acid Sequence , Animals , Phylogeny , Protein Precursors/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...