Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Prostate Cancer Prostatic Dis ; 24(3): 871-879, 2021 09.
Article in English | MEDLINE | ID: mdl-33746212

ABSTRACT

BACKGROUND: In 2004 docetaxel was the first life-prolonging drug (LPD) registered for metastatic castration-resistant prostate cancer (mCRPC) patients. Between 2011 and 2014 new LPDs for mCRPC (cabazitaxel, abiraterone, enzalutamide, and radium-223) were introduced in the Netherlands. The objective of this study is to assess the impact of the introduction of new LPDs on treatment patterns and overall survival (OS) over time. PATIENTS AND METHODS: CRPC patients diagnosed in the years 2010-2016 in the observational, retrospective CAPRI registry (20 hospitals) were included and followed up to 2018. Two subgroups were analyzed: treatment-naïve patients (subgroup 1, n = 3600) and post-docetaxel patients (subgroup 2, n = 1355). RESULTS: In both subgroups, the use of any LPD increased: from 57% (2010-2011) to 69% (2014-2015) in subgroup 1 and from 65% (2011-2012) to 79% (2015-2016) in subgroup 2. Chemotherapy as first mCRPC-treatment (i.e., docetaxel) and first post-docetaxel treatment (i.e., cabazitaxel or docetaxel rechallenge) decreased (46-29% and 20-9% in subgroup 1 and 2, respectively), while the use of androgen-receptor targeting treatments (ART) increased from 11% to 39% and 46% to 64% in subgroup 1 and 2, respectively. In subgroup 1, median OS (mOS) from diagnosis CRPC increased from 28.5 months to 31.0 months (p = 0.196). In subgroup 2, mOS from progression on docetaxel increased from 7.9 months to 12.5 months (p < 0.001). After multiple imputations of missing values, in multivariable cox-regression analysis with known prognostic parameters, the treatment period was independent significant for OS in subgroup 1 (2014-2015 vs. 2010-2011 with HR 0.749, p < 0.001) and subgroup 2 (2015-2016 vs. 2011-2012 with HR 0.811, p = 0.037). CONCLUSION: Since 2010, a larger proportion of mCRPC patients was treated with LPDs, which was related to an increased mOS.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemoradiotherapy/mortality , Prostatic Neoplasms, Castration-Resistant/mortality , Radium/therapeutic use , Aged , Aged, 80 and over , Androstenes/administration & dosage , Benzamides/administration & dosage , Docetaxel/administration & dosage , Follow-Up Studies , Humans , Male , Nitriles/administration & dosage , Phenylthiohydantoin/administration & dosage , Prognosis , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/therapy , Retrospective Studies , Survival Rate , Taxoids/administration & dosage
2.
Clin Transl Oncol ; 21(6): 774-780, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30465182

ABSTRACT

BACKGROUND: Health-related quality of life (HRQoL) is an important issue in the rapidly evolving field of adjuvant treatment for stage III melanoma. Dendritic cell vaccination is one of the adjuvant forms of therapy currently investigated. METHODS: We enrolled adults with stage III melanoma to receive adjuvant dendritic cell vaccination after a complete radical lymph node dissection. HRQoL assessment was one of the secondary endpoints of this trial and investigated with the EORTC-QLQ-C30 questionnaire at baseline and week 26. RESULTS: Fifteen patients with a median age of 50 years were included in the study, with twelve evaluable patients on study at time of the second questionnaire. Global health status and role functioning improved clinically relevant with a mean difference of 15 (p = 0.010) and 26 points (p = 0.005), respectively. DISCUSSION: Despite the small number of patients, we found a clinically relevant improved global health status. Besides, compared to the other investigated therapies, toxicity of dendritic cell vaccination is low, which supports our finding. CONCLUSION: This is the first description of HRQoL in melanoma patients receiving dendritic cell vaccination. We show the expected improvement in global health status after surgical treatment of stage III melanoma. Thus, adjuvant dendritic cell vaccination does not seem to hamper this improvement, as shown in our small explorative study.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Dendritic Cells/transplantation , Immunotherapy , Melanoma/therapy , Quality of Life , Adult , Aged , Dendritic Cells/immunology , Female , Follow-Up Studies , Humans , Male , Melanoma/immunology , Middle Aged , Neoplasm Staging , Prospective Studies , Surveys and Questionnaires , Young Adult
3.
Crit Rev Oncol Hematol ; 122: 157-163, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29458784

ABSTRACT

BACKGROUND: The purpose of this review was to assess the effectiveness of different strategies to implement physical activity during and after cancer treatment. DESIGN: We searched for studies containing strategies to implement physical activity in cancer care that meet the inclusion criteria of the Cochrane EPOC group. The primary outcome was physical activity uptake. We expressed the effectiveness of the strategies as the percentage of studies with improvement. RESULTS: Nine studies met the inclusion criteria. Patient groups doing physical activities via an implementation strategy had better outcomes than those receiving usual care: 83% of the studies showed improvement. Strategies showing significant improvement compared to usual care employed healthcare professionals to provide individual counselling or advice for exercise or interactive elements such as audit and feedback systems. When comparing the different strategies 1) interactive elements or 2) elements tailored to the needs of the patients had better physical activity uptake. CONCLUSIONS: Implementation strategies containing individual and interactive elements, tailored to the individual needs of patients, are more successful in improving physical activity uptake.


Subject(s)
Exercise/physiology , Neoplasms/therapy , Humans , Treatment Outcome
4.
Ann Oncol ; 23 Suppl 8: viii22-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22918924

ABSTRACT

The prognosis for men with metastatic, castration-resistant prostate cancer (CRPC) is limited, and patients have very few treatment options, particularly if the treatment failed with docetaxel (Taxotere). As a result, there is a requirement for novel approaches to therapy. Using immunotherapy to induce immune responses to prostate cancer in preclinical and clinical studies appears to be a valid therapeutic approach. In a pivotal phase III trial, treatment with sipuleucel-T, an autologous cellular vaccine consisting of activated antigen-presenting cells loaded with prostatic acid phosphatase (PAP), gave a median overall survival of 25.8 months compared with 21.7 months for placebo-treated patients, resulting in a 22% relative reduction in the risk of death. Based on these results, sipuleucel-T became the first therapeutic vaccine approved for any type of cancer in the USA. PROSTVAC(®)-VF, a poxvirus-based vaccine engineered to present prostate-specific antigen (PSA) and three immune costimulatory molecules, and GVAX, a vaccine consisting of two prostate cancer cell lines (LnCAP and PC3) and genetically modified to secrete granulocyte-macrophage colony-stimulating factor (GM-CSF), both showed promising results in phase II studies, although GVAX failed to meet its primary end point of overall survival when compared with docetaxel in a phase III study. T-cell modulation is another potential immunotherapeutic strategy for CRPC. Ipilimumab, an antibody against the cytotoxic T-lymphocyte-associated antigen-4, is being evaluated in phase I/II studies, both alone and in combination with chemotherapy, radiotherapy or GVAX, with activity in prostate cancer. CRPC is one of the few tumour types where immunotherapy is the current standard of care. Further research, however, will be necessary to improve antitumour responses and clinical benefits, including the use of novel combinatorial approaches.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Tissue Extracts/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CTLA-4 Antigen/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Docetaxel , Humans , Intercellular Adhesion Molecule-1/immunology , Ipilimumab , Male , Survival Rate , Taxoids/therapeutic use , Treatment Outcome
6.
Oncol Res ; 16(10): 471-7, 2007.
Article in English | MEDLINE | ID: mdl-18196871

ABSTRACT

Multicellular tumor spheroids are used as a model to assess the efficacy of replicating oncolytic adenoviruses. As most assays used to assess cellular viability are unsuitable for oncolytic viruses because of ongoing viral replication, we have used positron emission tomography (PET) to sequentially determine the incorporation of 18F-labeled deoxyglucose (18F-DG) as a measure of viability and compared the results to more commonly used assays for measuring the effect of oncolytic therapy. Glioma monolayer cultures and spheroids were infected with wild-type replicating adenovirus and viability was measured by 18F-DG incorporation, WST-1 assay, crystal violet assay, and spheroid volume 2 to 10 days following infection. Results show that volume measurements in adenovirus-infected spheroids are confounded by the cytopathic effect occurring in infected cells. 18F-DG PET provides a useful method to assess small differences in cell number and viability following oncolytic viral therapy in glioma monolayer cultures and spheroids without the need for disintegration of these cultures. Moreover, using 18F-DG PET, repeated sequential measurements of spheroid viability can be made, decreasing the required number of spheroids per experiment. This is a valuable feature when using spheroids derived from limited amounts of patient material.


Subject(s)
Adenoviridae/physiology , Fluorodeoxyglucose F18 , Glioma/diagnostic imaging , Glioma/therapy , Oncolytic Virotherapy/methods , Glioma/genetics , Glioma/virology , Humans , Positron-Emission Tomography/methods , Spheroids, Cellular , Tumor Cells, Cultured
8.
Br J Cancer ; 94(12): 1837-44, 2006 Jun 19.
Article in English | MEDLINE | ID: mdl-16736005

ABSTRACT

Despite improvements in treatment regimens for osteosarcoma (OS) patients, survival rate has not increased over the last two decades. New treatment modalities are therefore warranted. Preclinical results with conditionally replicative adenoviruses (CRAds) to treat OS are promising. One type of CRAd that was effective against OS cells is Ad5-Delta24RGD. In other types of cancer, CRAds have been shown to interact synergistically with chemotherapeutic agents. Chemotherapy for OS often includes doxorubicin and cisplatin. Therefore, we explored combination treatment of OS cell lines and primary OS cell cultures with Ad5-Delta24RGD and doxorubicin or cisplatin. On OS cell lines, combination treatment was additive to synergistic. Surprisingly, however, on seven of eight primary OS samples no such combination effects were observed. In contrast, in many cases chemotherapy even inhibited CRAd-mediated cell killing. The inhibitory effect of doxorubicin on Ad5-Delta24RGD in primary OS cells appeared to correlate with slow cell growth rate; reduced viral replication and absence of chemotherapy-induced G2 cell cycle arrest. Our results point to the possibility that, at least for OS, virotherapy and chemotherapy should best not be performed simultaneously. In general, our work underscores the importance of testing new genetic anticancer agents and treatment regimens on primary cancer specimens.


Subject(s)
Adenoviridae/physiology , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Doxorubicin/pharmacology , Oncolytic Virotherapy , Osteosarcoma/virology , Cell Cycle/drug effects , Cell Line, Tumor , Combined Modality Therapy , Humans , Osteosarcoma/therapy
9.
J Virol ; 80(3): 1250-60, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16415002

ABSTRACT

Murine hepatitis coronavirus (MHV)-A59 infection depends on the interaction of its spike (S) protein with the cellular receptor mCEACAM1a present on murine cells. Human cells lack this receptor and are therefore not susceptible to MHV. Specific alleviation of the tropism barrier by redirecting MHV to a tumor-specific receptor could lead to a virus with appealing properties for tumor therapy. To demonstrate that MHV can be retargeted to a nonnative receptor on human cells, we produced bispecific adapter proteins composed of the N-terminal D1 domain of mCEACAM1a linked to a short targeting peptide, the six-amino-acid His tag. Preincubation of MHV with the adapter proteins and subsequent inoculation of human cells expressing an artificial His receptor resulted in infection of these otherwise nonsusceptible cells and led to subsequent production of progeny virus. To generate a self-targeted virus able to establish multiround infection of the target cells, we subsequently incorporated the gene encoding the bispecific adapter protein as an additional expression cassette into the MHV genome through targeted RNA recombination. When inoculated onto murine LR7 cells, the resulting recombinant virus indeed expressed the adapter protein. Furthermore, inoculation of human target cells with the virus resulted in a His receptor-specific infection that was multiround. Extensive cell-cell fusion and rapid cell killing of infected target cells was observed. Our results show that MHV can be genetically redirected via adapters composed of the S protein binding part of mCEACAM1a and a targeting peptide recognizing a nonnative receptor expressed on human cells, consequently leading to rapid cell death. The results provide interesting leads for further investigations of the use of coronaviruses as antitumor agents.


Subject(s)
Murine hepatitis virus/physiology , Murine hepatitis virus/pathogenicity , Receptors, Virus/physiology , Animals , Base Sequence , Binding Sites , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/physiology , Cats , Cell Line , DNA, Recombinant/genetics , Gene Products, vif/genetics , Gene Products, vif/physiology , Humans , Membrane Fusion , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Murine hepatitis virus/genetics , Receptors, Virus/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics , Viral Envelope Proteins/physiology , Virus Replication
10.
J Virol ; 79(24): 15314-22, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16306602

ABSTRACT

The mouse hepatitis coronavirus (MHV) infects murine cells by binding of its spike (S) protein to murine CEACAM1a. The N-terminal part of this cellular receptor (soR) is sufficient for S binding and for subsequent induction of the conformational changes required for virus-cell membrane fusion. Here we analyzed whether these characteristics can be used to redirect MHV to human cancer cells. To this end, the soR domain was coupled to single-chain monoclonal antibody 425, which is directed against the human epidermal growth factor receptor (EGFR), resulting in a bispecific adapter protein (soR-425). The soR and soR-425 proteins, both produced with the vaccinia virus system, were able to neutralize MHV infection of murine LR7 cells. However, only soR-425 was able to target MHV to human EGFR-expressing cancer cells. Interestingly, the targeted infections induced syncytium formation. Furthermore, the soR-425-mediated infections were blocked by heptad repeat-mimicking peptides, indicating that virus entry requires the regular S protein fusion process. We conclude that the specific spike-binding property of the CEACAM1a N-terminal fragment can be exploited to direct the virus to selected cells by linking it to a moiety able to bind a receptor on those cells. This approach might be useful in the development of tumor-targeted coronaviruses.


Subject(s)
Coronavirus Infections/metabolism , ErbB Receptors/metabolism , Membrane Glycoproteins/physiology , Murine hepatitis virus/physiology , Viral Envelope Proteins/physiology , Animals , Coronavirus Infections/immunology , Humans , Membrane Glycoproteins/chemistry , Mice , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/chemistry
11.
Gene Ther ; 12(18): 1394-404, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15843808

ABSTRACT

To explore the potential of using non-human coronaviruses for cancer therapy, we first established their ability to kill human tumor cells. We found that the feline infectious peritonitis virus (FIPV) and a felinized murine hepatitis virus (fMHV), both normally incapable of infecting human cells, could rapidly and effectively kill human cancer cells artificially expressing the feline coronavirus receptor aminopeptidase N. Also 3-D multilayer tumor spheroids established from such cells were effectively eradicated. Next, we investigated whether FIPV and fMHV could be targeted to human cancer cells by constructing a bispecific single-chain antibody directed on the one hand against the feline coronavirus spike protein--responsible for receptor binding and subsequent cell entry through virus-cell membrane fusion--and on the other hand against the human epidermal growth factor receptor (EGFR). The targeting antibody mediated specific infection of EGFR-expressing human cancer cells by both coronaviruses. Furthermore, in the presence of the targeting antibody, infected cancer cells formed syncytia typical of productive coronavirus infection. By their potent cytotoxicity, the selective targeting of non-human coronaviruses to human cancer cells provides a rationale for further investigations into the use of these viruses as anticancer agents.


Subject(s)
Antibodies, Bispecific/administration & dosage , Gene Targeting/methods , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Animals , Antibodies, Bispecific/genetics , CD13 Antigens/genetics , Cats , Coronavirus Infections/immunology , Coronavirus Infections/metabolism , Coronavirus, Feline/genetics , Cytotoxicity, Immunologic , ErbB Receptors/immunology , Feline Infectious Peritonitis/metabolism , Humans , Membrane Glycoproteins/immunology , Mice , Murine hepatitis virus/genetics , Neoplasms/immunology , Neoplasms/virology , Protein Transport , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/immunology
12.
Br J Cancer ; 92(5): 882-7, 2005 Mar 14.
Article in English | MEDLINE | ID: mdl-15756257

ABSTRACT

CPT-11 (irinotecan or 7-ethyl-10[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecin) is an anticancer agent in use for the treatment of colon cancer. In order to be fully active, CPT-11 needs to be converted into SN-38 (7-ethyl-10-hydroxycamptothecin) by the enzyme carboxylesterase (CE). In humans, only a minority of CPT-11 is converted to SN-38. To increase the antitumour effect of CPT-11 by gene-directed enzyme prodrug therapy, we constructed a replication-deficient adenoviral vector Ad.C28-sCE2 containing a fusion gene encoding a secreted form of human liver CE2 targeted to the surface antigen epithelial cell adhesion molecule (EpCAM) that is highly expressed on most colon carcinoma cells. By targeting CE2 to EpCAM, the enzyme should accumulate specifically in tumours and leakage into the circulation should be minimised. Ad.C28-sCE2-transduced colon carcinoma cells expressed and secreted active CE that bound specifically to EpCAM-expressing cells. In sections of three-dimensional colon carcinoma spheroids transduced with Ad.C28-sCE2, it was shown that C28-sCE2 was capable of binding untransduced cells. Most importantly, treatment of these spheroids with nontoxic concentrations of CPT-11 resulted in growth inhibition comparable to treatment with SN-38. Therefore, Ad.C28-sCE2 holds promise in gene therapy approaches for the treatment of colon carcinoma.


Subject(s)
Adenoviridae/genetics , Camptothecin/analogs & derivatives , Camptothecin/toxicity , Carboxylesterase/genetics , Antineoplastic Agents, Phytogenic/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genetic Vectors , Humans , Irinotecan , Ovarian Neoplasms
13.
Gene Ther ; 12(12): 1011-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15729367

ABSTRACT

Conditionally replicating adenoviruses (CRAds) selectively replicate in and thereby kill cancer cells. The CRAd AdDelta24 with pRb-binding-deficient E1A kills cancer cells efficiently. Arming CRAds with genes encoding prodrug-converting enzymes could allow for enhanced anticancer efficacy by the combined effects of oncolytic replication and local prodrug activation. Here, we investigated combination treatment of human colon cancer cell lines with AdDelta24-type CRAds and gene-directed enzyme prodrug therapy (GDEPT) using two different enzyme/prodrug systems, that is, thymidine kinase/ganciclovir (TK/GCV) and carboxylesterase (CE)/CPT-11. On all three cell lines tested, GDEPT with TK/GCV made CRAd treatment less efficacious. In contrast, expression of a secreted form of CE (sCE2) combined with CPT-11 treatment markedly enhanced the efficacy of AdDelta24 virotherapy. Based on this observation, we constructed an AdDelta24 variant expressing sCE2. In the absence of CPT-11, this new CRAd Ad5-Delta24.E3-sCE2 was similarly effective as its parent in killing human colon cancer cells. Low concentrations of CPT-11 inhibited Ad5-Delta24.E3-sCE2 propagation. Nevertheless, CPT-11 specifically augmented the cytotoxicity of Ad5-Delta24.E3-sCE2 against all three-colon cancer cell lines. Hence, the positive contribution of sCE2/CPT-11 GDEPT to colon cancer cytotoxicity outweighed its negative influence on CRAd propagation. Therefore, CRAd-sCE2/CPT-11 combination therapy appears useful for more effective treatment of colon cancer.


Subject(s)
Adenoviridae/genetics , Carboxylesterase/genetics , Colonic Neoplasms/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Prodrugs/administration & dosage , Adenoviridae/physiology , Antiviral Agents/therapeutic use , Carboxylesterase/therapeutic use , Cell Line, Tumor , Colonic Neoplasms/virology , Combined Modality Therapy , Cytopathogenic Effect, Viral , Ganciclovir/therapeutic use , Gene Expression , Genetic Engineering , Genetic Vectors/genetics , Humans , Prodrugs/therapeutic use , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Thymidine Kinase/therapeutic use , Transduction, Genetic/methods , Virus Replication
14.
Br J Cancer ; 91(1): 171-7, 2004 Jul 05.
Article in English | MEDLINE | ID: mdl-15173860

ABSTRACT

TNF-related apoptosis-inducing ligand (TRAIL, also known as Apo-2L) is a promising novel anticancer agent that selectively induces apoptosis in tumour cells and the activity of which can be enhanced by combined treatment with chemo- or radiotherapy. For therapeutic purposes, the use of full-length TRAIL may be favourable to recombinant TRAIL based on its increased tumour cell killing potential, and the delivery of TRAIL at the tumour site by adenovirus vectors may provide an approach to overcome the short half-life of recombinant TRAIL and hepatocyte toxicity in vivo. Here, we constructed an adenoviral vector expressing full-length TRAIL (AdTRAIL) and studied the potential of chemo- and radiotherapy in enhancing AdTRAIL-induced apoptosis in non-small cell lung cancer (NSCLC) H460 cells and normal cells and, in addition, investigated the mechanism of AdTRAIL-induced apoptosis. AdTRAIL effectively killed H460 cells, which we previously showed to have a deficiency in mitochondria-dependent apoptosis by downstream activation of caspase-8 rather than caspase-9. Further analyses revealed that AdTRAIL induces death receptor- and mitochondria-dependent apoptosis that could be partially suppressed by Bcl2 overexpression. Combined treatment with doxorubicin (DOX), cisplatin (CDDP), paclitaxel (PTX) and radiation strongly enhanced AdTRAIL-induced cytotoxicity in a synergistic way. Synergy was accompanied by the cleavage of Bid and an increase in caspase-8 processing that was abolished by Bcl2 overexpression, indicating that the Bid-mitochondrial amplification loop is functional in H460 cells. Moreover, combination treatment did not alter the tumour selectivity of AdTRAIL since normal human fibroblasts (NHFs) remained resistant under these conditions. These findings further indicate that the combined use of chemo/radiotherapy and adenovirus-produced full-length TRAIL may provide a valuable treatment option for NSCLC.


Subject(s)
Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Membrane Glycoproteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Radiation Tolerance , Tumor Necrosis Factor-alpha/biosynthesis , Adenocarcinoma/pathology , Adenoviridae , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins , Fibroblasts/physiology , Humans , Ligands , Membrane Glycoproteins/pharmacology , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation , fas Receptor
15.
Gene Ther ; 10(23): 1982-91, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14528322

ABSTRACT

Conditionally replicative adenoviruses (CRAds) are potentially useful agents for anticancer virotherapy approaches. However, lack of coxsackievirus and adenovirus receptor (CAR) expression on many primary tumor cells limits the oncolytic potency of CRAds. This makes the concept of targeting, that is, redirecting infection via CAR-independent entry pathways, relevant for CRAd development. Bispecific adapter molecules constitute highly versatile means for adenovirus targeting. Here, we constructed a CRAd with the Delta24 E1A mutation that produces a bispecific single-chain antibody directed towards the adenovirus fiber knob and the epidermal growth factor receptor (EGFR). This EGFR-targeted CRAd exhibited increased infection efficiency and oncolytic replication on CAR-deficient cancer cells and augmented lateral spread in CAR-deficient 3-D tumor spheroids in vitro. When compared to its parent control with native tropism, the new CRAd exhibited similar cytotoxicity on CAR-positive cancer cells, but up to 1000-fold enhanced oncolytic potency on CAR-deficient, EGFR-positive cancer cells. In addition, EGFR-targeted CRAd killed primary human CAR-deficient brain tumor specimens that were refractory to the parent control virus. We conclude, therefore, that CRAds expressing bispecific targeting adapter molecules are promising agents for cancer treatment. Their use is likely to result in enhanced oncolytic replication in cancerous tissues and thus in more effective tumor regression.


Subject(s)
Adenovirus E1A Proteins/genetics , ErbB Receptors/metabolism , Genetic Therapy/methods , Genetic Vectors/genetics , Neoplasms/therapy , Receptors, Virus/genetics , Animals , Cell Line, Tumor , Coxsackie and Adenovirus Receptor-Like Membrane Protein , ErbB Receptors/immunology , Gene Targeting , Humans , Immunoglobulin Fc Fragments/immunology , Neoplasms/immunology , Neoplasms/metabolism , Receptors, Virus/deficiency , Virus Replication
16.
Br J Cancer ; 89(3): 577-84, 2003 Aug 04.
Article in English | MEDLINE | ID: mdl-12888833

ABSTRACT

In spite of aggressive surgery, irradiation and/or chemotherapy, treatment of malignant gliomas remains a major challenge in adults and children due to high treatment failure. We have demonstrated significant cell lysis and antitumour activity of the E1B-55 kDa-gene-deleted adenovirus ONYX-015 (dl1520, CI-1042; ONYX Pharmaceuticals) in subcutaneous human malignant glioma xenografts deriving from primary tumours. Here, we show the combined efficacy of this oncolytic therapy with radiation therapy. Total body irradiation (5 Gy) of athymic nude mice prior to intratumoral injections of ONYX-015 1 x 10(8) PFU daily for 5 consecutive days yielded additive tumour growth delays in the p53 mutant xenograft IGRG88. Radiation therapy was potentiated in the p53 functional tumour IGRG121 with a 'subtherapeutic' dose of 1 x 10(7) PFU daily for 5 consecutive days, inducing significant tumour growth delay, 90% tumour regression and 50% tumour-free survivors 4 months after treatment. These potentiating effects were not due to increased adenoviral infectivity or replication. Furthermore, cell lysis and induction of apoptosis, the major mechanisms for adenoviral antitumour activity, did not play a major role in the combined treatment strategy. Interestingly, the oncolytic adenovirus seemed to accelerate radiation-induced tumour fibrosis. Potentiating antitumour activity suggests the development of this combined treatment for these highly malignant tumours.


Subject(s)
Adenoviridae/genetics , Brain Neoplasms/radiotherapy , Brain Neoplasms/virology , Cell Death , Genetic Therapy , Glioma/radiotherapy , Glioma/virology , Radiation-Sensitizing Agents/pharmacology , Viral Vaccines/pharmacology , Animals , Brain Neoplasms/pathology , Female , Glioma/pathology , Mice , Mice, Nude , Neoplasms, Experimental , Transplantation, Heterologous
17.
J Control Release ; 87(1-3): 159-65, 2003 Feb 21.
Article in English | MEDLINE | ID: mdl-12618032

ABSTRACT

Recombinant adenoviral vectors are promising reagents for therapeutic interventions in humans, including gene therapy for biologically complex diseases like cancer and cardiovascular diseases. In this regard, the major advantage of adenoviral vectors is their superior in vivo gene transfer efficiency on a wide spectrum of both dividing and non-dividing cell types. However, this broad tropism at the same time represents an important limitation for their use in therapeutic applications where specific gene transfer is required. This limitation may be overcome by using targeting approaches. In this regard, targeting may be achieved at three levels: transductional targeting, translational targeting and targeting of the expressed transgene. Here we describe our research efforts towards cancer specific gene therapy using these different targeting approaches. The results show that targeting of adenoviral vectors may be achieved using cancer specific cell surface molecules for transductional and transgene targeting or cancer specific promoters for transcriptional targeting. Combinations of these targeting approaches should result in optimized cancer specific gene therapy.


Subject(s)
Adenoviridae/genetics , Drug Delivery Systems/methods , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/therapeutic use , Neoplasms/drug therapy , Animals , Humans , Neoplasms/genetics , Pliability
18.
J Gene Med ; 4(5): 510-6, 2002.
Article in English | MEDLINE | ID: mdl-12221644

ABSTRACT

BACKGROUND: Despite improvements in the treatment of osteosarcoma (OS) there are still too many patients who cannot benefit from current treatment modalities. Therefore, new therapeutic approaches are warranted. Here we explore the efficacy of targeted adenoviral based suicide gene therapy. METHODS AND RESULTS: Immunohistochemistry and FACS analysis detected low or absent expression levels of the primary adenovirus receptor CAR on human primary OS and human OS cell lines. These results predict a low infection efficiency and thus a reduced therapeutic effect. Targeting the adenoviruses to another receptor highly expressed on OS could overcome this limitation. We found epidermal growth factor receptor (EGFR) to be widely expressed on primary OS. Immunohistochemistry on primary tumor samples and FACS analysis on primary short-term cultures and four OS cell lines showed that EGFR was consistently expressed. The recombinant bispecific single-chain antibody 425-s11 redirects adenoviral vectors towards the EGFR. Adenovirus transduction experiments in the presence or absence of 425-s11 showed significantly enhanced gene transfer with the targeted adenoviral vector compared with the native vector (OS cell lines 2.5 to 7.2 times enhanced gene transfer and OS primary short term cultures 1.7 to 10 times enhanced gene transfer). On this basis, targeted suicide gene therapy experiments with AdCMVHSV-TK in combination with ganciclovir were performed. These experiments demonstrated up to 3.5-fold enhanced kill of OS cell lines and primary short-term cultures by the EGFR targeted vector. CONCLUSIONS: Suicide gene therapy with adenovirus targeted towards EGFR may have favorable therapeutic characteristics for future gene therapy applications in OS.


Subject(s)
Adenoviridae/genetics , ErbB Receptors/metabolism , Genetic Therapy , Osteosarcoma/therapy , Adolescent , Adult , Aged , Child , Coxsackie and Adenovirus Receptor-Like Membrane Protein , ErbB Receptors/genetics , Female , Genetic Vectors , Humans , Male , Middle Aged , Receptors, Virus/genetics , Receptors, Virus/metabolism
19.
Br J Cancer ; 87(6): 659-64, 2002 Sep 09.
Article in English | MEDLINE | ID: mdl-12237777

ABSTRACT

Irinotecan (CPT-11) is an anticancer agent for the treatment of colon cancer. CPT-11 can be considered as a prodrug, since it needs to be activated into the toxic drug SN-38 by the enzyme carboxylesterase. An approach to achieve tumour specific activation of CPT-11 is to transduce the cDNA encoding carboxylesterase into tumour cells. A secreted form of carboxylesterase may diffuse through a tumour mass and may activate CPT-11 extracellularly. This could enhance the antitumour efficacy by exerting a bystander effect on untransduced cells. In addition a secreted tumour-targeted form of carboxylesterase should prevent leakage of the enzyme from the site of the tumour into the circulation. We have constructed a secreted form of human liver carboxylesterase-2 by deletion of the cellular retention signal and by cloning the cDNA downstream of an Ig kappa leader sequence. The protein was secreted by transfected cells and showed both enzyme activity and efficient CPT-11 activation. To obtain a secreted, tumour-targeted form of carboxylesterase-2 the cDNA encoding the human scFv antibody C28 directed against the epithelial cell adhesion molecule EpCAM, was inserted between the leader sequence and carboxylesterase-2. This fusion protein showed CPT-11 activation and specific binding to EpCAM expressing cells. Importantly, in combination with CPT-11 both recombinant carboxylesterase proteins exerted strong antiproliferative effects on human colon cancer cells. They are, therefore, promising new tools for gene directed enzyme prodrug therapy approaches for the treatment of colon carcinoma with CPT-11.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Carboxylic Ester Hydrolases/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Prodrugs/pharmacology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Blotting, Western , COS Cells , Camptothecin/analogs & derivatives , Carboxylesterase , Carboxylic Ester Hydrolases/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Division/drug effects , Chlorocebus aethiops , Colonic Neoplasms/pathology , Drug Screening Assays, Antitumor , Gene Expression Regulation , Humans , Immunoenzyme Techniques , Irinotecan , Lymphokines/genetics , Lymphokines/metabolism , Plasmids , Polymerase Chain Reaction , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects
20.
J Urol ; 168(1): 266-72, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12050554

ABSTRACT

PURPOSE: We evaluated the delivery and oncolytic potential of targeted replication competent adenoviruses in bladder cancer lines. MATERIALS AND METHODS: Seven established human bladder cancer tumor lines (5637, SW800, TCCsup, J82, Scaber, T24 and 253J) were studied for the expression of integrins alpha(v)beta3, alpha(v)beta5, Coxsackievirus and adenovirus receptor, epidermal growth factor receptor (EGF-R) and epithelial cell adhesion molecule antigens using flow cytometry analysis. Bispecific single chain Fv fragments were used to target replication deficient luciferase reporter adenovirus to EGF-R (425-s11) or to epithelial cell adhesion molecule (C28-s11) antigens. Moreover, a fiber modified adenovirus targeting alpha(v)-integrins was studied. Replication competent serotype-5 adenoviruses attenuated to replicate specifically in retinoblastoma pRb (Ad5-d24) or p53 deficient (Ad5-d55K) cells were tested in vitro for oncolytic properties. RESULTS: Low to absent Coxsackievirus and adenovirus receptor expression was found in 5 of the 7 tumor lines (SW800, J82, T24, 5637 and Scaber). EGF-R expression was found in all cell lines, whereas elevated epithelial cell adhesion molecule expression was seen in 3 (5637, Scaber and TCCsup), alpha(v)beta3-integrin was found in 1 (Scaber) and alpha(v)beta5-integrin was found in 3 (TCCsup, 253J and T24). EGF-R targeting using 425-s11 improved transgene expression in all cell lines from 2.1 to 12.5 times over nontargeted viruses. Epithelial cell adhesion molecule and integrin targeting was inferior to EGF-R targeting with a maximal increase in transgene expression of 2 times for epithelial cell adhesion molecule in 5637cells and 1.6 times for integrin targeting in T24 cells. Comparison of the wild-type replication competent virus with conditionally replicating adenoviruses (Ad5-d55K and Ad5-d24) showed superior oncolytic activity for the latter 2 in all lines. Furthermore, improved cytotoxicity (29% to 33%) was obtained in 4 of the 7 lines after pre-incubation of Ad5-d24 with 425-s11. CONCLUSIONS: EGF-R directed bispecific single chain antibodies enhance adenovirus mediated transgene expression and oncolysis in bladder cancer lines.


Subject(s)
Adenoviridae/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Transitional Cell/genetics , Cytopathogenic Effect, Viral/genetics , ErbB Receptors/genetics , Gene Targeting , Genetic Therapy , Tumor Cells, Cultured/virology , Urinary Bladder Neoplasms/genetics , Virus Replication/genetics , Cell Adhesion Molecules/genetics , Cell Survival/genetics , Gene Expression Regulation, Viral/physiology , Humans , Integrins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...