Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Front Vet Sci ; 10: 1150590, 2023.
Article En | MEDLINE | ID: mdl-37396989

Introduction: In humans, gait speed is a crucial component in geriatric evaluation since decreasing speed can be a harbinger of cognitive decline and dementia. Aging companion dogs can suffer from age-related mobility impairment, cognitive decline and dementia known as canine cognitive dysfunction syndrome. We hypothesized that there would be an association between gait speed and cognition in aging dogs. Methods: We measured gait speed on and off leash in 46 adult and 49 senior dogs. Cognitive performance in senior dogs was assessed by means of the Canine Dementia Scale and a battery of cognitive tests. Results: We demonstrated that dogs' food-motivated gait speed off leash is correlated with fractional lifespan and cognitive performance in dogs, particularly in the domains of attention and working memory. Discussion: Food-motivated gait speed off leash represents a relatively easy variable to measure in clinical settings. Moreover, it proves to be a more effective indicator of age-related deterioration and cognitive decline than gait speed on leash.

2.
Doc Ophthalmol ; 147(1): 15-28, 2023 Aug.
Article En | MEDLINE | ID: mdl-37302110

PURPOSE: To determine the association between age and retinal full-field electroretinographic (ERG) measures in companion (pet) dogs, an important translational model species for human neurologic aging. METHODS: Healthy adult dogs with no significant ophthalmic abnormalities were included. Unilateral full-field light- and dark-adapted electroretinography was performed using a handheld device, with mydriasis and topical anaesthesia. Partial least squares effect screening analysis was performed to determine the effect of age, sex, body weight and use of anxiolytic medication on log-transformed ERG peak times and amplitudes; age and anxiolytic usage had significant effects on multiple ERG outcomes. Mixed model analysis was performed on data from dogs not receiving anxiolytic medications. RESULTS: In dogs not receiving anxiolytics, median age was 118 months (interquartile range 72-140 months, n = 77, 44 purebred, 33 mixed breed dogs). Age was significantly associated with prolonged peak times of a-waves (dark-adapted 3 and 10 cds/m2 flash p < 0.0001) and b-waves (cone flicker p = 0.03, dark-adapted 0.01 cds/m2 flash p = 0.001). Age was also significantly associated with reduced amplitudes of a-waves (dark-adapted 3 cds/m2 flash p < 0.0001, 10 cds/m2 flash p = 0.005) and b-waves (light-adapted 3 cds/m2 flash p < 0.0001, dark-adapted 0.01 cds/m2 flash p = 0.0004, 3 cds/m2 flash p < 0.0001, 10 cds/m2 flash p = 0.007) and flicker (light-adapted 30 Hz 3 cds/m2 p = 0.0004). Within the Golden Retriever breed, these trends were matched in a cross-sectional analysis of 6 individuals that received no anxiolytic medication. CONCLUSIONS: Aged companion dogs have slower and reduced amplitude responses in both rod- and cone-mediated ERG. Consideration of anxiolytic medication use should be made when conducting ERG studies in dogs.


Electroretinography , Pets , Adult , Humans , Animals , Dogs , Aged , Child , Cross-Sectional Studies , Dark Adaptation , Photic Stimulation
3.
J Vet Intern Med ; 36(5): 1708-1718, 2022 Sep.
Article En | MEDLINE | ID: mdl-35932193

BACKGROUND: Elderly people with presbycusis are at higher risk for dementia and depression than the general population. There is no information regarding consequences of presbycusis in dogs. OBJECTIVE: Evaluate the relationship between cognitive function, quality of life, and hearing loss in aging companion dogs. ANIMALS: Thirty-nine elderly companion dogs. METHODS: Prospective study. Hearing was evaluated using brainstem auditory evoked response (BAER) testing. Dogs were grouped by hearing ability. Owners completed the canine dementia scale (CADES) and canine owner-reported quality of life (CORQ) questionnaire. Cognitive testing was performed, and cognitive testing outcomes, CADES and CORQ scores and age were compared between hearing groups. RESULTS: Nineteen dogs could hear at 50 dB, 12 at 70 dB, and 8 at 90 dB with mean ages (months) of 141 ± 14, 160 ± 16, and 172 ± 15 for each group respectively (P = .0002). Vitality and companionship CORQ scores were significantly lower as hearing deteriorated (6.6-5.4, 50-90 dB group, P = .03 and 6.9-6.2, 50-90 dB group, P = .02, respectively). Cognitive classification by CADES was abnormal in all 90 dB group dogs and normal in 3/12 70 dB group and 11/19 50 dB group dogs (P = .0004). Performance on inhibitory control, detour and sustained gaze tasks decreased significantly with hearing loss (P = .001, P = .008, P = .002, respectively). In multivariate analysis, higher CADES score was associated with worse hearing (P = .01). CONCLUSIONS AND CLINICAL IMPORTANCE: Presbycusis negatively alters owner-pet interactions and is associated with poor executive performance and owner-assessed dementia severity.


Dementia , Dog Diseases , Presbycusis , Aging , Animals , Cognition , Dementia/epidemiology , Dogs , Hearing , Humans , Pets , Presbycusis/epidemiology , Presbycusis/veterinary , Prospective Studies , Quality of Life
5.
J Alzheimers Dis ; 87(3): 1367-1378, 2022.
Article En | MEDLINE | ID: mdl-35431246

BACKGROUND: Aging dogs may suffer from canine cognitive dysfunction syndrome (CCDS), a condition in which cognitive decline is associated with amyloid pathology and cortical atrophy. Presumptive diagnosis is made through physical examination, exclusion of systemic/metabolic conditions, and completion of screening questionnaires by owners. OBJECTIVE: This study aimed to determine whether cognitive function could be quantified in aging pet dogs, and to correlate cognitive testing with validated questionnaires and plasma neurofilament light chain (pNfL) concentration. METHODS: Thirty-nine dogs from fifteen breeds were recruited (9.3 to 15.3 years). Owners completed the Canine Dementia Scale (CADES) and Canine Cognitive Dysfunction Rating scale (CCDR). Executive control and social cues were tested, and pNfL was measured with single molecule array assay. Comparisons were made between cognitive testing scores, CADES, CCDR scores, and pNfL. RESULTS: CADES scoring classified five dogs as severe CCDS, six as moderate, ten as mild, and eighteen as normal. CCDR identified seven dogs at risk of CCDS and thirty-two as normal. Cognitive testing was possible in the majority of dogs, although severely affected dogs were unable to learn tasks. CADES score correlated with sustained attention duration (r = -0.47, p = 0.002), inhibitory control (r = -0.51, p = 0.002), detour (r = -0.43, p = 0.001), and pNfL (r = 0.41, p = 0.025). Concentration of pNfL correlated with inhibitory control (r = -0.7, p≤0.001). The CCDR scale correlated with performance on inhibitory control (r = -0.46, p = 0.005). CONCLUSION: Our findings suggest that a multi-dimensional approach using a combination of questionnaires, specific cognitive tests, and pNfL concentration can be used to quantify cognitive decline in aging pet dogs.


Cognitive Dysfunction , Aging , Animals , Biomarkers , Cognitive Dysfunction/diagnostic imaging , Dogs , Humans , Neuropsychological Tests , Surveys and Questionnaires
6.
BMC Vet Res ; 17(1): 296, 2021 Sep 06.
Article En | MEDLINE | ID: mdl-34488762

BACKGROUND: Canine elbow dysplasia (CED) is a complex developmental skeletal disorder associated with a number of pathological conditions within the cubital joint. Because CED is a heritable disease, it is important to identify and remove the affected animals from breeding. The first objective of this study was to describe the prevalence of medial coronoid process disease (MCPD) without (MCD) or with (FMCP) fragmented medial coronoid process, osteochondrosis (OC) and/or osteochondritis dissecans (OCD), ununited anconeal process (UAP), radio-ulnar incongruence (INC R-U) and humero-ulnar incongruence (INC H-U) in dogs with the use of CT imaging. The second aim was to determine the influence of demographics on the prevalence of investigated pathologies in dogs with clinical evidence of elbow dysplasia. RESULTS: In this retrospective study, CT data records of 169 dogs of different breeds presented to the small animal veterinary clinic from 2012 to 2018 were included. 69.23% of dogs diagnosed with CED were young (≤ 2 years old). The mean age of dogs presented with INC R-U was 1.68 ± 1.82 years, while in dogs without INC R-U the mean age was 2.64 ± 2.59 years. The mean age of dogs with INC H-U was 1.94 ± 2.06 years, while without INC H-U 3.29 ± 2.09 years. Labrador Retrievers, German Shepherd and Bernese Mountain dogs were most frequently presented with CED-associated lameness. In 122 dogs OA of varying severity was found. CONCLUSION: INC H-U, FMCP and MCD were among the most frequently found components of CED found in the present study. OCD and UAP were the least frequently diagnosed. Dogs presented with INC R-U and INC H-U were significantly younger than dogs without these CED components. Boxers, Dog de Bordeaux, American Staffordshire terriers and mixed-breed dogs were diagnosed later in life than the other breeds. OA of varying severity was found in 72.18% of dogs. Males accounted for more than 75% of the study population.


Dog Diseases/diagnostic imaging , Forelimb/diagnostic imaging , Joint Diseases/veterinary , Tomography, X-Ray Computed/veterinary , Age Factors , Animals , Dog Diseases/epidemiology , Dogs , Female , Joint Diseases/diagnostic imaging , Joint Diseases/epidemiology , Male , Prevalence , Retrospective Studies , Species Specificity
7.
Mol Neurobiol ; 58(2): 483-489, 2021 Feb.
Article En | MEDLINE | ID: mdl-32970242

Longevity-associated neurological disorders have been observed across human and canine aging populations. Alzheimer's disease (AD) and canine cognitive dysfunction syndrome (CDS) represent comparable diseases affecting both species as they age. Translational diagnostic and therapeutic research is needed for these incurable diseases. The amyloid ß (Aß) peptide family are AD-associated peptides with identical amino acid sequences between dogs and humans. Plasma Aß42 concentration increases with age and decreases with AD in humans, and cerebrospinal fluid (CSF) concentration decreases in AD and correlates inversely with the amyloid load within the brain. Similarly, CSF Aß42 concentrations decrease in dogs with CDS but there is limited and conflicting information on plasma Aß42 concentrations in aging dogs and dogs with CDS. We measured plasma concentrations of Aß42 and Aß40 with an ultrasensitive single-molecule array assay (SIMOA) in a population of healthy aging dogs of different life stages (n = 36) and dogs affected with CDS (n = 11). In addition, the ratio of Aß42/ß40 was calculated. The mean plasma concentrations of Aß42 and Aß40 increased significantly with age (r2 = 0.27, p = 0.001; and r2 = 0.42, p < 0.001, respectively) and with life stage: puppy/junior group (0.43-2 years): 1.23 ± 0.95 and 38.26 ± 49.43 pg/mL; adult/mature group (2.1-9 years): 10.99 ± 5.45 and 131.05 ± 80.17 pg/mL; geriatric/senior group (9.3-14.5 years): 18.65 ± 16.65 and 192.88 ± 146.38 pg/mL, respectively. Concentrations of Aß42 and Aß40 in dogs with CDS (11.0-15.6 years) were significantly lower than age-matched healthy dogs at 11.61 ± 6.39 and 150.23 ± 98.2 pg/mL (p = 0.0048 and p = 0.001), respectively. Our findings suggest the dynamics of canine plasma amyloid concentrations are analogous to that found in aging humans with and without AD.


Aging/blood , Amyloid beta-Peptides/blood , Cognitive Dysfunction/blood , Dogs/blood , Pets/blood , Animals , Female , Male
8.
Sci Transl Med ; 12(558)2020 08 26.
Article En | MEDLINE | ID: mdl-32848091

Metastases from primary breast cancer result in poor survival. ßIII-tubulin (TUBB3) has been established as a therapeutic target for breast cancer metastases specifically to the brain. In this study, we conducted a systematic analysis to determine the regulation of TUBB3 expression in breast cancer metastases to the brain and strategically target these metastases using vinorelbine (VRB), a drug approved by the U.S. Food and Drug Administration (FDA). We found that human epidermal growth factor receptor 2 (HER2) signaling regulates TUBB3 expression in both trastuzumab-sensitive and trastuzumab-resistant neoplastic cells. We further discovered that bromodomain and extra-terminal domain (BET) inhibition increases TUBB3 expression, rendering neoplastic cells more susceptible to apoptosis by VRB. Orthotopic xenograft assays using two different breast cancer cell models revealed a reduction in tumor volume with BET inhibition and VRB treatment. In addition, in vivo studies using a model of multiple brain metastasis (BM) showed improved survival with the combination of radiation + BET inhibitor (iBET-762) + VRB (75% long-term survivors, P < 0.05). Using in silico analysis and BET inhibition, we found that the transcription factor myeloid zinc finger-1 (MZF-1) protein binds to the TUBB3 promoter. BET inhibition decreases MZF-1 expression and subsequently increases TUBB3 expression. Overexpression of MZF-1 decreases TUBB3 expression and reduces BM in vivo, whereas its knockdown increases TUBB3 expression in breast cancer cells. In summary, this study demonstrates a regulatory mechanism of TUBB3 and provides support for an application of BET inhibition to sensitize breast cancer metastases to VRB-mediated therapy.


Breast Neoplasms , Tubulin , Brain/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Trastuzumab , Tubulin/metabolism , Vinorelbine
9.
Mol Neurobiol ; 57(7): 3143-3149, 2020 Jul.
Article En | MEDLINE | ID: mdl-32472519

Age is a primary risk factor for multiple comorbidities including neurodegenerative diseases. Pet dogs and humans represent two populations that have experienced a significant increase in average life expectancy over the last century. A higher prevalence of age-related neurodegenerative diseases has been observed across both species, and human diseases, such as Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS), have canine analogs, canine cognitive dysfunction (CCD), and degenerative myelopathy (DM) respectively. In humans, protein biomarkers have proved useful in the prediction and diagnosis of neurodegeneration. Molecular signatures of many proteins are highly conserved across species. In this study, we explored the potential of the neuronal cytoskeletal protein neurofilament light chain (NfL) as a biomarker of neuro-aging in dogs using an ultrasensitive single-molecule array assay to measure plasma concentrations. Healthy dogs of different ages and dogs affected with CCD and DM were evaluated. The mean plasma NfL concentrations in the different age groups of the healthy population were as follows: 4.55 ± 1.70 pg/mL in puppy/junior group (0.43-2 years), 13.51 ± 6.8 pg/mL in adult/mature group (2.1-9 years), and 47.1 ± 12.68 pg/mL in geriatric/senior group (9.3-14.5 years). Concentrations in dogs with DM (7.5-12.6 years) and CCD (11.0-15.6 years) were 84.17 ± 53.57 pg/mL and 100.73 ± 83.72 pg/mL, respectively. Plasma NfL increases in an age-dependent manner and is significantly elevated in dogs diagnosed with neurodegenerative disease. This work identified plasma NfL as a key clinical index of neuro-aging and neurodegeneration in pet dogs. Our findings mirror recent reports from human neurodegenerative diseases.


Aging/blood , Dog Diseases/diagnosis , Neurodegenerative Diseases/veterinary , Neurofilament Proteins/blood , Animals , Biomarkers/blood , Dog Diseases/blood , Dogs , Female , Male , Neurodegenerative Diseases/blood , Neurodegenerative Diseases/diagnosis
10.
Cancer Immunol Immunother ; 69(1): 81-94, 2020 Jan.
Article En | MEDLINE | ID: mdl-31844909

Amino acid deprivation is a strategy that malignancies utilize to blunt anti-tumor T-cell immune responses. It has been proposed that amino acid insufficiency in T-cells is detected by GCN2 kinase, which through phosphorylation of EIF2α, shuts down global protein synthesis leading to T-cell arrest. The role of this amino acid stress sensor in the context of malignant brain tumors has not yet been studied, and may elucidate important insights into the mechanisms of T-cell survival in this harsh environment. Using animal models of glioblastoma and animals with deficiency in GCN2, we explored the importance of this pathway in T-cell function within brain tumors. Our results show that GCN2 deficiency limited CD8+ T-cell activation and expression of cytotoxic markers in two separate murine models of glioblastoma in vivo. Importantly, adoptive transfer of antigen-specific T-cells from GCN2 KO mice did not control tumor burden as well as wild-type CD8+ T-cells. Our in vitro and in vivo data demonstrated that reduction in amino acid availability caused GCN2 deficient CD8+ T-cells to become rapidly necrotic. Mechanistically, reduced CD8+ T-cell activation and necrosis was due to a disruption in TCR signaling, as we observed reductions in PKCθ and phoshpo-PKCθ on CD8+ T-cells from GCN2 KO mice in the absence of tryptophan. Validating these observations, treatment of wild-type CD8+ T-cells with a downstream inhibitor of GCN2 activation also triggered necrosis of CD8+ T-cells in the absence of tryptophan. In conclusion, our data demonstrate the vital importance of intact GCN2 signaling on CD8+ T-cell function and survival in glioblastoma.


Brain Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Glioblastoma/immunology , Protein Serine-Threonine Kinases/metabolism , Tumor Escape/immunology , Adoptive Transfer , Animals , Brain Neoplasms/pathology , Brain Neoplasms/therapy , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor/transplantation , Cell Survival/immunology , Disease Models, Animal , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Lymphocyte Activation , Mice , Mice, Knockout , Necrosis/genetics , Necrosis/immunology , Phosphorylation/immunology , Protein Biosynthesis/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology
11.
Proc Natl Acad Sci U S A ; 116(47): 23714-23723, 2019 11 19.
Article En | MEDLINE | ID: mdl-31712430

Tumor-associated myeloid cells (TAMCs) are key drivers of immunosuppression in the tumor microenvironment, which profoundly impedes the clinical response to immune-dependent and conventional therapeutic modalities. As a hallmark of glioblastoma (GBM), TAMCs are massively recruited to reach up to 50% of the brain tumor mass. Therefore, they have recently been recognized as an appealing therapeutic target to blunt immunosuppression in GBM with the hope of maximizing the clinical outcome of antitumor therapies. Here we report a nano-immunotherapy approach capable of actively targeting TAMCs in vivo. As we found that programmed death-ligand 1 (PD-L1) is highly expressed on glioma-associated TAMCs, we rationally designed a lipid nanoparticle (LNP) formulation surface-functionalized with an anti-PD-L1 therapeutic antibody (αPD-L1). We demonstrated that this system (αPD-L1-LNP) enabled effective and specific delivery of therapeutic payload to TAMCs. Specifically, encapsulation of dinaciclib, a cyclin-dependent kinase inhibitor, into PD-L1-targeted LNPs led to a robust depletion of TAMCs and an attenuation of their immunosuppressive functions. Importantly, the delivery efficiency of PD-L1-targeted LNPs was robustly enhanced in the context of radiation therapy (RT) owing to the RT-induced up-regulation of PD-L1 on glioma-infiltrating TAMCs. Accordingly, RT combined with our nano-immunotherapy led to dramatically extended survival of mice in 2 syngeneic glioma models, GL261 and CT2A. The high targeting efficiency of αPD-L1-LNP to human TAMCs from GBM patients further validated the clinical relevance. Thus, this study establishes a therapeutic approach with immense potential to improve the clinical response in the treatment of GBM and warrants a rapid translation into clinical practice.


Brain Neoplasms/pathology , Glioblastoma/pathology , Myeloid Cells/pathology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cyclic N-Oxides , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Humans , Indolizines , Mice , Myeloid Cells/drug effects , Myeloid Cells/radiation effects , Nanoparticles , Pyridinium Compounds/administration & dosage , Pyridinium Compounds/therapeutic use , Tumor Microenvironment , Xenograft Model Antitumor Assays
12.
Oncogene ; 38(37): 6445-6460, 2019 09.
Article En | MEDLINE | ID: mdl-31324889

Overexpression of human epidermal growth factor receptor 2 (HER2) in breast cancer patients is associated with increased incidence of breast cancer brain metastases (BCBM), but the mechanisms underlying this phenomenon remain unclear. Here, to identify brain-predominant genes critical for the establishment of BCBM, we conducted an in silico screening analysis and identified that increased levels of fatty acid-binding protein 7 (FABP7) correlate with a lower survival and higher incidence of brain metastases in breast cancer patients. We validated these findings using HER2+ BCBM cells compared with parental breast cancer cells. Importantly, through knockdown and overexpression assays, we characterized the role of FABP7 in the BCBM process in vitro and in vivo. Our results uncover a key role of FABP7 in metabolic reprogramming of HER2 + breast cancer cells, supporting a glycolytic phenotype and storage of lipid droplets that enable their adaptation and survival in the brain microenvironment. In addition, FABP7 is shown to be required for upregulation of key metastatic genes and pathways, such as integrins-Src and VEGFA, and for the growth of HER2+ breast cancer cells in the brain microenvironment in vivo. Together, our results support FABP7 as a potential target for the treatment of HER2+ BCBM.


Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Fatty Acid-Binding Protein 7/physiology , Lipid Metabolism/genetics , Receptor, ErbB-2/genetics , Tumor Suppressor Proteins/physiology , Animals , Brain/metabolism , Brain/pathology , Brain Neoplasms/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Fatty Acid-Binding Protein 7/genetics , Female , Humans , Lipid Droplets/metabolism , Lipid Droplets/pathology , Mice , Oxidative Phosphorylation , Receptor, ErbB-2/metabolism , Tumor Microenvironment , Tumor Suppressor Proteins/genetics
13.
Cell Rep ; 27(1): 226-237.e4, 2019 04 02.
Article En | MEDLINE | ID: mdl-30943404

The mechanisms by which regulatory T cells (Tregs) migrate to and function within the hypoxic tumor microenvironment are unclear. Our studies indicate that specific ablation of hypoxia-inducible factor 1α (HIF-1α) in Tregs results in enhanced CD8+ T cell suppression versus wild-type Tregs under hypoxia, due to increased pyruvate import into the mitochondria. Importantly, HIF-1α-deficient Tregs are minimally affected by the inhibition of lipid oxidation, a fuel that is critical for Treg metabolism in tumors. Under hypoxia, HIF-1α directs glucose away from mitochondria, leaving Tregs dependent on fatty acids for mitochondrial metabolism within the hypoxic tumor. Indeed, inhibition of lipid oxidation enhances the survival of mice with glioma. Interestingly, HIF-1α-deficient-Treg mice exhibit significantly enhanced animal survival in a murine model of glioma, due to their stymied migratory capacity, explaining their reduced abundance in tumor-bearing mice. Thus HIF-1α acts as a metabolic switch for Tregs between glycolytic-driven migration and oxidative phosphorylation-driven immunosuppression.


Brain Neoplasms , Cell Movement/genetics , Energy Metabolism/genetics , Glioblastoma , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , T-Lymphocytes, Regulatory/immunology , Tumor Escape , Aged , Aged, 80 and over , Animals , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cells, Cultured , Female , Genes, Switch/physiology , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/metabolism , Glioblastoma/pathology , Glycolysis/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunosuppression Therapy , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Phosphorylation , T-Lymphocytes, Regulatory/metabolism , Tumor Escape/genetics , Tumor Escape/immunology , Tumor Microenvironment/genetics
14.
Mol Neurobiol ; 56(7): 5032-5040, 2019 Jul.
Article En | MEDLINE | ID: mdl-30460615

The immunosuppressive microenvironment is one of the major factors promoting the growth of glioblastoma multiforme (GBM). Infiltration of CD4+CD25+Foxp3+ regulatory T cells (Tregs) into the tumor microenvironment plays a significant role in the suppression of the anti-tumor immunity and portends a dismal prognosis for patients. Glioma-mediated secretion of chemo-attractant C-C motif ligand 2 and 22 (CCL2/22) has previously been shown by our group to promote Treg migration in vitro. In this study, we show that a local implantation of platelet-rich fibrin patch (PRF-P) into the brain of GL261 glioma-bearing mice prolonged the survival of affected animals by 42.85% (p = 0.0011). Analysis performed on brain tumor tissue harvested from PRF-P-treated mice revealed a specific decrease in intra-tumoral lymphocytes with a preferential depletion of immunosuppressive Tregs. Importantly, co-culture of GL261 or chemo-attractants (CCL2/22) with PRF-P abrogated Treg migration. Pharmacological blockade of the CCL2/22 interaction with their receptors potentiated the inhibitory effect of PRF-P on Tregs recruitment in culture. Moreover, our findings revealed the soluble CD40 ligand (sCD40L) as a major Treg inhibitory player produced by activated platelets entrapped within the fibrin matrix of the PRF-P. Blockade of sCD40L restored the migratory capacity of Tregs, emphasizing the role of PRF-P in preventing the Treg migration to glioma tissue. Our findings highlight autologous PRF-P as a personalized, Treg-selective suppression platform that can potentially supplement and enhance the efficacy of glioma therapies.


Autografts , Brain Neoplasms/therapy , Glioma/therapy , Platelet-Rich Fibrin/physiology , T-Lymphocytes, Regulatory/immunology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cells, Cultured , Craniotomy/methods , Glioma/immunology , Glioma/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Regulatory/metabolism , Tumor Microenvironment/immunology
15.
Neurotherapeutics ; 15(4): 1127-1138, 2018 10.
Article En | MEDLINE | ID: mdl-30027430

Antitumor immunotherapeutic strategies represent an especially promising set of approaches with rapid translational potential considering the dismal clinical context of high-grade gliomas. Dendritic cells (DCs) are the body's most professional antigen-presenting cells, able to recruit and activate T cells to stimulate an adaptive immune response. In this regard, specific loading of tumor-specific antigen onto dendritic cells potentially represents one of the most advanced strategies to achieve effective antitumor immunization. In this study, we developed a DC-specific adenoviral (Ad) vector, named Ad5scFvDEC205FF, targeting the DC surface receptor, DEC205. In vitro analysis shows that 60% of DCs was infected by this vector while the infectivity of other control adenoviral vectors was less than 10%, demonstrating superior infectivity on DCs. Moreover, an average of 14% of DCs were infected by Ad5scFvDEC205FF-GFP, while less than 3% of non-DCs were infected following in vivo administration, demonstrating highly selective in vivo DC infection. Importantly, vaccination with this vehicle expressing human glioma-specific antigen, Ad5scFvDEC205FF-CMV-IE, shows a prolonged survival benefit in GL261CMV-IE-implanted murine glioma models (p < 0.0007). Furthermore, when rechallenged, cancerous cells were completely rejected. In conclusion, our novel, viral-mediated, DC-based immunization approach has the significant therapeutic potential for patients with high-grade gliomas.


Adaptive Immunity/genetics , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Brain Neoplasms , Dendritic Cells/immunology , Glioma , Lectins, C-Type/metabolism , Minor Histocompatibility Antigens/metabolism , Receptors, Cell Surface/metabolism , Adenoviridae/genetics , Analysis of Variance , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Dendritic Cells/virology , Disease Models, Animal , Genetic Vectors/metabolism , Glioma/immunology , Glioma/pathology , Glioma/therapy , HEK293 Cells , Humans , Lymph Nodes/cytology , Mice , Spleen/cytology , Transduction, Genetic , Transfection , Xenograft Model Antitumor Assays
16.
Mol Ther ; 26(4): 986-995, 2018 04 04.
Article En | MEDLINE | ID: mdl-29503195

In order to fully harness the potential of immunotherapy with chimeric antigen receptor (CAR)-modified T cells, pre-clinical studies must be conducted in immunocompetent animal models that closely mimic the immunosuppressive malignant glioma (MG) microenvironment. Thus, the goal of this project was to study the in vivo fate of T cells expressing CARs specific for the MG antigen IL13Rα2 (IL13Rα2-CARs) in immunocompetent MG models. Murine T cells expressing IL13Rα2-CARs with a CD28.ζ (IL13Rα2-CAR.CD28.ζ) or truncated signaling domain (IL13Rα2-CAR.Δ) were generated by retroviral transduction, and their effector function was evaluated both in vitro and in vivo. IL13Rα2-CAR.CD28.ζ T cells' specificity toward IL13Rα2 was confirmed through cytokine production and cytolytic activity. In vivo, a single intratumoral injection of IL13Rα2-CAR.CD28.ζ T cells significantly extended the survival of IL13Rα2-expressing GL261 and SMA560 glioma-bearing mice; long-term survivors were resistant to re-challenge with IL13Rα2-negative and IL13Rα2-positive tumors. IL13Rα2-CAR.CD28.ζ T cells proliferated, produced cytokines (IFNγ, TNF-α), and promoted a phenotypically pro-inflammatory glioma microenvironment by inducing a significant increase in the number of CD4+ and CD8+ T cells and CD8α+ dendritic cells and a decrease in Ly6G+ myeloid-derived suppressor cells (MDSCs). Our data underline the significance of CAR T cell studies in immunocompetent hosts and further validate IL13Rα2-CAR T cells as an efficacious therapeutic strategy for MG.


Glioblastoma/immunology , Glioblastoma/metabolism , Immunotherapy, Adoptive , Interleukin-13 Receptor alpha2 Subunit/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , CD28 Antigens/immunology , CD28 Antigens/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Gene Expression , Genetic Vectors/genetics , Glioblastoma/genetics , Glioblastoma/therapy , Humans , Immunotherapy, Adoptive/methods , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-13 Receptor alpha2 Subunit/antagonists & inhibitors , Male , Mice , Receptors, Chimeric Antigen/genetics , Treatment Outcome , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
17.
Oncotarget ; 8(51): 89391-89405, 2017 Oct 24.
Article En | MEDLINE | ID: mdl-29179527

Glioblastoma is a highly aggressive malignant brain tumor with a poor prognosis and the median survival 14.6 months. Immunomodulatory proteins and oncolytic viruses represent two treatment approaches that have recently been developed for patients with glioblastoma that could extend patient survival and result in better treatment outcomes for patients with this disease. Together, these approaches could potentially augment the treatment efficacy and strength of these anti-tumor therapies. In addition to oncolytic activities, this combinatory approach introduces immunomodulation locally only where cancerous cells are present. This thereby results in the change of the tumor microenvironment from immune-suppressive to immune-vulnerable via activation of cytotoxic T cells or through the removal of glioma cells immune-suppressive capability. This review discusses the strengths and weaknesses of adenoviral oncolytic therapy, and highlights the genetic modifications that result in more effective and targeted viral agents. Additionally, the mechanism of action of immune-activating agents is described and the results of previous clinical trials utilizing these treatments in other solid tumors are reviewed. The feasibility, synergy, and limitations for treatments that combine these two approaches are outlined and areas for which more work is needed are considered.

19.
Proc Natl Acad Sci U S A ; 114(30): E6147-E6156, 2017 07 25.
Article En | MEDLINE | ID: mdl-28696296

Brain tumor-initiating cells (BTICs) have been identified as key contributors to therapy resistance, recurrence, and progression of diffuse gliomas, particularly glioblastoma (GBM). BTICs are elusive therapeutic targets that reside across the blood-brain barrier, underscoring the urgent need to develop novel therapeutic strategies. Additionally, intratumoral heterogeneity and adaptations to therapeutic pressure by BTICs impede the discovery of effective anti-BTIC therapies and limit the efficacy of individual gene targeting. Recent discoveries in the genetic and epigenetic determinants of BTIC tumorigenesis offer novel opportunities for RNAi-mediated targeting of BTICs. Here we show that BTIC growth arrest in vitro and in vivo is accomplished via concurrent siRNA knockdown of four transcription factors (SOX2, OLIG2, SALL2, and POU3F2) that drive the proneural BTIC phenotype delivered by multiplexed siRNA encapsulation in the lipopolymeric nanoparticle 7C1. Importantly, we demonstrate that 7C1 nano-encapsulation of multiplexed RNAi is a viable BTIC-targeting strategy when delivered directly in vivo in an established mouse brain tumor. Therapeutic potential was most evident via a convection-enhanced delivery method, which shows significant extension of median survival in two patient-derived BTIC xenograft mouse models of GBM. Our study suggests that there is potential advantage in multiplexed targeting strategies for BTICs and establishes a flexible nonviral gene therapy platform with the capacity to channel multiplexed RNAi schemes to address the challenges posed by tumor heterogeneity.


Glioblastoma/pathology , Nanoparticles/therapeutic use , RNA Interference , Animals , Carcinogenesis/genetics , Drug Resistance, Neoplasm , Female , Genetic Therapy/methods , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Male , Mice , Mice, Nude , Xenograft Model Antitumor Assays
20.
Mol Ther Oncolytics ; 5: 97-104, 2017 Jun 16.
Article En | MEDLINE | ID: mdl-28573184

Oncolytic virotherapy is a treatment approach with increasing clinical relevance, as indicated by the marked survival benefit seen in animal models and its current exploration in human patients with cancer. The use of an adenovirus vector for this therapeutic modality is common, has significant clinical benefit in animals, and its efficacy has recently been linked to an anti-tumor immune response that occurs following tumor antigen presentation. Here, we analyzed the adaptive immune system's response following viral infection by comparing replication-incompetent and replication-competent adenoviral vectors. Our findings suggest that cell death caused by replication-competent adenoviral vectors is required to induce a significant anti-tumor immune response and survival benefits in immunocompetent mice bearing intracranial glioma. We observed significant changes in the repertoire of immune cells in the brain and draining lymph nodes and significant recruitment of CD103+ dendritic cells (DCs) in response to oncolytic adenoviral therapy, suggesting the active role of the immune system in anti-tumor response. Our data suggest that the response to oncolytic virotherapy is accompanied by local and systemic immune responses and should be taken in consideration in the future design of the clinical studies evaluating oncolytic virotherapy in patients with glioblastoma multiforme (GBM).

...