Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
2.
Front Immunol ; 11: 1996, 2020.
Article in English | MEDLINE | ID: mdl-32903551

ABSTRACT

Lung cancer is one of the most commonly diagnosed cancer and despite therapeutic advances, mortality remains high. The long period of clinical latency associated with lung cancer provides an ideal window of opportunity to administer vaccines to at-risk individuals that can prevent tumor progression and initiate long-term anti-tumor immune surveillance. Here we describe a personalized vaccination regime that could be applied for both therapeutic and prophylactic prevention of lung cancer, based on the derivation of lung cancer cells from induced pluripotent stem cells. Stem cells from healthy mice were modified to express Cre-dependent KRASG12D and Trp53R172H prior to differentiation to lung progenitor cells. Subsequent viral delivery of Cre caused activation of exogenous driver mutations, resulting in transformation and development of lung cancer cells. iPSC-derived lung cancer cells were highly antigenically related to lung cancer cells induced in LSL-KRASG12D/+; Trp53R172H/+ transgenic mice and were antigenically unrelated to original pluripotent stem cells or pancreatic cancer cells derived using the same technological platform. For vaccination, induced lung cancer cells were infected with oncolytic Adenovirus or Vaccinia virus, to act as vaccine adjuvants, prior to delivery of vaccines sequentially to a murine inducible transgenic model of lung cancer. Application of this Virus-Infected, Reprogrammed Somatic cell-derived Tumor cell (VIReST) regime primed tumor-specific T cell responses that significantly prolonged survival in both subcutaneous post-vaccine challenge models and induced transgenic models of lung cancer, demonstrating that stem cell-derived prophylactic vaccines may be a feasible intervention for treatment or prevention of lung cancer development in at-risk individuals.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Lung Neoplasms/therapy , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Disease Models, Animal , Gene Expression , Genetic Vectors/genetics , Immunization , Lung Neoplasms/mortality , Lung Neoplasms/prevention & control , Male , Mice , Mice, Transgenic , Oncolytic Viruses/genetics , Survival , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transduction, Genetic , Treatment Outcome , Tumor Burden
3.
Clin Cancer Res ; 26(2): 465-476, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31767564

ABSTRACT

PURPOSE: Pancreatic cancer remains one of the most lethal cancers, and late detection renders most tumors refractory to conventional therapies. Development of cancer prophylaxis may be the most realistic option for improving mortality associated with this disease. Here, we develop a novel individualized prophylactic and therapeutic vaccination regimen using induced pluripotent stem cells (iPSC), gene editing, and tumor-targeted replicating oncolytic viruses. EXPERIMENTAL DESIGN: We created a Virus-Infected, Reprogrammed Somatic cell-derived Tumor cell (VIReST) regime. iPSCs from healthy cells were induced to pancreatic tumor cells using in situ gene editing via stable provision of KRas G12D and p53 R172H tumor driver mutations. These cells were preinfected with oncolytic Adenovirus (AdV) as prime or Vaccinia virus (VV) as boost, to improve vaccine immunogenicity, prior to delivery of vaccines in a sequential regime to young KPC transgenic mice, genetically programmed to develop pancreatic cancer, to prevent and delay disease development. RESULTS: Tumor cells preinfected with oncolytic AdV as prime or VV as boost were the best regime to induce tumor-specific immunity. iPSC-derived tumor cells were highly related in antigen repertoire to pancreatic cancer cells of KPC transgenic mice, suggesting that an individual's stem cells can provide an antigenically matched whole tumor cell vaccine. The VIReST vaccination primed tumor-specific T-cell responses, resulting in delayed disease emergence and progression and significantly prolonged survival of KPC transgenic mice. Importantly, this regime was well-tolerated and nontoxic. CONCLUSIONS: These results provide both proof of concept and a robust technology platform for the development of personalized prophylactic cancer vaccines to prevent pancreatic malignancies in at-risk individuals.


Subject(s)
Cancer Vaccines/administration & dosage , Induced Pluripotent Stem Cells/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Experimental/prevention & control , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Pancreatic Neoplasms/prevention & control , Animals , Cancer Vaccines/immunology , Chlorocebus aethiops , Disease Progression , Male , Mice , Mice, Transgenic , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Survival Rate , T-Lymphocytes/immunology , Treatment Outcome
4.
Oncotarget ; 7(13): 15703-24, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26872382

ABSTRACT

Adenovirus-mediated sensitization of cancer cells to cytotoxic drugs depends on simultaneous interactions of early viral genes with cell death and survival pathways. It is unclear what cellular factors mediate these interactions in the presence of DNA-damaging drugs. We found that adenovirus prevents Chk1-mediated checkpoint activation through inactivation of Mre11 and downregulation of the pChk1 adaptor-protein, Claspin, in cells with high levels of DNA-damage induced by the cytotoxic drugs gemcitabine and irinotecan. The mechanisms for Claspin downregulation involve decreased transcription and increased degradation, further attenuating pChk1-mediated signalling. Live cell imaging demonstrated that low doses of gemcitabine caused multiple mitotic aberrations including multipolar spindles, micro- and multi-nucleation and cytokinesis failure. A mutant virus with the anti-apoptotic E1B19K-gene deleted (AdΔ19K) further enhanced cell killing, Claspin downregulation, and potentiated drug-induced DNA damage and mitotic aberrations. Decreased Claspin expression and inactivation of Mre11 contributed to the enhanced cell killing in combination with DNA-damaging drugs. These results reveal novel mechanisms that are utilised by adenovirus to ensure completion of its life cycle in the presence of cellular DNA damage. Taken together, our findings reveal novel cellular targets that may be exploited when developing improved anti-cancer therapeutics.


Subject(s)
Adenocarcinoma , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Pancreatic Neoplasms , Adaptor Proteins, Signal Transducing/biosynthesis , Adenoviridae/genetics , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line, Tumor , DNA Damage/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Down-Regulation , Genes, Viral , Humans , Irinotecan , MRE11 Homologue Protein/biosynthesis , Gemcitabine
5.
Mol Ther Methods Clin Dev ; 2: 15035, 2015.
Article in English | MEDLINE | ID: mdl-26417609

ABSTRACT

The current method for creation of vaccinia virus (VACV) vectors involves using a selection and purification marker, however inclusion of a gene without therapeutic value in the resulting vector is not desirable for clinical use. The Cre-LoxP system has been used to make marker-free Poxviruses, but the efficiency was very low. To obtain a marker-free VACV vector, we developed marker gene excision systems to modify the thymidine kinase (TK) region and N1L regions using Cre-Loxp and Flp-FRET systems respectively. CRISPR-Cas9 system significantly resulted in a high efficiency (~90%) in generation of marker gene-positive TK-mutant VACV vector. The marker gene (RFP) could be excised from the recombinant virus using Cre recombinase. To make a marker-free VV vector with double gene deletions targeting the TK and N1L gene, we constructed a donor repair vector targeting the N1L gene, which can carry a therapeutic gene and the marker (RFP) that could be excised from the recombinant virus using Flp recombinase. The marker-free system developed here can be used to efficiently construct VACV vectors armed with any therapeutic genes in the TK region or N1L region without marker genes. Our marker-free system platform has significant potential for development of new marker-free VACV vectors for clinical application.

6.
Clin Cancer Res ; 21(15): 3512-21, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26240291

ABSTRACT

PURPOSE: Noninvasive biomarkers for early detection of pancreatic ductal adenocarcinoma (PDAC) are currently not available. Here, we aimed to identify a set of urine proteins able to distinguish patients with early-stage PDAC from healthy individuals. EXPERIMENTAL DESIGN: Proteomes of 18 urine samples from healthy controls, chronic pancreatitis, and patients with PDAC (six/group) were assayed using GeLC/MS/MS analysis. The selected biomarkers were subsequently validated with ELISA assays using multiple logistic regression applied to a training dataset in a multicenter cohort comprising 488 urine samples. RESULTS: LYVE-1, REG1A, and TFF1 were selected as candidate biomarkers. When comparing PDAC (n = 192) with healthy (n = 87) urine specimens, the resulting areas under the receiver-operating characteristic curves (AUC) of the panel were 0.89 [95% confidence interval (CI), 0.84-0.94] in the training (70% of the data) and 0.92 (95% CI, 0.86-0.98) in the validation (30% of the data) datasets. When comparing PDAC stage I-II (n = 71) with healthy urine specimens, the panel achieved AUCs of 0.90 (95% CI, 0.84-0.96) and 0.93 (95% CI, 0.84-1.00) in the training and validation datasets, respectively. In PDAC stage I-II and healthy samples with matching plasma CA19.9, the panel achieved a higher AUC of 0.97 (95% CI, 0.94-0.99) than CA19.9 (AUC = 0.88; 95% CI, 0.81-0.95, P = 0.005). Adding plasma CA19.9 to the panel increased the AUC from 0.97 (95% CI, 0.94-0.99) to 0.99 (95% CI, 0.97-1.00, P = 0.04), but did not improve the comparison of stage I-IIA PDAC (n = 17) with healthy urine. CONCLUSIONS: We have established a novel, three-protein biomarker panel that is able to detect patients with early-stage pancreatic cancer in urine specimens.


Subject(s)
Adenocarcinoma/urine , Biomarkers, Tumor/urine , Early Detection of Cancer , Lithostathine/urine , Pancreatic Neoplasms/urine , Tumor Suppressor Proteins/urine , Vesicular Transport Proteins/urine , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Antigens, Tumor-Associated, Carbohydrate/urine , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Staging , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proteome/genetics , Tandem Mass Spectrometry , Trefoil Factor-1
7.
J Virol ; 89(9): 5176-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25741005

ABSTRACT

Vaccinia virus (VACV) continues to be used in immunotherapy for the prevention of infectious diseases and treatment of cancer since its use for the eradication of smallpox. However, the current method of editing the VACV genome is not efficient. Here, we demonstrate that the CRISPR-Cas9 system can be used to edit the VACV genome rapidly and efficiently. Additionally, a set of 8,964 computationally designed unique guide RNAs (gRNAs) targeting all VACV genes will be valuable for the study of VACV gene functions.


Subject(s)
CRISPR-Cas Systems , Genome, Viral , Molecular Biology/methods , Recombination, Genetic , Vaccinia virus/genetics , Technology, Pharmaceutical/methods
8.
Onco Targets Ther ; 6: 1031-40, 2013.
Article in English | MEDLINE | ID: mdl-23940420

ABSTRACT

Oncolytic viruses (OVs) have the ability to selectively replicate in and lyse cancer cells. Angiogenesis is an essential requirement for tumor growth. Like OVs, the therapeutic effect of many angiogenesis inhibitors has been limited, leading to the development of more effective approaches to combine antiangiogenic therapy with OVs. Angiogenesis can be targeted either directly by OV infection of vascular endothelial cells, or by arming OVs with antiangiogenic transgenes, which are subsequently expressed locally in the tumor microenvironment. In this review, we describe the development and targeting of OVs, the role of angiogenesis in cancer, and the progress made in arming viruses with antiangiogenic transgenes. Future developments required to optimize this approach are addressed.

9.
PLoS One ; 7(10): e46617, 2012.
Article in English | MEDLINE | ID: mdl-23056370

ABSTRACT

Oncolytic adenoviruses have shown promising efficacy in clinical trials targeting prostate cancers that frequently develop resistance to all current therapies. The replication-selective mutants AdΔΔ and dl922-947, defective in pRb-binding, have been demonstrated to synergise with the current standard of care, mitoxantrone and docetaxel, in prostate cancer models. While expression of the early viral E1A gene is essential for the enhanced cell killing, the specific E1A-regions required for the effects are unknown. Here, we demonstrate that replicating mutants deleted in small E1A-domains, binding pRb (dl1108), p300/CBP (dl1104) and p400/TRRAP or p21 (dl1102) sensitize human prostate cancer cells (PC-3, DU145, 22Rv1) to mitoxantrone and docetaxel. Through generation of non-replicating mutants, we demonstrate that the small E1A12S protein is sufficient to potently sensitize all prostate cancer cells to the drugs even in the absence of viral replication and the E1A transactivating domain, conserved region (CR) 3. Furthermore, the p300/CBP-binding domain in E1ACR1 is essential for drug-sensitisation in the absence (AdE1A1104) but not in the presence of the E1ACR3 (dl1104) domain. AdE1A1104 also failed to increase apoptosis and accumulation of cells in G2/M. All E1AΔCR2 mutants (AdE1A1108, dl922-947) and AdE1A1102 or dl1102 enhance cell killing to the same degree as wild type virus. In PC-3 xenografts in vivo the dl1102 mutant significantly prolongs time to tumor progression that is further enhanced in combination with docetaxel. Neither dl1102 nor dl1104 replicates in normal human epithelial cells (NHBE). These findings suggest that additional E1A-deletions might be included when developing more potent replication-selective oncolytic viruses, such as the AdΔCR2-mutants, to further enhance potency through synergistic cell killing in combination with current chemotherapeutics.


Subject(s)
Adenoviridae/genetics , Adenovirus E1A Proteins/genetics , Antineoplastic Agents/therapeutic use , Mitoxantrone/therapeutic use , Taxoids/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel , Flow Cytometry , Humans , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mitoxantrone/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , Reverse Transcriptase Polymerase Chain Reaction , Taxoids/pharmacology , Xenograft Model Antitumor Assays
10.
Hum Gene Ther ; 23(9): 1003-15, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22788991

ABSTRACT

AdΔΔ is an oncolytic adenoviral mutant that has been engineered to selectively target tumors with deregulated cell cycle and apoptosis pathways. AdΔΔ potentiates apoptotic cell death induced by drugs, including mitoxantrone and docetaxel, which are commonly used to treat prostate cancer. Here, we demonstrate that AdΔΔ can also interact synergistically with dietary phytochemicals known to have anti-cancer activities, without incurring the toxic side effects of chemodrugs. Curcumin, genistein, epigallocatechin-gallate, equol, and resveratrol efficiently killed both androgen-receptor positive (22Rv1) and negative cell lines (PC-3, DU145) in combination with adenoviral mutants. Synergistic cell killing was demonstrated with wild-type virus (Ad5) and AdΔΔ in combination with equol and resveratrol. EC(50) values for both phytochemicals and viruses were reduced three- to eightfold in all three combination-treated cell lines. The most potent efficacy was achieved in the cytotoxic drug- and virus-insensitive PC-3 cells, both in vitro and in vivo, while cell killing in normal bronchial epithelial cells was not enhanced. Although equol and resveratrol induced only low levels of apoptosis when administered alone, in combination with wild-type virus or AdΔΔ, the level of apoptotic cell death was significantly increased in PC-3 and DU145 cells. In vivo studies using suboptimal doses of AdΔΔ and equol or resveratrol, showed reduced tumor growth without toxicity to normal tissue. These findings identify novel functions for AdΔΔ and phytochemicals in promoting cancer cell killing and apoptosis, suggesting the use of these natural nontoxic compounds might be a feasible and currently unexploited anti-cancer strategy.


Subject(s)
Adenoviridae , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Dietary Supplements , Equol/pharmacology , Mutation , Oncolytic Viruses , Prostatic Neoplasms/therapy , Stilbenes/pharmacology , Animals , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Phytoestrogens/pharmacology , Prostatic Neoplasms/pathology , Resveratrol , Transplantation, Heterologous
11.
Hum Gene Ther ; 22(9): 1101-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21361787

ABSTRACT

Oncolytic viral therapy represents a promising strategy for the treatment of head and neck squamous cell carcinoma (HNSCC), with dl1520 (ONYX-015) the most widely used oncolytic adenovirus in clinical trials. This study aimed to determine the effectiveness of the Lister vaccine strain of vaccinia virus as well as a vaccinia virus armed with the endostatin-angiostatin fusion gene (VVhEA) as a novel therapy for HNSCC and to compare them with dl1520. The potency and replication of the Lister strain and VVhEA and the expression and function of the fusion protein were determined in human HNSCC cells in vitro and in vivo. Finally, the efficacy of VVhEA was compared with dl1520 in vivo in a human HNSCC model. The Lister vaccine strain of vaccinia virus was more effective than the adenovirus against all HNSCC cell lines tested in vitro. Although the potency of VVhEA was attenuated in vitro, the expression and function of the endostatin-angiostatin fusion protein was confirmed in HNSCC models both in vitro and in vivo. This novel vaccinia virus (VVhEA) demonstrated superior antitumor potency in vivo compared with both dl1520 and the control vaccinia virus. This study suggests that the Lister strain vaccinia virus armed with an endostatin-angiostatin fusion gene may be a potential therapeutic agent for HNSCC.


Subject(s)
Angiostatins/genetics , Carcinoma, Squamous Cell/therapy , Endostatins/genetics , Genetic Vectors/genetics , Head and Neck Neoplasms/therapy , Oncolytic Viruses/genetics , Vaccinia virus/genetics , Adenoviridae/genetics , Angiostatins/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cytopathogenic Effect, Viral , Endostatins/metabolism , Female , Gene Expression Regulation, Viral , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Nude , Oncolytic Virotherapy , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck , Viral Vaccines , Virus Replication , Xenograft Model Antitumor Assays
12.
Gut ; 59(11): 1535-44, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20947888

ABSTRACT

BACKGROUND AND AIMS: Metastasis accounts for the poor outcome of patients with pancreatic cancer. We recently discovered PRSS3 to be over-expressed in metastatic human pancreatic cancer cells. This study aimed to elucidate the role of PRSS3 in the growth and metastasis of human pancreatic cancer. METHODS: PRSS3 expression in human pancreatic cancer cell lines was detected by qPCR and immunoblotting. The effect of PRSS3 on cancer cell proliferation, migration and invasion in vitro, tumour growth and metastasis in vivo were investigated by manipulation of PRSS3 expression in human pancreatic cancer cell lines. VEGF expression was detected by ELISA, and the pathway through which PRSS3 regulates VEGF expression was investigated. The therapeutic effect of targeting this pathway on metastasis was assessed in vivo. Immunohistochemistry was employed to detect PRSS3 expression in human pancreatic cancer tissues. RESULTS: PRSS3 was over-expressed in the metastatic PaTu8988s cell line, but not in the non-metastatic PaTu8988t cell line. Over-expression of PRSS3 promoted pancreatic cancer cell proliferation as well as invasion in vitro, and tumour progression and metastasis in vivo. Stepwise investigations demonstrated that PRSS3 upregulates VEGF expression via the PAR1-mediated ERK pathway. ERK inhibitor significantly delayed the progression of metastases of pancreatic cancer and prolonged the survival of animals bearing metastatic pancreatic cancer (p<0.05). 40.54% of human pancreatic cancers (n=74) were positive for PRSS3 protein. A significant correlation was observed between PRSS3 expression and metastasis (p<0.01). Multivariate Cox regression analysis indicated that patients with PRSS3 expression in their tumours had a shorter survival time compared to those without PRSS3 expression (p<0.05). CONCLUSION: PRSS3 plays an important role in the progression, metastasis and prognosis of human pancreatic cancer. Targeting the PRSS3 signalling pathway may be an effective and feasible approach for treatment of this lethal cancer.


Subject(s)
Neoplasm Proteins/physiology , Pancreatic Neoplasms/pathology , Trypsin/physiology , Adult , Aged , Animals , Butadienes/therapeutic use , Cell Proliferation , Disease Progression , Enzyme Inhibitors/therapeutic use , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/physiology , Female , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Nitriles/therapeutic use , Pancreatic Neoplasms/enzymology , Prognosis , Survival Analysis , Trypsin/pharmacology , Tumor Cells, Cultured , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
13.
Cancer Res ; 70(13): 5207-12, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20516122

ABSTRACT

Prostate cancer is significantly more common in Western men than in Asian men, but the basis for this difference remains unknown. Because genomic studies of Asian prostate cancer are very limited, we used a genome-wide approach to reveal the genomic alterations in Chinese prostate cancers. We found a significant reduction in the frequency of certain somatic genomic changes that are commonly found in Western prostate cancers, including the 21q22.2-22.3 deletion, which involves the TMPRSS2:ERG fusion gene, and 10q deletion, which causes PTEN inactivation. Array results were confirmed by PCR-based molecular copy-number counting in selected samples. The different frequencies of these genomic changes were further evaluated by fluorescent in situ hybridization and immunohistochemistry analyses of tissue microarray samples. These alterations might be key genetic changes underlying the regional/ethnic difference in clinical incidence and might be induced by specific environmental and/or genetic risk factors that Western men are exposed to. Our findings suggest that tumors arise in Western and Chinese populations by alternative pathogenetic mechanisms.


Subject(s)
Asian People/genetics , Prostatic Neoplasms/ethnology , Prostatic Neoplasms/genetics , White People/genetics , China , Gene Rearrangement , Genome, Human , Humans , Male , Oncogene Proteins, Fusion/genetics , PTEN Phosphohydrolase/genetics , Polymorphism, Single Nucleotide , Prostatic Neoplasms/pathology , Trans-Activators/genetics , Transcriptional Regulator ERG , United Kingdom
14.
Clin Cancer Res ; 16(2): 541-53, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20068104

ABSTRACT

PURPOSE: Replication-selective oncolytic adenoviruses are a promising class of tumor-targeting agents with proven safety in hundreds of patients. However, clinical responses have been limited and viral mutants with higher potency are needed. Here, we report on the generation of a novel set of mutants with improved efficacy in prostate and pancreatic carcinoma models. Currently, no curative treatments are available for late-stage metastatic prostate or rapidly progressing pancreatic cancers. EXPERIMENTAL DESIGN: Adenovirus type 5 mutants were created with deletions in the E1ACR2 region for tumor selectivity and/or the E1B19K gene for attenuated replication in vivo; all constructs retain the E3 genes intact. Cell-killing efficacy, replication, and cytotoxicity in combination with chemotherapeutics were investigated in normal cells (PrEC and NHBE), seven carcinoma cell lines, and human (PC3 and DU145) and murine (TRAMPC, CMT-64, and CMT-93) tumor models in vivo. RESULTS: The double-deleted AdDeltaDelta (DeltaE1ACR2 and DeltaE1B19K) mutant had high cell-killing activity in prostate, pancreatic, and lung carcinomas. Replication was similar to wild-type in all tumor cells and was attenuated in normal cells to levels less than the single-deleted AdDeltaCR2 mutant. AdDeltaDelta combined with the chemotherapeutics docetaxel and mitoxantrone resulted in synergistically enhanced cell killing and greatly improved antitumor efficacy in prostate xenografts in vivo. In murine immunocompetent in vivo models efficacy was greater for mutants with the E3B genes intact even in the absence of viral replication, indicating attenuated macrophage-dependent clearance. CONCLUSIONS: These data suggest that the novel oncolytic mutant AdDeltaDelta is a promising candidate for targeting of solid tumors specifically in combination with chemotherapeutics.


Subject(s)
Adenovirus E1A Proteins/genetics , Adenovirus E1B Proteins/genetics , Carcinoma/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Pancreatic Neoplasms/therapy , Adenoviridae/genetics , Animals , Cell Line, Tumor , Gene Deletion , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Oncolytic Viruses/physiology , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/physiology , Substrate Specificity , Treatment Outcome , Xenograft Model Antitumor Assays
15.
Cancer Res ; 69(22): 8645-51, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19903855

ABSTRACT

Lung cancer is the commonest cancer killer. Small cell lung cancer (SCLC) is initially chemosensitive, but rapidly relapses in a chemoresistant form with an overall survival of <5%. Consequently, novel therapies are urgently required and will likely arise from an improved understanding of the disease biology. Our previous work showed that fibroblast growth factor-2 induces proliferation and chemoresistance in SCLC cells. Here, we show that the selective fibroblast growth factor receptor (FGFR) inhibitor PD173074 blocks H-510 and H-69 SCLC proliferation and clonogenic growth in a dose-dependent fashion and prevents FGF-2-induced chemoresistance. These effects correlate with the inhibition of both FGFR1 and FGFR2 transphosphorylation. We then determined the efficacy of daily oral administration of PD173074 for 28 days in two human SCLC models. In the H-510 xenograft, tumor growth was impaired similar to that seen with single-agent cisplatin administration, increasing median survival compared with control sham-treated animals. Crucially, the effect of cisplatin was significantly potentiated by coadministration of PD173074. More dramatically, in H-69 xenografts, PD173074 induced complete responses lasting >6 months in 50% of mice. These effects were not a consequence of disrupted tumor vasculature but instead correlated with increased apoptosis (caspase 3 and cytokeratin 18 cleavage) in excised tumors. Moreover, in vivo imaging with 3'-deoxy-3'-[(18)F]fluorothymidine-positron emission tomography ([(18)F]FLT-PET) showed decreased intratumoral proliferation in live animals treated with the compound at 7 to 14 days. Our results suggest that clinical trials of FGFR inhibitors should be undertaken in patients with SCLC and that [(18)F]FLT-PET imaging could provide early in vivo evidence of response.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/physiology , Lung Neoplasms/drug therapy , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/drug effects , Small Cell Lung Carcinoma/drug therapy , Animals , Blotting, Western , Cell Proliferation/drug effects , Cisplatin/pharmacology , Drug Synergism , Fibroblast Growth Factor 2/drug effects , Fibroblast Growth Factor 2/metabolism , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mice , Positron-Emission Tomography , Receptors, Fibroblast Growth Factor/metabolism , Transfection , Xenograft Model Antitumor Assays
16.
Clin Cancer Res ; 15(21): 6595-601, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19861465

ABSTRACT

PURPOSE: The Na/I symporter (hNIS) promotes concentration of iodine in cells. In cancer gene therapy, this transgene has potential as a reporter gene for molecular imaging of viral biodistribution and as a therapeutic protein promoting (131)I-mediated radiotherapy. Here, we combined the imaging and therapeutic potential of hNIS in an oncolytic adenoviruses targeting colorectal cancer cells. EXPERIMENTAL DESIGN: We generated an adenovirus (AdIP2) encoding hNIS and capable of selective replication in colorectal carcinoma cells. The selectivity of this virus was verified in vitro and in vivo. Its spread in tumors was monitored in vivo using single-photon emission computed tomography/CT imaging upon (99m)TcO(4)(-) injection and confirmed by immunohistochemistry. Metabolic radiotherapy was done through injection of therapeutic doses of (131)I(-). RESULTS: We showed in vitro and in vivo the selectivity of AdIP2 and that hNIS expression is restricted to the target cells. Imaging and immunohistochemical data showed that viral spread is limited and that the point of maximal hNIS expression is reached 48 hours after a single intratumoral injection. Administration of a single therapeutic dose of (131)I at this time point led to a dramatic reduction in tumor size not observed in hNIS-negative viruses. CONCLUSIONS: This report showed for the first time that the combination of the imaging and therapeutic potentials of hNIS can be applied to oncolytic adenoviruses in experimental models of cancer.


Subject(s)
Adenoviridae/genetics , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/therapy , Genes, Reporter , Genetic Therapy/methods , Iodine Radioisotopes/therapeutic use , Symporters/genetics , Wnt Proteins/metabolism , Animals , Cell Line, Tumor , Diagnostic Imaging , Gene Targeting , Genetic Vectors , Mice , Mice, Nude , Radionuclide Imaging , Symporters/metabolism , Transgenes
17.
Clin Cancer Res ; 15(5): 1730-40, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19223497

ABSTRACT

PURPOSE: Pancreatic adenocarcinoma is a rapidly progressive malignancy that is highly resistant to current chemotherapeutic modalities and almost uniformly fatal. We show that a novel targeting strategy combining oncolytic adenoviral mutants with the standard cytotoxic treatment, gemcitabine, can markedly improve the anticancer potency. EXPERIMENTAL DESIGN: Adenoviral mutants with the E1B19K gene deleted with and without E3B gene expression (AdDeltaE1B19K and dl337 mutants, respectively) were assessed for synergistic interactions in combination with gemcitabine. Cell viability, mechanism of cell death, and antitumor efficacy in vivo were determined in the pancreatic carcinoma cells PT45 and Suit2, normal human bronchial epithelial cells, and in PT45 xenografts. RESULTS: The DeltaE1B19K-deleted mutants synergized with gemcitabine to selectively kill cultured pancreatic cancer cells and xenografts in vivo with no effect in normal cells. The corresponding wild-type virus (Ad5) stimulated drug-induced cell killing to a lesser degree. Gemcitabine blocked replication of all viruses despite the enhanced cell killing activity due to gemcitabine-induced delay in G1/S-cell cycle progression, with repression of cyclin E and cdc25A, which was not abrogated by viral E1A-expression. Synergistic cell death occurred through enhancement of gemcitabine-induced apoptosis in the presence of both AdDeltaE1B19K and dl337 mutants, shown by increased cell membrane fragmentation, caspase-3 activation, and mitochondrial dysfunction. CONCLUSIONS: Our data suggest that oncolytic mutants lacking the antiapoptotic E1B19K gene can improve efficacy of DNA-damaging drugs such as gemcitabine through convergence on cellular apoptosis pathways. These findings imply that less toxic doses than currently practiced in the clinic could efficiently target pancreatic adenocarcinomas when combined with adenoviral mutants.


Subject(s)
Adenocarcinoma/pathology , Adenoviridae/genetics , Adenovirus E1B Proteins/genetics , Apoptosis/drug effects , Deoxycytidine/analogs & derivatives , Gene Deletion , Mutation/genetics , Oncolytic Viruses/physiology , Pancreatic Neoplasms/pathology , Adenocarcinoma/genetics , Adenovirus E1B Proteins/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Combined Modality Therapy , Deoxycytidine/therapeutic use , Drug Synergism , Flow Cytometry , Genetic Therapy , Genetic Vectors , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Nude , Pancreatic Neoplasms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , S Phase/drug effects , S Phase/physiology , Virus Replication , Xenograft Model Antitumor Assays , Gemcitabine
18.
Curr Opin Mol Ther ; 11(6): 664-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20072943

ABSTRACT

Angiogenesis is essential for development and tissue repair, and is controlled by a balance of inhibitors and promoters. Overactive angiogenesis promotes tumor progression and other chronic disorders, including diabetic retinopathy and rheumatoid arthritis. The discovery of angiogenesis inhibitors has resulted in a promising therapeutic approach to these diseases. However, the benefits of anti-angiogenic drugs have been modest, stimulating interest in developing more effective approaches by combining anti-angiogenic therapy with other therapeutics. Oncolytic virotherapies are attractive therapeutics for cancer, but virotherapy alone has had similar problems to anti-angiogenic therapy, with few examples of clinical efficacy. This review summarizes the progress of the emerging field of combinations of anti-angiogenic therapy and virotherapy, and highlights future challenges in experimental and translational research that need to be addressed in order for these therapeutics to advance into the clinic.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Oncolytic Viruses/metabolism , Chemokines/metabolism , Combined Modality Therapy , Fibroblast Growth Factors/metabolism , Humans , Interleukins/metabolism , Matrix Metalloproteinases/metabolism , Neoplasms/therapy , Signal Transduction/physiology , Translational Research, Biomedical , Vascular Endothelial Growth Factor A/metabolism
19.
20.
Int J Cancer ; 120(11): 2344-52, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17311258

ABSTRACT

Pancreatic cancer is one of the most aggressive human tumors with a 5-year survival rate of only 3% and a striking resistance to chemotherapy and radiotherapy. The search for new therapeutic approaches includes strategies exploiting the deregulation of apoptotic pathways commonly found in cancer cells. The IAP proteins are inhibitors of apoptosis that have altered activity in numerous cancer types and are implicated in resistance to chemotherapy, and therefore are potentially interesting as therapeutic targets. We investigated alterations in the expression of IAPs and their inhibitors in pancreatic adenocarcinoma by using real-time PCR, in situ hybridization and immunohistochemistry. We found differential expression of various IAPs in this malignancy, and particularly we observed overexpression of cIAP-2, survivin, livin and XIAP. We also looked for correlations between the expression of IAPs and resistance to paclitaxel, doxorubicin, CDDP and 5-fluorouracil, and found that resistance to these drugs correlates most significantly with expression of cIAP-2. Using RNAi to downregulate these proteins we further confirmed that the levels of cIAP-2 and XIAP influence the response to the anti-cancer drugs, although only marginally for 5-FU. We conclude that anti-tumor strategies based on the inhibition of particular IAPs can be useful in targeting pancreatic adenocarcinoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Inhibitor of Apoptosis Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/physiopathology , Area Under Curve , Base Sequence , DNA Primers , Drug Resistance, Neoplasm , Humans , Immunohistochemistry , In Situ Hybridization , Inhibitor of Apoptosis Proteins/physiology , Polymerase Chain Reaction , RNA Interference
SELECTION OF CITATIONS
SEARCH DETAIL
...