Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Front Oncol ; 11: 719091, 2021.
Article in English | MEDLINE | ID: mdl-34336705

ABSTRACT

Glioblastoma (GBM) is an aggressive brain malignancy with a dismal prognosis. With emerging evidence to disprove brain-immune privilege, there has been much interest in examining immunotherapy strategies to treat central nervous system (CNS) cancers. Unfortunately, the limited success of clinical studies investigating immunotherapy regimens, has led to questions about the suitability of immunotherapy for these cancers. Inadequate inherent populations of tumor infiltrating lymphocytes (TILs) and limited trafficking of systemic, circulating T cells into the CNS likely contribute to the poor response to immunotherapy. This paucity of TILs is in concert with the finding of epigenetic silencing of genes that promote immune cell movement (chemotaxis) to the tumor. In this study we evaluated the ability of GSK126, a blood-brain barrier (BBB) permeable small molecule inhibitor of EZH2, to reverse GBM immune evasion by epigenetic suppression of T cell chemotaxis. We also evaluated the in vivo efficacy of this drug in combination with anti-PD-1 treatment on tumor growth, survival and T cell infiltration in syngeneic mouse models. GSK126 reversed H3K27me3 in murine and human GBM cell lines. When combined with anti-PD-1 treatment, a significant increase in activated T cell infiltration into the tumor was observed. This resulted in decreased tumor growth and enhanced survival both in sub-cutaneous and intracranial tumors of immunocompetent, syngeneic murine models of GBM. Additionally, a significant increase in CXCR3+ T cells was also seen in the draining lymph nodes, suggesting their readiness to migrate to the tumor. Closer examination of the mechanism of action of GSK126 revealed its ability to promote the expression of IFN-γ driven chemokines CXCL9 and CXCL10 from the tumor cells, that work to traffic T cells without directly affecting T maturation and/or proliferation. The loss of survival benefit either with single agent or combination in immunocompromised SCID mice, suggest that the therapeutic efficacy of GSK126 in GBM is primarily driven by lymphocytes. Taken together, our data suggests that in glioblastoma, epigenetic modulation using GSK126 could improve current immunotherapy strategies by reversing the epigenetic changes that enable immune cell evasion leading to enhanced immune cell trafficking to the tumor.

2.
Curr Pharm Teach Learn ; 13(9): 1221-1227, 2021 09.
Article in English | MEDLINE | ID: mdl-34330402

ABSTRACT

BACKGROUND AND PURPOSE: Teaching and learning the spectrum of activity (SOA) of antimicrobial agents can be a challenge in pharmacy education. This study describes the implementation and assessment of a novel tool to aid in the instruction of SOA. Physical manipulatives were used as an active-learning technique to model bacterial pathogens for antimicrobial SOA in an infectious diseases (ID) integrated medication therapy management course. EDUCATIONAL ACTIVITY AND SETTING: Pharmacy students enrolled in two consecutive years of the ID course were provided the opportunity to utilize a set of manipulatives for in-class activities and out-of-class practice. The manipulatives were small, colored building blocks that could be used to model bacterial pathogens for antimicrobial SOA. A key was included with each set of blocks, color-coding each block to represent a different bacterial pathogen or pathogen group. Blocks were used during classroom instruction to model the SOA of antimicrobial agents, compare/contrast SOA between medications, and model bacterial pathogens requiring empiric coverage for various infections, allowing students to produce "bug-drug" matches. Course data from the previous year was utilized to compare pre-implementation aggregate performance with post-implementation data. Performance on SOA-related questions was assessed during the course, using an independent samples t-test. FINDINGS: The intervention group exhibited a statistically significant increased mean score on test questions relating to SOA as compared to the control group. SUMMARY: The use of manipulatives was associated with improved performance on SOA-related questions in an integrated ID course of pharmacy students.


Subject(s)
Education, Pharmacy , Students, Pharmacy , Curriculum , Humans , Learning , Problem-Based Learning
3.
J Pharm Pract ; 34(5): 703-709, 2021 Oct.
Article in English | MEDLINE | ID: mdl-31884876

ABSTRACT

BACKGROUND: Antibiotics are commonly prescribed for uncomplicated urinary tract infection (UTI) and acute otitis media (AOM) and may be unnecessary at times. The aim of this study was to evaluate prescribing practices for UTIs and AOM in a rural ambulatory care setting and to identify areas for improvement. METHODS: In a single-center, retrospective review conducted at a rural clinic, patients diagnosed with uncomplicated UTI and AOM were included. Patients were identified by International Classification of Diseases, Tenth (ICD-10) codes, and data were collected for visits between January 1, 2017, and December 31, 2017. The primary outcome was to assess adherence of antimicrobial prescribing to current treatment guidelines. RESULTS: Of the 76 patients identified, 28 met inclusion criteria. Of the 28 patients, 75% received an agent recommended first line in the treatment guidelines, and 18 of the 21 received a recommended dose. Only 17% of patients were prescribed an appropriate duration of treatment. CONCLUSION: Opportunities exist for antimicrobial stewardship interventions for uncomplicated UTIs and AOM. Prescribers are not consistently adhering to guidelines in regard to antibiotic choice, dose, or duration. Additional education and stewardship interventions are crucial considering the increased prevalence of antimicrobial resistance.


Subject(s)
Antimicrobial Stewardship , Urinary Tract Infections , Ambulatory Care , Ambulatory Care Facilities , Anti-Bacterial Agents/therapeutic use , Humans , Practice Patterns, Physicians' , Retrospective Studies , Urinary Tract Infections/diagnosis , Urinary Tract Infections/drug therapy , Urinary Tract Infections/epidemiology
4.
Biophys J ; 119(12): 2378-2390, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33189686

ABSTRACT

We have developed a novel, to our knowledge, in vitro instrument that can deliver intermediate-frequency (100-400 kHz), moderate-intensity (up to and exceeding 6.5 V/cm pk-pk) electric fields (EFs) to cell and tissue cultures generated using induced electromagnetic fields (EMFs) in an air-core solenoid coil. A major application of these EFs is as an emerging cancer treatment modality. In vitro studies by Novocure reported that intermediate-frequency (100-300 kHz), low-amplitude (1-3 V/cm) EFs, which they called "tumor-treating fields (TTFields)," had an antimitotic effect on glioblastoma multiforme (GBM) cells. The effect was found to increase with increasing EF amplitude. Despite continued theoretical, preclinical, and clinical study, the mechanism of action remains incompletely understood. All previous in vitro studies of "TTFields" have used attached, capacitively coupled electrodes to deliver alternating EFs to cell and tissue cultures. This contacting delivery method suffers from a poorly characterized EF profile and conductive heating that limits the duration and amplitude of the applied EFs. In contrast, our device delivers EFs with a well-characterized radial profile in a noncontacting manner, eliminating conductive heating and enabling thermally regulated EF delivery. To test and demonstrate our system, we generated continuous, 200-kHz EMF with an EF amplitude profile spanning 0-6.5 V/cm pk-pk and applied them to exemplar human thyroid cell cultures for 72 h. We observed moderate reduction in cell density (<10%) at low EF amplitudes (<4 V/cm) and a greater reduction in cell density of up to 25% at higher amplitudes (4-6.5 V/cm). Our device can be readily extended to other EF frequency and amplitude regimes. Future studies with this device should contribute to the ongoing debate about the efficacy and mechanism(s) of action of "TTFields" by better isolating the effects of EFs and providing access to previously inaccessible EF regimes.


Subject(s)
Electric Stimulation Therapy , Glioblastoma , Electric Conductivity , Electromagnetic Fields , Glioblastoma/therapy , Humans
5.
J Immunother Cancer ; 8(1)2020 02.
Article in English | MEDLINE | ID: mdl-32051289

ABSTRACT

Immunomodulatory therapies targeting inhibitory checkpoint molecules have revolutionized the treatment of solid tumor malignancies. Concerns about whether systemic administration of an immune checkpoint inhibitor could impact primary brain tumors were answered with the observation of definitive responses in pediatric patients harboring hypermutated gliomas. Although initial clinical results in patients with glioblastoma (GBM) were disappointing, recently published results have demonstrated a potential survival benefit in patients with recurrent GBM treated with neoadjuvant programmed cell death protein 1 blockade. While these findings necessitate verification in subsequent studies, they support the possibility of achieving clinical meaningful immune responses in malignant primary brain tumors including GBM, a disease in dire need of additional therapeutic options. There are several challenges involved in treating glioma with immune checkpoint modulators including the immunosuppressive nature of GBM itself with high inhibitory checkpoint expression, the immunoselective blood brain barrier impairing the ability for peripheral lymphocytes to traffic to the tumor microenvironment and the high prevalence of corticosteroid use which suppress lymphocyte activation. However, by simultaneously targeting multiple costimulatory and inhibitory pathways, it may be possible to achieve an effective antitumoral immune response. To this end, there are now several novel agents targeting more recently uncovered "second generation" checkpoint molecules. Given the multiplicity of drugs being considered for combination regimens, an increased understanding of the mechanisms of action and resistance combined with more robust preclinical and early clinical testing will be needed to be able to adequately test these agents. This review summarizes our current understanding of T lymphocyte-modulating checkpoint molecules as it pertains to glioma with the hope for a renewed focus on the most promising therapeutic strategies.


Subject(s)
Brain Neoplasms/drug therapy , Glioma/drug therapy , Immunotherapy/methods , T-Lymphocytes/metabolism , Brain Neoplasms/pathology , Glioma/pathology , Humans
6.
Pharmacy (Basel) ; 7(3)2019 Sep 11.
Article in English | MEDLINE | ID: mdl-31514363

ABSTRACT

As one of the earliest developed antimicrobial classes, sulfonamides remain important therapeutic options for the empiric and definitive treatment of various infectious diseases. In the general population, approximately 3-8% of patients are reported to experience a sulfonamide allergy. Sulfonamide allergies can result in various physical manifestations; however, rash is reported as the most frequently observed. In patients with human immunodeficiency virus (HIV), dermatologic reactions to sulfonamide antimicrobial agents occur 10 to 20 times more frequently compared to immunocompetent patients. This article describes the incidence, manifestations, and risk factors associated with sulfonamide allergies. The potential for cross-reactivity of allergies to sulfonamide antimicrobials with nonantimicrobial sulfonamide medications is also reviewed. Data suggest that substitutions at the N1 and N4 positions are the primary determinants of drug allergy instead of the common sulfonamide moiety. For patients with an indication for a sulfonamide antimicrobial with a listed allergy, it is important for healthcare practitioners to adequately assess the allergic reaction to determine appropriate management. Rechallenge and desensitization strategies may be appropriate for patients with delayed maculopapular eruptions, while alternative treatment options may be prudent for more severe reactions. Available data suggests a low risk of cross-allergenicity between sulfonamide antimicrobial and nonantimicrobial agents.

7.
Cancer Res ; 79(20): 5218-5232, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31488423

ABSTRACT

Cancer cells rely on mitochondrial functions to regulate key survival and death signals. How cancer cells regulate mitochondrial autophagy (mitophagy) in the tumor microenvironment as well as utilize mitophagy as a survival signal is still not well understood. Here, we elucidate a key survival mechanism of mitochondrial NIX-mediated mitophagy within the hypoxic region of glioblastoma, the most malignant brain tumor. NIX was overexpressed in the pseudopalisading cells that envelop the hypoxic-necrotic regions, and mitochondrial NIX expression was robust in patient-derived glioblastoma tumor tissues and glioblastoma stem cells. NIX was required for hypoxia and oxidative stress-induced mitophagy through NFE2L2/NRF2 transactivation. Silencing NIX impaired mitochondrial reactive oxygen species clearance, cancer stem cell maintenance, and HIF/mTOR/RHEB signaling pathways under hypoxia, resulting in suppression of glioblastoma survival in vitro and in vivo. Clinical significance of these findings was validated by the compelling association between NIX expression and poor outcome for patients with glioblastoma. Taken together, our findings indicate that the NIX-mediated mitophagic pathway may represent a key therapeutic target for solid tumors, including glioblastoma. SIGNIFICANCE: NIX-mediated mitophagy regulates tumor survival in the hypoxic niche of glioblastoma microenvironment, providing a potential therapeutic target for glioblastoma.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/20/5218/F1.large.jpg.


Subject(s)
Brain Neoplasms/metabolism , Cell Hypoxia/physiology , Glioblastoma/metabolism , Membrane Proteins/physiology , Mitochondria/metabolism , Mitophagy/physiology , Neoplasm Proteins/physiology , Proto-Oncogene Proteins/physiology , Tumor Suppressor Proteins/physiology , Animals , Brain Neoplasms/pathology , Glioblastoma/pathology , Glioma/metabolism , Glioma/pathology , Heterografts , Humans , Hypoxia-Inducible Factor 1/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, SCID , NF-E2-Related Factor 2/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Oxidative Stress , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Ras Homolog Enriched in Brain Protein/physiology , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology , Tumor Microenvironment , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
8.
JCI Insight ; 4(20)2019 10 17.
Article in English | MEDLINE | ID: mdl-31536478

ABSTRACT

Meningiomas are the most common adult primary tumor of the central nervous system, but there are no known effective medical therapies for recurrent meningioma, particularly for World Health Organization grade II and III tumors. Meningiomas arise from the meninges, located outside the blood-brain barrier, and therefore may be directly targeted by antibody-mediated immunotherapy. We found that programmed cell death ligand 1 (PD-L1) was highly expressed in multiple human malignant meningioma cell lines and patient tumor samples. PD-L1 was targeted with the anti-PD-L1 antibody avelumab and directed natural killer cells to mediate antibody-dependent cellular cytotoxicity (ADCC) of PD-L1-expressing meningioma tumors both in vitro and in vivo. ADCC of meningioma cells was significantly increased in target cells that upregulated PD-L1 expression and, conversely, abrogated in tumor cells that were depleted of PD-L1. Additionally, the high-affinity natural killer cell line, haNK, outperformed healthy donor NK cells in meningioma ADCC. Together, these data support a clinical trial designed to target PD-L1 with avelumab and haNK cells, potentially offering a novel immunotherapeutic approach for patients with malignant meningioma.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Immunotherapy/methods , Killer Cells, Natural/transplantation , Meningeal Neoplasms/therapy , Meningioma/therapy , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cell Line, Tumor , Female , Gene Knockdown Techniques , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Meningeal Neoplasms/immunology , Meningioma/immunology , Mice , Treatment Outcome , Xenograft Model Antitumor Assays
9.
Neuro Oncol ; 21(1): 37-46, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29771386

ABSTRACT

Glioblastoma (GBM) is a highly malignant CNS tumor with very poor survival despite intervention with conventional therapeutic strategies. Although the CNS is separated from the immune system by the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier, emerging evidence of immune surveillance and the selective infiltration of GBMs by immune suppressive cells indicates that there is breakdown or compromise of these physical barriers. This in turn offers hope that immunotherapy can be applied to specifically target and reduce tumor burden. One of the major setbacks in translating immunotherapy strategies is the hostile microenvironment of the tumor that inhibits trafficking of effector immune cells such as cytotoxic T lymphocytes into the CNS. Incorporating important findings from autoimmune disorders such as multiple sclerosis to understand and thereby enhance cytotoxic lymphocyte infiltration into GBM could augment immunotherapy strategies to treat this disease. However, although these therapies are designed to evoke a potent immune response, limited space in the brain and cranial vault reduces tolerance for immune therapy-induced inflammation and resultant brain edema. Therefore, successful immunotherapy requires that a delicate balance be maintained between activating and retaining lasting antitumor immunity.


Subject(s)
Antineoplastic Agents/therapeutic use , Central Nervous System Neoplasms/therapy , Immune Tolerance/immunology , Immunotherapy/methods , Tumor Microenvironment/immunology , Animals , Blood-Brain Barrier , Central Nervous System Neoplasms/immunology , Humans
10.
Neurosurgery ; 85(2): 280-289, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30011045

ABSTRACT

BACKGROUND: Checkpoint inhibition has demonstrated clinical efficacy in a variety of solid tumors. Reports of programmed death ligand 1 (PD-L1) expression in glioblastoma are highly variable (ranging from 6% to 88%) and its role as a prognostic marker has yielded conflicting results. OBJECTIVE: To validate the prevalence and prognostic role of PD-L1 expression in a large cohort of diffuse gliomas according to the 2016 revised WHO classification. METHODS: Using tissue microarrays, we compared 5 PD-L1 monoclonal antibodies (n = 56) and validated expression (n = 183) using quantitative immunohistochemistry (IHC) and RNA in situ hybridization (RISH). Expression data from The Cancer Genome Atlas (TCGA) and published studies were compared with clinical outcome. Multiplexed immunophenotyping was used to identify PD-L1+ cell populations in post-treatment glioblastoma. RESULTS: Using a 5% cut-off, PD-L1 expression was significantly associated with a poor prognosis in both histologically defined (n = 125, log-rank P < .001) and recurrent isocitrate dehydrogenase (IDH)-wildtype glioblastoma (n = 60, log-rank P = .015). PD-L1 remained a significant negative prognosticator in Cox regression analysis (hazard ratio: 1.96, P = .021). Analysis of TCGA data confirmed decreased overall survival in recurrent non-glioma CpG island methylator phenotype (G-CIMP) glioblastoma (n = 12, log-rank P = .023), but not in glioblastoma as a group (n = 444, log-rank P = .135). PD-L1 RISH showed a significant correlation with IHC (P < .0001). PD-L1 was observed in the proliferating perivascular stem cell and immune niche of post-treatment glioblastoma. CONCLUSION: A 5% PD-L1 expression cut-off identified a subset of glioblastoma that is associated with a worse clinical outcome. This association remained significant within the newly defined IDH-wildtype classification. These findings could have implications for patient stratification in future clinical trials of PD-1/PD-L1 blockade.


Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor/analysis , Brain Neoplasms/pathology , Glioblastoma/pathology , Adolescent , Adult , B7-H1 Antigen/analysis , Brain Neoplasms/genetics , Child , Child, Preschool , Cohort Studies , Female , Glioblastoma/genetics , Humans , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Prognosis , Proportional Hazards Models , Retrospective Studies , Young Adult
11.
Clin Infect Dis ; 69(2): 259-265, 2019 07 02.
Article in English | MEDLINE | ID: mdl-30339217

ABSTRACT

BACKGROUND: Vancomycin-resistant enterococci are an important cause of healthcare-associated infections and are inherently resistant to many commonly used antibiotics. Linezolid is the only drug currently approved by the US Food and Drug Administration to treat vancomycin-resistant enterococci; however, resistance to this antibiotic appears to be increasing. Although outbreaks of linezolid- and vancomycin-resistant Enterococcus faecium (LR-VRE) in solid organ transplant recipients remain uncommon, they represent a major challenge for infection control and hospital epidemiology. METHODS: We describe a cluster of 4 LR-VRE infections among a group of liver and multivisceral transplant recipients in a single intensive care unit. Failure of treatment with linezolid in 2 cases led to a review of standard clinical laboratory methods for susceptibility determination. Testing by alternative methods including whole genome sequencing (WGS) and a comprehensive outbreak investigation including sampling of staff members and surfaces was performed. RESULTS: Review of laboratory testing methods revealed a limitation in the VITEK 2 system with regard to reporting resistance to linezolid. Linezolid resistance in all cases was confirmed by E-test method. The use of WGS identified a resistant subpopulation with the G2376C mutation in the 23S ribosomal RNA. Sampling of staff members' dominant hands as well as sampling of surfaces in the unit identified no contaminated sources for transmission. CONCLUSIONS: This cluster of LR-VRE in transplant recipients highlights the possible shortcomings of standard microbiology laboratory methods and underscores the importance of WGS to identify resistance mechanisms that can inform patient care, as well as infection control and antibiotic stewardship measures.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Enterococcus faecium/drug effects , Gram-Positive Bacterial Infections/microbiology , Linezolid/pharmacology , Transplant Recipients , Vancomycin-Resistant Enterococci/drug effects , Aged , Antimicrobial Stewardship , Disease Management , Disease Outbreaks , Enterococcus faecium/genetics , Enterococcus faecium/isolation & purification , Gram-Positive Bacterial Infections/epidemiology , Humans , Infection Control/methods , Intensive Care Units , Male , Middle Aged , Point Mutation , RNA, Ribosomal, 23S/genetics , Sequence Analysis, DNA , Vancomycin-Resistant Enterococci/genetics , Vancomycin-Resistant Enterococci/isolation & purification , Whole Genome Sequencing
12.
Neurosurgery ; 84(4): 945-953, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30189044

ABSTRACT

BACKGROUND: Glioblastoma is the most common primary malignancy of the brain, with a dismal prognosis. Immunomodulation via checkpoint inhibition has provided encouraging results in non-CNS malignancies, but prediction of responders has proven to be challenging in glioblastoma patients. OBJECTIVE: To determine the proportion of patients who have a measurable increase of interferon gamma levels in brain tumor tissue after their first dose of nivolumab, and to evaluate the safety of using brain tumor microdialysis to monitor for immune response while evaluating the safety of the combination of anti-programmed death 1 (PD-1) and anti-lymphocyte activation gene 3 (LAG-3) checkpoint inhibition. METHODS: The study design is a single-center, nonrandomized phase 1 clinical trial. Up to 15 adult patients with recurrent glioblastoma will be enrolled with the goal of 10 patients completing the trial over an anticipated 18 mo. Patients will undergo biopsy; placement of microdialysis catheters and lumbar drains; treatment with anti-PD-1 checkpoint inhibition; comprehensive immune biomarker collection; tumor resection; and then treatment with anti-PD-1 and anti-LAG-3 checkpoint inhibition until progression. EXPECTED OUTCOMES: We expect interferon gamma levels to increase in the brain as measured via microdialysis in treated patients. Based on published reports, microdialysis in this patient population is expected to be safe, and anti-LAG-3 and anti-PD-1 combined will likely have a similar side effect profile to other checkpoint inhibitor combinations. DISCUSSION: The failure of recent trials of immune therapies in glioblastoma underscores the need to appropriately measure response in the treated tissue. This trial may provide insight on indicators of which patients will respond to immune therapy.


Subject(s)
Brain Neoplasms , Cytokines , Glioblastoma , Microdialysis , Monitoring, Immunologic , Adult , Brain Chemistry , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Cytokines/analysis , Cytokines/isolation & purification , Glioblastoma/immunology , Glioblastoma/metabolism , Glioblastoma/therapy , Humans , Interferon-gamma/analysis , Interferon-gamma/isolation & purification , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/therapy
14.
J Immunother Cancer ; 6(1): 51, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29891009

ABSTRACT

BACKGROUND: Corticosteroids are routinely utilized to alleviate edema in patients with intracranial lesions and are first-line agents to combat immune-related adverse events (irAEs) that arise with immune checkpoint blockade treatment. However, it is not known if or when corticosteroids can be administered without abrogating the efforts of immunotherapy. The purpose of this study was to evaluate the impact of dexamethasone on lymphocyte activation and proliferation during checkpoint blockade to provide guidance for corticosteroid use while immunotherapy is being implemented as a cancer treatment. METHODS: Lymphocyte proliferation, differentiation, and cytokine production were evaluated during dexamethasone exposure. Human T cells were stimulated through CD3 ligation and co-stimulated either directly by CD28 ligation or by providing CD80, a shared ligand for CD28 and CTLA-4. CTLA-4 signaling was inhibited by antibody blockade using ipilimumab which has been approved for the treatment of several solid tumors. The in vivo effects of dexamethasone during checkpoint blockade were evaluated using the GL261 syngeneic mouse intracranial model, and immune populations were profiled by flow cytometry. RESULTS: Dexamethasone upregulated CTLA-4 mRNA and protein in CD4 and CD8 T cells and blocked CD28-mediated cell cycle entry and differentiation. Naïve T cells were most sensitive, leading to a decrease of the development of more differentiated subsets. Resistance to dexamethasone was conferred by blocking CTLA-4 or providing strong CD28 co-stimulation prior to dexamethasone exposure. CTLA-4 blockade increased IFNγ expression, but not IL-2, in stimulated human peripheral blood T cells exposed to dexamethasone. Finally, we found that CTLA-4 blockade partially rescued T cell numbers in mice bearing intracranial gliomas. CTLA-4 blockade was associated with increased IFNγ-producing tumor-infiltrating T cells and extended survival of dexamethasone-treated mice. CONCLUSIONS: Dexamethasone-mediated T cell suppression diminishes naïve T cell proliferation and differentiation by attenuating the CD28 co-stimulatory pathway. However, CTLA-4, but not PD-1 blockade can partially prevent some of the inhibitory effects of dexamethasone on the immune response.


Subject(s)
Dexamethasone/therapeutic use , Immunosuppression Therapy/methods , Immunotherapy/methods , Animals , Dexamethasone/pharmacology , Disease Models, Animal , Female , Humans , Mice
15.
Clin Cancer Res ; 24(17): 4175-4186, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29437767

ABSTRACT

Purpose: T-cell dysfunction is a hallmark of glioblastoma (GBM). Although anergy and tolerance have been well characterized, T-cell exhaustion remains relatively unexplored. Exhaustion, characterized in part by the upregulation of multiple immune checkpoints, is a known contributor to failures amid immune checkpoint blockade, a strategy that has lacked success thus far in GBM. This study is among the first to examine, and credential as bona fide, exhaustion among T cells infiltrating human and murine GBM.Experimental Design: Tumor-infiltrating and peripheral blood lymphocytes (TILs and PBLs) were isolated from patients with GBM. Levels of exhaustion-associated inhibitory receptors and poststimulation levels of the cytokines IFNγ, TNFα, and IL2 were assessed by flow cytometry. T-cell receptor Vß chain expansion was also assessed in TILs and PBLs. Similar analysis was extended to TILs isolated from intracranial and subcutaneous immunocompetent murine models of glioma, breast, lung, and melanoma cancers.Results: Our data reveal that GBM elicits a particularly severe T-cell exhaustion signature among infiltrating T cells characterized by: (1) prominent upregulation of multiple immune checkpoints; (2) stereotyped T-cell transcriptional programs matching classical virus-induced exhaustion; and (3) notable T-cell hyporesponsiveness in tumor-specific T cells. Exhaustion signatures differ predictably with tumor identity, but remain stable across manipulated tumor locations.Conclusions: Distinct cancers possess similarly distinct mechanisms for exhausting T cells. The poor TIL function and severe exhaustion observed in GBM highlight the need to better understand this tumor-imposed mode of T-cell dysfunction in order to formulate effective immunotherapeutic strategies targeting GBM. Clin Cancer Res; 24(17); 4175-86. ©2018 AACRSee related commentary by Jackson and Lim, p. 4059.


Subject(s)
Glioblastoma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/immunology , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Interferon-gamma/genetics , Interleukin-2/genetics , Lymphocytes, Tumor-Infiltrating/pathology , Male , Mice , Middle Aged , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/pathology , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/genetics
16.
Neuro Oncol ; 20(6): 799-809, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29294092

ABSTRACT

Background: Standard therapy for chordoma consists of surgical resection followed by high-dose irradiation. Protein phosphatase 2A (PP2A) is a ubiquitously expressed serine/threonine phosphatase involved in signal transduction, cell cycle progression, cell differentiation, and DNA repair. LB100 is a small-molecule inhibitor of PP2A designed to sensitize cancer cells to DNA damage from irradiation and chemotherapy. A recently completed phase I trial of LB100 in solid tumors demonstrated its safety. Here, we show the therapeutic potential of LB100 in chordoma. Methods: Three patient-derived chordoma cell lines were used: U-CH1, JHC7, and UM-Chor1. Cell proliferation was determined with LB100 alone and in combination with irradiation. Cell cycle progression was assessed by flow cytometry. Quantitative γ-H2AX immunofluorescence and immunoblot evaluated the effect of LB100 on radiation-induced DNA damage. Ultrastructural evidence for nuclear damage was investigated using Raman imaging and transmission electron microscopy. A xenograft model was established to determine potential clinical utility of adding LB100 to irradiation. Results: PP2A inhibition in concert with irradiation demonstrated in vitro growth inhibition. The combination of LB100 and radiation also induced accumulation at the G2/M phase of the cell cycle, the stage most sensitive to radiation-induced damage. LB100 enhanced radiation-induced DNA double-strand breaks. Animals implanted with chordoma cells and treated with the combination of LB100 and radiation demonstrated tumor growth delay. Conclusions: Combining LB100 and radiation enhanced DNA damage-induced cell death and delayed tumor growth in an animal model of chordoma. PP2A inhibition by LB100 treatment may improve the effectiveness of radiation therapy for chordoma.


Subject(s)
Biomarkers, Tumor/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Chordoma/drug therapy , Chordoma/pathology , Piperazines/pharmacology , Protein Phosphatase 2/antagonists & inhibitors , Radiation Tolerance/drug effects , Animals , Apoptosis , Cell Movement , Cell Proliferation , Chordoma/enzymology , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Oncotarget ; 8(20): 33316-33328, 2017 May 16.
Article in English | MEDLINE | ID: mdl-28410196

ABSTRACT

EGFR pathway is upregulated in malignant gliomas, and its downstream signaling is important for self-renewal of glioma cancer stem-like cells (GSC). p38 mitogen-activated protein kinase (MAPK) signaling, a stress-activated signaling cascade with suppressive and permissive effects on tumorigenesis, can promote internalization and ubiquitin ligase mediated degradation of EGFR. In this study, we investigated the role of p38 MAPK signaling on the self-renewal of GSCs with the hypothesis that inhibition may lead to enhanced self-renewal capacity by retention of EGFR. Inhibition of p38 MAPK pathway led to increase in EGFR expression but surprisingly, reduced proliferation. Additional functional evaluation revealed that p38 inhibition was associated with decrease in cell death and maintenance of undifferentiated state. Further probing the effect of p38 inhibition demonstrated attenuation of EGFR downstream signaling activity in spite of prolonged surface expression of the receptor. In vitro observations were confirmed in xenograft in vivo experiments. These data suggest that p38 MAPK control of EGFR signaling activity may alter GSC cell cycle state by regulating quiescence and passage into transit amplifying state.


Subject(s)
ErbB Receptors/metabolism , Glioma/metabolism , Neoplastic Stem Cells/metabolism , Resting Phase, Cell Cycle , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Apoptosis , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Self Renewal , ErbB Receptors/genetics , Gene Expression , Glioma/genetics , Glioma/pathology , Humans , Ligands , Phosphorylation , Protein Binding , Protein Transport
18.
Oncotarget ; 8(25): 40233-40245, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28410215

ABSTRACT

Tissue hypoxia contributes to solid tumor pathogenesis by activating a series of adaptive programs. We previously showed that hypoxia promotes the preferential expansion and maintenance of CD133 positive human glioma stem cells (GSC) in a hypoxia inducible factor 1 alpha (HIF-1α)-dependent mechanism. Here, we examined the activity of digitoxin (DT), a cardiac glycoside and a putative inhibitor of HIF-1α, on human GSC in vitro and in vivo. During hypoxic conditions (1% O2), we observed the effect of DT on the intracellular level of HIF-1α and the extracellular level of vascular endothelial growth factor (VEGF) in human GSC. We found that DT at clinically achievable concentrations, suppressed HIF-1α accumulation during hypoxic conditions in human GSC and established glioma cell lines. DT treatment also significantly attenuated hypoxia-induced expression of VEGF, a downstream target of HIF-1α. Exposure to DT also reduced hypoxia-induced activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway. Furthermore, DT potently inhibited neurosphere formation, and decreased CD133 expression even at concentrations that were not overtly cytotoxic. Lastly, treatment with DT reduced GSC engraftment in an in vivo xenograft model of glioblastoma. Intraperitoneal injections of DT significantly inhibited the growth of established glioblastoma xenografts, and suppressed expression of HIF-1α and carbonic anhydrase (CA9), a surrogate marker of hypoxia. Taken together, these results suggest that DT at clinically achievable concentration functions as an inhibitor of HIF-1α, worthy of further investigations in the therapy of glioblastoma.


Subject(s)
Carbonic Anhydrases/biosynthesis , Digitoxin/pharmacology , Glioblastoma/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Neoplastic Stem Cells/pathology , AC133 Antigen/metabolism , Animals , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mice , Mice, SCID , RNA Interference , RNA, Small Interfering/genetics , Spheroids, Cellular , Tumor Cells, Cultured , Tumor Hypoxia/physiology , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
19.
Neuro Oncol ; 19(7): 887-896, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28339582

ABSTRACT

Glioblastoma is the most common and aggressive malignant primary brain tumor. Cellular heterogeneity is a characteristic feature of the disease and contributes to the difficulty in formulating effective therapies. Glioma stem-like cells (GSCs) have been identified as a subpopulation of tumor cells that are thought to be largely responsible for resistance to treatment. Intratumoral hypoxia contributes to maintenance of the GSCs by supporting the critical stem cell traits of multipotency, self-renewal, and tumorigenicity. This review highlights the interaction of GSCs with the hypoxic tumor microenvironment, exploring the mechanisms underlying the contribution of GSCs to tumor vessel dynamics, immune modulation, and metabolic alteration.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Tumor Hypoxia , Tumor Microenvironment , Animals , Brain Neoplasms/complications , Glioblastoma/complications , Humans , Neovascularization, Pathologic/complications , Neovascularization, Pathologic/metabolism , Phenotype
20.
Pharmacol Ther ; 168: 53-60, 2016 12.
Article in English | MEDLINE | ID: mdl-27595927

ABSTRACT

Hematopoietic cells are increasingly recognized as playing key roles in tumor growth and metastatic progression. Although many studies have focused on the functional interaction of hematopoietic cells with tumor cells, few have examined the regulation of hematopoiesis by the hematopoietic stem cell (HSC) niche in the setting of cancer. Hematopoiesis occurs primarily in the bone marrow, and processes including expansion, mobilization, and differentiation of hematopoietic progenitors are tightly regulated by the specialized stem cell niche. Loss of niche components or the ability of stem cells to localize to the stem cell niche relieves HSCs of the restrictions imposed under normal homeostasis. In this review, we discuss how tumor-derived factors and therapeutic interventions disrupt structural and regulatory properties of the stem cell niche, resulting in niche invasion by hematopoietic malignancies, extramedullary hematopoiesis, myeloid skewing by peripheral tissue microenvironments, and lymphopenia. The key regulatory roles played by the bone marrow niche in hematopoiesis has implications for therapy-related toxicity and the successful development of immune-based therapies for cancer.


Subject(s)
Hematopoietic Stem Cells/metabolism , Neoplasms/metabolism , Stem Cell Niche , Animals , Disease Progression , Hematologic Neoplasms/immunology , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/therapy , Hematopoiesis/physiology , Humans , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/therapy , Neoplastic Stem Cells/metabolism , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL