Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 9137, 2024 04 21.
Article in English | MEDLINE | ID: mdl-38644422

ABSTRACT

To investigate the therapeutic potential of photodynamic therapy (PDT) for malignant gliomas arising in unresectable sites, we investigated the effect of tumor tissue damage by interstitial PDT (i-PDT) using talaporfin sodium (TPS) in a mouse glioma model in which C6 glioma cells were implanted subcutaneously. A kinetic study of TPS demonstrated that a dose of 10 mg/kg and 90 min after administration was appropriate dose and timing for i-PDT. Performing i-PDT using a small-diameter plastic optical fiber demonstrated that an irradiation energy density of 100 J/cm2 or higher was required to achieve therapeutic effects over the entire tumor tissue. The tissue damage induced apoptosis in the area close to the light source, whereas vascular effects, such as fibrin thrombus formation occurred in the area slightly distant from the light source. Furthermore, when irradiating at the same energy density, irradiation at a lower power density for a longer period of time was more effective than irradiation at a higher power density for a shorter time. When performing i-PDT, it is important to consider the rate of delivery of the irradiation light into the tumor tissue and to set irradiation conditions that achieve an optimal balance between cytotoxic and vascular effects.


Subject(s)
Glioma , Lasers, Semiconductor , Photochemotherapy , Photosensitizing Agents , Porphyrins , Animals , Photochemotherapy/methods , Glioma/drug therapy , Glioma/pathology , Porphyrins/pharmacology , Porphyrins/therapeutic use , Mice , Lasers, Semiconductor/therapeutic use , Cell Line, Tumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Disease Models, Animal , Allografts , Apoptosis/drug effects , Male
2.
Cytokine Growth Factor Rev ; 56: 69-82, 2020 12.
Article in English | MEDLINE | ID: mdl-32893095

ABSTRACT

In the past two decades there have been substantial advances in understanding the anti-cancer mechanisms of oncolytic viruses (OVs). OVs can mediate their effects directly, by preferentially infecting and killing tumour cells. Additionally, OVs can indirectly generate anti-tumour immune responses. These differing mechanisms have led to a paradoxical divergence in strategies employed to further increase the potency of oncolytic virotherapies. On one hand, the tumour neovasculature is seen as a vital lifeline to the survival of the tumour, leading some to use OVs to target the tumour vasculature in hopes to starve cancers. Therapeutics causing vascular collapse can potentiate tumour hypoxia, nutrient restriction and pro-inflammatory cytokine release, which has shown promise in oncological studies. On the other hand, the same vasculature plays an important role for the dissemination of OVs, trafficking of effector cells and other therapeutics, which has prompted researchers to find ways of normalizing the vasculature to enhance infiltration of leukocytes and delivery of therapeutic agents. This article describes the recent developments of therapies aimed to shut down versus normalize tumour vasculature in order to inform researchers striving to optimize OV-based therapies.


Subject(s)
Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Humans , Immunotherapy , Neoplasms/therapy
3.
Cancers (Basel) ; 12(9)2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32825648

ABSTRACT

Photodynamic therapy (PDT) is an attractive cancer treatment modality. Talaporfin sodium, a second-generation photosensitizer, results in lower systemic toxicity and relatively better selective tumor destruction than first-generation photosensitizers. However, the mechanism through which PDT induces vascular shutdown is unclear. In this study, the in vitro effects of talaporfin sodium-based PDT on human umbilical vein endothelial cells (HUVECs) were determined through cell viability and endothelial tube formation assays, and evaluation of the tubulin and F-actin dynamics and myosin light chain (MLC) phosphorylation. Additionally, the effects on tumor blood flow and tumor vessel destruction were assessed in vivo. In the HUVECs, talaporfin sodium-based PDT induced endothelial tube destruction and microtubule depolymerization, triggering the formation of F-actin stress fibers and a significant increase in MLC phosphorylation. However, pretreatment with the Rho-associated protein kinase (ROCK) inhibitor, Y27632, completely prevented PDT-induced stress fiber formation and MLC phosphorylation. The in vivo analysis and pathological examination revealed that the PDT had significantly decreased the tumor blood flow and the active area of the tumor vessel. We concluded that talaporfin sodium-based PDT induces the shutdown of existing tumor vessels via the RhoA/ROCK pathway by activating the Rho-GTP pathway and decreasing the tumor blood flow.

4.
Photodiagnosis Photodyn Ther ; 26: 436-441, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31054334

ABSTRACT

Indocyanine green lactosome (ICG-lactosome) is an attractive new-generation agent for photodynamic therapy (PDT) that is characterized by a near-infrared excitation wavelength and high stability in the bloodstream. Fluorescence imaging has been used to examine its pharmacokinetics in vivo, but no depth-resolved information can be obtained with this method. In this study, we applied photoacoustic (PA) imaging to visualize the depth distribution of ICG-lactosome in a mouse subcutaneous tumor model. With this method, the depth distribution of blood vessels can also be visualized, enabling detection of vascular shutdown effects due to PDT. We performed PA imaging of both the distributions of ICG-lactosome and blood vessels in a tumor before and after PDT, and we found that PA signals originating from ICG-lactosome were greatly increased at 18 h after drug injection but rapidly decreased after PDT. These results indicate efficient accumulation of ICG-lactosome and rapid photobleaching due to the PDT reaction in the tumor, respectively. After PDT, PA amplitudes of hemoglobin were significantly decreased, being attributable to vascular shutdown effects. These results show the usefulness of PA imaging for monitoring not only photosensitizer accumulation and bleaching but also vascular responses in PDT with ICG-lactosome. This method can be applied to the diagnosis of many types of PDT processes.


Subject(s)
Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/drug therapy , Indocyanine Green/pharmacology , Photoacoustic Techniques , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Disease Models, Animal , Female , Indocyanine Green/pharmacokinetics , Mice , Mice, Nude , Photosensitizing Agents/pharmacokinetics
5.
Photodiagnosis Photodyn Ther ; 20: 95-106, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28865875

ABSTRACT

BACKGROUND: Photochemical internalization (PCI) is a novel technique for delivery of active macromolecules into cancerous cells, via light activation of a specific photosensitizer and a low dose systemic drug. Numerous pre-clinical studies and one clinical trial have confirmed the treatment potential in carcinomas. Soft tissue sarcomas are rare and generally resistant to radio- and chemotherapy. Due to treatment resistance and surgical morbidity in sarcoma care, we seek to increase knowledge on PCI effects in sarcomas by studying two different, but closely related leiomyosarcomas. METHODS: MES-SA and SK-LMS-1 tumours were established in the leg muscles of athymic mice. Treatment effects after AlPcS2a-PCI of bleomycin, PCI with no drug (photodynamic therapy, PDT) and control groups were evaluated by: 1) assessment of tumour growth, 2) uptake of contrast agent during MRI and 3) histopathology. RESULTS: PCI of bleomycin induced a similar and significant increase in time to reach the end point in both tumour models, while neither responded to AlPcS2a-PDT. In the MES-SA tumours PCI reduced the growth rate, while in the SK-LMS-1 tumours the growth was blocked for 12days followed by exponential growth close to that of untreated tumours. SK-LMS-1 tumours were more homogenously and better vascularized than MES-SA. After PCI the vascular shutdown was more complete in the SK-LMS-1 tumours than in the MES-SA tumours. CONCLUSIONS: AlPcS2a-based PCI, but not PDT, induced significant tumour growth delay in the evaluated sarcomas. Cellular responsiveness to bleomycin and tumour vascularity are identified as predictive markers for PCI treatment effects.


Subject(s)
Bleomycin/pharmacology , Indoles/pharmacology , Leiomyosarcoma/drug therapy , Organometallic Compounds/pharmacology , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Genes, p53/genetics , Lasers, Semiconductor , Leiomyosarcoma/genetics , Mice , Mice, Knockout , Mice, Nude , Neovascularization, Pathologic
6.
Bioorg Med Chem ; 25(5): 1630-1642, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28143677

ABSTRACT

5,7-Dihydro-3,9,10,11-tetramethoxybenz[c,e]oxepin-4-ol 1, prepared from a dibenzyl ether precursor via Pd-catalysed intramolecular direct arylation, possesses broad-spectrum in vitro cytotoxicity towards various tumour cell lines, and induces vascular shutdown, necrosis and growth delay in tumour xenografts in mice at sub-toxic doses. The biological properties of 1 and related compounds can be attributed to their ability to inhibit microtubule assembly at the micromolar level, by binding reversibly to the same site of the tubulin αß-heterodimer as colchicine 2 and the allocolchinol, N-acetylcolchinol 4.


Subject(s)
Dibenzoxepins/metabolism , Neoplasms/blood supply , Tubulin/metabolism , Animals , Cell Line, Tumor , Dibenzoxepins/chemistry , Dibenzoxepins/pharmacology , Dose-Response Relationship, Drug , Heterografts , Humans , Mice , Molecular Structure
7.
Biochim Biophys Acta ; 1830(10): 4594-603, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23747302

ABSTRACT

BACKGROUND: Broader clinical acceptance of photodynamic therapy is currently hindered by (a) poor depth efficacy, and (b) predisposition towards establishment of an angiogenic environment during the treatment. Improved depth efficacy is being sought by exploiting the NIR tissue transparency window and by photo-activation using two-photon absorption (2PA). Here, we use two-photon activation of PDT sensitizers, untargeted and targeted to SST2 receptors or EGF receptors, to achieve deep tissue treatment. METHODS: Human tumor lines, positive or negative for SST2r expression were used, as well as murine 3LL cells and bovine aortic endothelial cells. Expression of SST2 receptors on cancer cells and tumor vasculature was evaluated in vitro and frozen xenograft sections. PDT effects on tumor blood flow were followed using in vivo scanning after intravenous injection of FITC conjugated dextran 150K. Dependence of the PDT efficacy on the laser pulse duration was evaluated. Effectiveness of targeting to vascular SST2 receptors was compared to that of EGF receptors, or no targeting. RESULTS: Tumor vasculature stained for SST2 receptors even in tumors from SST2 receptor negative cell lines, and SST2r targeted PDT led to tumor vascular shutdown. Stretching the pulse from ~120fs to ~3ps led to loss of the PDT efficacy especially at greater depth. PDT targeted to SST2 receptors was much more effective than untargeted PDT or PDT targeted to EGF receptors. GENERAL SIGNIFICANCE: The use of octreotate to target SST2 receptors expressed on tumor vessels is an excellent approach to PDT with few recurrences and some long term cures.


Subject(s)
Blood Vessels/drug effects , Neoplasms/drug therapy , Photochemotherapy , Receptors, Somatostatin/drug effects , Spectroscopy, Near-Infrared , Cell Line, Tumor , Humans , Photons
8.
Article in Korean | WPRIM (Western Pacific) | ID: wpr-120710

ABSTRACT

OBJECTIVE: Arsenic trioxide (As2O3) is known to have potent anti-vascular activity and significantly suppress solid tumor growth. The present study was conducted to investigate the vascular shutdown effects of a novel arsenic compound, tetraasrsenic oxide (As4O6), in comparison with As2O3 using cervical cancer animal model. METHODS: Mice tumor challenge model was used C57BL/6 mice transplanted with TC-1 cells. After the growth of tumors was reached up 200~250 mm3, mice were divided into 3 groups randomly for control and treatment of either As2O3 or As4O6. As2O3 and As4O6 was treated by i.p. injection. The tumor size was caliperated in twice for weeks and anti-vascular effect were assessed by Evans blue extraction assay and Hoechst 33342 staining. In tumor tissue, histopathological feaure was obserevd by hematoxylin and eosin (H&E) staining. RESULTS: In mice treated with either As2O3 and As4O6 (i.p.), both of As2O3 and As4O6 was significantly suppressed the tumor growth compared with control group. Moreover, effect of As4O6 is more pronounced. These tumor growth inhibition is led to the massive necrosis and vacular shutdown in tumor tissue. CONCLUSION: This study suggests that As4O6 may have potential anticancer activity via vascular shutdown in C57BL/6 mice transplanted with TC-1 cells.


Subject(s)
Animals , Mice , Arsenic , Arsenicals , Benzimidazoles , Eosine Yellowish-(YS) , Evans Blue , Hematoxylin , Models, Animal , Necrosis , Oxides , Transplants , Uterine Cervical Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL
...