Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3552, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670972

ABSTRACT

Chimeric antigen receptor (CAR)-T cell therapy for solid tumors faces significant hurdles, including T-cell inhibition mediated by the PD-1/PD-L1 axis. The effects of disrupting this pathway on T-cells are being actively explored and controversial outcomes have been reported. Here, we hypothesize that CAR-antigen affinity may be a key factor modulating T-cell susceptibility towards the PD-1/PD-L1 axis. We systematically interrogate CAR-T cells targeting HER2 with either low (LA) or high affinity (HA) in various preclinical models. Our results reveal an increased sensitivity of LA CAR-T cells to PD-L1-mediated inhibition when compared to their HA counterparts by using in vitro models of tumor cell lines and supported lipid bilayers modified to display varying PD-L1 densities. CRISPR/Cas9-mediated knockout (KO) of PD-1 enhances LA CAR-T cell cytokine secretion and polyfunctionality in vitro and antitumor effect in vivo and results in the downregulation of gene signatures related to T-cell exhaustion. By contrast, HA CAR-T cell features remain unaffected following PD-1 KO. This behavior holds true for CD28 and ICOS but not 4-1BB co-stimulated CAR-T cells, which are less sensitive to PD-L1 inhibition albeit targeting the antigen with LA. Our findings may inform CAR-T therapies involving disruption of PD-1/PD-L1 pathway tailored in particular for effective treatment of solid tumors.


Subject(s)
B7-H1 Antigen , Immunotherapy, Adoptive , Programmed Cell Death 1 Receptor , Receptors, Chimeric Antigen , T-Lymphocytes , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , Animals , Humans , Immunotherapy, Adoptive/methods , Mice , Cell Line, Tumor , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/immunology , Xenograft Model Antitumor Assays , Female , CRISPR-Cas Systems , Mice, Inbred NOD
2.
Nat Commun ; 13(1): 6840, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36369429

ABSTRACT

The human transcriptome contains thousands of small open reading frames (sORFs) that encode microproteins whose functions remain largely unexplored. Here, we show that TINCR lncRNA encodes pTINCR, an evolutionary conserved ubiquitin-like protein (UBL) expressed in many epithelia and upregulated upon differentiation and under cellular stress. By gain- and loss-of-function studies, we demonstrate that pTINCR is a key inducer of epithelial differentiation in vitro and in vivo. Interestingly, low expression of TINCR associates with worse prognosis in several epithelial cancers, and pTINCR overexpression reduces malignancy in patient-derived xenografts. At the molecular level, pTINCR binds to SUMO through its SUMO interacting motif (SIM) and to CDC42, a Rho-GTPase critical for actin cytoskeleton remodeling and epithelial differentiation. Moreover, pTINCR increases CDC42 SUMOylation and promotes its activation, triggering a pro-differentiation cascade. Our findings suggest that the microproteome is a source of new regulators of cell identity relevant for cancer.


Subject(s)
Neoplasms , RNA, Long Noncoding , Sumoylation , Humans , Neoplasms/genetics , rho GTP-Binding Proteins/metabolism , Ubiquitins/metabolism , RNA, Long Noncoding/genetics
3.
J Control Release ; 332: 517-528, 2021 04 10.
Article in English | MEDLINE | ID: mdl-33675877

ABSTRACT

Tumor targeting and intratumoral virus spreading are key features for successful oncolytic virotherapy. VCN-11 is a novel oncolytic adenovirus, genetically modified to express hyaluronidase (PH20) and display an albumin-binding domain (ABD) on the hexon. ABD allows the virus to self-coat with albumin when entering the bloodstream and evade neutralizing antibodies (NAbs). Here, we validate VCN-11 mechanism of action and characterize its toxicity. VCN-11 replication, hyaluronidase activity and binding to human albumin to evade NAbs was evaluated. Toxicity and efficacy of VCN-11 were assessed in mice and hamsters. Tumor targeting, and antitumor activity was analyzed in the presence of NAbs in several tumor models. VCN-11 induced 450 times more cytotoxicity in tumor cells than in normal cells. VCN-11 hyaluronidase production was confirmed by measuring PH20 activity in vitro and in virus-infected tumor areas in vivo. VCN-11 evaded NAbs from different sources and tumor targeting was demonstrated in the presence of high levels of NAbs in vivo, whereas the control virus without ABD was neutralized. VCN-11 showed a low toxicity profile in athymic nude mice and Syrian hamsters, allowing treatments with high doses and fractionated administrations without major toxicities (up to 1.2x1011vp/mouse and 7.5x1011vp/hamster). Fractionated intravenous administrations improved circulation kinetics and tumor targeting. VCN-11 antitumor efficacy was demonstrated in the presence of NAbs against Ad5 and itself. Oncolytic adenovirus VCN-11 disrupts tumor matrix and displays antitumor effects even in the presence of NAbs. These features make VCN-11 a safe promising candidate to test re-administration in clinical trials.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses , Adenoviridae , Animals , Antibodies, Neutralizing , Cell Line, Tumor , Cricetinae , Hyaluronoglucosaminidase , Mice , Mice, Nude , Oncolytic Viruses/genetics , Virus Replication , Xenograft Model Antitumor Assays
4.
Front Cell Dev Biol ; 9: 747667, 2021.
Article in English | MEDLINE | ID: mdl-35036403

ABSTRACT

Long noncoding RNAs (lncRNAs) are regulatory molecules which have been traditionally considered as "non-coding". Strikingly, recent evidence has demonstrated that many non-coding regions, including lncRNAs, do in fact contain small-open reading frames that code for small proteins that have been called microproteins. Only a few of them have been characterized so far, but they display key functions in a wide variety of cellular processes. Here, we show that TUNAR lncRNA encodes an evolutionarily conserved microprotein expressed in the nervous system that we have named pTUNAR. pTUNAR deficiency in mouse embryonic stem cells improves their differentiation potential towards neural lineage both in vitro and in vivo. Conversely, pTUNAR overexpression impairs neuronal differentiation by reduced neurite formation in different model systems. At the subcellular level, pTUNAR is a transmembrane protein that localizes in the endoplasmic reticulum and interacts with the calcium transporter SERCA2. pTUNAR overexpression reduces cytoplasmatic calcium, consistent with a possible role of pTUNAR as an activator of SERCA2. Altogether, our results suggest that our newly discovered microprotein has an important role in neural differentiation and neurite formation through the regulation of intracellular calcium. From a more general point of view, our results provide a proof of concept of the role of lncRNAs-encoded microproteins in neural differentiation.

5.
J Immunother Cancer ; 9(11)2021 11.
Article in English | MEDLINE | ID: mdl-35149591

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is characterized by dense desmoplastic stroma that limits the delivery of anticancer agents. VCN-01 is an oncolytic adenovirus designed to replicate in cancer cells with a dysfunctional RB1 pathway and express hyaluronidase. Here, we evaluated the mechanism of action of VCN-01 in preclinical models and in patients with pancreatic cancer. METHODS: VCN-01 replication and antitumor efficacy were evaluated alone and in combination with standard chemotherapy in immunodeficient and immunocompetent preclinical models using intravenous or intratumoral administration. Hyaluronidase activity was evaluated by histochemical staining and by measuring drug delivery into tumors. In a proof-of-concept clinical trial, VCN-01 was administered intratumorally to patients with PDAC at doses up to 1×1011 viral particles in combination with chemotherapy. Hyaluronidase expression was measured in serum by an ELISA and its activity within tumors by endoscopic ultrasound elastography. RESULTS: VCN-01 replicated in PDAC models and exerted antitumor effects which were improved when combined with chemotherapy. Hyaluronidase expression by VCN-01 degraded tumor stroma and facilitated delivery of a variety of therapeutic agents such as chemotherapy and therapeutic antibodies. Clinically, treatment was generally well-tolerated and resulted in disease stabilization of injected lesions. VCN-01 was detected in blood as secondary peaks and in post-treatment tumor biopsies, indicating virus replication. Patients had increasing levels of hyaluronidase in sera over time and decreased tumor stiffness, suggesting stromal disruption. CONCLUSIONS: VCN-01 is an oncolytic adenovirus with direct antitumor effects and stromal disruption capabilities, representing a new therapeutic agent for cancers with dense stroma. TRIAL REGISTRATION NUMBER: EudraCT number: 2012-005556-42 and NCT02045589.


Subject(s)
Adenoviridae/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/therapy , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/therapy , Stromal Cells/drug effects , Albumins/administration & dosage , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Female , Humans , Male , Mesocricetus , Mice , Mice, Inbred C57BL , Paclitaxel/administration & dosage , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Prognosis , Gemcitabine
6.
Sci Transl Med ; 11(476)2019 01 23.
Article in English | MEDLINE | ID: mdl-30674657

ABSTRACT

Retinoblastoma is a pediatric solid tumor of the retina activated upon homozygous inactivation of the tumor suppressor RB1 VCN-01 is an oncolytic adenovirus designed to replicate selectively in tumor cells with high abundance of free E2F-1, a consequence of a dysfunctional RB1 pathway. Thus, we reasoned that VCN-01 could provide targeted therapeutic activity against even chemoresistant retinoblastoma. In vitro, VCN-01 effectively killed patient-derived retinoblastoma models. In mice, intravitreous administration of VCN-01 in retinoblastoma xenografts induced tumor necrosis, improved ocular survival compared with standard-of-care chemotherapy, and prevented micrometastatic dissemination into the brain. In juvenile immunocompetent rabbits, VCN-01 did not replicate in retinas, induced minor local side effects, and only leaked slightly and for a short time into the blood. Initial phase 1 data in patients showed the feasibility of the administration of intravitreous VCN-01 and resulted in antitumor activity in retinoblastoma vitreous seeds and evidence of viral replication markers in tumor cells. The treatment caused local vitreous inflammation but no systemic complications. Thus, oncolytic adenoviruses targeting RB1 might provide a tumor-selective and chemotherapy-independent treatment option for retinoblastoma.


Subject(s)
Adenoviridae/physiology , Molecular Targeted Therapy , Oncolytic Viruses/physiology , Retinoblastoma Protein/metabolism , Retinoblastoma/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Mice , Neoplasm Metastasis , Rabbits , Retinoblastoma/immunology , Retinoblastoma/pathology , Survival Analysis , Tissue Distribution , Translational Research, Biomedical , Treatment Outcome , Virus Replication , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 21(6): 1406-18, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25391696

ABSTRACT

PURPOSE: Tumor targeting upon intravenous administration and subsequent intratumoral virus dissemination are key features to improve oncolytic adenovirus therapy. VCN-01 is a novel oncolytic adenovirus that combines selective replication conditional to pRB pathway deregulation, replacement of the heparan sulfate glycosaminoglycan putative-binding site KKTK of the fiber shaft with an integrin-binding motif RGDK for tumor targeting, and expression of hyaluronidase to degrade the extracellular matrix. In this study, we evaluate the safety and efficacy profile of this novel oncolytic adenovirus. EXPERIMENTAL DESIGN: VCN-01 replication and potency were assessed in a panel of tumor cell lines. VCN-01 tumor-selective replication was evaluated in human fibroblasts and pancreatic islets. Preclinical toxicity, biodistribution, and efficacy studies were conducted in mice and Syrian hamsters. RESULTS: Toxicity and biodistribution preclinical studies support the selectivity and safety of VCN-01. Antitumor activity after intravenous or intratumoral administration of the virus was observed in all tumor models tested, including melanoma and pancreatic adenocarcinoma, both in immunodeficient mice and immunocompetent hamsters. CONCLUSIONS: Oncolytic adenovirus VCN-01 characterized by the expression of hyaluronidase and the RGD shaft retargeting ligand shows an efficacy-toxicity prolife in mice and hamsters by intravenous and intratumoral administration that warrants clinical testing.


Subject(s)
Adenocarcinoma/therapy , Melanoma/therapy , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/therapy , Adenoviridae/genetics , Animals , Binding Sites/genetics , Carrier Proteins/genetics , Cell Line , Cell Line, Tumor , Cricetinae , Extracellular Matrix/metabolism , Female , HEK293 Cells , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Humans , Hyaluronoglucosaminidase/biosynthesis , Hyaluronoglucosaminidase/genetics , Islets of Langerhans/virology , Male , Mesocricetus , Mice , Mice, Inbred BALB C , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/genetics , Protein Binding , Xenograft Model Antitumor Assays
8.
J Control Release ; 192: 148-56, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25037019

ABSTRACT

Selective tumor targeting of oncolytic adenovirus at the level of cell entry remains a major challenge to improve efficacy and safety. Matrix metalloproteases (MMPs) are overexpressed in a variety of tumors and in particular in pancreatic cancer. In the current work, we have exploited the expression of MMPs together with the penetration capabilities of a TAT-like peptide to engineer tumor selective adenoviruses. We have generated adenoviruses containing CAR-binding ablated fibers further modified with a C-terminus TAT-like peptide linked to a blocking domain by an MMP-cleavable sequence. This linker resulted in a MMP-dependent cell transduction of the reporter MMP-activatable virus AdTATMMP and in efficient transduction of neoplastic cells and cancer-associated fibroblasts. Intravenous and intraductal administration of AdTATMMP into mice showed very low AdTATMMP activity in the normal pancreas, whereas increased transduction was observed in pancreatic tumors of transgenic Ela-myc mice. Intraductal administration of AdTATMMP into mice bearing orthotopic tumors led to a 25-fold increase in tumor targeting compared to the wild type fiber control. A replication competent adenovirus, Ad(RC)MMP, with the MMP-activatable fiber showed oncolytic efficacy and increased antitumor activity compared to Adwt in a pancreatic orthotopic model. Reduced local and distant metastases were observed in Ad(RC)MMP treated-mice. Moreover, no signs of pancreatic toxicity were detected. We conclude that MMP-activatable adenovirus may be beneficial for pancreatic cancer treatment.


Subject(s)
Adenoviridae/genetics , Matrix Metalloproteinases/genetics , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/therapy , Animals , Base Sequence , Cell Line, Tumor , Enzyme Activation , HEK293 Cells , Humans , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , NIH 3T3 Cells , Oncolytic Virotherapy/methods , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Transduction, Genetic
9.
Hum Gene Ther ; 23(9): 960-79, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22708837

ABSTRACT

Achieving high-efficiency tumor targeting after systemic delivery is a considerable challenge facing oncolytic gene therapists. Efficient retargeting should be combined with efforts to improve in vivo safety, reduce hepatotoxicity, minimize off-target interactions, and improve antitumoral potency and efficacy. We previously described the successful retargeting of adenovirus serotype 5 (Ad5) to α(v)ß(6), an integrin that is highly overexpressed in numerous human carcinomas. In this study, we have further modified this construct by introducing mutations that ablate coxsackievirus-adenovirus receptor (CAR) binding and putative interactions with factor IX (FIX)/C4b-binding protein (C4BP). We have found that the resulting vector, Ad5-477dlTAYT(A20), displays a desirable in vivo safety profile. This vector does not agglutinate human erythrocytes, fails to cause thrombocytopenia after intravenous delivery, has limited induction of proinflammatory cytokines, and results in low-level toxicity (aspartate aminotransferase/alanine aminotransferase) when compared with Ad5-EGFP(WT). Furthermore, it has reduced accumulation in Kupffer cells (1 hr) and limited hepatocyte transduction at later time points (24 and 96 hr). The parental vector, Ad5-EGFP(A20), also displayed many of these desirable properties. As a result of the improved safety profile of both A20-modified vectors, we escalated the dose from 2×10(10) to 4×10(10) viral particles in an antitumoral efficacy study. We observed improvements in reducing percent tumor growth at early time points (96 hr) when compared with Ad5-EGFP(WT), although increasing the dose did not affect the therapeutic outcome beneficially. On completion of the experiment, we detected increased E1A staining in the tumors of all A20-treated groups and we determined that E1A expression was localized largely within α(v)ß(6)(+) tumor cells. However, in spite of apparently efficient tumor transduction, this did not result in enhanced antitumoral efficacy as the virus failed to disseminate effectively throughout the tumor mass, presumably due to physical intratumoral restrictions. This highlights a remaining challenge that needs to be overcome before such vectors can be developed for future cancer gene therapy applications.


Subject(s)
Adenoviridae , Antigens, Neoplasm/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Integrins/metabolism , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses , Viral Tropism , Adenovirus E1A Proteins/biosynthesis , Animals , Antigens, Neoplasm/genetics , Binding Sites , CHO Cells , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Cricetinae , Cricetulus , Female , Gene Expression Regulation, Viral/genetics , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Integrins/genetics , Kupffer Cells/metabolism , Kupffer Cells/virology , Male , Mice , Mice, Inbred BALB C , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/virology , Transduction, Genetic
10.
Methods Mol Biol ; 797: 21-34, 2012.
Article in English | MEDLINE | ID: mdl-21948466

ABSTRACT

Adenovirus represents a valuable tool for the treatment of cancer, but tumor targeting remains a pending issue. Most common procedures to modify adenovirus genome are time-consuming due to the requirement of multiple cloning steps, and the low efficacy of the recombination process. Here, we present a new method for homologous recombination in yeast to fast construct recombinant adenoviruses. Also, an alternative procedure to purify viral stocks, based on iodixanol gradient is described. Compared to classical methods, iodixanol is nontoxic to cells, which avoids desalting to use in vitro and in vivo. Moreover, viral stocks are more viable and it can be used for large-scale purifications. Finally, a protocol for analyzing blood persistence of modified vector in in vivo biodistribution is presented.


Subject(s)
Adenoviridae/genetics , Adenoviridae/isolation & purification , Genetic Engineering/methods , Homologous Recombination , Oncolytic Virotherapy , Saccharomyces cerevisiae/genetics , Triiodobenzoic Acids/chemistry , Capsid/chemistry , Capsid Proteins/blood , Capsid Proteins/genetics , Genetic Vectors , Humans
11.
Hum Gene Ther ; 20(10): 1214-21, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19537946

ABSTRACT

Tumor targeting on systemic adenovirus administration is key to improve the prospects of adenovirus-mediated gene therapy and virus therapy of cancer. Despite many genetic and ligand conjugation approaches this objective remains elusive. Ablation of human adenovirus type 5 (Ad5) binding to its natural receptors in airway epithelial cells, that is, the coxsackievirus and adenovirus receptor (CAR) and integrins, does not impact on the preeminent liver tropism of adenovirus in the bloodstream. This is explained by a distinct entry pathway mediated by blood factors and heparan sulfates. Mutation of the KKTK heparin sulfate-binding domain of the fiber shaft to GATK results in liver transduction detargeting, but it is not compatible with otherwise useful HI-loop tumor-targeting ligand insertions such as the insertion of Arg-Gly-Asp (RGD). To circumvent this problem we have mutated the KKTK domain to RGDK, and analyzed the liver-detargeting and tumor-targeting transduction properties of this replacement mutant. Similar to RGD at the HI-loop, RGD at this new shaft location efficiently enhances the infectivity of adenovirus and improves the tumor-to-liver transduction ratio in vivo.


Subject(s)
Adenoviridae/genetics , Heparan Sulfate Proteoglycans/chemistry , Neoplasms/therapy , Neoplasms/virology , Oligopeptides/chemistry , Animals , Cell Line, Tumor , Genetic Vectors/genetics , Humans , Liver/enzymology , Liver/virology , Luciferases/metabolism , Mice , Protein Structure, Tertiary , Recombination, Genetic/genetics , Tissue Distribution , Transduction, Genetic
12.
Hum Gene Ther ; 19(12): 1415-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18795826

ABSTRACT

A critical obstacle for efficient gene therapy and virotherapy of cancer with adenoviral vectors and oncolytic adenoviruses is to target tumor cells in vivo. Recent reports indicate that, contrary to the natural airborne infection of epithelial cells with adenovirus type 5 mediated by coxsackievirus B and adenovirus receptor (CAR) and integrins, blood-borne adenovirus infects hepatocytes mainly through an indirect pathway that involves blood coagulation factors. In this report we have studied whether adenovirus also infects tumor cells in vivo by this pathway. In vitro and in vivo analyses show that vitamin K-dependent coagulation zymogens mediate tumor transduction and that the elimination of these factors abrogates tumor transduction. This finding imposes new challenges to retarget adenoviruses in vivo.


Subject(s)
Neoplasms/genetics , Transduction, Genetic , Vitamin K/pharmacology , Adenoviridae/genetics , Blood Coagulation/drug effects , Enterovirus B, Human/genetics , Genetic Therapy , Genetic Vectors , Humans , Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...