Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Blood ; 130(14): 1628-1638, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28830887

ABSTRACT

Downregulation of CD20, a molecular target for monoclonal antibodies (mAbs), is a clinical problem leading to decreased efficacy of anti-CD20-based therapeutic regimens. The epigenetic modulation of CD20 coding gene (MS4A1) has been proposed as a mechanism for the reduced therapeutic efficacy of anti-CD20 antibodies and confirmed with nonselective histone deacetylase inhibitors (HDACis). Because the use of pan-HDACis is associated with substantial adverse effects, the identification of particular HDAC isoforms involved in CD20 regulation seems to be of paramount importance. In this study, we demonstrate for the first time the role of HDAC6 in the regulation of CD20 levels. We show that inhibition of HDAC6 activity significantly increases CD20 levels in established B-cell tumor cell lines and primary malignant cells. Using pharmacologic and genetic approaches, we confirm that HDAC6 inhibition augments in vitro efficacy of anti-CD20 mAbs and improves survival of mice treated with rituximab. Mechanistically, we demonstrate that HDAC6 influences synthesis of CD20 protein independently of the regulation of MS4A1 transcription. We further demonstrate that translation of CD20 mRNA is significantly enhanced after HDAC6 inhibition, as shown by the increase of CD20 mRNA within the polysomal fraction, indicating a new role of HDAC6 in the posttranscriptional mechanism of CD20 regulation. Collectively, our findings suggest HDAC6 inhibition is a rational therapeutic strategy to be implemented in combination therapies with anti-CD20 monoclonal antibodies and open up novel avenues for the clinical use of HDAC6 inhibitors.


Subject(s)
Antigens, CD20/genetics , Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Rituximab/pharmacology , Animals , Antigens, CD20/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase 6 , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/pathology , Mice, Inbred BALB C , Mice, SCID , RNA, Messenger/genetics , Tumor Cells, Cultured , Up-Regulation/drug effects
2.
BMC Cancer ; 18(1): 210, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29463237

ABSTRACT

BACKGROUND: Accumulating evidence suggest that autophagy plays a pivotal role in various anticancer therapies, including photodynamic therapy (PDT), acting as a pro-death or pro-survival mechanism in a context-dependent manner. Therefore, we aimed to determine the role of autophagy in Photofrin-based PDT. METHODS: In vitro cytotoxic/cytostatic effects of PDT were evaluated with crystal violet cell viability assay. Autophagy induction was analyzed by immunoblotting and immunofluorescence using anti-LC3 antibody. Autophagy was inhibited by shRNA-mediated ATG5 knockdown or CRISPR/Cas9-mediated ATG5 knockout. Apoptosis was assessed by flow cytometry analysis of propidium iodide and anexin V-positive cells as well as by detection of cleaved PARP and caspase 3 proteins using immunoblotting. Protein carbonylation was evaluated by the 2,4-dinitrophenylhydrazine (DNPH) method. RESULTS: Photofrin-PDT leads to robust autophagy induction in two cancer cell lines, Hela and MCF-7. shRNA-mediated knockdown of ATG5 only partially blocks autophagic response and only marginally affects the sensitivity of Hela and MCF-7 cells to PDT. ATG5 knockout in HeLa cell line utilizing CRISPR/Cas9 genome editing results in increased PDT-mediated cytotoxicity, which is accompanied by an enhanced apoptotic response and increased accumulation of carbonylated proteins. CONCLUSIONS: Altogether, these observations imply that autophagy contributes to Photofrin-PDT resistance by enabling clearance of carbonylated and other damaged proteins. Therefore, autophagy inhibition may serve as a strategy to improve PDT efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Autophagy/radiation effects , Dihematoporphyrin Ether/pharmacology , Photosensitizing Agents/pharmacology , Autophagy/genetics , Autophagy-Related Protein 5/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Gene Expression , Gene Knockdown Techniques , Gene Targeting , Humans , Light , Photochemotherapy , RNA Interference , RNA, Small Interfering/genetics
3.
Mol Med Rep ; 16(3): 3041-3048, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28713922

ABSTRACT

Gene transfer into normal quiescent human B cells is a challenging procedure. The present study aimed to investigate whether it is possible to increase the levels of transgene expression by using various types of promoters to drive the expression of selected genes­of­interest. To produce lentiviral particles, the present study used the 2nd generation psPAX2 packaging vector and the vesicular stomatitis virus ­expressing envelope vector pMD2.G. Subsequently, lentiviral vectors were generated containing various promoters, including cytomegalovirus (CMV), elongation factor­1 alpha (EF1α) and spleen focus­forming virus (SFFV). The present study was unable to induce satisfactory transduction efficiency in quiescent normal B cells; however, infection of normal B cells with Epstein­Barr virus resulted in increased susceptibility to lentiviral transduction. In addition, the SFFV promoter resulted in a higher level of transgene expression compared with CMV or EF1α promoters. As a proof­of concept that this approach allows for stable gene expression in normal B cells, the present study used bicistronic lentiviral vectors with genes encoding fluorescent reporter proteins, as well as X­box binding protein­1 and binding immunoglobulin protein.


Subject(s)
B-Lymphocytes/metabolism , Gene Transfer Techniques , Promoter Regions, Genetic , Adult , B-Lymphocytes/virology , Biomarkers/metabolism , CD40 Ligand/metabolism , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Feeder Cells/metabolism , Female , Fluorescence , Gene Expression , HEK293 Cells , Herpesvirus 4, Human/physiology , Humans , Internal Ribosome Entry Sites/genetics , Lentivirus/genetics , Male , Middle Aged , Spleen Focus-Forming Viruses/physiology , Transduction, Genetic , Transgenes
4.
Eur J Cancer ; 83: 19-27, 2017 09.
Article in English | MEDLINE | ID: mdl-28709135

ABSTRACT

Photodynamic therapy (PDT) has been shown to destroy tumour-associated lymphatic vessels. Therefore, we sought to investigate the functional outcomes of PDT-mediated damage to the lymphatic vessels. We observed that PDT with verteporfin, completely but transiently, blocks the functional lymphatic drainage in the orthotopic mammary tumour models. Sustained inhibition of lymphatic vessels regeneration induced by lenalidomide or the soluble form of vascular endothelial growth factor receptor 3 (sVEGFR3) that neutralises lymphangiogenic vascular endothelial growth factor C (VEGF-C), significantly impaired antitumour efficacy of PDT. Antilymphangiogenic compounds also significantly inhibited the ability of intratumourally inoculated dendritic cells (DCs) to translocate to local lymph nodes and diminished the number of tumour-infiltrating interferon-γ-secreting or tumour antigen-specific CD8+ T cells. Lenalidomide also abrogated antitumour effects of the combination immunotherapy with PDT and anti-programmed death-ligand 1 (PD-L1) antibodies. Altogether, these findings indicate that PDT-mediated damage to the lymphatic vessels negatively affects development of antitumour immunity, and that drugs that impair lymphatic vessel regeneration might not be suitable for the use in combination with PDT.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Cycle Checkpoints/drug effects , Lymphangiogenesis/drug effects , Photochemotherapy , Porphyrins/metabolism , Porphyrins/pharmacology , Thalidomide/analogs & derivatives , Animals , Disease Models, Animal , Female , Lenalidomide , Lymphangiogenesis/radiation effects , Lymphatic Vessels/drug effects , Lymphatic Vessels/pathology , Mice , Mice, Inbred C57BL , Photosensitizing Agents/pharmacology , Thalidomide/pharmacology , Vascular Endothelial Growth Factor Receptor-3/pharmacology , Verteporfin
5.
Photodiagnosis Photodyn Ther ; 14: 57-65, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26868051

ABSTRACT

BACKGROUND: Photodynamic therapy (PDT) has been shown to induce ablation and functional occlusion of tumor-associated lymphatic vessels. However, direct effects of PDT on lymphatic endothelial cells (LECs) have not been studied so far. The aim of this study was to elucidate molecular mechanisms of cell death induced by PDT in human LECs. METHODS: Verteporfin was used as a photosensitizer to investigate PDT-mediated damage of lymphatic vessels in mice using immunofluorescent staining and stereomicroscopy. In vitro dose-response studies were carried-out with crystal violet staining. Immunofluorescence, flow cytometry, immunoblotting and DNA electrophoresis were used to investigate the mechanisms of cell death in human LECs undergoing PDT. RESULTS: PDT induced an increase in the number of propidium iodide positive lymphatic endothelial cells in the mouse dermis. In in vitro studies dose-dependent cytotoxic effects of PDT towards LECs were observed. Typical hallmarks of apoptotic cell death, including Annexin V binding, loss of mitochondrial membrane potential, caspase activation, cleavage of PARP as well as DNA fragmentation were observed in LECs when PDT was used at high irradiation conditions, causing >80% cell death. At lower light fluencies causing <50% cell death PDT induced autophagy rather than apoptosis, as revealed by conversion of LC3-I to the autophagosomal LC3-II and formation of LC3 puncta. Z-VAD-FMK, a caspase inhibitor, prevented cell death induced by high-dose PDT only, while 3-methyladenine, an autophagy suppressor, inhibited cell death induced by low-dose PDT. CONCLUSIONS: Both apoptosis and autophagy are involved in cell death induced by verteporfin-PDT in LECs.


Subject(s)
Endothelial Cells/drug effects , Lymph/cytology , Photochemotherapy , Porphyrins/pharmacology , Animals , Blotting, Western , Cell Death/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Fluorescent Antibody Technique , Humans , Mice , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Porphyrins/therapeutic use , Verteporfin
6.
Oncol Rep ; 35(6): 3151-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27035643

ABSTRACT

Photodynamic therapy (PDT) exerts direct cytotoxic effects on tumor cells, destroys tumor blood and lymphatic vessels and induces local inflammation. Although PDT triggers the release of immunogenic antigens from tumor cells, the degree of immune stimulation is regimen-dependent. The highest immunogenicity is achieved at sub-lethal doses, which at the same time trigger cytoprotective responses, that include increased expression of glucose-regulated protein 78 (GRP78). To mitigate the cytoprotective effects of GRP78 and preserve the immunoregulatory activity of PDT, we investigated the in vivo efficacy of PDT in combination with EGF-SubA cytotoxin that was shown to potentiate in vitro PDT cytotoxicity by inactivating GRP78. Treatment of immunocompetent BALB/c mice with EGF-SubA improved the efficacy of PDT but only when mice were treated with a dose of EGF-SubA that exerted less pronounced effects on the number of T and B lymphocytes as well as dendritic cells in mouse spleens. The observed antitumor effects were critically dependent on CD8+ T cells and were completely abrogated in immunodeficient SCID mice. All these results suggest that GRP78 targeting improves in vivo PDT efficacy provided intact T-cell immune system.


Subject(s)
Antineoplastic Agents/administration & dosage , Epidermal Growth Factor/administration & dosage , Escherichia coli Proteins/administration & dosage , Heat-Shock Proteins/metabolism , Subtilisins/administration & dosage , Animals , Cell Line, Tumor , Combined Modality Therapy , Dihematoporphyrin Ether/pharmacology , Endoplasmic Reticulum Chaperone BiP , Female , Humans , Liver/drug effects , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, SCID , Photochemotherapy , Photosensitizing Agents/pharmacology , Recombinant Fusion Proteins/administration & dosage , Xenograft Model Antitumor Assays
8.
Biochem Pharmacol ; 93(4): 418-27, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25573101

ABSTRACT

Thioredoxins (Trx) together with thioredoxin reductases (TrxR) participate in the maintenance of protein thiol homeostasis and play cytoprotective roles in tumor cells. Therefore, thioredoxin-thioredoxin reductase system is considered to be a promising therapeutic target in cancer treatment. We have previously reported that SK053, a peptidomimetic compound targeting the thioredoxin-thioredoxin reductase system, induces oxidative stress and demonstrates antitumor activity in mice. In this study, we investigated the mechanisms of SK053-mediated tumor cell death. Our results indicate that SK053 induces apoptosis of Raji cells accompanied by the activation of the endoplasmic reticulum (ER) stress and induction of unfolded protein response. Incubation of tumor cells with SK053 induces increase in BiP, CHOP, and spliced XBP-1 levels, which precede induction of apoptosis. CHOP-deficient (CHOP(-/-)) mouse embryonic fibroblasts are more resistant to SK053-induced apoptosis as compared with normal fibroblasts indicating that the apoptosis of tumor cells depends on the expression of this transcription factor. Additionally, the ER-stress-induced apoptosis, caused by SK053, is strongly related with Trx expression levels. Altogether, our results indicate that SK053 induces ER stress-associated apoptosis and reveal a link between thioredoxin inhibition and induction of UPR in tumor cells.


Subject(s)
Apoptosis/physiology , Burkitt Lymphoma/metabolism , Dipeptides/toxicity , Endoplasmic Reticulum Stress/physiology , Methacrylates/toxicity , Oxidative Stress/physiology , Animals , Apoptosis/drug effects , Burkitt Lymphoma/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/drug effects , HEK293 Cells , Humans , Mice , Mice, Knockout , Oxidative Stress/drug effects
9.
Oncoimmunology ; 3: e28837, 2014.
Article in English | MEDLINE | ID: mdl-25057447

ABSTRACT

Photodynamic therapy has been shown to induce strong immunity against tumor cells expressing exogenous tumor-associated antigens (TAAs), including P1A antigen. Cancer cells can evade the immune system by epigenetic silencing of TAAs, while DNA methyltransferase inhibitors, such as 5-aza-2'-deoxycytidine (5-aza-dC) can restore the expression of silenced or downregulated TAA. Thus, epigenetic remodeling with 5-aza-dC combined with PDT can elicit robust and durable antitumor immunity.

10.
Eur J Cancer ; 50(7): 1370-81, 2014 May.
Article in English | MEDLINE | ID: mdl-24559534

ABSTRACT

Photodynamic therapy (PDT) of tumours is based on administration of a photosensitiser followed by irradiation of the tumour with visible light leading to production of reactive oxygen species that cause direct tumour cell death and vascular damage. PDT also initiates acute local inflammation, which facilitates the development of adaptive antitumour immunity. It has recently been reported that PDT can induce strong antitumour immunity towards tumours cells expressing P1A, tumour-associated antigen. Using four different tumour models, we show that antitumour immune response can be further improved when PDT is combined with a clinically approved epigenetic agent that induces expression of a silenced P1A antigen. Induction of P1A with 5-aza-2'-deoxycytidine, a methyltransferase inhibitor, resulted in potentiated antitumour effects in mice with Lewis lung carcinoma and 4T1 mammary carcinoma when combined with PDT treatment. In CT26 colon carcinoma and EMT6 mammary carcinoma models the combination therapy resulted in complete responses and long-term survival. All long-term surviving mice were resistant to re-inoculation with the same tumour cells. Antitumour efficacy of the combination treatment was severely impaired by depletion of CD8(+) cytotoxic T cells, whereas adoptive transfer of CD8(+) T cells from long-term surviving mice allowed for significant tumour growth delay in tumour-bearing mice. Taken together, these findings show that PDT leads to strong specific antitumour immune responses, and that epigenetic modification of tumour antigens levels may be a novel approach to further enhance the effectiveness of PDT. The present results provide a strong rationale for clinical development of this therapeutic approach.


Subject(s)
Antigens, Neoplasm/metabolism , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Carcinoma, Lewis Lung/drug therapy , Lung Neoplasms/drug therapy , Photochemotherapy , Animals , Azacitidine/pharmacology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Lewis Lung/immunology , Cell Line, Tumor , Decitabine , Disease Models, Animal , Female , Flow Cytometry , Lung Neoplasms/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL