Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Cancers (Basel) ; 13(15)2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34359704

ABSTRACT

Tumor-associated macrophages (TAMs) in breast cancer regulate inflammation, immunosuppression, angiogenesis, and metastasis. However, TAM imaging remains a clinical challenge. Ferumoxytol has long been an FDA-approved superparamagnetic iron oxide nanoparticle (SPION) preparation used as an intravenous (IV) treatment for iron-deficiency anemia. Given its high transverse relaxivity, ferumoxytol produces a negative image contrast upon cellular uptake in T2-weighted magnetic resonance imaging (MRI) studies. Here we evaluated ferumoxytol as a contrast agent to image/quantify TAMs in an aggressive mouse model of breast cancer: We developed [Fe]MRI to measure the 5-dimensional function c(x,y,z,t), where c is the concentration of nanoparticle iron and {x,y,z,t} is the 4-dimensional set of tumor space-time coordinates. Ferumoxytol SPIONs are readily phagocytosed (~104/cell) by the F4/80+CD11b+ TAMs within breast tumors. Quantitative [Fe]MRIs served to determine both the spatial and the temporal distribution of the SPION iron, and hence to measure [Fe] = c(x,y,z,t), a surrogate for TAM density. In single-dose pharmacokinetic studies, after an IV dose of 5 mg/Kg iron, [Fe]MRI measurements showed that c(x,y,z,t) within breast tumors peaked around [Fe] = 70 µM at 42 h post-administration, and decayed below the [Fe]MRI detection limit (~2 µM) by day 7. There was no SPION uptake in control organs (muscle and adipose tissue). Optical microscopy of tissue sections confirmed that F4/80+CD11b+ TAMs infiltrated the tumors and accumulated SPION iron. Our methodology and findings have translational applications for breast cancer patients.

2.
J Immunother Cancer ; 8(1)2020 04.
Article in English | MEDLINE | ID: mdl-32312906

ABSTRACT

BACKGROUND: Programmed death 1/programmed death ligand 1 (PD-1/PD-L1) targeted immunotherapy affords clinical benefit in ~20% of unselected patients with lung cancer. The factor(s) that determine whether a tumor responds or fails to respond to immunotherapy remains an active area of investigation. We have previously defined divergent responsiveness of two KRAS-mutant cell lines to PD-1/PD-L1 blockade using an orthotopic, immunocompetent mouse model. Responsiveness to PD-1/PD-L1 checkpoint blockade correlates with an interferon gamma (IFNγ)-inducible gene signature and major histocompatibility complex class II (MHC II) expression by cancer cells. In the current study, we aim to identify therapeutic targets that can be manipulated in order to enhance cancer-cell-specific MHC II expression. METHODS: Responsiveness to IFNγ and induction of MHC II expression was assessed after various treatment conditions in mouse and human non-small cell lung cancer (NSCLC) cell lines using mass cytometric and flow cytometric analysis. RESULTS: Single-cell analysis using mass and flow cytometry demonstrated that IFNγ consistently induced PD-L1 and MHC class I (MHC I) across multiple murine and human NSCLC cell lines. In contrast, MHC II showed highly variable induction following IFNγ treatment both between lines and within lines. In mouse models of NSCLC, MHC II induction was inversely correlated with basal levels of phosphorylated extracellular signal-regulated kinase (ERK) 1/2, suggesting potential mitogen-activated protein (MAP) kinase-dependent antagonism of MHC II expression. To test this, cell lines were subjected to varying levels of stimulation with IFNγ, and assessed for MHC II expression in the presence or absence of mitogen-activated protein kinase kinase (MEK) inhibitors. IFNγ treatment in the presence of MEK inhibitors significantly enhanced MHC II induction across multiple lung cancer lines, with minimal impact on expression of either PD-L1 or MHC I. Inhibition of histone deacetylases (HDACs) also enhanced MHC II expression to a more modest extent. Combined MEK and HDAC inhibition led to greater MHC II expression than either treatment alone. CONCLUSIONS: These studies emphasize the active inhibitory role that epigenetic and ERK signaling cascades have in restricting cancer cell-intrinsic MHC II expression in NSCLC, and suggest that combinatorial blockade of these pathways may engender new responsiveness to checkpoint therapies.


Subject(s)
B7-H1 Antigen/metabolism , Epigenesis, Genetic , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class II/metabolism , Lung Neoplasms/pathology , Mitogen-Activated Protein Kinases/metabolism , Animals , Antiviral Agents/pharmacology , B7-H1 Antigen/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Histocompatibility Antigens Class II/genetics , Humans , Interferon-gamma/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Mice , Mitogen-Activated Protein Kinases/genetics , Tumor Cells, Cultured
3.
J Immunol ; 204(8): 2295-2307, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32179637

ABSTRACT

MHC class II (MHCII) expression is usually restricted to APC but can be expressed by cancer cells. We examined the effect of cancer cell-specific MHCII (csMHCII) expression in lung adenocarcinoma on T cell recruitment to tumors and response to anti-PD-1 therapy using two orthotopic immunocompetent murine models of non-small cell lung cancer: CMT167 (CMT) and Lewis lung carcinoma (LLC). We previously showed that CMT167 tumors are eradicated by anti-PD1 therapy, whereas LLC tumors are resistant. RNA sequencing analysis of cancer cells recovered from tumors revealed that csMHCII correlated with response to anti-PD1 therapy, with immunotherapy-sensitive CMT167 cells being csMHCII positive, whereas resistant LLC cells were csMHCII negative. To test the functional effects of csMHCII, MHCII expression was altered on the cancer cells through loss- and gain-of-function of CIITA, a master regulator of the MHCII pathway. Loss of CIITA in CMT167 decreased csMHCII and converted tumors from anti-PD-1 sensitive to anti-PD-1 resistant. This was associated with lower levels of Th1 cytokines, decreased T cell infiltration, increased B cell numbers, and decreased macrophage recruitment. Conversely, overexpression of CIITA in LLC cells resulted in csMHCII in vitro and in vivo. Enforced expression of CIITA increased T cell infiltration and sensitized tumors to anti-PD-1 therapy. csMHCII expression was also examined in a subset of surgically resected human lung adenocarcinomas by multispectral imaging, which provided a survival benefit and positively correlated with T cell infiltration. These studies demonstrate a functional role for csMHCII in regulating T cell infiltration and sensitivity to anti-PD-1.


Subject(s)
Adenocarcinoma of Lung/therapy , Histocompatibility Antigens Class II/genetics , Lung Neoplasms/therapy , Nuclear Proteins/genetics , Trans-Activators/genetics , Tumor Microenvironment/genetics , Adenocarcinoma of Lung/immunology , Animals , Disease Models, Animal , Histocompatibility Antigens Class II/immunology , Lung Neoplasms/immunology , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/immunology , Programmed Cell Death 1 Receptor/immunology , Trans-Activators/immunology , Tumor Microenvironment/immunology
4.
Life Sci Alliance ; 2(3)2019 06.
Article in English | MEDLINE | ID: mdl-31133614

ABSTRACT

Targeting PD-1/PD-L1 is only effective in ∼20% of lung cancer patients, but determinants of this response are poorly defined. We previously observed differential responses of two murine K-Ras-mutant lung cancer cell lines to anti-PD-1 therapy: CMT167 tumors were eliminated, whereas Lewis Lung Carcinoma (LLC) tumors were resistant. The goal of this study was to define mechanism(s) mediating this difference. RNA sequencing analysis of cancer cells recovered from lung tumors revealed that CMT167 cells induced an IFNγ signature that was blunted in LLC cells. Silencing Ifngr1 in CMT167 resulted in tumors resistant to IFNγ and anti-PD-1 therapy. Conversely, LLC cells had high basal expression of SOCS1, an inhibitor of IFNγ. Silencing Socs1 increased response to IFNγ in vitro and sensitized tumors to anti-PD-1. This was associated with a reshaped tumor microenvironment, characterized by enhanced T cell infiltration and enrichment of PD-L1hi myeloid cells. These studies demonstrate that targeted enhancement of tumor-intrinsic IFNγ signaling can induce a cascade of changes associated with increased therapeutic vulnerability.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Interferon-gamma/pharmacology , Lung Neoplasms/pathology , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Chemokine CXCL9/metabolism , Disease Models, Animal , Gene Silencing , Humans , Immunohistochemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism
5.
Cancer Res ; 78(1): 143-156, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29118090

ABSTRACT

The complement cascade is a part of the innate immune system that acts primarily to remove pathogens and injured cells. However, complement activation is also peculiarly associated with tumor progression. Here we report mechanistic insights into this association in multiple immunocompetent orthotopic models of lung cancer. After tumor engraftment, we observed systemic activation of the complement cascade as reflected by elevated levels of the key regulator C3a. Notably, growth of primary tumors and metastases was both strongly inhibited in C3-deficient mice (C3-/- mice), with tumors undetectable in many subjects. Growth inhibition was associated with increased numbers of IFNγ+/TNFα+/IL10+ CD4+ and CD8+ T cells. Immunodepletion of CD4+ but not CD8+ T cells in tumor-bearing subjects reversed the inhibitory effects of C3 deletion. Similarly, antagonists of the C3a or C5a receptors inhibited tumor growth. Investigations using multiple tumor cell lines in the orthotopic model suggested the involvement of a C3/C3 receptor autocrine signaling loop in regulating tumor growth. Overall, our findings offer functional evidence that complement activation serves as a critical immunomodulator in lung cancer progression, acting to drive immune escape via a C3/C5-dependent pathway.Significance: This provocative study suggests that inhibiting complement activation may heighten immunotherapeutic responses in lung cancer, offering findings with immediate implications, given the existing clinical availability of complement antagonists. Cancer Res; 78(1); 143-56. ©2017 AACR.


Subject(s)
Adenocarcinoma/immunology , CD4-Positive T-Lymphocytes/immunology , Complement Activation , Lung Neoplasms/pathology , Receptors, Complement/immunology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , CD4-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Complement C3/genetics , Complement C3d/metabolism , Female , Humans , Immunoglobulin M/metabolism , Lung Neoplasms/immunology , Male , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins, Fusion/genetics , Receptors, Complement/metabolism , Xenograft Model Antitumor Assays
6.
CNS Oncol ; 3(4): 267-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25286038

ABSTRACT

AIMS: Prophylaxis against Pneumocystis jiroveci pneumonia (PJP) is currently recommended for patients receiving chemoradiation with temozolomide for newly diagnosed glioblastoma multiforme. At our institution, PJP prophylaxis during temozolomide treatment has not been routinely given because of the paucity of supporting data. We investigated the rate of PJP infections in our patients. PATIENTS & METHODS: We conducted a retrospective chart review of 240 brain tumor patients treated between 1999 and 2012 with temozolomide and no PJP prophylaxis, 127 of which received concurrent chemoradiation. RESULTS: One in 240 patients (0.4%; 95% CI: 0.01-2.00; median total dose: 7375 mg/m(2); interquartile range: 1300) were diagnosed with PJP. CONCLUSION: There was a <1% rate of PJP for brain tumor patients treated with temozolomide until progression without PJP prophylaxis.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Dacarbazine/analogs & derivatives , Pneumonia, Pneumocystis/chemically induced , Pneumonia, Pneumocystis/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/drug therapy , Child , Dacarbazine/adverse effects , Female , Glioblastoma/drug therapy , Humans , Longitudinal Studies , Male , Middle Aged , Retrospective Studies , T-Lymphocytes/pathology , Temozolomide , Young Adult
7.
Pediatr Blood Cancer ; 61(1): 120-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23956023

ABSTRACT

BACKGROUND: Atypical teratoid rhabdoid tumors (AT-RT) are pediatric tumors of the central nervous system with limited treatment options and poor survival rate. We investigated whether enhancing chemotherapy toxicity by depleting intracellular glutathione (GSH; a key molecule in cisplatin resistance) with high dose acetaminophen (AAP), may improve therapeutic efficacy in AT-RT in vitro. PROCEDURE: BT16 (cisplatin-resistant) and BT12 (cisplatin-sensitive) AT-RT cell lines were treated with combinations of AAP, cisplatin, and the anti-oxidant N-acetylcysteine (NAC). Cell viability, GSH and peroxide concentrations, mitochondrial damage, and apoptosis were evaluated in vitro. RESULTS: AAP enhanced cisplatin cytotoxicity in cisplatin-resistant BT16 cells but not cisplatin-sensitive BT12 cells. Baseline GSH levels were elevated in BT16 cells compared to BT12 cells, and AAP decreased GSH to a greater magnitude in BT16 cells than BT12 cells. Unlike BT12 cells, BT16 cells did not have elevated peroxide levels upon treatment with cisplatin alone, but did have elevated levels when treated with AAP + cisplatin. Both cell lines had markedly increased mitochondrial injury when treated with AAP + cisplatin relative to either drug treatment alone. The enhanced toxic effects were partially reversed with concurrent administration of NAC. CONCLUSIONS: Our results suggest that AAP could be used as a chemo-enhancement agent to potentiate cisplatin chemotherapeutic efficacy particularly in cisplatin-resistant AT-RT tumors with high GSH levels in clinical settings.


Subject(s)
Acetaminophen/administration & dosage , Acetylcysteine/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cisplatin/administration & dosage , Rhabdoid Tumor , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Drug Evaluation, Preclinical , Glutathione/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Rhabdoid Tumor/metabolism , Rhabdoid Tumor/pathology
8.
Neurol Clin Pract ; 3(6): 469-474, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24353921

ABSTRACT

Diagnosing dystonia can be challenging and depends on the recognition of subtle clinical signs. Due to clinical heterogeneity, variable age at presentation, and overlapping features with other disorders, dystonia is under-recognized. The presence of dystonic tremor is often a reason for misdiagnosis. We report an illustrative case of a patient with DYT1 dystonia who was originally misdiagnosed with Parkinson disease. Careful physical examination and history-taking can reveal dystonia and prompt appropriate diagnostic studies, which, in turn, can lead to potentially life-changing treatment. Our report illustrates typical challenges in the recognition and diagnosis of dystonia, and serves to increase clinicians' awareness of this disabling, but treatable, condition.

9.
Anticancer Res ; 33(6): 2391-400, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23749887

ABSTRACT

BACKGROUND: Ovarian cancer is commonly treated with cisplatin/paclitaxel but many tumors become resistant. Acetaminophen reduced glutathione and enhanced chemotherapy efficacy in hepatic cancer treatment. The objective of this study was to examine if acetaminophen enhances the cytotoxicity of cisplatin/paclitaxel in ovarian cancer. MATERIALS AND METHODS: SKOV3 human ovarian carcinoma cells in vitro and a subcutaneous tumor nude rat model were used and treated with cisplatin/paclitaxel with or without acetaminophen. RESULTS: In vitro, acetaminophen enhanced apoptosis induced by cisplatin and paclitaxel with similar effects on glutathione, reactive oxygen species and mitochondrial membrane potential, but different effects on nuclear factor erythroid 2-related factor 2 (NRF2) translocation. In vivo, acetaminophen was uniformly distributed in tissues and significantly reduced hepatic glutathione. Acetaminophen enhanced the cisplatin chemotherapeutic effect by reducing tumor recurrence. CONCLUSION: Our results suggest that acetaminophen as a chemoenhancing adjuvant could improve the efficacy of cisplatin and paclitaxel in treating patients with ovarian carcinoma and other tumor types.


Subject(s)
Acetaminophen/pharmacology , Cisplatin/pharmacology , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , Acetaminophen/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/therapeutic use , Female , Glutathione/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , NF-E2-Related Factor 2/metabolism , Neoplasm Transplantation , Paclitaxel/therapeutic use , Rats , Rats, Nude , Reactive Oxygen Species , Xenograft Model Antitumor Assays
10.
Sensors (Basel) ; 10(1): 280-91, 2010.
Article in English | MEDLINE | ID: mdl-22315540

ABSTRACT

The direct and accurate estimation of nitric dioxide levels is an extremely laborious and technically demanding procedure in the molecular diagnostics of inflammatory processes. The aim of this work is to demonstrate that a stop-flow technique utilizing a specific spectroscopic biosensor can be used for detection of nanomolar quantities of NO(2) in biological milieu. The use of novel compound cis-[Cr(C(2)O(4))(AaraNH(2))(OH(2))(2)](+) increases NO(2) estimation accuracy by slowing down the rate of NO(2) uptake. In this study, an animal model of pancreatitis, where nitrosative stress is induced by either 3g/kg bw or 1.5 g/kg bw dose of L-arginine, was used. Biochemical parameters and morphological characteristics of acute pancreatitis were monitored, specifically assessing pancreatic acinar cell death mode, NO(2) generation and cellular glutathione level. The severity of the process correlated positively with NO(2) levels in pancreatic acinar cell cytosol samples, and negatively with cellular glutathione levels.


Subject(s)
Biosensing Techniques/instrumentation , Flow Injection Analysis/instrumentation , Nitrogen Dioxide/metabolism , Pancreatitis/metabolism , Pancreatitis/pathology , Acute Disease , Animals , Apoptosis , Equipment Design , Equipment Failure Analysis , Male , Necrosis/metabolism , Nitrogen Dioxide/analysis , Rats , Rats, Wistar
11.
Neoplasia ; 11(10): 1003-11, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19794959

ABSTRACT

BACKGROUND/AIMS: Acetaminophen overdose causes hepatotoxicity mediated by toxic metabolites generated through the cytochrome P450 enzyme. The objective of this study was to investigate whether acetaminophen (AAP) can enhance cisplatin (CDDP) cytotoxicity against human hepatocarcinoma and hepatoblastoma cells in vitro and whether this effect can be prevented by N-acetylcysteine (NAC). METHODS: In vitro studies (glutathione [GSH] level, cell viability, and immunoblot assays) were performed using human hepatocarcinoma and hepatoblastoma cells cultured in AAP, CDDP, and the combination of both with or without delayed NAC administration. The pharmacology and toxicology of high-dose AAP in rats were also examined. RESULTS: Acetaminophen decreased GSH levels in liver cancer cells in a dose- and time-dependent manner. Acetaminophen combined with CDDP had enhanced cytotoxicity over CDDP alone. The cytotoxicity caused by AAP plus CDDP was decreased by NAC, with the effectiveness being time-dependent. The GSH level was lowered in the liver but not in the blood or the brain in rats treated with a high dose of AAP (1000 mg/kg). The expression of CYP2E1 protein, a key cytochrome P450 enzyme, varies among species but is not correlated to AAP sensitivity in liver cancer cells. CONCLUSIONS: Our results suggest that a chemotherapeutic regimen containing both AAP and CDDP with delayed NAC rescue has the potential to enhance chemotherapeutic efficacy while decreasing adverse effects. This would be a promising approach particularly for hepatoblastomas regardless of cellular CYP2E1 protein level but could also be beneficial in other malignancies.


Subject(s)
Acetaminophen/pharmacology , Cisplatin/pharmacology , Glutathione/metabolism , Acetylcysteine/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cytochrome P-450 CYP2E1/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Free Radical Scavengers/pharmacology , Hepatoblastoma/metabolism , Hepatoblastoma/pathology , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL