Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Surg Res ; 298: 240-250, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38631173

ABSTRACT

INTRODUCTION: Neuroblastoma (NB) is the most common extra-cranial malignancy in children. Poor survival in high-risk NB is attributed to recurrent metastatic disease. To better study metastatic disease, we used a novel mouse model to investigate differential gene expression between primary tumor cells and metastatic cells. We hypothesized that metastatic NB cells have a different gene expression profile from primary tumor cells and cultured cells. METHODS: Using three human NB cell lines (NGP, CHLA255, and SH-SY5Y), orthotopic xenografts were established in immunodeficient nod/scid gamma mice via subcapsular renal injection. Mice were sacrificed and NB cells were isolated from the primary tumor and from sites of metastasis (bone marrow, liver). RNA sequencing, gene set analysis, and pathway analysis were performed to identify differentially expressed genes and molecular pathways in the metastatic cells compared to primary tumor cells. RESULTS: There were 266 differentially expressed genes in metastatic tumor cells (bone marrow and liver combined) compared to primary tumor cells. The top upregulated gene was KCNK1 and the top downregulated genes were PDE7B and NEBL. Top upregulated pathways in the metastatic cells were involved in ion transport, cell signaling, and cell proliferation. Top downregulated pathways were involved in DNA synthesis, transcription, and cellular metabolism. CONCLUSIONS: In metastatic NB cells, our study identified the upregulation of biologic processes involved in cell cycle regulation, cell proliferation, migration, and invasion. Ongoing studies aim to validate downstream translation of these genomic alterations, as well as target these pathways to more effectively suppress and inhibit recurrent metastatic disease in NB.


Subject(s)
Gene Expression Regulation, Neoplastic , Mice, Inbred NOD , Mice, SCID , Neuroblastoma , Animals , Neuroblastoma/pathology , Neuroblastoma/genetics , Neuroblastoma/metabolism , Humans , Mice , Cell Line, Tumor , Liver Neoplasms/secondary , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Bone Marrow Neoplasms/secondary , Bone Marrow Neoplasms/genetics , Gene Expression Profiling , Transcriptome
2.
Sci Rep ; 13(1): 19915, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37964011

ABSTRACT

C-C motif chemokine ligand 2 (CCL2) is a monocyte chemoattractant that promotes metastatic disease and portends a poor prognosis in many cancers. To determine the potential of anti-CCL2 inhibition as a therapy for recurrent metastatic disease in neuroblastoma, a mouse model of minimal residual disease was utilized in which residual disease was treated with anti-CCL2 monoclonal antibody with etoposide. The effect of anti-CCL2 antibody on neuroblastoma cells was determined in vitro with cell proliferation, transwell migration, and 2-dimensional chemotaxis migration assays. The in vivo efficacy of anti-CCL2 antibody and etoposide against neuroblastoma was assessed following resection of primary tumors formed by two cell lines or a patient-derived xenograft (PDX) in immunodeficient NOD-scid gamma mice. In vitro, anti-CCL2 antibody did not affect cell proliferation but significantly inhibited neuroblastoma cell and monocyte migration towards an increasing CCL2 concentration gradient. Treatment of mice with anti-CCL2 antibody combined with etoposide significantly increased survival of mice after resection of primary tumors, compared to untreated mice.


Subject(s)
Neuroblastoma , Humans , Animals , Mice , Etoposide/pharmacology , Etoposide/therapeutic use , Ligands , Neoplasm, Residual/drug therapy , Mice, Inbred NOD , Neuroblastoma/pathology , Chemokines , Chemokine CCL2 , Cell Line, Tumor
3.
J Immunother Cancer ; 10(1)2022 01.
Article in English | MEDLINE | ID: mdl-35091455

ABSTRACT

BACKGROUND: Mesothelin (MSLN) is a classic tumor-associated antigen that is expressed in lung cancer and many other solid tumors. However, MSLN is also expressed in normal mesothelium which creates a significant risk of serious inflammation for MSLN-directed therapeutics. We have developed a dual-receptor (Tmod™) system that exploits the difference between tumor and normal tissue in a subset of patients with defined heterozygous gene loss (LOH) in their tumors. METHODS: T cells engineered with the MSLN CAR Tmod construct described here contain (1) a novel MSLN-activated CAR and (2) an HLA-A*02-gated inhibitory receptor (blocker). A*02 binding is intended to override T-cell cytotoxicity, even in the presence of MSLN. The Tmod system is designed to treat heterozygous HLA class I patients, selected for HLA LOH. When A*02 is absent from tumors selected for LOH, the MSLN Tmod cells are predicted to mediate potent killing of the MSLN(+)A*02(-) malignant cells. RESULTS: The sensitivity of the MSLN Tmod cells is comparable with a benchmark MSLN CAR-T that was active but toxic in the clinic. Unlike MSLN CAR-T cells, the Tmod system robustly protects surrogate "normal" cells even in mixed-cell populations in vitro and in a xenograft model. The MSLN CAR can also be paired with other HLA class I blockers, supporting extension of the approach to patients beyond A*02 heterozygotes. CONCLUSIONS: The Tmod mechanism exemplified by the MSLN CAR Tmod construct provides an alternative route to leverage solid-tumor antigens such as MSLN in safer, more effective ways than previously possible.


Subject(s)
HLA-A2 Antigen/genetics , Immunotherapy, Adoptive/methods , Mesothelin/immunology , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , Animals , Cell Line, Tumor , Female , HLA-A2 Antigen/immunology , Humans , Loss of Heterozygosity , Mice , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
4.
Cancer Res Commun ; 2(1): 58-65, 2022 01.
Article in English | MEDLINE | ID: mdl-36860694

ABSTRACT

Neoantigens are among the most intriguing potential immuno-oncology targets because, unlike many cancer targets that are expressed on normal tissues, they are by definition restricted to cancer cells. Medicines directed at common neoantigens such as mutant KRAS are especially interesting because they may offer the convenience and cost of an off-the-shelf therapy. However, all common KRAS mutations produce proteins that differ from the wild type at a single amino acid, creating challenges for molecular discrimination. We have undertaken an effort to optimize single-chain variable fragments (scFv) against peptide/major histocompatibility antigen complexes composed of HLA-A*11 and either G12V- or G12D-mutant KRAS peptides. These scFvs could in principle be used in chimeric antigen receptor (CAR) T-cell therapies for selected patients whose tumors bear either of these mutations. Here we show that optimization of such CARs involves a trade-off between potency and selectivity. We further show that targeting this family without high selectivity engenders risks of cross-reactivity against other members of the G-protein family to which KRAS belongs. Significance: We report an effort to generate high potency, selective CARs directed at mutant KRAS peptides. Although the heavily optimized CARs maintain high selectivity against wild-type KRAS, they lose selectivity against other KRAS-related peptides derived from human proteins. To our knowledge, this work is the first to examine the trade-off between potency and selectivity with regard to KRAS pMHC-directed CARs, illustrating the challenge to achieve both sufficient potency and high selectivity.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Single-Chain Antibodies , Humans , Receptors, Chimeric Antigen/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Immunotherapy, Adoptive , Single-Chain Antibodies/genetics
5.
Mol Immunol ; 128: 298-310, 2020 12.
Article in English | MEDLINE | ID: mdl-33012527

ABSTRACT

We describe an approach to cancer therapy based on exploitation of common losses of genetic material in tumor cells (loss of heterozygosity) (Basilion et al., 1999; Beroukhim et al., 2010). This therapeutic concept addresses the fundamental problem of discrimination between tumor and normal cells and can be applied in principle to the large majority of tumors. It utilizes modular activator/blocker elements that integrate signals related to the presence and absence of ligands displayed on the cell surface (Fedorov et al., 2013). We show that the targeting system works robustly in vitro and in a mouse cancer model where absence of the HLA-A*02 allele releases a brake on engineered T cells activated by the CD19 surface antigen. This therapeutic approach potentially opens a route toward a large, new source of cancer targets.


Subject(s)
Loss of Heterozygosity/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Alleles , Animals , Antigens, CD19/immunology , Cell Line, Tumor , Female , HLA-A Antigens/immunology , Humans , Jurkat Cells , Ligands , Mice , Mice, Inbred NOD
6.
Mol Immunol ; 126: 56-64, 2020 10.
Article in English | MEDLINE | ID: mdl-32768859

ABSTRACT

Chimeric antigen receptors (CARs) and their parent signaling molecule, the T cell receptor (TCR), are fascinating proteins of increasing relevance to disease therapy. Here we use a collection of 1221 pMHC-directed CAR constructs representing 10 pMHC targets to study aspects of CAR structure-activity relationships (SAR), with particular focus on the extracellular and transmembrane structural components. These experiments that involve pMHC targets whose number/cell can be manipulated by peptide dosing in vitro enable systematic analysis of the SAR of CARs in carefully controlled experimental situations (Harris and Kranz, 2016). We find that CARs tolerate a wide range of structural variation, with the ligand-binding domains (LBDs) dominating the SAR of CAR antigen sensitivity. Notwithstanding the critical role of the LBD, CAR antigen-binding on the cell surface, measured by pMHC tetramer staining, is not an effective predictor of functional sensitivity. These results have important implications for the design and testing of CARs aimed toward the clinic.


Subject(s)
HLA-A Antigens/immunology , Receptors, Chimeric Antigen/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Binding Sites/immunology , HLA-A Antigens/metabolism , Humans , Jurkat Cells , Ligands , MCF-7 Cells , Protein Domains/immunology , Protein Multimerization/immunology , Receptors, Chimeric Antigen/immunology , Structure-Activity Relationship , T-Lymphocytes/metabolism
7.
Clin Cancer Res ; 25(1): 325-333, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30232225

ABSTRACT

PURPOSE: Immunotherapy of neuroblastoma that remains after myeloablative chemotherapy with anti-GD2 antibody dinutuximab has increased the two-year event-free and overall survival of high-risk neuroblastoma patients; however, 40% of patients develop recurrent disease during or after this treatment. To determine the potential of such antibody-based immunotherapy earlier in treatment, a mouse model was developed in which surgical resection of the primary tumor was followed by therapy of residual disease with dinutuximab combined with ex vivo-activated human natural killer (aNK) cells. EXPERIMENTAL DESIGN: The effect of combining dinutuximab with human aNK cells was determined in vitro with cellular cytotoxicity and Matrigel invasion assays. The in vivo efficacy of dinutuximab and aNK cells against neuroblastoma was assessed following resection of primary tumors formed by two cell lines or a patient-derived xenograft (PDX) in immunodeficient NOD-scid gamma mice. RESULTS: In vitro, the combination of aNK cells and dinutuximab caused cytotoxicity and decreased invasiveness of three human neuroblastoma cell lines. Treatment of mice with dinutuximab combined with aNK cells after surgical resection of primary intrarenal tumors formed by two cell lines or a PDX decreased tumor cells in liver and bone marrow as evaluated by histopathology and bioluminescence imaging. Survival of mice after resection of these tumors was most significantly increased by treatment with dinutuximab combined with aNK cells compared with that of untreated mice. CONCLUSIONS: The combination of dinutuximab and adoptively transferred human aNK cells following surgical resection of primary neuroblastomas significantly improves survival of immunodeficient mice.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Antibodies, Monoclonal/pharmacology , N-Acetylgalactosaminyltransferases/genetics , Neuroblastoma/therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Combined Modality Therapy , Cytotoxicity, Immunologic/drug effects , Disease Models, Animal , Heterografts , Humans , Immunotherapy , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Mice , N-Acetylgalactosaminyltransferases/antagonists & inhibitors , Neuroblastoma/immunology , Neuroblastoma/surgery
8.
Front Pediatr ; 6: 143, 2018.
Article in English | MEDLINE | ID: mdl-29868530

ABSTRACT

Perianal abscess and fistula-in-ano are well-described in the pediatric population. They are most common in infants less than 1 year of age and often resolve with oral antibiotics; occasionally they require drainage or fistulotomy. The etiology is commonly associated with cryptoglandular obstruction and subsequent infection, however alternative diagnoses should be considered in cases of recurrent abscesses and fistulae that are refractory to standard treatments. In this report, we present the case of an 8-year-old boy with a complex, recurrent fistula-in-ano that resulted from a rare congenital perirectal dermoid cyst.

9.
J Surg Res ; 229: 351-356, 2018 09.
Article in English | MEDLINE | ID: mdl-29937013

ABSTRACT

BACKGROUND: With the advent of minimally invasive techniques, laparoscopic Ladd's procedure is increasingly used to treat children with malrotation, yet evidence regarding its safety and efficacy is lacking. We hypothesize that operative and postoperative outcomes with the open technique are superior to the laparoscopic Ladd's procedure. METHODS: We conducted a 5-y retrospective chart review of all patients who underwent Ladd's procedure at our institution from 2010-2015. Exclusion of patients included those with concomitant conditions, such as poor gut perfusion, significant reflux, tracheoesophageal fistula, failure to thrive requiring concomitant gastrostomy, and biliary atresia. Kruskal-Wallis and Mann-Whitney tests were used where appropriate. RESULTS: Between 2010 and 2015, of 130 patients who underwent Ladd's procedure, 77 met inclusion criteria. Sixty-two patients underwent initial open surgery, 15 patients underwent laparoscopy, seven of which were converted to open. Patients undergoing open surgery were younger compared to the laparoscopic groups. Thirty-three of the 77 malrotation patients (43%) presented with volvulus, 27 underwent open surgery, four had laparoscopic converted to open procedures, and two patients underwent laparoscopic Ladd's without incident. Laparoscopy resulted in increased operative time and clinic visits. Patients undergoing laparoscopic to open surgery had longer operative times, time to resume diet, and length of hospital stay. No difference was noted in complications among the groups. CONCLUSIONS: Although minimally invasive approaches are becoming increasingly used, no evidence supports laparoscopic superiority over open Ladd's procedure. We found that open surgery was associated with shorter operating times and fewer clinic visits. Furthermore, laparotomy remains the favored procedure for patients presenting with volvulus.


Subject(s)
Conversion to Open Surgery/statistics & numerical data , Intestinal Obstruction/surgery , Intestinal Volvulus/surgery , Laparoscopy/adverse effects , Postoperative Complications/epidemiology , Adolescent , Child , Child, Preschool , Humans , Incidence , Infant , Infant, Newborn , Intestinal Obstruction/etiology , Intestinal Volvulus/complications , Intestines/abnormalities , Intestines/surgery , Laparoscopy/methods , Length of Stay/statistics & numerical data , Operative Time , Postoperative Complications/etiology , Retrospective Studies , Time Factors , Treatment Outcome
10.
Proc Natl Acad Sci U S A ; 111(25): 9115-20, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24927526

ABSTRACT

The role of the nuclear receptor TLX in hippocampal neurogenesis and cognition has just begun to be explored. In this study, we generated a transgenic mouse model that expresses TLX under the control of the promoter of nestin, a neural precursor marker. Transgenic TLX expression led to mice with enlarged brains with an elongated hippocampal dentate gyrus and increased numbers of newborn neurons. Specific expression of TLX in adult hippocampal dentate gyrus via lentiviral transduction increased the numbers of BrdU(+) cells and BrdU(+)NeuN(+) neurons. Furthermore, the neural precursor-specific expression of the TLX transgene substantially rescued the neurogenic defects of TLX-null mice. Consistent with increased neurogenesis in the hippocampus, the TLX transgenic mice exhibited enhanced cognition with increased learning and memory. These results suggest a strong association between hippocampal neurogenesis and cognition, as well as significant contributions of TLX to hippocampal neurogenesis, learning, and memory.


Subject(s)
Dentate Gyrus/metabolism , Gene Expression , Memory , Neurogenesis , Neurons/metabolism , Receptors, Cytoplasmic and Nuclear/biosynthesis , Animals , Cognition , Dentate Gyrus/cytology , Mice , Nestin/genetics , Nestin/metabolism , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/genetics
11.
Mol Cell Biol ; 33(13): 2551-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23629626

ABSTRACT

Although Wnt7a has been implicated in axon guidance and synapse formation, investigations of its role in the early steps of neurogenesis have just begun. We show here that Wnt7a is essential for neural stem cell self-renewal and neural progenitor cell cycle progression in adult mouse brains. Loss of Wnt7a expression dramatically reduced the neural stem cell population and increased the rate of cell cycle exit in neural progenitors in the hippocampal dentate gyrus of adult mice. Furthermore, Wnt7a is important for neuronal differentiation and maturation. Loss of Wnt7a expression led to a substantial decrease in the number of newborn neurons in the hippocampal dentate gyrus. Wnt7a(-/-) dentate granule neurons exhibited dramatically impaired dendritic development. Moreover, Wnt7a activated ß-catenin and its downstream target genes to regulate neural stem cell proliferation and differentiation. Wnt7a stimulated neural stem cell proliferation by activating the ß-catenin-cyclin D1 pathway and promoted neuronal differentiation and maturation by inducing the ß-catenin-neurogenin 2 pathway. Thus, Wnt7a exercised critical control over multiple steps of neurogenesis by regulating genes involved in both cell cycle control and neuronal differentiation.


Subject(s)
Neural Stem Cells/physiology , Neurogenesis/physiology , Wnt Proteins/metabolism , Age Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle/physiology , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Cyclin D1/genetics , Cyclin D1/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/physiology , Female , Gene Expression Regulation , Male , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Prosencephalon/cytology , Prosencephalon/growth & development , Wnt Proteins/genetics , beta Catenin/metabolism
13.
J Mol Cell Biol ; 4(4): 190-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22371436

ABSTRACT

The field of regenerative medicine is rapidly gaining momentum as an increasing number of reports emerge concerning the induced conversions observed in cellular fate reprogramming. While in recent years, much attention has been focused on the conversion of fate-committed somatic cells to an embryonic-like or pluripotent state, there are still many limitations associated with the applications of induced pluripotent stem cell reprogramming, including relatively low reprogramming efficiency, the times required for the reprogramming event to take place, the epigenetic instability, and the tumorigenicity associated with the pluripotent state. On the other hand, lineage reprogramming involves the conversion from one mature cell type to another without undergoing conversion to an unstable intermediate. It provides an alternative approach in regenerative medicine that has a relatively lower risk of tumorigenesis and increased efficiency within specific cellular contexts. While lineage reprogramming provides exciting potential, there is still much to be assessed before this technology is ready to be applied in a clinical setting.


Subject(s)
Alchemy , Cell Lineage , Cellular Reprogramming , Regenerative Medicine/methods , Animals , Cell Differentiation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...