Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Publication year range
1.
Cancer Immunol Immunother ; 73(1): 5, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38180524

ABSTRACT

Cancer immunotherapies strive to overcome tumor-induced immune suppression and activate antitumor immune responses. Although cytotoxic T lymphocytes (CTLs) play a pivotal role in this process, natural killer (NK) cells have also demonstrated remarkable tumor-killing abilities, given their ability to discriminate tumor cells from normal cells and mediate specific antitumoral cytotoxicity. NK cells activation depends on a balance between activation and inhibition signals from several ligands/receptors. Among them, MICA/NKG2D axis is a master regulator of NK activation. MHC class I chain-related polypeptide A (MICA) expression is upregulated by many tumor cell lines and primary tumors and serves as a ligand for the activating NK group 2D (NKG2D) receptor on NK cells and subpopulations of T cells. However, cancer cells can cleave MICA, making it soluble and de-targeting tumor cells from NK cells, leading to tumor immune escape.In this study, we present ICOVIR15KK-MICAMut, an oncolytic adenovirus (OAdv) armed with a transgene encoding a non-cleavable MICA to promote NK-mediated cell-killing capacity and activate the immune response against cancer cells. We first demonstrated the correct MICA overexpression from infected cells. Moreover, our MICA-expressing OAdv promotes higher NK activation and killing capacity than the non-armed virus in vitro. In addition, the armed virus also demonstrated significant antitumor activity in immunodeficient mice in the presence of human PBMCs, indicating the activation of human NK cells. Finally, OAdv-MICA overexpression in immunocompetent tumor-bearing mice elicits tumor-specific immune response resulting in a greater tumor growth control.In summary, this study highlights the significance of NK cells in cancer immunotherapy and presents an innovative approach using a modified oncolytic virus to enhance NK cell activation and antitumor immune response. These findings suggest promising potential for future research and clinical applications.


Subject(s)
Adenoviridae , NK Cell Lectin-Like Receptor Subfamily K , Humans , Animals , Mice , Adenoviridae/genetics , NK Cell Lectin-Like Receptor Subfamily K/genetics , Lymphocyte Activation , Genes, MHC Class I , Tumor Escape
2.
BMC Urol ; 21(1): 12, 2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33509164

ABSTRACT

BACKGROUND: Radical prostatectomy is the gold standard treatment for men with localized prostate cancer. This technique is associated with post-operative urinary incontinence. Pelvic floor physiotherapy is a conservative, painless and economical treatment for this specific situation. Kegel exercises and perineal electrostimulation are common techniques to train pelvic floor muscles. The perineal electrostimulation can be applied to the patient with surface electrodes or by an intra-cavitary anal probe. This study proposes that transcutaneous perineal electrostimulation is as effective as intra-cavitary electrostimulation in reducing urinary incontinence secondary to radical prostatectomy. The main objective is to compare the efficacy of the treatment with transcutaneous perineal electrostimulation versus the same intra-cavitary treatment to reduce the magnitude of urinary incontinence after radical prostatectomy, and the impact on the quality of life. METHODS: This single-blind equivalence randomized controlled trial will include 70 man who suffer urinary incontinence post radical prostatectomy. Participants will be randomized into surface electrodes group and intra-anal probe group. The groups will receive treatment for 10 consecutive weeks. Outcomes include changes in the 24-h Pad Test, and ICIQ-SF, SF-12 and I-QoL questionnaires. Clinical data will be collected at baseline, 6 and 10 weeks after the first session, and 6 months after the end of treatment. DISCUSSION: The results will allow us to prescribe the most beneficial perineal electrostimulation technique in the treatment of urinary incontinence derived from radical prostatectomy. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT03587402. 27/06/2018.


Subject(s)
Electric Stimulation Therapy/methods , Postoperative Complications/therapy , Prostatectomy , Prostatic Neoplasms/surgery , Randomized Controlled Trials as Topic/methods , Urinary Incontinence/therapy , Equivalence Trials as Topic , Humans , Male , Perineum , Prostatectomy/methods , Single-Blind Method , Transcutaneous Electric Nerve Stimulation , Treatment Outcome
3.
Gene Ther ; 24(2): 92-103, 2017 02.
Article in English | MEDLINE | ID: mdl-27906162

ABSTRACT

CD40 is an interesting target in cancer immunotherapy due to its ability to stimulate T-helper 1 immunity via maturation of dendritic cells and to drive M2 to M1 macrophage differentiation. Pancreatic cancer has a high M2 content that has shown responsive to anti-CD40 agonist therapy and CD40 may thus be a suitable target for immune activation in these patients. In this study, a novel oncolytic adenovirus armed with a trimerized membrane-bound extracellular CD40L (TMZ-CD40L) was evaluated as a treatment of pancreatic cancer. Further, the CD40L mechanisms of action were elucidated in cancer models. The results demonstrated that the virus transferring TMZ-CD40L had oncolytic capacity in pancreatic cancer cells and could control tumor progression. TMZ-CD40L was a potent stimulator of human myeloid cells and T-cell responses. Further, CD40L-mediated stimulation increased tumor-infiltrating T cells in vivo, which may be due to a direct activation of endothelial cells to upregulate receptors for lymphocyte attachment and transmigration. In conclusion, CD40L-mediated gene therapy is an interesting concept for the treatment of tumors with high levels of M2 macrophages, such as pancreatic cancer, and an oncolytic virus as carrier of CD40L may further boost tumor killing and immune activation.


Subject(s)
CD40 Ligand/genetics , Endothelium, Vascular/immunology , Genetic Therapy , Myeloid Cells/immunology , Oncolytic Viruses/genetics , Pancreatic Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Movement/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Female , Genetic Vectors/administration & dosage , Humans , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Tumor Cells, Cultured , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
4.
Gene Ther ; 22(7): 596-601, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25994521

ABSTRACT

Oncolytic adenoviruses can promote immune responses against tumors by expressing and/or displaying tumor-associated antigens. However, the strong immunodominance of viral antigens mask responses against tumor epitopes. In addition, defects in major histocompatibility complex class I antigen presentation pathway such as the downregulation of the transporter-associated with antigen processing (TAP) are frequently associated with immune evasion of tumor cells. To promote the immunogenicity of exogenous epitopes in the context of an oncolytic adenovirus, we have taken advantage of the ER localization of the viral protein E3-19K. We have inserted tumor-associated epitopes after the N-terminal signal sequence for membrane insertion of this protein and flanked them with linkers cleavable by the protease furin to facilitate their TAP-independent presentation. This strategy allowed an enhanced presentation of the exogenous epitopes in TAP-deficient tumor cells in vitro and the generation of higher specific immune responses in vivo that were able to significantly control tumor growth.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenovirus E3 Proteins/genetics , Adenoviruses, Human/genetics , Epitopes/genetics , Mutagenesis, Insertional , Neoplasms/therapy , Oncolytic Viruses/genetics , Adenoviruses, Human/metabolism , Animals , Antigen Presentation , Cell Line, Tumor , Female , HEK293 Cells , Humans , Mice, Inbred C57BL
5.
Gene Ther ; 21(8): 767-74, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24942629

ABSTRACT

Endovenously administered oncolytic viruses extravasate and penetrate poorly into tumors. iRGD is a cyclic peptide that enhances tumor penetration when conjugated or coadministered with different types of molecules such as drugs, nanoparticles or phages. iRGD-mediated tumor penetration occurs in three steps: binding to αv-integrins on tumor vasculature or tumor cells, exposure by proteolysis of a C-terminal motif that binds to neuropilin-1 (NRP-1) and cell internalization. We have genetically inserted the iRGD peptide in the fiber C terminus of ICOVIR15K, an oncolytic tumor-retargeted adenovirus to increase its tumor penetration. In vitro, NRP-1 interaction improved binding and internalization of the virus in different cancer cells overexpressing integrins and NRP-1. However, such NRP-1-mediated internalization did not affect transduction or cytotoxicity. In vivo, iRGD did not change the normal organ transduction pattern, with liver and spleen as main targeted organs. In tumors, however, iRGD enhanced transduction and early adenovirus dissemination through the tumor mass leading to an improved antitumor efficacy.


Subject(s)
Genetic Therapy/methods , Oligopeptides/therapeutic use , Oncolytic Virotherapy/methods , Amino Acid Motifs , Animals , Cell Line, Tumor , Drug Delivery Systems/methods , Female , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , MCF-7 Cells , Mice, Inbred BALB C , Neuropilin-1/metabolism , Virus Internalization , Xenograft Model Antitumor Assays
6.
Gene Ther ; 19(4): 453-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21776023

ABSTRACT

Retargeting oncolytic adenoviruses from their systemic preeminent liver tropism to disseminated tumor foci would highly improve the efficacy of these agents at eradicating tumors. We have replaced the KKTK fiber shaft heparan sulfate glycosaminoglycan-binding domain with an RGDK motif in order to achieve simultaneously liver detargeting and tumor targeting. When inserted into a wild-type backbone, this mutation palliated liver transaminase elevation and hematological alterations in mice. Importantly, when tested in a backbone that redirects E1A transcription towards pRB pathway deregulation, RGD at this novel shaft location also improved significantly systemic antitumor therapy compared with the broadly used RGD location at the HI-loop of the fiber knob domain.


Subject(s)
Adenoviridae/genetics , Neoplasms/therapy , Oligopeptides , Oncolytic Virotherapy/methods , Animals , Binding Sites , Cell Line, Tumor , Gene Transfer Techniques , Genetic Vectors , Heparitin Sulfate/metabolism , Mice , Mice, Inbred BALB C , Receptors, Cell Surface/metabolism
7.
Gene Ther ; 19(11): 1048-57, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22113313

ABSTRACT

The limitations of the current oncolytic adenoviruses for cancer therapy include insufficient potency and poor distribution of the virus throughout the tumor mass. To address these problems, we generated an oncolytic adenovirus expressing the hyperfusogenic form of the gibbon-ape leukemia virus (GALV) envelope glycoprotein under the control of the adenovirus major late promoter. The oncolytic properties of the new fusogenic adenovirus, ICOVIR16, were analyzed both in vitro and in vivo, and compared with that of its non-fusogenic counterpart, ICOVIR15. Our results indicate that GALV expression by ICOVIR16 induced extensive syncytia formation and enhanced tumor cell killing in a variety of tumor cell types. When injected intratumorally or intravenously into mice with large pre-established melanoma or pancreatic tumors, ICOVIR16 rapidly reduced tumor burden, and in some cases, resulted in complete eradication of the tumors. Importantly, GALV expression induced tumor cell fusion in vivo and enhanced the spreading of the virus throughout the tumor. Taken together, these results indicate that GALV expression can improve the antitumoral potency of an oncolytic adenovirus and suggest that ICOVIR16 is a promising candidate for clinical evaluation in patients with cancer.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Giant Cells , Leukemia Virus, Gibbon Ape/genetics , Oncolytic Viruses , Adenoviridae/metabolism , Animals , Cell Line, Tumor , Cricetinae , Female , Gene Expression Regulation, Viral , Gene Order , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Genetic Vectors/metabolism , Giant Cells/virology , Humans , Injections , Male , Mice , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Tumor Burden , Xenograft Model Antitumor Assays
8.
Nat Med ; 4(6): 685-90, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9623977

ABSTRACT

The transfer of apoptosis genes to tumors is one of the most promising strategies for cancer gene therapy. We have shown that massive apoptosis occurs when wild-type p53 expression is induced in glioma cells carrying a p53 gene mutation. However, adenovirus-mediated p53 gene transfer is ineffective in causing apoptosis in glioma cells that retain a wild-type p53 genotype. We evaluated the effect of E2F-1 overexpression on the growth of gliomas in vitro and in vivo. In the in vitro study, the adenovirus-mediated transfer of exogenous E2F-1 protein precipitated generalized apoptosis in gliomas. The treatment with Ad5CMV-E2F-1 of nude mice carrying subcutaneous gliomas arrested tumor growth. Our results indicate that E2F-1 has anti-glioma activity in vitro and in vivo.


Subject(s)
Apoptosis/physiology , Carrier Proteins , Glioma/genetics , Transcription Factors/genetics , Adenoviruses, Human/genetics , Animals , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Death/genetics , Cell Death/physiology , Cell Survival/genetics , Cell Survival/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Disease Models, Animal , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression/genetics , Genes, Tumor Suppressor , Genetic Therapy , Genetic Vectors/genetics , Glioma/physiopathology , Glioma/therapy , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Oncogene Protein p21(ras)/metabolism , Recombinant Fusion Proteins/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/physiology , Transfection/genetics , Tumor Cells, Cultured
9.
Trials ; 22(1): 356, 2021 May 20.
Article in English | MEDLINE | ID: mdl-34016168

ABSTRACT

BACKGROUND: Chronic pelvic pain syndrome (CPPS) is a multifactorial disorder that affects 5.7% to 26.6% of women and 2.2% to 9.7% of men, characterized by hypersensitivity of the central and peripheral nervous system affecting bladder and genital function. People with CPPS have much higher rates of psychological disorders (anxiety, depression, and catastrophizing) that increase the severity of chronic pain and worsen quality of life. Myofascial therapy, manual therapy, and treatment of trigger points are proven therapeutic options for this syndrome. This study aims to evaluate the efficacy of capacitive resistive monopolar radiofrequency (CRMRF) at 448 kHz as an adjunct treatment to other physiotherapeutic techniques for reducing pain and improving the quality of life of patients with CPPS. METHODS: This triple-blind (1:1) randomized controlled trial will include 80 women and men with CPPS. Participants will be randomized into a CRMRF activated group or a CRMRF deactivated group and receive physiotherapeutic techniques and pain education. The groups will undergo treatment for 10 consecutive weeks. At the beginning of the trial there will be an evaluation of pain intensity (using VAS), quality of life (using the SF-12), kinesiophobia (using the TSK-11), and catastrophism (using the PCS), as well as at the sixth and tenth sessions. DISCUSSION: The results of this study will show that CRMRF benefits the treatment of patients with CPPS, together with physiotherapeutic techniques and pain education. These results could offer an alternative conservative treatment option for these patients. TRIAL REGISTRATION: ClinicalTrials.gov NCT03797911 . Registered on 8 January 2019.


Subject(s)
Chronic Pain , Chronic Pain/diagnosis , Chronic Pain/therapy , Female , Humans , Male , Pain Measurement , Pelvic Pain/diagnosis , Pelvic Pain/therapy , Quality of Life , Randomized Controlled Trials as Topic , Syndrome
10.
Gene Ther ; 16(12): 1441-51, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19710704

ABSTRACT

The E2F-1 promoter has been used to confer tumor-selective E1A expression in oncolytic adenoviruses. Tumor specificity is mainly conferred by a unique structure of E2F-responsive sites organized in palindromes. Binding of the E2F-pRb complex to these palindromes results in repression of transcription in normal cells. Owing to deregulation of the Rb/p16 pathway in tumor cells, binding of free E2F activates transcription and initiates an autoactivation loop involving E1A and E4-6/7. ICOVIR-7 is a new oncolytic adenovirus designed to increase the E2F dependency of E1A gene expression. It incorporates additional palindromes of E2F-responsive sites in an insulated E2F-1 promoter controlling E1A-Delta24. The E2F palindromes inhibited replication in normal cells, resulting in a low systemic toxicity at high doses in immunocompetent mice. The Delta24 deletion avoids a loop of E2F-mediated self-activation in nontumor cells. Importantly, the additional E2F-binding hairpins boost the positive feedback loop on the basis of E1A-mediated transcriptional regulation of E4-6/7 turned on in cancer cells and increased antitumoral potency as shown in murine subcutaneous xenograft models treated by intravenous injection. These results suggest that the unique genetic combination featured in ICOVIR-7 may be promising for treating disseminated neoplasias.


Subject(s)
Adenoviridae/genetics , Adenovirus E1A Proteins/biosynthesis , E2F1 Transcription Factor/genetics , Oncolytic Viruses/genetics , Promoter Regions, Genetic , Animals , Cell Line , Cell Line, Tumor , Gene Expression Regulation, Viral , Humans , Male , Mice , Mice, Nude , Oncolytic Virotherapy/methods , Virus Replication
11.
Gene Ther ; 15(17): 1240-5, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18509378

ABSTRACT

Fusogenic membrane glycoproteins (FMGs) may enhance the cytotoxicity of conditionally replicative adenoviruses. However, expression at early stages of infection impairs virus replication. We have inserted the hyperfusogenic form of the gibbon ape leukemia virus (GALV) envelope glycoprotein as a new splice unit of the major late promoter (MLP) to generate a replication-competent adenovirus expressing this protein. At high multiplicity of infection (MOI), this virus replicated efficiently forming clumps of fused cells and showing a faster release. In contrast, at low MOI, infected cells formed syncytia where only one nucleus contained virus DNA, decreasing total virus production but increasing cytotoxicity.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Giant Cells/physiology , Leukemia Virus, Gibbon Ape/physiology , Oncolytic Virotherapy/methods , Viral Fusion Proteins/genetics , Cell Line, Tumor , Gene Expression , Genetic Engineering , Humans , Leukemia Virus, Gibbon Ape/genetics , Promoter Regions, Genetic , Transgenes , Virus Replication
12.
Int J Obes (Lond) ; 32(3): 464-73, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18059405

ABSTRACT

OBJECTIVE: To investigate the relationship between chemical structure and physiological effect, the efficacy and the molecular mechanisms involved in the reduction of body weight by C18 fatty acids (stearic, elaidic, oleic, linoleic and 2-hydroxyoleic acids (2-OHOA)). DESIGN: Ad libitum fed, lean Wistar Kyoto rats treated orally with up to 600 mg kg(-1) of the fatty acids or vehicle every 12 h for 7 days. Besides, starved rats and rats pairfed to the 2-OHOA-treated group served as additional controls under restricted feeding conditions. MEASUREMENTS: Body weight, food intake, weight of various fat depots, plasma leptin, hypothalamic neuropeptides, uncoupling proteins (UCP) in white (WAT) and brown adipose tissue (BAT) and phosphorylation level of cyclic AMP (cAMP) response element-binding protein (CREB) in WAT. RESULTS: Only treatment with oleic acid and 2-OHOA induced body weight loss (3.3 and 11.4%, respectively) through reduction of adipose fat mass. Food intake in these rats was lower, although hypothalamic neuropeptide and plasma leptin levels indicated a rise in orexigenic status. Rats pairfed to the 2-hydroxyoleic group only lost 6.3% body weight. UCP1 expression and phosphorylation of CREB was drastically increased in WAT, but not BAT of 2-OHOA-treated rats, whereas no UCP1 expression could be detected in WAT of rats treated with oleic acid. CONCLUSION: Both cis-configured monounsaturated C18 fatty acids (oleic acid and 2-OHOA) reduce body weight, but the introduction of a hydroxyl group in position 2 drastically increases loss of adipose tissue mass. The novel molecular mechanism unique to 2-hydroxyoleic, but not oleic acid, implies induction of UCP1 expression in WAT by the cAMP/PKA pathway-dependent transcription factor CREB, most probably as part of a transdifferentiation process accompanied by enhanced energy expenditure.


Subject(s)
Adipose Tissue/physiology , Body Weight/physiology , Fatty Acids/administration & dosage , Adipose Tissue/metabolism , Animals , Feeding Behavior , Immunoblotting , Leptin/metabolism , Linoleic Acid/administration & dosage , Neuropeptides/metabolism , Oleic Acid/administration & dosage , Oleic Acids/administration & dosage , Random Allocation , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Stearic Acids/administration & dosage , Structure-Activity Relationship
13.
Cancer Gene Ther ; 14(8): 756-61, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17557108

ABSTRACT

Novel therapies are clearly needed for gliomas, and the combination of oncolytic vectors with chemotherapy possesses a significant hope for the treatment of this malignancy. In addition, combination with chemotherapy allows for lower virus doses to achieve anticancer effect, thus resulting in lower undesirable toxicities due to viral proteins. In this work, we sought to determine whether combination of an oncolytic adenovirus ICOVIR-5, with RAD001 or temozolomide (TMZ) could result in enhanced anti-glioma effect in vivo. We assessed the in vitro cytotoxic effect and replication properties of ICOVIR-5 in combination with RAD001 or TMZ in U87 MG glioma cell line by MTT and TCID(50), respectively. Our data showed that in vitro treatment with RAD001 or TMZ not only interfered with adenovirus replication but, in addition, enhanced its oncolytic properties. To evaluate the in vivo anticancer effect, athymic mice bearing glioma xenografts (5 x 10(5) U87 MG cells/animal) received a single intratumoral injection of ICOVIR-5 (10(7) PFU/animal). RAD001 was given as a regimen of 5 mg/kg 5 days per week until the end of the experiment and TMZ was administered for 5 days at 7.5 mg/kg/mice. Of significance, combination of ICOVIR-5 with RAD001 or TMZ showed a potent anti-glioma effect in vivo, resulting in a dramatic extension of the median animal survival and in 20-40% animals becoming free of disease beyond 90 days.


Subject(s)
Adenoviridae , Antineoplastic Agents, Alkylating/pharmacology , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Oncolytic Virotherapy , Sirolimus/analogs & derivatives , Animals , Cell Line, Tumor , Dacarbazine/pharmacology , Everolimus , Glioma/therapy , Humans , Mice , Mice, Nude , Neoplasms, Experimental/drug therapy , Sirolimus/pharmacology , Temozolomide
14.
Nat Biotechnol ; 18(7): 723-7, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10888838

ABSTRACT

Rapid advances are being made in the engineering of replication-competent viruses to treat cancer. Adenovirus is a mildly pathogenic human virus that propagates prolifically in epithelial cells, the origin of most human cancers. While virologists have revealed many details about its molecular interactions with the cell, applied scientists have developed powerful technologies to genetically modify or regulate every viral protein. In tandem, the limited success of nonreplicative adenoviral vectors in cancer gene therapy has brought the old concept of adenovirus oncolysis back into the spotlight. Major efforts have been directed toward achieving selective replication by the deletion of viral functions dispensable in tumor cells or by the regulation of viral genes with tumor-specific promoters. However, the predicted replication selectivity has not been realized because of incomplete knowledge of the complex virus-cell interactions and the leakiness of cellular promoters in the viral genome. Capsid modifications are being developed to achieve tumor targeting and enhance infectivity. Cellular and viral functions that confer greater oncolytic potency are also being elucidated. Ultimately, the interplay of the virus with the immune system will likely dictate the success of this approach as a cancer therapy.


Subject(s)
Adenoviridae/genetics , Adenoviridae/physiology , Genetic Therapy/methods , Neoplasms/therapy , Animals , Genetic Vectors , Humans , Models, Biological , Neoplasms/immunology
15.
Stem Cells Int ; 2017: 3615729, 2017.
Article in English | MEDLINE | ID: mdl-28781596

ABSTRACT

Antitumor efficacy of systemically administered oncolytic adenoviruses (OAdv) is limited due to diverse factors such as liver sequestration, neutralizing interactions in blood, elimination by the immune system, and physical barriers in tumors. It is therefore of clinical relevance to improve OAdv bioavailability and tumor delivery. Among the variety of tumor-targeting strategies, the use of stem cells and specifically bone marrow-derived mesenchymal stem cells (BM-MSCs) is of particular interest due to their tumor tropism and immunomodulatory properties. Nonetheless, the invasive methods to obtain these cells, the low number of MSCs present in the bone marrow, and their restricted in vitro expansion represent major obstacles for their use in cancer treatments, pointing out the necessity to identify an alternative source of MSCs. Here, we have evaluated the use of menstrual blood-derived mesenchymal stem cells (MenSCs) as cell carriers for regional delivery of an OAdv in the tumor. Our results indicate that MenSCs can be isolated without invasive methods, they have an increased proliferation rate compared to BM-MSCs, and they can be efficiently infected with different serotype 5-based capsid-modified adenoviruses, leading to viral replication and release. In addition, our in vivo studies confirmed the tumor-homing properties of MenSCs after regional administration.

16.
Cancer Gene Ther ; 13(7): 696-705, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16498429

ABSTRACT

We previously described Ad-Delta24RGD as an enhanced-infectivity oncolytic adenovirus that targets tumors with an impaired RB pathway. The common alteration of this pathway in cancer eliminates the interaction of pRB with E2F and releases free E2F to activate E2F-responsive promoters, including the E2F-1 promoter. To improve the selectivity towards RB pathway-defective tumors and reduce the toxicity of Ad-Delta24RGD we aimed to control E1A-Delta24 expression under the E2F-1 promoter. A polyA signal was inserted upstream of the E2F-1 promoter to stop transcription initiated at the adenovirus ITR and packaging signal. The human myotonic dystropy locus insulator (DM-1) was also located between the E1a enhancers and the E2F-1 promoter to further insulate the promoter. The Ad-Delta24RGD derivative containing these insulation sequences expressed less E1a-Delta24 in normal cells and resulted less toxic while maintaining the potent oncolytic activity of the parental virus. These results demonstrate that the human DM-1 inslulator can function in an adenovirus context to maintain heterologous promoter selectivity. The new oncolytic adenovirus presented here may represent a valuable therapeutic option for a broad range of tumors with a deregulated E2F/pRB pathway.


Subject(s)
Adenoviridae/genetics , Adenovirus E1A Proteins/genetics , E2F1 Transcription Factor/genetics , Gene Expression Regulation, Viral/genetics , Promoter Regions, Genetic/genetics , Cell Line, Transformed , Cell Line, Tumor , Codon, Terminator/genetics , Genetic Therapy , Humans , Neoplasms/genetics , Neoplasms/therapy , Polyadenylation/genetics
17.
Cancer Res ; 61(17): 6377-81, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11522629

ABSTRACT

Cancer gene therapy with conditionally replicating adenoviruses is a powerful way of overcoming low tumor transduction. However, one of the main remaining obstacles is the highly variable level of the coxsackie-adenovirus receptor expression on human primary cancers. In contrast, the epidermal growth factor receptor (EGFR) is overexpressed in various tumor types, and its expression correlates with metastatic behavior and poor prognosis. We constructed an adenovirus expressing a secretory adaptor capable of retargeting adenovirus to EGFR, resulting in a more than 150-fold increase in gene transfer. A replication-competent dual-virus system secreting the adaptor displayed increased oncolytic potency in vitro and therapeutic gain in vivo. This approach could translate into increased efficacy and specificity in the treatment of EGFR overexpressing human cancers.


Subject(s)
Adenoviridae/genetics , ErbB Receptors/metabolism , Genetic Therapy/methods , Recombinant Fusion Proteins/metabolism , Adenoviridae/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/therapy , Coxsackie and Adenovirus Receptor-Like Membrane Protein , ErbB Receptors/genetics , HeLa Cells , Humans , Mice , Mice, Nude , Receptors, Virus/genetics , Receptors, Virus/metabolism , Recombinant Fusion Proteins/genetics , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/therapy , Tumor Cells, Cultured , Virus Replication , Xenograft Model Antitumor Assays
18.
Cancer Res ; 56(13): 3047-54, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8674061

ABSTRACT

In most cervical cancers, the function of p53 is down regulated. To explore the potential use of p53 in gene therapy for cervical cancer, we introduced wild-type p53 into cervical cancer cell lines via a recombinant adenoviral vector, Ad5CMV-p53, and analyzed its effects on cell and tumor growth. The transduction efficiencies of all cell lines were 100% at a multiplicity of infection of 100 or greater. The p53 protein was detected in Ad5CMV-p53-infected cells. Protein expression peaked at day 3 after infection and lasted 15 days. The Ad5CMV-p53-infected cells underwent apoptosis, and cell growth was greatly suppressed. The Ad5CMV-p53 treatment significantly reduced the volumes of established s.c. tumors in vivo. These results indicate that transfection of cervical cancer cells with the wild-type p53 gene via Ad5CMV-p53 is a potential novel approach to the therapy of cervical cancer.


Subject(s)
Apoptosis/physiology , Genes, p53 , Genetic Therapy , Uterine Cervical Neoplasms/therapy , Adenoviridae/genetics , Adenoviridae/metabolism , Adenovirus Infections, Human/genetics , Animals , Cell Division/physiology , Female , Gene Expression , Genetic Vectors , Humans , Mice , Mice, Nude , Transduction, Genetic , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology
19.
Cancer Gene Ther ; 23(12): 411-414, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27767087

ABSTRACT

There is great skepticism in the capability of adenovirus vectors and oncolytic adenoviruses to reach specific organs or tumors upon systemic administration. Besides antibodies, the presence of CAR (coxsackie and adenovirus receptor) in human erythrocytes has been postulated to sequester CAR-binding adenoviruses, commonly used in gene therapy and oncolytic applications. The use of non-CAR-binding fibers or serotypes has been postulated to solve this limitation. Given the lack of integrins in erythrocytes and therefore of internalization of the CAR-bound virus, we hypothesized that the interaction of adenovirus type 5 (Ad5) with CAR in human erythrocytes could be reversible. In this work, we have studied the effects of Ad5 interaction with human erythrocytes via CAR. Although erythrocyte binding was observed, it did not reduce viral transduction of tumor cells in vitro after long-term incubations. Transplantation of human erythrocytes into nude mice did not reduce Ad5 extravasation and transduction of liver and human xenograft tumors after systemic administration. These findings indicate that despite human erythrocytes are able to bind to Ad5, this binding is reversible and does not prevent extravasation and organ transduction after systemic delivery. Thus, the poor bioavailability of systemically delivered CAR-binding adenoviruses in humans is likely due to other factors such as liver sequestration or neutralizing antibodies.


Subject(s)
Adenoviridae/physiology , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Erythrocytes/metabolism , Erythrocytes/virology , Transduction, Genetic , Virus Attachment , Adenoviridae Infections/metabolism , Adenoviridae Infections/virology , Adenoviruses, Human/physiology , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Virus Internalization
20.
Oncogene ; 19(1): 2-12, 2000 Jan 06.
Article in English | MEDLINE | ID: mdl-10644974

ABSTRACT

Effective anti cancer strategies necessitate the use of agents that target tumor cells rather than normal tissues. In this study, we constructed a tumor-selective adenovirus, Delta24, that carries a 24-bp deletion in the E1A region responsible for binding Rb protein. Immunoprecipitation analyses verified that this deletion rendered Delta24 unable to bind the Rb protein. However, titration experiments in 293 cells demonstrated that the Delta24 adenovirus could replicate in and lyse cancer cells with great efficiency. Lysis of most human glioma cells was observed within 10 - 14 days after infection with Delta24 at 10 PFU/cell. In vivo, a single dose of the Delta24 virus induced a 66.3% inhibition (P<0.005) and multiple injections, an 83.8% inhibition (P<0.01) of tumor growth in nude mice. However, normal fibroblasts or cancer cells with restored Rb activity were resistant to the Delta24 adenovirus. These results suggest that the E1A-mutant Delta24 adenovirus may be clinically and therapeutically useful against gliomas and possibly other cancers with disrupted Rb pathway.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Glioma/therapy , Retinoblastoma Protein/physiology , Animals , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , S Phase , Tumor Cells, Cultured , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL