Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Cancer Res ; 81(23): 6044-6057, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34645608

ABSTRACT

CD271 (NGFR) is a neurotrophin receptor that belongs to the tumor necrosis receptor (TNFR) family. Upon ligand binding, CD271 can mediate either survival or cell death. Although the role of CD271 as a marker of tumor-initiating cells is still a matter of debate, its role in melanoma progression has been well documented. Moreover, CD271 has been shown to be upregulated after exposure to both chemotherapy and targeted therapy. In this study, we demonstrate that activation of CD271 by a short ß-amyloid-derived peptide (Aß(25-35)) in combination with either chemotherapy or MAPK inhibitors induces apoptosis in 2D and 3D cultures of eight melanoma cell lines. This combinatorial treatment significantly reduced metastasis in a zebrafish xenograft model and led to significantly decreased tumor volume in mice. Administration of Aß(25-35) in ex vivo tumors from immunotherapy- and targeted therapy-resistant patients significantly reduced proliferation of melanoma cells, showing that activation of CD271 can overcome drug resistance. Aß(25-35) was specific to CD271-expressing cells and induced CD271 cleavage and phosphorylation of JNK (pJNK). The direct protein-protein interaction of pJNK with CD271 led to PARP1 cleavage, p53 and caspase activation, and pJNK-dependent cell death. Aß(25-35) also mediated mitochondrial reactive oxygen species (mROS) accumulation, which induced CD271 overexpression. Finally, CD271 upregulation inhibited mROS production, revealing the presence of a negative feedback loop in mROS regulation. These results indicate that targeting CD271 can activate cell death pathways to inhibit melanoma progression and potentially overcome resistance to targeted therapy. SIGNIFICANCE: The discovery of a means to specifically activate the CD271 death domain reveals unknown pathways mediated by the receptor and highlights new treatment possibilities for melanoma.


Subject(s)
Amyloid beta-Peptides/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Melanoma/drug therapy , Molecular Targeted Therapy , Nerve Tissue Proteins/agonists , Receptors, Nerve Growth Factor/agonists , Animals , Apoptosis , Cell Proliferation , Drug Therapy, Combination , Female , Humans , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Zebrafish
2.
Cells ; 10(8)2021 07 23.
Article in English | MEDLINE | ID: mdl-34440640

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disorder and results in severe neurodegeneration and progressive cognitive decline. Neurotrophins are growth factors involved in the development and survival of neurons, but also in underlying mechanisms for memory formation such as hippocampal long-term potentiation. Our aim was to identify small molecules with stimulatory effects on the signaling of two neurotrophins, the nerve growth factor (NGF) and the brain derived neurotrophic factor (BDNF). To identify molecules that could potentiate neurotrophin signaling, 25,000 molecules were screened, which led to the identification of the triazinetrione derivatives ACD855 (Ponazuril) and later on ACD856, as positive allosteric modulators of tropomyosin related kinase (Trk) receptors. ACD855 or ACD856 potentiated the cellular signaling of the neurotrophin receptors with EC50 values of 1.9 and 3.2 or 0.38 and 0.30 µM, respectively, for TrkA or TrkB. ACD855 increased acetylcholine levels in the hippocampus by 40% and facilitated long term potentiation in rat brain slices. The compounds acted as cognitive enhancers in a TrkB-dependent manner in several different behavioral models. Finally, the age-induced cognitive dysfunction in 18-month-old mice could be restored to the same level as found in 2-month-old mice after a single treatment of ACD856. We have identified a novel mechanism to modulate the activity of the Trk-receptors. The identification of the positive allosteric modulators of the Trk-receptors might have implications for the treatment of Alzheimer's diseases and other diseases characterized by cognitive impairment.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Cognition/drug effects , Cognitive Dysfunction/drug therapy , Nootropic Agents/pharmacology , Receptors, Nerve Growth Factor/agonists , Age Factors , Animals , Brain/enzymology , Brain/physiopathology , Cell Line, Tumor , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Disease Models, Animal , Humans , Male , Maze Learning/drug effects , Membrane Glycoproteins , Mice, Inbred C57BL , Motor Activity/drug effects , Protein-Tyrosine Kinases , Rats, Sprague-Dawley , Receptor, trkA/agonists , Receptor, trkA/metabolism , Receptor, trkB/agonists , Receptor, trkB/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Small Molecule Libraries , Triazines/pharmacology
3.
Pharmacol Res ; 121: 129-137, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28438600

ABSTRACT

In the last few years, exciting properties have emerged regarding the activation, signaling, mechanisms of action, and therapeutic targeting of the two types of neurotrophin receptors: the p75NTR with its intracellular and extracellular peptides, the Trks, their precursors and their complexes. This review summarizes these new developments, with particular focus on neurodegenerative diseases. Based on the evolving knowledge, innovative concepts have been formulated regarding the pathogenesis of these diseases, especially the Alzheimer's and two other, the Parkinson's and Huntington's diseases. The medical progresses include original procedures of diagnosis, started from studies in mice and now investigated for human application, based on innovative classes of receptor agonists and blockers. In parallel, comprehensive studies have been and are being carried out for the development of drugs. The relevance of these studies is based on the limitations of the therapies employed until recently, especially for the treatment of Alzheimer's patients. Starting from well known drugs, previously employed for non-neurodegenerative diseases, the ongoing progress has lead to the development of small molecules that cross rapidly the blood-brain barrier. Among these molecules the most promising are specific blockers of the p75NTR receptor. Additional drugs, that activate Trk receptors, were shown effective against synaptic loss and memory deficits. In the near future such approaches, coordinated with treatments with monoclonal antibodies and with developments in the microRNA field, are expected to improve the therapy of neurodegenerative diseases, and may be relevant also for other human disease conditions.


Subject(s)
Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Receptors, Nerve Growth Factor/metabolism , Animals , Drug Discovery , Humans , Molecular Targeted Therapy , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/drug therapy , Receptors, Nerve Growth Factor/agonists , Receptors, Nerve Growth Factor/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
4.
Vitam Horm ; 104: 263-311, 2017.
Article in English | MEDLINE | ID: mdl-28215298

ABSTRACT

Age-related neurodegenerative diseases, such as Alzheimer's disease, will represent one of the largest future burdens on worldwide healthcare systems due to the increasing proportion of elderly in our society. As deficiencies in neurotrophins are implicated in the pathogenesis of many age-related neurodegenerative disorders, it is reasonable to consider that global neurotrophin resistance may also become a major healthcare threat. Central nervous system networks are effectively maintained through aging by neuroprotective and neuroplasticity signaling mechanisms which are predominantly controlled by neurotrophin receptor signaling. Neurotrophin receptors are single pass receptor tyrosine kinases that form dimeric structures upon ligand binding to initiate cellular signaling events that control many protective and plasticity-related pathways. Declining functionality of the neurotrophin ligand-receptor system is considered one of the hallmarks of neuropathological aging. Therefore, it is imperative to develop effective therapeutic strategies to contend with this significant issue. While the therapeutic applications of cognate ligands for neurotrophin receptors are limited, the development of nonpeptidergic, small-molecule ligands can overcome these limitations, and productively regulate this important receptor system with beneficial effects. Using our advanced knowledge of the high-dimensionality complexity of receptor systems, the future generation of precision medicines targeting these systems will be an attainable goal.


Subject(s)
Drug Design , Drugs, Investigational/therapeutic use , Nerve Growth Factors/therapeutic use , Neurodegenerative Diseases/drug therapy , Protein Precursors/therapeutic use , Receptors, Nerve Growth Factor/agonists , Animals , Central Nervous System/drug effects , Central Nervous System/growth & development , Central Nervous System/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/prevention & control , Dimerization , Drugs, Investigational/chemistry , Humans , Ligands , Molecular Targeted Therapy , Nerve Growth Factors/chemistry , Nerve Growth Factors/metabolism , Neurodegenerative Diseases/metabolism , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Neuroprotection/drug effects , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Neuroprotective Agents/therapeutic use , Nootropic Agents/chemistry , Nootropic Agents/metabolism , Nootropic Agents/therapeutic use , Protein Precursors/chemistry , Protein Precursors/metabolism , Receptors, Nerve Growth Factor/metabolism , Signal Transduction/drug effects
5.
Vitam Horm ; 104: 313-341, 2017.
Article in English | MEDLINE | ID: mdl-28215300

ABSTRACT

Allergic inflammation is the result of a specific pattern of cellular and humoral responses leading to the activation of the innate and adaptive immune system, which, in turn, results in physiological and structural changes affecting target tissues such as the airways and the skin. Eosinophil activation and the production of soluble mediators such as IgE antibodies are pivotal features in the pathophysiology of allergic diseases. In the past few years, however, convincing evidence has shown that neurons and other neurosensory structures are not only a target of the inflammatory process but also participate in the regulation of immune responses by actively releasing soluble mediators. The main products of these activated sensory neurons are a family of protein growth factors called neurotrophins. They were first isolated in the central nervous system and identified as important factors for the survival and differentiation of neurons during fetal and postnatal development as well as neuronal maintenance later in life. Four members of this family have been identified and well defined: nerve growth factor, brain-derived neurotrophic factor, neurotrophin 3, and neurotrophin 4/5. Neurotrophins play a critical role in the bidirectional signaling mechanisms between immune cells and the neurosensory network structures in the airways and the skin. Pruritus and airway hyperresponsiveness, two major features of atopic dermatitis and asthma, respectively, are associated with the disruption of the neurosensory network activities. In this chapter, we provide a comprehensive description of the neuroimmune interactions underlying the pathophysiological mechanisms of allergic and inflammatory diseases.


Subject(s)
Hypersensitivity/metabolism , Models, Immunological , Models, Neurological , Nerve Growth Factors/metabolism , Neurogenic Inflammation/metabolism , Neurons/metabolism , Receptors, Nerve Growth Factor/agonists , Animals , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , Humans , Hypersensitivity/immunology , Hypersensitivity/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/pathology , Neurogenic Inflammation/immunology , Neurogenic Inflammation/pathology , Neurons/immunology , Neurons/pathology , Receptors, Nerve Growth Factor/metabolism
6.
Vitam Horm ; 104: 367-404, 2017.
Article in English | MEDLINE | ID: mdl-28215302

ABSTRACT

The p75 neurotrophin receptor (p75NTR, a.k.a. CD271), a transmembrane glycoprotein and a member of the tumor necrosis family (TNF) of receptors, was originally identified as a nerve growth factor receptor in the mid-1980s. While p75NTR is recognized to have important roles during neural development, its presence in both neural and nonneural tissues clearly supports the potential to mediate a broad range of functions depending on cellular context. Using an unbiased in vivo selection paradigm for genes underlying the invasive behavior of glioma, a critical characteristic that contributes to poor clinical outcome for glioma patients, we identified p75NTR as a central regulator of glioma invasion. Herein we review the expanding role that p75NTR plays in glioma progression with an emphasis on how p75NTR may contribute to the treatment refractory nature of glioma. Based on the observation that p75NTR is expressed and functional in two critical glioma disease reservoirs, namely, the highly infiltrative cells that evade surgical resection, and the radiation- and chemotherapy-resistant brain tumor-initiating cells (also referred to as brain tumor stem cells), we propose that p75NTR and its myriad of downstream signaling effectors represent rationale therapeutic targets for this devastating disease. Lastly, we provide the provocative hypothesis that, in addition to the well-documented cell autonomous signaling functions, the neurotrophins, and their respective receptors, contribute in a cell nonautonomous manner to drive the complex cellular and molecular composition of the brain tumor microenvironment, an environment that fuels tumorigenesis.


Subject(s)
Carcinogenesis/metabolism , Glioma/metabolism , Models, Neurological , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/agonists , Receptors, Nerve Growth Factor/agonists , Signal Transduction , Animals , Antineoplastic Agents/therapeutic use , Carcinogenesis/pathology , Drug Resistance, Neoplasm , Glioma/drug therapy , Glioma/immunology , Glioma/pathology , Humans , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Microglia/cytology , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Neoplasm Invasiveness , Neoplasm Proteins/agonists , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/immunology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Receptors, Nerve Growth Factor/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects
7.
Vitam Horm ; 104: 459-473, 2017.
Article in English | MEDLINE | ID: mdl-28215304

ABSTRACT

Neurotrophins (NTs) have been implicated in generation and modulation of nociceptive pathways. Change in NTs levels is associated with painful conditions and neurological diseases such as migraine. Currently, it is generally recognized that migraine headaches result from the activation and sensitization of trigeminal sensory afferent fibers leading to neuropeptides release such as calcitonin gene-related peptide (CGRP) and substance P (SP). This triggers an inflammatory cascade causing a neurogenic inflammation. The agents responsible for trigeminal activation and release of neuropeptides are still unclear. It is known that the transient receptor potential vanilloid receptor-1 (TRPV1) is an important mediator of CGRP and SP release. TRPV1 is closely associated with tyrosine receptors kinases (Trk), which are NTs receptors. NTs can act on TRPV1 increasing its sensitivity to painful stimuli, therefore predisposing to hyperalgesia. Upregulation of ion channels and pain receptors in dorsal root ganglion neurons may be alternative mechanisms by which NTs contribute to pain development. Only a few studies have been performed to investigate the role of NTs in migraine. These studies have reported changes in NTs levels in migraine patients either during the migraine attack or in free-headache periods.


Subject(s)
Brain/metabolism , Migraine Disorders/metabolism , Models, Neurological , Nerve Growth Factors/metabolism , Neurons/metabolism , Receptors, Nerve Growth Factor/agonists , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Brain/drug effects , Brain/immunology , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , Ganglia, Spinal/drug effects , Ganglia, Spinal/immunology , Ganglia, Spinal/metabolism , Humans , Migraine Disorders/drug therapy , Migraine Disorders/immunology , Migraine Disorders/physiopathology , Nerve Growth Factors/antagonists & inhibitors , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurogenic Inflammation/etiology , Neurogenic Inflammation/prevention & control , Neurons/drug effects , Neurons/immunology , Neurons, Afferent/drug effects , Neurons, Afferent/immunology , Neurons, Afferent/metabolism , Nociceptive Pain/etiology , Nociceptive Pain/prevention & control , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Nerve Growth Factor/metabolism , Signal Transduction/drug effects , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism
8.
Vitam Horm ; 104: 497-531, 2017.
Article in English | MEDLINE | ID: mdl-28215306

ABSTRACT

In this chapter, we parse the literature on neurotrophins that have been implicated in the pathogenesis of perineural invasion (PNI) in select lineage-unrelated malignancies. We also detail evidence linking neurotrophins and their receptors (TrkA, RET, p75NGFR, and NCAM) to the pathogenesis of PNI in desmoplastic melanoma and cutaneous squamous cell carcinoma-both malignancies with an established propensity for PNI. Lastly, the clinical potential of neurotrophins as receptors for targeted therapies is explored.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Melanoma/metabolism , Models, Neurological , Nerve Growth Factors/metabolism , Peripheral Nerves/metabolism , Receptors, Nerve Growth Factor/agonists , Skin Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , Humans , Melanoma/diagnosis , Melanoma/drug therapy , Melanoma/pathology , Molecular Targeted Therapy , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Nerve Growth Factors/antagonists & inhibitors , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Peripheral Nerves/drug effects , Peripheral Nerves/pathology , Prognosis , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Nerve Growth Factor/metabolism , Schwann Cells/drug effects , Schwann Cells/metabolism , Schwann Cells/pathology , Signal Transduction/drug effects , Skin Neoplasms/diagnosis , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology
9.
Neurobiol Dis ; 97(Pt B): 139-155, 2017 01.
Article in English | MEDLINE | ID: mdl-27546056

ABSTRACT

Neurodegenerative disorders are prevalent, complex and devastating conditions, with very limited treatment options currently available. While they manifest in many forms, there are commonalities that link them together. In this review, we will focus on neurotrophins - a family of related factors involved in neuronal development and maintenance. Neurodegenerative diseases often present with a neurotrophin imbalance, in which there may be decreases in trophic signaling through Trk receptors for example, and/or increases in pro-apoptotic activity through p75. Clinical trials with neurotrophins have continuously failed due to their poor pharmacological properties as well as the unavoidable activation of p75. Thus, there is a need for drugs without such setbacks. Small molecule neurotrophin mimetics are favorable options since they can selectively activate Trks or inactivate p75. In this review, we will initially present a brief outline of how these molecules are synthesized and their mechanisms of action; followed by an update in the current state of neurotrophins and small molecules in major neurodegenerative diseases. Although there has been significant progress in the development of potential therapeutics, more studies are needed to establish clear mechanisms of action and target specificity in order to transition from animal models to the assessment of safety and use in humans.


Subject(s)
Neurodegenerative Diseases/drug therapy , Receptors, Nerve Growth Factor/agonists , Receptors, Nerve Growth Factor/antagonists & inhibitors , Animals , Humans , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Receptors, Nerve Growth Factor/metabolism
10.
Oncotarget ; 7(49): 81026-81048, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27835587

ABSTRACT

Nerve growth factor (NGF)/nerve growth factor receptors (NGFRs) axis and canonical WNT/ß-catenin pathway have shown to play crucial roles in tumor initiation, progression and prognosis. But little did we know the relationship between them in modulation of tumor progress. In this report, we found that NGF/NGFRs and ß-catenin were coexpression in ovarian cancer cell lines, and NGF can decrease the expression level of ß-catenin and affect its activities, which may be related to the NGF-induced down-regulation of B-cell CLL/lymphoma 9-like (BCL9L, BCL9-2). Furthermore, NGF can also increase or decrease the downstream target gene expression levels of WNT/ß-catenin depending on the cell types. Especially, we created a novel in vitro cell growth model based on a microfluidic device to intuitively observe the effects of NGF/NGFRs on the motility behaviors of ovarian cancer cells. The results showed that the migration area and maximum distance into three dimensional (3D) matrigel were decreased in CAOV3 and OVCAR3 cells, but increased in SKOV3 cells following the stimulation with NGF. In addition, we found that the cell colony area was down-regulated in CAOV3 cells, however, it was augmented in OVCAR3 cells after treatment with NGF. The inhibitors of NGF/NGFRs, such as Ro 08-2750, K252a and LM11A-31,can all block NGF-stimulated changes of gene expression or migratory behavior on ovarian cancer cells. The different results among ovarian cancer cells illustrated the heterogeneity and complexity of ovarian cancer. Collectively, our results suggested for the first time that NGF is functionally linked to ß-catenin in the migration of human ovarian cancer cells, which may be a novel therapeutic perspective to prevent the spread of ovarian carcinomas by studying the interaction between NGF/NGFRs and canonical WNT/ß-catenin signaling.


Subject(s)
Cell Movement/drug effects , Nerve Growth Factor/pharmacology , Ovarian Neoplasms/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Female , Humans , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Neoplasm Invasiveness , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phosphorylation , Receptors, Nerve Growth Factor/agonists , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Time Factors , Transcription Factors/metabolism , beta Catenin/genetics
11.
PLoS One ; 8(11): e80113, 2013.
Article in English | MEDLINE | ID: mdl-24244623

ABSTRACT

Nerve growth factor is a neurotrophin that promotes cell growth, differentiation, survival and death through two different receptors: TrkA(NTR) and p75(NTR). Nerve growth factor serum concentrations increase during many inflammatory and autoimmune diseases, glomerulonephritis, chronic kidney disease, end-stage renal disease and, particularly, in renal transplant. Considering that nerve growth factor exerts beneficial effects in the treatment of major central and peripheral neurodegenerative diseases, skin and corneal ulcers, we asked whether nerve growth factor could also exert a role in Cyclosporine A-induced graft nephrotoxicity. Our hypothesis was raised from basic evidence indicating that Cyclosporine A-inhibition of calcineurin-NFAT pathway increases nerve growth factor expression levels. Therefore, we investigated the involvement of nerve growth factor and its receptors in the damage exerted by Cyclosporine A in tubular renal cells, HK-2. Our results showed that in HK-2 cells combined treatment with Cyclosporine A + nerve growth factor induced a significant reduction in cell vitality concomitant with a down-regulation of Cyclin D1 and up-regulation of p21 levels respect to cells treated with Cyclosporine A alone. Moreover functional experiments showed that the co-treatment significantly up-regulated human p21promoter activity by involvement of the Sp1 transcription factor, whose nuclear content was negatively regulated by activated NFATc1. In addition we observed that the combined exposure to Cyclosporine A + nerve growth factor promoted an up-regulation of p75 (NTR) and its target genes, p53 and BAD leading to the activation of intrinsic apoptosis. Finally, the chemical inhibition of p75(NTR) down-regulated the intrinsic apoptotic signal. We describe two new mechanisms by which nerve growth factor promotes growth arrest and apoptosis in tubular renal cells exposed to Cyclosporine A.


Subject(s)
Apoptosis/drug effects , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Kidney Tubules, Proximal/drug effects , Nerve Growth Factor/pharmacology , Cell Line , Cell Survival/drug effects , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Cyclin D1/metabolism , Drug Synergism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , NFATC Transcription Factors/agonists , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic , Receptors, Nerve Growth Factor/agonists , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Tumor Suppressor Protein p53/agonists , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-Associated Death Protein/agonists , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
12.
Anticancer Drugs ; 24(9): 899-910, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23872911

ABSTRACT

Nerve growth factor (NGF) receptors, TrKA and p75(NTR), are being investigated in cancer therapy. Our previous data show that, in HTB114 uterine leiomyosarcoma cells, p75(NTR)-dependent apoptosis is inducible by cytotoxic drugs and can suppress nerve growth factor-dependent growth. Although amitriptyline can kill cancer cells and bind TrKA/B, its effects on p75-dependent apoptosis are unknown. The aim of this paper was to evaluate the antineoplastic potential of amitriptyline, and the role of p75(NTR)-dependent apoptosis in the chemoresistant uterine HTB114 leiomyosarcoma. Using proliferation assays and fluorescence-activated cell sorting analysis, we found that amitriptyline caused a marked reduction in HTB114 cell viability, associated with the parallel upregulation of p75(NTR) expression. This converted the TrKA⁺-proliferating cells into TrKA⁺/p75(NTR⁺), leading to downregulation of TrKA-prosurvival signaling (AKT) and activation of p75(NTR)-dependent apoptosis (through caspase-3). Overall, we provide novel evidence that HTB114 uterine leiomyosarcoma cells are highly sensitive to amitriptyline, supporting the role of p75(NTR)-dependent apoptosis as a novel cytotoxic mechanism of this drug and of p75(NTR) as an inducible stress receptor and a novel target in clinical oncology.


Subject(s)
Amitriptyline/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Leiomyosarcoma/drug therapy , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Uterine Neoplasms/drug therapy , Caspase 3/chemistry , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Enzyme Activation/drug effects , Female , Humans , Leiomyosarcoma/metabolism , Molecular Targeted Therapy , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/agonists , Receptors, Nerve Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Up-Regulation/drug effects , Uterine Neoplasms/metabolism
13.
Anesthesiology ; 116(2): 352-61, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22198221

ABSTRACT

BACKGROUND: Propofol exposure to neurons during synaptogenesis results in apoptosis, leading to cognitive dysfunction in adulthood. Previous work from our laboratory showed that isoflurane neurotoxicity occurs through p75 neurotrophin receptor (p75(NTR)) and subsequent cytoskeleton depolymerization. Given that isoflurane and propofol both suppress neuronal activity, we hypothesized that propofol also induces apoptosis in developing neurons through p75(NTR). METHODS: Days in vitro 5-7 neurons were exposed to propofol (3 µM) for 6 h and apoptosis was assessed by cleaved caspase-3 (Cl-Csp3) immunoblot and immunofluorescence microscopy. Primary neurons from p75(NTR-/-) mice or wild-type neurons were treated with propofol, with or without pretreatment with TAT-Pep5 (10 µM, 15 min), a specific p75(NTR) inhibitor. P75(NTR-/-) neurons were transfected for 72 h with a lentiviral vector containing the synapsin-driven p75(NTR) gene (Syn-p75(NTR)) or control vector (Syn-green fluorescent protein) before propofol. To confirm our in vitro findings, wild-type mice and p75(NTR-/-) mice (PND5) were pretreated with either TAT-Pep5 or TAT-ctrl followed by propofol for 6 h. RESULTS: Neurons exposed to propofol showed a significant increase in Cl-Csp3, an effect attenuated by TAT-Pep5 and hydroxyfasudil. Apoptosis was significantly attenuated in p75(NTR-/-) neurons. In p75(NTR-/-) neurons transfected with Syn-p75(NTR), propofol significantly increased Cl-Csp3 in comparison with Syn-green fluorescent protein-transfected p75(NTR-/-) neurons. Wild-type mice exposed to propofol exhibited increased Cl-Csp3 in the hippocampus, an effect attenuated by TAT-Pep5. By contrast, propofol did not induce apoptosis in p75(NTR-/-) mice. CONCLUSION: These results demonstrate that propofol induces apoptosis in developing neurons in vivo and in vitro and implicate a role for p75(NTR) and the downstream effector RhoA kinase.


Subject(s)
Apoptosis/drug effects , Neurons/drug effects , Neurons/metabolism , Propofol/toxicity , Receptors, Nerve Growth Factor/metabolism , Animals , Animals, Newborn , Apoptosis/physiology , Cells, Cultured , Mice , Mice, Inbred BALB C , Mice, Knockout , Neurons/enzymology , Receptors, Nerve Growth Factor/agonists , Receptors, Nerve Growth Factor/physiology , rhoA GTP-Binding Protein/physiology
14.
Curr Top Microbiol Immunol ; 344: 211-44, 2011.
Article in English | MEDLINE | ID: mdl-20563707

ABSTRACT

The rational manipulation of antigen-specific T cells to reignite a tumor-specific immune response in cancer patients is a challenge for cancer immunotherapy. Targeting coinhibitory and costimulatory T cell receptors with specific antibodies in cancer patients is an emerging approach to T cell manipulation, namely "immune modulation." Cytotoxic T-lymphocyte antigen-4 (CTLA-4) and glucocorticoid-induced tumor necrosis factor family receptor (GITR) are potential targets for immune modulation through anti-CTLA-4 blocking antibodies and anti-GITR agonistic antibodies, respectively. In this review, we first discuss preclinical findings key to the understanding of the mechanisms of action of these immunomodulatory antibodies and the preclinical evidence of antitumor activity which preceded translation into the clinic. We next describe the outcomes and immune related adverse effects associated with anti-CTLA-4 based clinical trials with particular emphasis on specific biomarkers used to elucidate the mechanisms of tumor immunity in patients. The experience with anti-CTLA-4 therapy and the durable clinical benefit observed provide proof of principle to effective antitumor immune modulation and the promise of future clinical immune modulatory antibodies.


Subject(s)
Antigens, CD/physiology , Immunotherapy , Neoplasms/therapy , Receptors, Nerve Growth Factor/agonists , Receptors, Tumor Necrosis Factor/agonists , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , CTLA-4 Antigen , Glucocorticoid-Induced TNFR-Related Protein , Humans , Ipilimumab , Lymphocyte Activation , Neoplasms/immunology
15.
J Immunother ; 33(8): 789-97, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20842058

ABSTRACT

An agonistic antibody DTA-1, to glucocorticoid-induced TNFR-related protein (GITR), induces T-cell activation and antitumor immunity. CD4(+) effector T cells are essential in initiating GITR-induced immune activation, and the sequentially activated cytolytic CD8(+) T cells are sufficient to induce tumor rejection. Administration of DTA-1 to a tumor-bearing mouse also induces B-cell activation illustrated by CD69 expression. Substantial evidence suggests that resting B cells are tumor promoting, which has prompted the idea of B-cell depletion by Rituximab, to be combined with other agents in the clinic to augment antitumor response. In this study, we have found that mature B cells are needed for the mechanism of anti-GITR agonist to kill tumors. The treatment of GITR agonist induces profound B-cell activation, differentiation, and antibody production. In a mature B-cell-deficient mouse (JHD), DTA-1 fails to induce tumor regression with a reduced early activation of CD4(+) and CD8(+) T cells. B-cell deficiency disables the capability of the DTA-1 in generating cytolytic CD8(+) T cells and significantly reduces the cytokine production in tumor bearing mice. The tumor-killing activities of DTA-1 are still present albeit reduced in the CD40(-/-) mice, in which IgG production is impaired. We have also shown that the dependence on B cells to kill tumors differentiates GITR costimulation from CTLA4 blockade and OX40 agonism in tumor immunotherapy. The findings underscore the reciprocal T-cell-B-cell interaction to enhance antitumor immunity upon GITR costimulation. The results provide the insight that attenuating B-cell functions may not be beneficial in cancer immunotherapy based on GITR agonism.


Subject(s)
Antibodies, Monoclonal/administration & dosage , B-Lymphocytes/metabolism , Colorectal Neoplasms/immunology , Immunotherapy , T-Lymphocytes/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibody Formation/genetics , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD40 Antigens/genetics , Cell Communication , Cell Differentiation , Cell Line, Tumor , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Cytotoxicity, Immunologic/genetics , Female , Glucocorticoid-Induced TNFR-Related Protein , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, Nerve Growth Factor/agonists , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/agonists , Receptors, Tumor Necrosis Factor/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
16.
PLoS One ; 5(5): e10436, 2010 May 03.
Article in English | MEDLINE | ID: mdl-20454651

ABSTRACT

In vivo GITR ligation has previously been shown to augment T-cell-mediated anti-tumor immunity, yet the underlying mechanisms of this activity, particularly its in vivo effects on CD4+ foxp3+ regulatory T cells (Tregs), have not been fully elucidated. In order to translate this immunotherapeutic approach to the clinic it is important gain better understanding of its mechanism(s) of action. Utilizing the agonist anti-GITR monoclonal antibody DTA-1, we found that in vivo GITR ligation modulates regulatory T cells (Tregs) directly during induction of melanoma tumor immunity. As a monotherapy, DTA-1 induced regression of small established B16 melanoma tumors. Although DTA-1 did not alter systemic Treg frequencies nor abrogate the intrinsic suppressive activity of Tregs within the tumor-draining lymph node, intra-tumor Treg accumulation was significantly impaired. This resulted in a greater Teff:Treg ratio and enhanced tumor-specific CD8+ T-cell activity. The decreased intra-tumor Treg accumulation was due both to impaired infiltration, coupled with DTA-1-induced loss of foxp3 expression in intra-tumor Tregs. Histological analysis of B16 tumors grown in Foxp3-GFP mice showed that the majority of GFP+ cells had lost Foxp3 expression. These "unstable" Tregs were absent in IgG-treated tumors and in DTA-1 treated TDLN, demonstrating a tumor-specific effect. Impairment of Treg infiltration was lost if Tregs were GITR(-/-), and the protective effects of DTA-1 were reduced in reconstituted RAG1(-/-) mice if either the Treg or Teff subset were GITR-negative and absent if both were negative. Our results demonstrate that DTA-1 modulates both Teffs and Tregs during effective tumor treatment. The data suggest that DTA-1 prevents intra-tumor Treg accumulation by altering their stability, and as a result of the loss of foxp3 expression, may modify their intra-tumor suppressive capacity. These findings provide further support for the continued development of agonist anti-GITR mAbs as an immunotherapeutic strategy for cancer.


Subject(s)
Antibodies, Monoclonal/immunology , Immunity/immunology , Melanoma/immunology , Receptors, Nerve Growth Factor/agonists , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/agonists , Receptors, Tumor Necrosis Factor/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation , Forkhead Transcription Factors/metabolism , Glucocorticoid-Induced TNFR-Related Protein , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Up-Regulation/immunology
17.
J Immunother ; 33(2): 136-45, 2010.
Article in English | MEDLINE | ID: mdl-20145549

ABSTRACT

We generated an adenovirus-based T-cell vaccine (Ad-p14) that reliably elicits T-cell responses to human papillomavirus (HPV) oncogenes of the 2 most common high-risk HPV serotypes. The artificial gene used to create the vaccine comprising 415 aa (1248 bp) was cloned by fusing 14 polymerase chain reaction fragments of HPV16 and HPV18 E6 and E7 oncogenes devoid of sequences with transforming potential. Although ensuring maximal biologic safety, the construct includes approximately 70% of the relevant T-cell epitopes. In a tumor model for cervical cancer (C3), therapeutic vaccination led to complete eradication in 100% of the mice. In a second model (TC1), it induced initial tumor mass reduction, but 90% of the animals showed delayed tumor progression. To further improve the therapeutic effect, vaccination was combined with systemic application of imiquimod, anti-CD4, alpha-interferon, or anti-GITR. Although adding alpha-interferon improved the therapeutic potential of Ad-p14 by 40%, the combination with anti-GITR resulted in complete and permanent eradication of all TC1 tumors. Ad-p14 has clinical potential for treating HPV-induced lesions, and the added effect of immune response modifiers stresses the importance of combined protocols for immunotherapy of malignant tumors.


Subject(s)
Cancer Vaccines , Fibrosarcoma/therapy , Human papillomavirus 16/immunology , Human papillomavirus 18/immunology , Uterine Cervical Neoplasms/therapy , Adenoviridae/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Female , Fibrosarcoma/immunology , Fibrosarcoma/pathology , Forkhead Transcription Factors/biosynthesis , Genetic Engineering , Glucocorticoid-Induced TNFR-Related Protein , Human papillomavirus 16/genetics , Human papillomavirus 18/genetics , Humans , Interferon-gamma/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/metabolism , Receptors, Nerve Growth Factor/agonists , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/agonists , Receptors, Tumor Necrosis Factor/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Burden/drug effects , Tumor Burden/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology
18.
J Neurosci Res ; 88(7): 1406-19, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20029969

ABSTRACT

We use immunohistochemistry to describe the localization of brain-derived neurotrophic factor (BDNF) and its receptors trkB and p75(NTR) in the neuromuscular synapses of postnatal rats (P6-P7) during the synapse elimination period. The receptor protein p75(NTR) is present in the nerve terminal, muscle cell and glial Schwann cell whereas BDNF and trkB proteins can be detected mainly in the pre- and postsynaptic elements. Exogenously applied BDNF (10 nM for 3 hr or 50 nM for 1 hr) increases ACh release from singly and dually innervated synapses. This effect may be specific for BDNF because the neurotrophin NT-4 (2-8 nM) does not modulate release at P6-P7. Blocking the receptors trkB and p75(NTR) (with K-252a and anti-p75-192-IgG, respectively) completely abolishes the potentiating effect of exogenous BDNF. In addition, exogenous BDNF transiently recruits functionally depressed silent terminals, and this effect seems to be mediated by trkB. Calcium ions, the L-type voltage-dependent calcium channels and protein kinase C are involved in BDNF-mediated nerve ending recruitment. Blocking experiments suggest that endogenous BDNF could operate through p75(NTR) receptors coupled to potentiate ACh release in all nerve terminals because the anti-p75-192-IgG reduces release. However, blocking the trkB receptor (K-252a) or neutralizing endogenous BDNF with the trkB-IgG fusion protein reveals a trkB-mediated release inhibition on almost mature strong endings in dual junctions. Taken together these results suggest that a BDNF-induced p75(NTR)-mediated ACh release potentiating mechanism and a BDNF-induced trkB-mediated release inhibitory mechanism may contribute to developmental synapse disconnection.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Neuromuscular Junction/growth & development , Neuromuscular Junction/metabolism , Neuronal Plasticity/physiology , Presynaptic Terminals/metabolism , Synapses/metabolism , Acetylcholine/metabolism , Animals , Animals, Newborn , Antibodies, Blocking/pharmacology , Brain-Derived Neurotrophic Factor/pharmacology , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Nerve Tissue Proteins , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neuromuscular Junction/ultrastructure , Neuronal Plasticity/drug effects , Presynaptic Terminals/drug effects , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Receptor, trkB/agonists , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/metabolism , Receptors, Growth Factor , Receptors, Nerve Growth Factor/agonists , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Nerve Growth Factor/metabolism , Recombinant Fusion Proteins/pharmacology , Synapses/drug effects , Synapses/ultrastructure
19.
Respir Res ; 10: 93, 2009 Oct 07.
Article in English | MEDLINE | ID: mdl-19811658

ABSTRACT

BACKGROUND: Allergic asthma is characterized by airway hyperresponsiveness (AHR) and allergic inflammation of the airways, driven by allergen-specific Th2 cells. The asthma phenotypes and especially AHR are sensitive to the presence and activity of regulatory T (Treg) cells in the lung. Glucocorticoid-induced tumor necrosis factor receptor (GITR) is known to have a co-stimulatory function on effector CD4+ T cells, rendering these cells insensitive to Treg suppression. However, the effects of GITR signaling on polarized Th1 and Th2 cell effector functions are not well-established. We sought to evaluate the effect of GITR signaling on fully differentiated Th1 and Th2 cells and to determine the effects of GITR activation at the time of allergen provocation on AHR and airway inflammation in a Th2-driven mouse model of asthma. METHODS: CD4+CD25- cells were polarized in vitro into Th1 and Th2 effector cells, and re-stimulated in the presence of GITR agonistic antibodies to assess the effect on IFNgamma and IL-4 production. To evaluate the effects of GITR stimulation on AHR and allergic inflammation in a mouse asthma model, BALB/c mice were sensitized to OVA followed by airway challenges in the presence or absence of GITR agonist antibodies. RESULTS: GITR engagement potentiated cytokine release from CD3/CD28-stimulated Th2 but not Th1 cells in vitro. In the mouse asthma model, GITR triggering at the time of challenge induced enhanced airway hyperresponsiveness, serum IgE and ex vivo Th2 cytokine release, but did not increase BAL eosinophilia. CONCLUSION: GITR exerts a differential effect on cytokine release of fully differentiated Th1 and Th2 cells in vitro, potentiating Th2 but not Th1 cytokine production. This effect on Th2 effector functions was also observed in vivo in our mouse model of asthma, resulting in enhanced AHR, serum IgE responses and Th2 cytokine production. This is the first report showing the effects of GITR activation on cytokine production by polarized primary Th1 and Th2 populations and the relevance of this pathway for AHR in mouse models for asthma. Our data provides crucial information on the mode of action of the GITR signaling, a pathway which is currently being considered for therapeutic intervention.


Subject(s)
Asthma/immunology , Bronchial Hyperreactivity/immunology , Bronchoconstriction , Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Th2 Cells/immunology , Animals , Asthma/physiopathology , Bronchial Hyperreactivity/physiopathology , Bronchial Provocation Tests , Bronchoalveolar Lavage Fluid/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , Cells, Cultured , Disease Models, Animal , Glucocorticoid-Induced TNFR-Related Protein , Humans , Immunoglobulin E/blood , Interferon-gamma/metabolism , Interleukin-2 Receptor alpha Subunit/immunology , Interleukins/metabolism , Male , Mice , Mice, Inbred BALB C , Ovalbumin , Pulmonary Eosinophilia/immunology , Rats , Receptors, Nerve Growth Factor/agonists , Receptors, Tumor Necrosis Factor/agonists , Recombinant Proteins/metabolism , Th1 Cells/immunology
20.
Cancer Sci ; 100(7): 1317-25, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19432889

ABSTRACT

We have reported for the first time the significance of effector T-cell multifunctionality in antitumor immunity, suggesting that the appearance of multifunctional/polyfunctional tumor-specific CD8(+) T cells in vivo is a critical determinant of the success of antitumor immunotherapy, and a strategy to induce multifunctionality in effector cells is required for the successful immunotherapy of hosts with progressing tumor. Glucocorticoid-induced tumor necrosis factor receptor (GITR) stimulation has been shown to enhance antitumor immune response. However, its functional impact on adoptively transferred T cells remains unclear. Here, we analyzed the impact of GITR stimulation in vivo on the functional profiles of adoptively transferred CD8(+) T cells specific for murine fibrosarcoma CMS5. GITR stimulation was found to enhance multifunctionality (interferon (IFN)-gamma and tumor necrosis factor (TNF)-alpha production and CD107a mobilization as a degranulation marker) in transferred cells at the single-cell level. These cells exhibited upregulated expression of CD25 in draining lymph nodes and increased infiltration in tumor. Mice that received T-cell therapy with GITR stimulation showed reduced Foxp3(+)CD4(+) T cells among tumor infiltrating lymphocytes and increased in vivo cytotoxic T lymphocytes (CTL) activity even with progressing tumor, resulting in enhanced tumor regression. These data strengthen the idea that effector T-cell multifunctionality is a sensitive immune correlate for successful immunotherapy against malignancy and provide an immunological rationale for effective T-cell therapy combined with GITR stimulation.


Subject(s)
CD8-Positive T-Lymphocytes/transplantation , Immunotherapy, Adoptive , Neoplasms, Experimental/therapy , Receptors, Nerve Growth Factor/agonists , Receptors, Tumor Necrosis Factor/agonists , Animals , CD8-Positive T-Lymphocytes/immunology , Combined Modality Therapy , Female , Glucocorticoid-Induced TNFR-Related Protein , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology
SELECTION OF CITATIONS
SEARCH DETAIL