Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Article in English | MEDLINE | ID: mdl-33075494

ABSTRACT

MicroRNA-221-3p (miR-221-3p) is associated with both metabolic diseases and cancers. However, its role in terminal adipocyte differentiation and lipid metabolism are uncharacterized. miR-221-3p or its inhibitor was transfected into differentiating or mature human adipocytes. Triglyceride (TG) content and adipogenic gene expression were monitored, global lipidome analysis was carried out, and mechanisms underlying the effects of miR-221-3p were investigated. Finally, cross-talk between miR-221-3p expressing adipocytes and MCF-7 breast carcinoma (BC) cells was studied, and miR-221-3p expression in tumor-proximal adipose biopsies from BC patients analyzed. miR-221-3p overexpression inhibited terminal differentiation of adipocytes, as judged from reduced TG storage and gene expression of the adipogenic markers SCD1, GLUT4, FAS, DGAT1/2, AP2, ATGL and AdipoQ, whereas the miR-221-3p inhibitor increased TG storage. Knockdown of the predicted miR-221-3p target, 14-3-3γ, had similar antiadipogenic effects as miR-221-3p overexpression, indicating it as a potential mediator of mir-221-3p function. Importantly, miR-221-3p overexpression inhibited de novo lipogenesis but increased the concentrations of ceramides and sphingomyelins, while reducing diacylglycerols, concomitant with suppression of sphingomyelin phosphodiesterase, ATP citrate lyase, and acid ceramidase. miR-221-3p expression was elevated in tumor proximal adipose tissue from patients with invasive BC. Conditioned medium of miR-221-3p overexpressing adipocytes stimulated the invasion and proliferation of BC cells, while medium of the BC cells enhanced miR-221-3p expression in adipocytes. Elevated miR-221-3p impairs adipocyte lipid storage and differentiation, and modifies their ceramide, sphingomyelin, and diacylglycerol content. These alterations are relevant for metabolic diseases but may also affect cancer progression.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lipid Droplets/metabolism , MicroRNAs/genetics , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Adipocytes/pathology , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adult , Aged , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Case-Control Studies , Cell Differentiation , Cell Proliferation , Ceramides/classification , Ceramides/metabolism , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Humans , Lipase/genetics , Lipase/metabolism , MCF-7 Cells , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Middle Aged , Signal Transduction , Sphingolipids/classification , Sphingolipids/metabolism , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Triglycerides/classification , Triglycerides/metabolism , fas Receptor/genetics , fas Receptor/metabolism
2.
Oncoimmunology ; 6(2): e1265717, 2017.
Article in English | MEDLINE | ID: mdl-28344872

ABSTRACT

Vaccination with dendritic cells (DCs), the most potent professional antigen-presenting cells in the body, is a promising approach in cancer immunotherapy. However, tumors induce immunosuppression in their microenvironment that suppresses and impairs the function of DCs. Therefore, human clinical trials with DC therapy have often been disappointing. To improve the therapeutic efficacy and to overcome the major obstacles of DC therapy, we generated a novel adenovirus, Ad3-hTERT-CMV-hCD40L, which is fully serotype 3 and expresses hCD40L for induction of antitumor immune response. The specific aim is to enhance DCs function. Data from a human cancer patient indicated that this capsid allows effective transduction of distant tumors through the intravenous route. Moreover, patient data suggested that virally produced hCD40L can activate DCs in situ. The virus was efficient in vitro and had potent antitumor activity in vivo. In a syngeneic model, tumors treated with Ad5/3-CMV-mCD40L virus plus DCs elicited greater antitumor effect as compared with either treatment alone. Moreover, virally coded CD40L induced activation of DCs, which in turn, lead to the induction of a Th1 immune response and increased tumor-specific T cells. In conclusion, Ad3-hTERT-CMV-hCD40L is promising for translation into human trials. In particular, this virus could enable successful dendritic cell therapy in cancer patients.

4.
J Immunother ; 39(9): 343-354, 2016.
Article in English | MEDLINE | ID: mdl-27741089

ABSTRACT

The immunosuppressive microenvironment of solid tumors renders adoptively transferred T cells hypofunctional. However, adenoviral delivery of immunostimulatory cytokines IL2 and TNFα can significantly improve the efficacy of adoptive T-cell therapy. Using ret transgenic mice that spontaneously develop skin malignant melanoma, we analyzed the mechanism of action of adenoviruses coding for IL2 and TNFα in combination with adoptive transfer of TCR-transgenic TRP-2-specific T cells. Following T-cell therapy and intratumoral virus injection, a significant increase in antigen-experienced, tumor-reactive PD-1 CD8 T cells was seen in both cutaneous lesions and in metastatic lymph nodes. A reverse correlation between tumor weight and the number of tumor-reactive PD-1 tumor-infiltrating lymphocytes (TILs) was observed, suggesting that these T cells could target and kill tumor cells. It is interesting to note that, local expression of cytokines did not affect intratumoral levels of T-regulatory cells (Tregs), which had previously been associated with systemic IL2 therapy. Instead, Ad5-IL2 induced upregulation of IL2 receptor α-chain (CD25) on conventional CD4CD25Foxp3 cells, indicating that these CD4 T cells may contribute to CD8 T-cell activation and/or homing. Signs of therapy-induced resistance were also observed as the expression of PD-L1 on tumor-infiltrating granulocytic myeloid-derived suppressor cells was upregulated as a reaction to PD-1+ TILs. Finally, beneficial ratios between tumor-reactive PD-1 CD8 TILs and immunosuppressive cell subsets (Tregs and nitric oxide-producing myeloid-derived suppressor cells) were observed in primary and secondary tumor sites, indicating that local delivery of IL2 and TNFα coding adenoviruses can systemically modify the cellular composition of the tumor microenvironment in favor of adoptively transferred T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Melanoma/therapy , Skin Neoplasms/therapy , T-Lymphocyte Subsets/immunology , Adenoviridae/genetics , Animals , CD8-Positive T-Lymphocytes/transplantation , Cell Proliferation , Cells, Cultured , Genetic Vectors/genetics , Immunomodulation , Interleukin-2/genetics , Interleukin-2/metabolism , Melanoma/genetics , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-ret/genetics , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Tumor Burden , Tumor Microenvironment , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
Mol Ther ; 24(8): 1435-43, 2016 08.
Article in English | MEDLINE | ID: mdl-27357626

ABSTRACT

Adoptive T-cell transfer is a promising treatment approach for metastatic cancer, but efficacy in solid tumors has only been achieved with toxic pre- and postconditioning regimens. Thus, adoptive T-cell therapies would benefit from complementary modalities that enable their full potential without excessive toxicity. We aimed to improve the efficacy and safety of adoptive T-cell transfer by using adenoviral vectors for direct delivery of immunomodulatory murine cytokines into B16.OVA melanoma tumors with concomitant T-cell receptor transgenic OT-I T-cell transfer. Armed adenoviruses expressed high local and low systemic levels of cytokine when injected into B16.OVA tumors, suggesting safety of virus-mediated cytokine delivery. Antitumor efficacy was significantly enhanced with adenoviruses coding for murine interleukin-2 (mIL-2) and tumor necrosis factor-α (mTNFα) when compared with T-cell transfer alone or viruses alone. Further improvement in efficacy was achieved with a triple combination of mIL-2, mTNFα, and OT-I T-cells. Mechanistic studies suggest that mIL-2 has an important role in activating T-cells at the tumor, while mTNFα induces chemokine expression. Furthermore, adenovirus treatments enhanced tumor-infiltration of OT-I T-cells as demonstrated by SPECT/CT imaging of (111)In-labeled cells. Our results suggest the utility of cytokine-coding adenoviruses for improving the efficacy of adoptive T-cell therapies.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Immunotherapy, Adoptive , Interleukin-2/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Tumor Necrosis Factor-alpha/genetics , Animals , B7-H1 Antigen/metabolism , Cell- and Tissue-Based Therapy , Disease Models, Animal , Gene Expression , Genetic Therapy , Genetic Vectors/administration & dosage , Immunocompromised Host , Injections, Intralesional , Interleukin-2/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/diagnosis , Melanoma, Experimental/therapy , Mice , Programmed Cell Death 1 Receptor/metabolism , Single Photon Emission Computed Tomography Computed Tomography , Tumor Necrosis Factor-alpha/metabolism
6.
Oncoimmunology ; 5(5): e1136046, 2016 May.
Article in English | MEDLINE | ID: mdl-27467954

ABSTRACT

Adoptive transfer of tumor-infiltrating lymphocytes (TIL) has shown promising yet sometimes suboptimal results in clinical trials for advanced cancer, underscoring the need for approaches improving efficacy and safety. Six implantable syngeneic tumor cell lines of the Syrian hamster were used to initiate TIL cultures. TIL generated from tumor fragments cultured in human interleukin-2 (IL-2) for 10 d were adoptively transferred into tumor-bearing hamsters with concomitant intratumoral injections of oncolytic adenovirus (Ad5-D24) for the assessment of antitumor efficacy. Pancreatic cancer (HapT1) and melanoma (RPMI 1846) TIL exhibited potent and tumor-specific cytotoxicity in effector-to-target (E/T) assays. MHC Class I blocking abrogated the cell killing of RPMI 1846 TIL, indicating cytotoxic CD8(+) T-cell activity. When TIL were combined with Ad5-D24 in vitro, HapT1 tumor cell killing was significantly enhanced over single agents. In vivo, the intratumoral administration of HapT1 TIL and Ad5-D24 resulted in improved tumor growth control compared with either treatment alone. Additionally, splenocytes derived from animals treated with the combination of Ad5-D24 and TIL killed autologous tumor cells more efficiently than monotherapy-derived splenocytes, suggesting that systemic antitumor immunity was induced. For the first time, TIL of the Syrian hamster have been cultured, characterized and used therapeutically together with oncolytic adenovirus for enhancing the efficacy of TIL therapy. Our results support human translation of oncolytic adenovirus as an enabling technology for adoptive T-cell therapy of solid tumors.

7.
PLoS One ; 10(6): e0131242, 2015.
Article in English | MEDLINE | ID: mdl-26107883

ABSTRACT

Unfavorable ratios between the number and activation status of effector and suppressor immune cells infiltrating the tumor contribute to resistance of solid tumors to T-cell based therapies. Here, we studied the capacity of FDA and EMA approved recombinant cytokines to manipulate this balance in favor of efficient anti-tumor responses in B16.OVA melanoma bearing C57BL/6 mice. Intratumoral administration of IFN-α2, IFN-γ, TNF-α, and IL-2 significantly enhanced the anti-tumor effect of ovalbumin-specific CD8+ T-cell (OT-I) therapy, whereas GM-CSF increased tumor growth in association with an increase in immunosuppressive cell populations. None of the cytokines augmented tumor trafficking of OT-I cells significantly, but injections of IFN-α2, IFN-γ and IL-2 increased intratumoral cytokine secretion and recruitment of endogenous immune cells capable of stimulating T-cells, such as natural killer and maturated CD11c+ antigen-presenting cells. Moreover, IFN-α2 and IL-2 increased the levels of activated tumor-infiltrating CD8+ T-cells concomitant with reduction in the CD8+ T-cell expression of anergy markers CTLA-4 and PD-1. In conclusion, intratumoral administration of IFN-α2, IFN-γ and IL-2 can lead to immune sensitization of the established tumor, whereas GM-CSF may contribute to tumor-associated immunosuppression. The results described here provide rationale for including local administration of immunostimulatory cytokines into T-cell therapy regimens. One appealing embodiment of this would be vectored delivery which could be advantageous over direct injection of recombinant molecules with regard to efficacy, cost, persistence and convenience.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cytokines/metabolism , Neoplasms/therapy , Tumor Microenvironment , Animals , Antigen-Presenting Cells/cytology , Female , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immune Tolerance , Immunotherapy/methods , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Interleukin-2/pharmacology , Lymphocyte Activation/immunology , Lymphocyte Subsets , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Recombinant Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology
8.
PLoS One ; 10(3): e0120496, 2015.
Article in English | MEDLINE | ID: mdl-25799430

ABSTRACT

Vaccinia virus is a large, enveloped virus of the poxvirus family. It has broad tropism and typically virus replication culminates in accumulation and lytic release of intracellular mature virus (IMV), the most abundant form of infectious virus, as well as release by budding of extracellular enveloped virus (EEV). Vaccinia viruses have been modified to replicate selectively in cancer cells and clinically tested as oncolytic agents. During preclinical screening of relevant cancer targets for a recombinant Western Reserve strain deleted for both copies of the thymidine kinase and vaccinia growth factor genes, we noticed that confluent monolayers of SCCF1 cat squamous carcinoma cells were not destroyed even after prolonged infection. Interestingly, although SCCF1 cells were not killed, they continuously secreted virus into the cell culture supernatant. To investigate this finding further, we performed detailed studies by electron microscopy. Both intracellular and secreted virions showed morphological abnormalities on ultrastructural inspection, suggesting compromised maturation and morphogenesis of vaccinia virus in SCCF1 cells. Our data suggest that SCCF1 cells produce a morphologically abnormal virus which is nevertheless infective, providing new information on the virus-host cell interactions and intracellular biology of vaccinia virus.


Subject(s)
Oncolytic Viruses , Vaccinia virus/physiology , Virion/physiology , Animals , Cats , Cell Line, Tumor , Humans , Transduction, Genetic , Vaccinia virus/genetics , Vaccinia virus/metabolism , Vaccinia virus/ultrastructure , Virion/genetics , Virion/metabolism , Virion/ultrastructure
9.
Oncotarget ; 6(6): 4467-81, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25714011

ABSTRACT

Oncolytic viruses that selectively replicate in tumor cells can be used for treatment of cancer. Accumulating data suggests that virus induced oncolysis can enhance anti-tumor immunity and break immune tolerance. To capitalize on the immunogenic nature of oncolysis, we generated a quadruple modified oncolytic adenovirus expressing granulocyte-macrophage colony-stimulating factor (GMCSF). Ad5/3-E2F-Δ24-GMCSF (CGTG-602) was engineered to contain a tumor specific E2F1 promoter driving an E1 gene deleted at the retinoblastoma protein binding site ("Δ24"). The fiber features a knob from serotype 3 for enhanced gene delivery to tumor cells. The virus was tested preclinically in vitro and in vivo and then 13 patients with solid tumors refractory to standard therapies were treated. Treatments were well tolerated and frequent tumor- and adenovirus-specific T-cell immune responses were seen. Overall, with regard to tumor marker or radiological responses, signs of antitumor efficacy were seen in 9/12 evaluable patients (75%). The radiological disease control rate with positron emission tomography was 83% while the response rate (including minor responses) was 50%. Tumor biopsies indicated accumulation of immunological cells, especially T-cells, to tumors after treatment. RNA expression analyses of tumors indicated immunological activation and metabolic changes secondary to virus replication.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Immunotherapy/methods , Neoplasms/therapy , Oncolytic Virotherapy/methods , Adult , Aged , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cricetinae , E2F1 Transcription Factor/genetics , Female , Humans , Male , Mesocricetus , Middle Aged , Oncolytic Viruses , Promoter Regions, Genetic , Xenograft Model Antitumor Assays
10.
Hum Gene Ther ; 26(3): 134-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25557131

ABSTRACT

For long it has been recognized that tumor necrosis factor alpha (TNFa) has anticancer characteristics, and its use as a cancer therapeutic was proposed already in the 1980s. However, its systemic toxicity has limited its usability. Oncolytic viruses, selectively cancer-killing viruses, have shown great potency, and one of their most useful aspects is their ability to produce high amounts of transgene products locally, resulting in high local versus systemic concentrations. Therefore, the overall magnitude of tumor cell killing results from the combination of oncolysis, transgene-mediated direct effect such as TNFa-mediated apoptosis, and, perhaps most significantly, from activation of the host immune system against the tumor. We generated a novel chimeric oncolytic adenovirus expressing human TNFa, Ad5/3-D24-hTNFa, whose efficacy and immunogenicity were tested in vitro and in vivo. The hTNFa-expressing adenovirus showed increased cancer-eradicating potency, which was shown to be because of elevated apoptosis and necrosis rates and induction of various immune responses. Interestingly, we saw increase in immunogenic cell death markers in Ad5/3-d24-hTNFa-treated cells. Moreover, tumors treated with Ad5/3-D24-hTNFa displayed enhanced presence of OVA-specific cytotoxic T cells. We thus can conclude that tumor eradication and antitumor immune responses mediated by Ad5/3-d24-hTNFa offer a new potential drug candidate for cancer therapy.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/therapeutic use , Neoplasms/genetics , Neoplasms/therapy , Oncolytic Virotherapy/methods , Tumor Necrosis Factor-alpha/metabolism , Apoptosis/immunology , Cell Line, Tumor , Genetic Vectors/immunology , HEK293 Cells , Humans , Neoplasms/immunology , Statistics, Nonparametric , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor-alpha/genetics
11.
Int J Cancer ; 136(5): 1065-72, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25042001

ABSTRACT

Oncolytic Western Reserve strain vaccinia virus selective for epidermal growth factor receptor pathway mutations and tumor-associated hypermetabolism was armed with human granulocyte-macrophage colony-stimulating factor (GMCSF) and a tdTomato fluorophore. As the assessment of immunological responses to human transgenes is challenging in the most commonly used animal models, we used immunocompetent Syrian golden hamsters, known to be sensitive to human GMCSF and semipermissive to vaccinia virus. Efficacy was initially tested in vitro on various human and hamster cell lines and oncolytic potency of transgene-carrying viruses was similar to unarmed virus. The hGMCSF-encoding virus was able to completely eradicate subcutaneous pancreatic tumors in hamsters, and to fully protect the animals from subsequent rechallenge with the same tumor. Induction of specific antitumor immunity was also shown by ex vivo co-culture experiments with hamster splenocytes. In addition, histological examination revealed increased infiltration of neutrophils and macrophages in GMCSF-virus-treated tumors. These findings help clarify the mechanism of action of GMCSF-armed vaccinia viruses undergoing clinical trials.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Oncolytic Virotherapy , Pancreatic Neoplasms/immunology , Vaccinia virus/genetics , Virus Replication/immunology , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Chlorocebus aethiops , Coculture Techniques , Cricetinae , DNA, Viral/genetics , Humans , Immunoenzyme Techniques , Macrophages , Mesocricetus , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes , Vaccinia virus/immunology , Vero Cells , Xenograft Model Antitumor Assays
12.
Int J Cancer ; 136(4): 945-54, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-24975392

ABSTRACT

Despite originating from several different tissues, soft-tissue sarcomas (STS) are often grouped together as they share mesenchymal origin and treatment guidelines. Also, with some exceptions, a common denominator is that when the tumor cannot be cured with surgery, the efficacy of current therapies is poor and new treatment modalities are thus needed. We have studied the combination of a capsid-modified oncolytic adenovirus CGTG-102 (Ad5/3-D24-GMCSF) with doxorubicin, with or without ifosfamide, the preferred first-line chemotherapeutic options for most types of STS. We show that CGTG-102 and doxorubicin plus ifosfamide together are able to increase cell killing of Syrian hamster STS cells over single agents, as well as upregulate immunogenic cell death markers. When tested in vivo against established STS tumors in fully immunocompetent Syrian hamsters, the combination was highly effective. CGTG-102 and doxorubicin (without ifosfamide) resulted in synergistic antitumor efficacy against human STS xenografts in comparison with single agent treatments. Doxorubicin increased adenoviral replication in human and hamster STS cells, potentially contributing to the observed therapeutic synergy. In conclusion, the preclinical data generated here support clinical translation of the combination of CGTG-102 and doxorubicin, or doxorubicin plus ifosfamide, for the treatment of STS, and provide clues on the mechanisms of synergy.


Subject(s)
Adenoviridae/immunology , Antibiotics, Antineoplastic/therapeutic use , Doxorubicin/therapeutic use , Leiomyosarcoma/therapy , Melanoma, Experimental/therapy , Oncolytic Viruses/immunology , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Survival , Combined Modality Therapy , Cricetinae , Doxorubicin/pharmacology , Drug Synergism , Female , Humans , Ifosfamide/pharmacology , Ifosfamide/therapeutic use , Leiomyosarcoma/immunology , Male , Melanoma, Experimental/immunology , Mesocricetus , Mice , Mice, Inbred C57BL , Mice, Nude , Oncolytic Virotherapy , Sarcoma , Virus Replication , Xenograft Model Antitumor Assays
13.
Mol Ther Oncolytics ; 1: 14002, 2014.
Article in English | MEDLINE | ID: mdl-27119092

ABSTRACT

We evaluated adverse events, biodistribution and shedding of oncolytic vaccinia virus encoding CD40 ligand in two Beagles, in preparation for a phase 1 trial in canine cancer patients. Dog 1 received one dose of vaccinia virus and was euthanized 24 hours afterwards, while dog 2 received virus four times once weekly and was euthanized 7 days after that. Dogs were monitored for adverse events and underwent a detailed postmortem examination. Blood, saliva, urine, feces, and organs were collected for virus detection. Dog 1 had mild fever and lethargy while dog 2 experienced a possible seizure 5.5 hours after first virus administration. Viral DNA declined quickly in the blood after virus administration in both dogs but was still detectable 1 week later by quantitative polymerase chain reaction. Only samples taken directly after virus infusion contained infectious virus. Small amounts of viral DNA, but no infectious virus, were detected in a few saliva and urine samples. Necropsies did not reveal any relevant pathological changes and virus DNA was detected mainly in the spleen. The dogs in the study did not have cancer, and thus adverse events could be more common and viral load higher in dogs with tumors which allow viral amplification.

14.
Int J Cancer ; 135(3): 720-30, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24374597

ABSTRACT

Sarcomas are a relatively rare cancer, but often incurable at the late metastatic stage. Oncolytic immunotherapy has gained attention over the past years, and a wide range of oncolytic viruses have been delivered via intratumoral injection with positive safety and promising efficacy data. Here, we report preclinical and clinical results from treatment of sarcoma with oncolytic adenovirus Ad5/3-D24-GMCSF (CGTG-102). Ad5/3-D24-GMCSF is a serotype chimeric oncolytic adenovirus coding for human granulocyte-macrophage colony-stimulating factor (GM-CSF). The efficacy of Ad5/3-D24-GMCSF was evaluated on a panel of soft-tissue sarcoma (STS) cell lines and in two animal models. Sarcoma specific human data were also collected from the Advanced Therapy Access Program (ATAP), in preparation for further clinical development. Efficacy was seen in both in vitro and in vivo STS models. Fifteen patients with treatment-refractory STS (13/15) or primary bone sarcoma (2/15) were treated in ATAP, and treatments appeared safe and well-tolerated. A total of 12 radiological RECIST response evaluations were performed, and two cases of minor response, six cases of stable disease and four cases of progressive disease were detected in patients progressing prior to virus treatment. Overall, the median survival time post treatment was 170 days. One patient is still alive at 1,459 days post virus treatment. In summary, Ad5/3-D24-GMCSF appears promising for the treatment of advanced STS; a clinical trial for treatment of refractory injectable solid tumors including STS is ongoing.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Oncolytic Virotherapy , Sarcoma/therapy , Animals , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Injections, Intralesional , Mesocricetus , Mice , Mice, Nude , Prognosis , Sarcoma/blood , Sarcoma/mortality , Survival Rate , Tumor Cells, Cultured , Virus Replication , Xenograft Model Antitumor Assays
15.
Cancer Res ; 72(9): 2327-38, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22396493

ABSTRACT

Oncolytic adenovirus is an attractive platform for immunotherapy because virus replication is highly immunogenic and not subject to tolerance. Although oncolysis releases tumor epitopes and provides costimulatory danger signals, arming the virus with immunostimulatory molecules can further improve efficacy. CD40 ligand (CD40L, CD154) induces apoptosis of tumor cells and triggers several immune mechanisms, including a T-helper type 1 (T(H)1) response, which leads to activation of cytotoxic T cells and reduction of immunosuppression. In this study, we constructed a novel oncolytic adenovirus, Ad5/3-hTERT-E1A-hCD40L, which features a chimeric Ad5/3 capsid for enhanced tumor transduction, a human telomerase reverse transcriptase (hTERT) promoter for tumor selectivity, and human CD40L for increased efficacy. Ad5/3-hTERT-E1A-hCD40L significantly inhibited tumor growth in vivo via oncolytic and apoptotic effects, and (Ad5/3-hTERT-E1A-hCD40L)-mediated oncolysis resulted in enhanced calreticulin exposure and HMGB1 and ATP release, which were suggestive of immunogenicity. In two syngeneic mouse models, murine CD40L induced recruitment and activation of antigen-presenting cells, leading to increased interleukin-12 production in splenocytes. This effect was associated with induction of the T(H)1 cytokines IFN-γ, RANTES, and TNF-α. Tumors treated with Ad5/3-CMV-mCD40L also displayed an enhanced presence of macrophages and cytotoxic CD8(+) T cells but not B cells. Together, our findings show that adenoviruses coding for CD40L mediate multiple antitumor effects including oncolysis, apoptosis, induction of T-cell responses, and upregulation of T(H)1 cytokines.


Subject(s)
CD40 Ligand/genetics , CD40 Ligand/immunology , Neoplasms/immunology , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Animals , Apoptosis/immunology , Cell Line, Tumor , Cytokines/immunology , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/virology , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasms/genetics , Neoplasms/virology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/virology , Xenograft Model Antitumor Assays
16.
Int J Cancer ; 131(1): 253-63, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-21834073

ABSTRACT

Conventional cancer treatments often have little impact on the course of advanced pancreatic cancer. Although cancer gene therapy with adenoviruses is a promising developmental approach, the primary receptor is poorly expressed in pancreatic cancers which might compromise efficacy and thus targeting to other receptors could be beneficial. Extended stealth delivery, combination with standard chemotherapy or circumvention of host antiadenoviral immune response might improve efficacy further. In this work, capsid-modified adenoviruses were studied for transduction of cell lines and clinical normal and tumor tissue samples. The respective oncolytic viruses were tested for oncolytic activity in vitro and in vivo. Survival was studied in a peritoneally disseminated pancreas cancer model, with or without concurrent gemcitabine while silica implants were utilized for extended intraperitoneal virus delivery. Immunocompetent mice and Syrian hamsters were used to study the effect of silica mediated delivery on antiviral immune responses and subsequent in vivo gene delivery. Capsid modifications selectively enhanced gene transfer to malignant pancreatic cancer cell lines and clinical samples. The respective oncolytic viruses resulted in increased cell killing in vitro, which translated into a survival benefit in mice. Early proinfammatory cytokine responses and formation of antiviral neutralizing antibodies was partially avoided with silica implants. The implant also shielded the virus from pre-existing neutralizing antibodies, while increasing the pancreas/liver gene delivery ratio six-fold. In conclusion, capsid modified adenoviruses would be useful for testing in pancreatic cancer trials. Silica implants might increase the safety and efficacy of the approach.


Subject(s)
Adenoviruses, Human , Deoxycytidine/analogs & derivatives , Oncolytic Virotherapy/methods , Oncolytic Viruses , Pancreatic Neoplasms/therapy , Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Animals , Antibodies, Neutralizing , Capsid/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line, Tumor , Combined Modality Therapy , Cricetinae , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Female , Humans , Mice , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Silica Gel , Transfection , Gemcitabine
17.
PLoS One ; 5(9)2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20927353

ABSTRACT

BACKGROUND: Adenoviruses are attractive vectors for gene therapy because of their stability in vivo and the possibility of production at high titers. Despite exciting preclinical data with various approaches, there are only a few examples of clear efficacy in clinical trials. Effective gene delivery to target cells remains the key variable determining efficacy and thus enhanced transduction methods are important. METHODS/RESULTS: We found that heated serum could enhance adenovirus 5 mediated gene delivery up to twentyfold. A new protein-level interaction was found between fiber knob and serum transthyretin, but this was not responsible for the observed effect. Instead, we found that heating caused the calcium and phosphate present in the serum mix to precipitate, and this was responsible for enhanced gene delivery. This finding could have relevance for designing preclinical experiments with adenoviruses, since calcium and phosphate are present in many solutions. To translate this into an approach potentially testable in patients, we used calcium gluconate in phosphate buffered saline, both of which are clinically approved, to increase adenoviral gene transfer up to 300-fold in vitro. Gene transfer was increased with or without heating and in a manner independent from the coxsackie-adenovirus receptor. In vivo, in mouse studies, gene delivery was increased 2-, 110-, 12- and 13-fold to tumors, lungs, heart and liver and did not result in increased pro-inflammatory cytokine induction. Antitumor efficacy of a replication competent virus was also increased significantly. CONCLUSION: In summary, adenoviral gene transfer and antitumor efficacy can be enhanced by calcium gluconate in phosphate buffered saline.


Subject(s)
Adenoviridae/genetics , Calcium Gluconate/pharmacology , Gene Transfer Techniques , Genetic Vectors/genetics , Phosphates/pharmacology , Adenoviridae/drug effects , Adenoviridae/metabolism , Animals , Buffers , Cell Line, Tumor , Cricetinae , Gene Transfer Techniques/instrumentation , Genetic Therapy/instrumentation , Genetic Vectors/drug effects , Genetic Vectors/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Nude
SELECTION OF CITATIONS
SEARCH DETAIL
...