Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cancer Immunol Res ; 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38842347

ABSTRACT

Despite clinical evidence of antitumor activity, the development of cytokine therapies has been hampered by a narrow therapeutic window and limited response rates. Two cytokines of high interest for clinical development are interleukin 2 (IL-2) and interleukin 12 (IL-12), which potently synergize to promote the activation and proliferation of T cells and natural killer (NK) cells. However, the only approved human IL-2 therapy, Proleukin, is rarely used in the clinic due to systemic toxicities, and no IL-12 product has been approved to date due to severe dose-limiting toxicities. Here, we describe CLN-617, a first-in-class therapeutic for intratumoral (IT) injection that co-delivers IL-2 and IL-12 on a single molecule in a safe and effective manner. CLN-617 is a single-chain fusion protein comprised of IL-2, leukocyte-associated immunoglobulin-like receptor 2 (LAIR2), human serum albumin (HSA), and IL-12. LAIR2 and HSA function to retain CLN-617 in the treated tumor by binding collagen and increasing molecular weight, respectively. We found that IT administration of a murine surrogate of CLN-617, mCLN-617, eradicated established treated and untreated tumors in syngeneic models, significantly improved response to anti-PD1 checkpoint therapy, and generated a robust abscopal response dependent on cellular immunity and antigen cross-presentation. CLN-617 is being evaluated in a clinical trial in patients with advanced solid tumors (NCT06035744).

2.
MAbs ; 15(1): 2208697, 2023.
Article in English | MEDLINE | ID: mdl-37165468

ABSTRACT

The field of immuno-oncology has revolutionized cancer patient care and improved survival and quality of life for patients. Much of the focus in the field has been on exploiting the power of the adaptive immune response through therapeutic targeting of T cells. While these approaches have markedly advanced the field, some challenges remain, and the clinical benefit of T cell therapies does not extend to all patients or tumor indications. Alternative strategies, such as engaging the innate immune system, have become an intense area of focus in the field. In particular, the engagement of natural killer (NK) cells as potent effectors of the innate immune response has emerged as a promising modality in immunotherapy. Here, we review therapeutic approaches for selective engagement of NK cells for cancer therapy, with a particular focus on targeting the key activating receptors NK Group 2D (NKG2D) and cluster of differentiation 16A (CD16A).


Subject(s)
NK Cell Lectin-Like Receptor Subfamily K , Neoplasms , Humans , Quality of Life , Killer Cells, Natural , Neoplasms/therapy , Immunotherapy
3.
Methods Mol Biol ; 2318: 297-312, 2021.
Article in English | MEDLINE | ID: mdl-34019298

ABSTRACT

The tetracycline regulatory system provides a tractable strategy to interrogate the role of oncogenes in the initiation, maintenance, and regression of tumors through both spatial and temporal control of expression. This approach has several potential advantages over conventional methods to generate genetically engineered mouse models. First, continuous constitutive overexpression of an oncogene can be lethal to the host impeding further study. Second, constitutive overexpression fails to model adult onset of disease. Third, constitutive deletion does not permit, whereas conditional overexpression of an oncogene enables the study of the consequences of restoring expression of an oncogene back to endogenous levels. Fourth, the conditional activation of oncogenes enables examination of specific and/or developmental state-specific consequences.Hence, by allowing precise control of when and where a gene is expressed, the tetracycline regulatory system provides an ideal approach for the study of putative oncogenes in the initiation as well as the maintenance of tumorigenesis and the examination of the mechanisms of oncogene addiction. In this protocol, we describe the methods involved in the development of a conditional mouse model of MYC-induced T-cell acute lymphoblastic leukemia.


Subject(s)
Genetic Engineering/methods , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , Animals, Genetically Modified/genetics , Apoptosis , Carcinogenesis/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , DNA/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Genes, myc/genetics , Genes, myc/physiology , Humans , Mice , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Synthesis Inhibitors , Proto-Oncogene Proteins c-myc/genetics , T-Lymphocytes/metabolism , Tetracycline/pharmacology
4.
Sci Immunol ; 6(57)2021 03 19.
Article in English | MEDLINE | ID: mdl-33741657

ABSTRACT

Tissue-resident memory T cells (TRMs) can profoundly enhance mucosal immunity, but parameters governing TRM induction by vaccination remain poorly understood. Here, we describe an approach exploiting natural albumin transport across the airway epithelium to enhance mucosal TRM generation by vaccination. Pulmonary immunization with albumin-binding amphiphile conjugates of peptide antigens and CpG adjuvant (amph-vaccines) increased vaccine accumulation in the lung and mediastinal lymph nodes (MLNs). Amph-vaccines prolonged antigen presentation in MLNs over 2 weeks, leading to 25-fold increased lung-resident T cell responses over traditional immunization and enhanced protection from viral or tumor challenge. Mimicking such prolonged exposure through repeated administration of soluble vaccine revealed that persistence of both antigen and adjuvant was critical for optimal TRM induction, mediated through T cell priming in MLNs after prime, and directly in the lung tissue after boost. Thus, vaccine persistence strongly promotes TRM induction, and amph-conjugates may provide a practical approach to achieve such kinetics in mucosal vaccines.


Subject(s)
Adjuvants, Immunologic , Albumins/immunology , Immunity, Mucosal , Immunologic Memory , Lung/immunology , Memory T Cells/immunology , Animals , Biomarkers , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunization , Immunophenotyping , Lung/metabolism , Lymphocyte Activation/immunology , Memory T Cells/metabolism , Mice , Mice, Knockout , Organ Specificity/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Vaccines/immunology
5.
Nat Biomed Eng ; 4(6): 636-648, 2020 06.
Article in English | MEDLINE | ID: mdl-32483299

ABSTRACT

The formulations of peptide-based antitumour vaccines being tested in clinical studies are generally associated with weak potency. Here, we show that pharmacokinetically tuning the responses of peptide vaccines by fusing the peptide epitopes to carrier proteins optimizes vaccine immunogenicity in mice. In particular, we show in immunized mice that the carrier protein transthyretin simultaneously optimizes three factors: efficient antigen uptake in draining lymphatics from the site of injection, protection of antigen payloads from proteolytic degradation and reduction of antigen presentation in uninflamed distal lymphoid organs. Optimizing these factors increases vaccine immunogenicity by up to 90-fold and maximizes the responses to viral antigens, tumour-associated antigens, oncofetal antigens and shared neoantigens. Protein-peptide epitope fusions represent a facile and generalizable strategy for enhancing the T-cell responses elicited by subunit vaccines.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Immunogenicity, Vaccine/immunology , T-Lymphocytes/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics , Albumins/immunology , Animals , Antigens, Neoplasm , Basic-Leucine Zipper Transcription Factors , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Epitopes , Immunity, Cellular , Immunotherapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/genetics
7.
Nat Commun ; 8: 14069, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28094297

ABSTRACT

Inorganic nanoparticles (NPs) are studied as drug carriers, radiosensitizers and imaging agents, and characterizing nanoparticle biodistribution is essential for evaluating their efficacy and safety. Tracking NPs at the single-cell level with current technologies is complicated by the lack of reliable methods to stably label particles over extended durations in vivo. Here we demonstrate that mass cytometry by time-of-flight provides a label-free approach for inorganic nanoparticle quantitation in cells. Furthermore, mass cytometry can enumerate AuNPs with a lower detection limit of ∼10 AuNPs (3 nm core size) in a single cell with tandem multiparameter cellular phenotyping. Using the cellular distribution insights, we selected an amphiphilic surface ligand-coated AuNP that targeted myeloid dendritic cells in lymph nodes as a peptide antigen carrier, substantially increasing the efficacy of a model vaccine in a B16-OVA melanoma mouse model. This technology provides a powerful new level of insight into nanoparticle fate in vivo.


Subject(s)
Gold/analysis , Mass Spectrometry/methods , Metal Nanoparticles/analysis , Single-Cell Analysis/methods , Animals , Dendritic Cells/chemistry , Dendritic Cells/metabolism , Drug Carriers/chemistry , Female , Melanoma/drug therapy , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Tissue Distribution , Vaccines/administration & dosage , Vaccines/chemistry , Vaccines/metabolism
8.
Nat Med ; 22(12): 1402-1410, 2016 12.
Article in English | MEDLINE | ID: mdl-27775706

ABSTRACT

Checkpoint blockade with antibodies specific for cytotoxic T lymphocyte-associated protein (CTLA)-4 or programmed cell death 1 (PDCD1; also known as PD-1) elicits durable tumor regression in metastatic cancer, but these dramatic responses are confined to a minority of patients. This suboptimal outcome is probably due in part to the complex network of immunosuppressive pathways present in advanced tumors, which are unlikely to be overcome by intervention at a single signaling checkpoint. Here we describe a combination immunotherapy that recruits a variety of innate and adaptive immune cells to eliminate large tumor burdens in syngeneic tumor models and a genetically engineered mouse model of melanoma; to our knowledge tumors of this size have not previously been curable by treatments relying on endogenous immunity. Maximal antitumor efficacy required four components: a tumor-antigen-targeting antibody, a recombinant interleukin-2 with an extended half-life, anti-PD-1 and a powerful T cell vaccine. Depletion experiments revealed that CD8+ T cells, cross-presenting dendritic cells and several other innate immune cell subsets were required for tumor regression. Effective treatment induced infiltration of immune cells and production of inflammatory cytokines in the tumor, enhanced antibody-mediated tumor antigen uptake and promoted antigen spreading. These results demonstrate the capacity of an elicited endogenous immune response to destroy large, established tumors and elucidate essential characteristics of combination immunotherapies that are capable of curing a majority of tumors in experimental settings typically viewed as intractable.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer Vaccines/pharmacology , Cytokines/drug effects , Immunotherapy/methods , Interleukin-2/pharmacology , Lung Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Melanoma, Experimental/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/drug effects , Adaptive Immunity , Animals , Cell Line, Tumor , Cytokines/immunology , Drug Therapy, Combination , Flow Cytometry , Gene Knockout Techniques , Immunity, Innate , Immunoblotting , Intramolecular Oxidoreductases/genetics , Mice , T-Lymphocytes/immunology
10.
Article in English | MEDLINE | ID: mdl-24110741

ABSTRACT

Understanding the complex dynamics between the tumor cells and the host immune system will be key to improved therapeutic strategies against cancer. We propose an ODE-based mathematical model of both the tumor and immune system and how they respond to inactivation of the driving oncogene. Our model supports experimental results showing that cellular senescence of tumor cells is dependent on CD4+ T helper cells, leading to relapse of tumors in immunocompromised hosts.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Models, Theoretical , Neoplasms/pathology , T-Lymphocytes, Helper-Inducer/immunology , CD4-Positive T-Lymphocytes/metabolism , Cellular Senescence/immunology , Humans , Immune System/immunology , Oncogenes , T-Lymphocytes, Helper-Inducer/metabolism
11.
Methods Mol Biol ; 1012: 221-35, 2013.
Article in English | MEDLINE | ID: mdl-24006068

ABSTRACT

The tetracycline regulatory system provides a tractable strategy to interrogate the role of oncogenes in the initiation and maintenance of tumorigenesis through both spatial and temporal control of expression. This approach has several potential advantages over conventional methods to generate genetically engineered mouse models. First, continuous constitutive overexpression of an oncogene can be lethal to the host impeding further study. Second, constitutive overexpression fails to model adult onset of disease. Third, constitutive deletion does not permit, whereas conditional overexpression of an oncogene enables the study of the consequences of restoring expression of an oncogene back to endogenous levels. Fourth, the conditional activation of oncogenes enables examination of specific and/or developmental state-specific consequences. Hence, by allowing precise control of when and where a gene is expressed, the tetracycline regulatory system provides an ideal approach for the study of putative oncogenes in both the initiation and maintenance of tumorigenesis. In this protocol, we describe the methods involved in the development of a conditional mouse model of MYC-induced T-cell acute lymphoblastic leukemia.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Tetracycline/pharmacology , Animals , Cell Line , Disease Models, Animal , Female , Gene Expression , Gene Targeting/methods , Genetic Vectors/genetics , Isografts , Male , Mice , Mice, Transgenic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism
12.
Comput Math Methods Med ; 2013: 802512, 2013.
Article in English | MEDLINE | ID: mdl-23573174

ABSTRACT

The dependence on the overexpression of a single oncogene constitutes an exploitable weakness for molecular targeted therapy. These drugs can produce dramatic tumor regression by targeting the driving oncogene, but relapse often follows. Understanding the complex interactions of the tumor's multifaceted response to oncogene inactivation is key to tumor regression. It has become clear that a collection of cellular responses lead to regression and that immune-mediated steps are vital to preventing relapse. Our integrative mathematical model includes a variety of cellular response mechanisms of tumors to oncogene inactivation. It allows for correct predictions of the time course of events following oncogene inactivation and their impact on tumor burden. A number of aspects of our mathematical model have proven to be necessary for recapitulating our experimental results. These include a number of heterogeneous tumor cell states since cells following different cellular programs have vastly different fates. Stochastic transitions between these states are necessary to capture the effect of escape from oncogene addiction (i.e., resistance). Finally, delay differential equations were used to accurately model the tumor growth kinetics that we have observed. We use this to model oncogene addiction in MYC-induced lymphoma, osteosarcoma, and hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular/immunology , Liver Neoplasms/immunology , Lymphoma/immunology , Neoplasms/pathology , Oncogenes/genetics , Osteosarcoma/immunology , Apoptosis , CD4-Positive T-Lymphocytes/cytology , Carcinoma, Hepatocellular/metabolism , Cellular Senescence , Computational Biology/methods , Computer Simulation , Humans , Liver Neoplasms/metabolism , Lymphoma/metabolism , Models, Biological , Neoplasms/metabolism , Osteosarcoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Recurrence , Stochastic Processes
13.
Proc Natl Acad Sci U S A ; 108(42): 17432-7, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-21969595

ABSTRACT

The suppression of oncogenic levels of MYC is sufficient to induce sustained tumor regression associated with proliferative arrest, differentiation, cellular senescence, and/or apoptosis, a phenomenon known as oncogene addiction. However, after prolonged inactivation of MYC in a conditional transgenic mouse model of Eµ-tTA/tetO-MYC T-cell acute lymphoblastic leukemia, some of the tumors recur, recapitulating what is frequently observed in human tumors in response to targeted therapies. Here we report that these recurring lymphomas express either transgenic or endogenous Myc, albeit in many cases at levels below those in the original tumor, suggesting that tumors continue to be addicted to MYC. Many of the recurring lymphomas (76%) harbored mutations in the tetracycline transactivator, resulting in expression of the MYC transgene even in the presence of doxycycline. Some of the remaining recurring tumors expressed high levels of endogenous Myc, which was associated with a genomic rearrangement of the endogenous Myc locus or activation of Notch1. By gene expression profiling, we confirmed that the primary and recurring tumors have highly similar transcriptomes. Importantly, shRNA-mediated suppression of the high levels of MYC in recurring tumors elicited both suppression of proliferation and increased apoptosis, confirming that these tumors remain oncogene addicted. These results suggest that tumors induced by MYC remain addicted to overexpression of this oncogene.


Subject(s)
Genes, myc , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/therapy , Neoplasm Recurrence, Local/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Animals , Gene Expression , Gene Knockdown Techniques , Humans , Mice , Mice, Transgenic , Mutation , Phenotype , RNA, Small Interfering/genetics , Tetracycline Resistance/genetics , Trans-Activators/genetics
14.
Cancer Cell ; 18(5): 485-98, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21035406

ABSTRACT

Oncogene addiction is thought to occur cell autonomously. Immune effectors are implicated in the initiation and restraint of tumorigenesis, but their role in oncogene inactivation-mediated tumor regression is unclear. Here, we show that an intact immune system, specifically CD4(+) T cells, is required for the induction of cellular senescence, shutdown of angiogenesis, and chemokine expression resulting in sustained tumor regression upon inactivation of the MYC or BCR-ABL oncogenes in mouse models of T cell acute lymphoblastic lymphoma and pro-B cell leukemia, respectively. Moreover, immune effectors knocked out for thrombospondins failed to induce sustained tumor regression. Hence, CD4(+) T cells are required for the remodeling of the tumor microenvironment through the expression of chemokines, such as thrombospondins, in order to elicit oncogene addiction.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Oncogenes/genetics , Tumor Microenvironment/immunology , Animals , Apoptosis/physiology , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Chemokines/genetics , Chemokines/metabolism , Cyclosporine/pharmacology , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Immunosuppressive Agents/pharmacology , Leukemia, Experimental/genetics , Leukemia, Experimental/immunology , Mice , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Proto-Oncogene Proteins c-myc/genetics , Remission Induction , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
16.
Acta Pharm ; 56(3): 259-72, 2006 Sep.
Article in English | MEDLINE | ID: mdl-19831276

ABSTRACT

Several 2,4-dimethylphenyl substituted semicarbazones were synthesized in three steps involving aryl urea and aryl semicarbazide formation. The structures were confirmed by spectral and elemental analyses. All the compounds were evaluated for anticonvulsant activity by using a series of test models, including maximal electroshock seizure, subcutaneous pentylenetetrazole and subcutaneous strychnine seizure threshold tests. The compounds were also evaluated for behavioural impairement and depression activity. In the neurochemical investigation, potent compounds were evaluated for their effects on rat brain gamma-aminobutyric acid (GABA) levels and in vitro gamma-aminobutyrate transaminase (Pseudomonas fluorescens) activity. Preliminary studies suggest that these compounds exhibit anticonvulsant activity via a GABA-mediated mechanism.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Anticonvulsants/pharmacology , Seizures/drug therapy , Semicarbazones/pharmacology , Animals , Anticonvulsants/chemical synthesis , Behavior, Animal/drug effects , Brain/metabolism , Disease Models, Animal , Male , Mice , Rats , Rats, Sprague-Dawley , Rats, Wistar , Semicarbazones/chemical synthesis , gamma-Aminobutyric Acid/drug effects , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...