Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cancer Immunol Immunother ; 72(1): 151-168, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35776159

ABSTRACT

The complex immunosuppressive nature of solid tumor microenvironments poses a significant challenge to generating efficacious and durable anticancer responses. Photoimmunotherapy is a cancer treatment strategy by which an antibody is conjugated with a non-toxic light-activatable dye. Following administration of the conjugate and binding to the target tumor, subsequent local laser illumination activates the dye, resulting in highly specific target cell membrane disruption. Here we demonstrate that photoimmunotherapy treatment elicited tumor necrosis, thus inducing immunogenic cell death characterized by the release of damage-associated molecular patterns (DAMPs). Photoimmunotherapy-killed tumor cells activated dendritic cells (DC), leading to the production of proinflammatory cytokines, T cell stimulation, priming antigen-specific T cells, and durable memory T cell responses, which led complete responder mice to effectively reject new tumors upon rechallenge. PD-1 blockade in combination with photoimmunotherapy enhanced overall anticancer efficacy, including against anti-PD-1-resistant tumors. The combination treatment also elicited abscopal anticancer activity, as observed by reduction of distal, non-illuminated tumors, further demonstrating the ability of photoimmunotherapy to harness local and peripheral T cell responses. With this work we therefore delineate the immune mechanisms of action for photoimmunotherapy and demonstrate the potential for cancer-targeted photoimmunotherapy to be combined with other immunotherapy approaches for augmented, durable anticancer efficacy. Moreover, we demonstrate responses utilizing various immunocompetent mouse models, as well as in vitro data from human cells, suggesting broad translational potential.


Subject(s)
Immunotherapy , Neoplasms , Humans , Animals , Mice , Immunotherapy/methods , Phototherapy/methods , Neoplasms/therapy , Disease Models, Animal , Cell Line, Tumor , Tumor Microenvironment
2.
Comput Biol Med ; 152: 106337, 2023 01.
Article in English | MEDLINE | ID: mdl-36502695

ABSTRACT

Immunotherapy targeting immune checkpoint proteins, such as programmed cell death ligand 1 (PD-L1), has shown impressive outcomes in many clinical trials but only 20%-40% of patients benefit from it. Utilizing Combined Positive Score (CPS) to evaluate PD-L1 expression in tumour biopsies to identify patients with the highest likelihood of responsiveness to anti-PD-1/PD-L1 therapy has been approved by the Food and Drug Administration for several solid tumour types. Current CPS workflow requires a pathologist to manually score the two-colour PD-L1 chromogenic immunohistochemistry image. Multiplex immunofluorescence (mIF) imaging reveals the expression of an increased number of immune markers in tumour biopsies and has been used extensively in immunotherapy research. Recent rapid progress of Artificial Intelligence (AI)-based imaging analysis, particularly Deep Learning, provides cost effective and high-quality solutions to healthcare. In this article, we propose an imaging pipeline that utilizes three-colour mIF images (DAPI, PD-L1, and Pan-cytokeratin) as input and predicts the CPS using AI techniques. Our novel pipeline is composed of three modules employing algorithms of image processing, machine learning, and deep learning techniques. The first module of quality check (QC) detects and removes the image regions contaminated with sectioning and staining artefacts. The QC module ensures that only image regions free of the three common artefacts are used for downstream analysis. The second module of nuclear segmentation uses deep learning to segment and count nuclei in the DAPI images wherein our specialized method can accurately separate touching nuclei. The third module of cell phenotyping calculates CPS by identifying and counting PD-L1 positive cells and tumour cells. These modules are data-efficient and require only few manual annotations for training purposes. Using tumour biopsies from a clinical trial, we found that the CPS from the AI-based models shows a high Spearman correlation (78%, p = 0.003) to the pathologist-scored CPS.


Subject(s)
Artificial Intelligence , Neoplasms , Humans , B7-H1 Antigen/metabolism , Neoplasms/diagnostic imaging , Immunohistochemistry , Fluorescent Antibody Technique , Biomarkers, Tumor/metabolism
3.
Head Neck ; 43(12): 3875-3887, 2021 12.
Article in English | MEDLINE | ID: mdl-34626024

ABSTRACT

BACKGROUND: Recurrent head and neck squamous cell carcinoma (rHNSCC) represents a significant global health burden with an unmet medical need. In this study we determined the safety and efficacy of RM-1929 photoimmunotherapy in patients with heavily pretreated rHNSCC. METHODS: RM-1929 (anti-EGFR-IR700 dye conjugate) was infused, followed by tumor illumination. We evaluated safety, tumor response, and pharmacokinetics. RESULTS: Nine patients were enrolled in Part 1 (dose-finding) and 30 patients in Part 2 (safety and efficacy). No dose-limiting toxicities were experienced in Part 1; 640 mg/m2 with fixed light dose (50 J/cm2 or 100 J/cm) was recommended for Part 2. Adverse events (AEs) in Part 2 were mostly mild to moderate but 19 (63.3%) patients had AE ≥Grade 3, including 3 (10.0%) with serious AEs leading to death (not treatment related). Efficacy in Part 2: unconfirmed objective response rate (ORR) 43.3% (95% CI 25.46%-62.57%); confirmed ORR 26.7% (95% CI 12.28%-45.89%); median overall survival 9.30 months (95% CI 5.16-16.92 months). CONCLUSIONS: Treatment was well tolerated. Responses and survival following RM-1929 photoimmunotherapy in heavily pretreated patients with rHNSCC were clinically meaningful and warrant further investigation. CLINICAL TRIAL INFORMATION: NCT02422979.


Subject(s)
Head and Neck Neoplasms , Immunotherapy , Neoplasm Recurrence, Local , Squamous Cell Carcinoma of Head and Neck , Antineoplastic Combined Chemotherapy Protocols , Cetuximab/therapeutic use , Head and Neck Neoplasms/therapy , Humans , Neoplasm Recurrence, Local/drug therapy , Phototherapy , Squamous Cell Carcinoma of Head and Neck/therapy
4.
Angew Chem Int Ed Engl ; 55(37): 11193-11197, 2016 09 05.
Article in English | MEDLINE | ID: mdl-27404798

ABSTRACT

Palmitoylethanolamide (PEA) and oleoylethanolamide (OEA) are endogenous lipid mediators that suppress inflammation. Their actions are terminated by the intracellular cysteine amidase, N-acylethanolamine acid amidase (NAAA). Even though NAAA may offer a new target for anti-inflammatory therapy, the lipid-like structures and reactive warheads of current NAAA inhibitors limit the use of these agents as oral drugs. A series of novel benzothiazole-piperazine derivatives that inhibit NAAA in a potent and selective manner by a non-covalent mechanism are described. A prototype member of this class (8) displays high oral bioavailability, access to the central nervous system (CNS), and strong activity in a mouse model of multiple sclerosis (MS). This compound exemplifies a second generation of non-covalent NAAA inhibitors that may be useful in the treatment of MS and other chronic CNS disorders.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Disease Models, Animal , Endocannabinoids/pharmacology , Enzyme Inhibitors/pharmacology , Ethanolamines/pharmacology , Multiple Sclerosis/drug therapy , Oleic Acids/pharmacology , Palmitic Acids/pharmacology , Administration, Oral , Amides , Amidohydrolases/metabolism , Animals , Dose-Response Relationship, Drug , Endocannabinoids/administration & dosage , Endocannabinoids/chemistry , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Ethanolamines/administration & dosage , Ethanolamines/chemistry , Mice , Molecular Structure , Multiple Sclerosis/metabolism , Oleic Acids/administration & dosage , Oleic Acids/chemistry , Palmitic Acids/administration & dosage , Palmitic Acids/chemistry , Structure-Activity Relationship
5.
PLoS One ; 10(3): e0121989, 2015.
Article in English | MEDLINE | ID: mdl-25799218

ABSTRACT

Photoimmunotherapy (PIT) of cancer utilizes tumor-specific monoclonal antibodies conjugated to a photosensitizer phthalocyanine dye IR700 which becomes cytotoxic upon irradiation with near infrared light. In this study, we aimed to evaluate the efficacy of PIT on human pancreatic cancer cells in vitro and in vivo in an orthotopic nude mouse model. The binding capacity of anti-CEA antibody to BxPC-3 human pancreatic cancer cells was determined by FACS analysis. An in vitro cytotoxicity assay was used to determine cell death following treatment with PIT. For in vivo determination of PIT efficacy, nude mice were orthotopically implanted with BxPC-3 pancreatic tumors expressing green fluorescent protein (GFP). After tumor engraftment, the mice were divided into two groups: (1) treatment with anti-CEA-IR700 + 690 nm laser and (2) treatment with 690 nm laser only. Anti-CEA-IR700 (100 µg) was administered to group (1) via tail vein injection 24 hours prior to therapy. Tumors were then surgically exposed and treated with phototherapy at an intensity of 150 mW/cm2 for 30 minutes. Whole body imaging was done subsequently for 5 weeks using an OV-100 small animal imaging system. Anti-CEA-IR700 antibody bound to the BxPC3 cells to a high degree as shown by FACS analysis. Anti-CEA-IR700 caused extensive cancer cell killing after light activation compared to control cells in cytotoxicity assays. In the orthotopic models of pancreatic cancer, the anti-CEA-IR700 group had significantly smaller tumors than the control after 5 weeks (p<0.001). There was no significant difference in the body weights of mice in the anti-CEA-IR700 and control groups indicating that PIT was well tolerated by the mice.


Subject(s)
Carcinoembryonic Antigen/immunology , Immunotherapy , Pancreatic Neoplasms/therapy , Phototherapy , Animals , Antibodies, Monoclonal , Cell Line, Tumor , Humans , Infrared Rays , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Radiation-Sensitizing Agents/therapeutic use , Tumor Burden , Xenograft Model Antitumor Assays
6.
J Surg Res ; 197(1): 5-11, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25799527

ABSTRACT

BACKGROUND: Photoimmunotherapy (PIT) is based on the use of a monoclonal antibody specific to cancer epitopes conjugated to a photosensitizer near-infrared phthalocyanine dye (IR700). In this study, PIT with IR700 conjugated to anti-carcinoembryonic antigen (CEA) was used as an adjunct to surgery in orthotopically-implanted human pancreatic cancer in a nude mouse model to eliminate microscopic disease in the post-surgical tumor bed and prevent local as well as metastatic recurrence. MATERIALS AND METHODS: Athymic nude mice were orthotopically implanted with the human pancreatic cancer cell line BxPC3 expressing green fluorescent protein. After tumor engraftment, the mice were divided into two groups as follows: bright light surgery (BLS) + anti-CEA-IR700 + 690 nm laser (PIT); and BLS only. Anti-CEA-IR700 (100 µg) was administered to the treatment group via tail-vein injection 24 h before therapy. Tumors were resected, and the surgical bed was treated with intraoperative phototherapy at an intensity of 150 mW/cm(2) for 30 min. Mice were imaged noninvasively for 8 wk using an OV-100 small animal fluorescence imager. RESULTS: BLS + PIT reduced local recurrence to 1/7 mice from 7/7 mice with BLS-only (P = 0.001) and metastatic recurrence to 2/7 mice compared with 6/7 mice with BLS-only (P = 0.03). Local tumor growth continued at a rapid rate after BLS-only compared with BLS + PIT where almost no local growth occurred. There was a significant difference in tumor size between mice in the BLS + PIT (2.14 mm(2), 95% confidence interval [CI] [-2.06 to 6.34] and BLS-only groups (115.2 mm(2), 95% CI [88.8-141.6]) at 6 wk after surgery (P < 0.001). There was also a significant difference in tumor weight between the BLS + PIT group (6.65 mg, 95% CI [-6.35 to 19.65] and BLS-only group (1100 mg, 95% CI [794-1406] at 8 wk after surgery (P < 0.001). CONCLUSIONS: PIT holds promise in the treatment of pancreatic cancer and may serve as a useful adjunct to surgery in the eradication of microscopic residual disease that can lead to both local and metastatic recurrence. Further studies are warranted to investigate the potential toxicities of PIT, especially with regard to anastomoses, such as those involved in pancreaticoduodenectomy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Indoles/therapeutic use , Neoplasm Recurrence, Local/prevention & control , Pancreatectomy , Pancreatic Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Animals , Carcinoembryonic Antigen/immunology , Cell Line, Tumor , Chemotherapy, Adjuvant , Humans , Isoindoles , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/surgery , Treatment Outcome
7.
Bioorg Med Chem Lett ; 24(3): 845-9, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24405707
8.
Handb Exp Pharmacol ; (208): 191-221, 2012.
Article in English | MEDLINE | ID: mdl-22222700

ABSTRACT

The antinociceptive and analgesic effects of muscarinic receptor ligands in human and nonhuman species have been evident for more than half a century. In this review, we describe the current understanding of the roles of different muscarinic subtypes in pain modulation and their mechanism of action along the pain signaling pathway, including peripheral nociception, spinal cord pain processing, and supraspinal analgesia. Extensive preclinical and clinical validation of these mechanisms points to the development of selective muscarinic agonists as one of the most exciting and promising avenues toward novel pain medications.


Subject(s)
Analgesics/pharmacology , Muscarinic Agonists/pharmacology , Pain Threshold/drug effects , Pain/drug therapy , Receptors, Muscarinic/drug effects , Acetylcholine/metabolism , Animals , Humans , Ligands , Neural Pathways/drug effects , Neural Pathways/metabolism , Pain/metabolism , Pain/physiopathology , Receptors, Muscarinic/metabolism , Signal Transduction/drug effects
9.
Structure ; 18(9): 1083-93, 2010 Sep 08.
Article in English | MEDLINE | ID: mdl-20826335

ABSTRACT

Dysregulation of the calcitonin gene-related peptide (CGRP), a potent vasodilator, is directly implicated in the pathogenesis of migraine. CGRP binds to and signals through the CGRP receptor (CGRP-R), a heterodimer containing the calcitonin receptor-like receptor (CLR), a class B GPCR, and RAMP1, a receptor activity-modifying protein. We have solved the crystal structure of the CLR/RAMP1 N-terminal ectodomain heterodimer, revealing how RAMPs bind to and potentially modulate the activities of the CLR GPCR subfamily. We also report the structures of CLR/RAMP1 in complex with the clinical receptor antagonists olcegepant (BIBN4096BS) and telcagepant (MK0974). Both drugs act by blocking access to the peptide-binding cleft at the interface of CLR and RAMP1. These structures illustrate, for the first time, how small molecules bind to and modulate the activity of a class B GPCR, and highlight the challenges of designing potent receptor antagonists for the treatment of migraine and other class B GPCR-related diseases.


Subject(s)
Azepines/chemistry , Imidazoles/chemistry , Piperazines/chemistry , Quinazolines/chemistry , Receptors, Calcitonin Gene-Related Peptide/chemistry , Azepines/pharmacology , Binding Sites , Calcitonin Gene-Related Peptide/chemistry , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide Receptor Antagonists , Calcitonin Receptor-Like Protein/chemistry , Calcitonin Receptor-Like Protein/metabolism , Crystallography, X-Ray , Imidazoles/pharmacology , Piperazines/pharmacology , Protein Structure, Tertiary , Quinazolines/pharmacology , Receptors, Calcitonin Gene-Related Peptide/metabolism
10.
Biochemistry ; 49(9): 1862-72, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20099900

ABSTRACT

The calcitonin gene-related peptide (CGRP) receptor is a heterodimer of two membrane proteins: calcitonin receptor-like receptor (CLR) and receptor activity-modifying protein 1 (RAMP1). CLR is a class B G-protein-coupled receptor (GPCR), possessing a characteristic large amino-terminal extracellular domain (ECD) important for ligand recognition and binding. Dimerization of CLR with RAMP1 provides specificity for CGRP versus related agonists. Here we report the expression, purification, and refolding of a soluble form of the CGRP receptor comprising a heterodimer of the CLR and RAMP1 ECDs. The extracellular protein domains corresponding to residues 23-133 of CLR and residues 26-117 of RAMP1 were shown to be sufficient for formation of a stable, monodisperse complex. The binding affinity of the purified ECD complex for the CGRP peptide was significantly lower than that of the native receptor (IC(50) of 12 microM for the purified ECD complex vs 233 pM for membrane-bound CGRP receptor), indicating that other regions of CLR and/or RAMP1 are important for peptide agonist binding. However, high-affinity binding to known potent and specific nonpeptide antagonists of the CGRP receptor, including olcegepant and telcagepant (K(D) < 0.02 muM), as well as N-terminally truncated peptides and peptide analogues (140 nM to 1.62 microM) was observed.


Subject(s)
Extracellular Space/chemistry , Protein Folding , Receptors, Calcitonin Gene-Related Peptide/chemistry , Receptors, Calcitonin/chemistry , Amino Acid Sequence , Binding, Competitive , Calcitonin Receptor-Like Protein , Cell Line, Tumor , Crystallography, X-Ray , Dimerization , Extracellular Space/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Ligands , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Magnetic Resonance Spectroscopy , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Receptor Activity-Modifying Protein 1 , Receptor Activity-Modifying Proteins , Receptors, Calcitonin/metabolism , Receptors, Calcitonin Gene-Related Peptide/biosynthesis , Receptors, Calcitonin Gene-Related Peptide/genetics , Receptors, Calcitonin Gene-Related Peptide/isolation & purification , Solubility
11.
Biochem Biophys Res Commun ; 324(3): 993-8, 2004 Nov 19.
Article in English | MEDLINE | ID: mdl-15485652

ABSTRACT

The docking protein p130Cas becomes phosphorylated upon cell adhesion to extracellular matrix proteins, and is thought to play an essential role in cell transformation. Cas transmits signals through interactions with the Src-homology 3 (SH3) and Src-homology 2 domains of FAK or v-Crk signaling molecules, or with 14-3-3 protein, as well as phosphatases PTP1B and PTP-PEST. The large (130kDa), multi-domain Cas molecule contains an SH3 domain, a Src-binding domain, a serine-rich protein interaction region, and a C-terminal region that participates in protein interactions implicated in antiestrogen resistance in breast cancer. In this study, as part of a long-term goal to examine the protein interactions of Cas by X-ray crystallography and nuclear magnetic resonance spectroscopy, molecular constructs were designed to express two adjacent domains, the serine-rich domain and the Src-binding domain, that each participate in intermolecular contacts dependent on protein phosphorylation. The protein products are soluble, homogeneous, monodisperse, and highly suitable for structural studies to define the role of Cas in integrin-mediated cell signaling.


Subject(s)
Proteins/chemistry , 14-3-3 Proteins/chemistry , Animals , Cell Transformation, Neoplastic , Circular Dichroism , Crk-Associated Substrate Protein , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/metabolism , Integrins , Light , Magnetic Resonance Spectroscopy , Phosphorylation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 12 , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Proteins/metabolism , Rats , Retinoblastoma-Like Protein p130 , Scattering, Radiation , Serine/chemistry , Signal Transduction , src Homology Domains , src-Family Kinases/metabolism
12.
Biochem Genet ; 40(11-12): 359-78, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12463345

ABSTRACT

Transdominant genetic selections can yield protein fragment and peptide modulators of specific biochemical pathways. In yeast, such screens have been highly successful in targeting the MAP (mitogen-activated protein) kinase growth-control pathway. We performed a similar type of selection aimed at recovery of modulators of the mammalian MAP kinase cascade. Two pathway activators were identified, fragments of the TrkB and Raf-1 kinases. In a second selection directed at the beta-catenin growth-control pathway, three different clones encoding cadherin fragments were recovered. In neither selection were peptide inhibitors observed. We conclude that some transdominant selections in mammalian cells can readily yield high-penetrance protein fragments, but may be less amenable to isolation of peptide inhibitors.


Subject(s)
Cytoskeletal Proteins/genetics , Mitogen-Activated Protein Kinases/genetics , Selection, Genetic , Signal Transduction/genetics , Trans-Activators/genetics , 3T3 Cells , Animals , Biological Assay , Cytoskeletal Proteins/physiology , In Vitro Techniques , Mice , Mitogen-Activated Protein Kinases/physiology , Signal Transduction/physiology , Trans-Activators/physiology , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...