Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(7)2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38612758

ABSTRACT

The prevention of tumor recurrence by the successful targeting of glioma stem cells endowed with a tumor-initiating capacity is deemed the key to the long-term survival of glioblastoma patients. Glioma stem cells are characterized by their marked therapeutic resistance; however, recent evidence suggests that they have unique vulnerabilities that may be therapeutically targeted. We investigated MDM2 expression levels in glioma stem cells and their non-stem cell counterparts and the effects of the genetic and pharmacological inhibition of MDM2 on the viability of these cells as well as downstream molecular pathways. The results obtained showed that MDM2 expression was substantially higher in glioma stem cells than in their non-stem cell counterparts and also that the inhibition of MDM2, either genetically or pharmacologically, induced a more pronounced activation of the p53 pathway and apoptotic cell death in the former than in the latter. Specifically, the inhibition of MDM2 caused a p53-dependent increase in the expression of BAX and PUMA and a decrease in the expression of survivin, both of which significantly contributed to the apoptotic death of glioma stem cells. The present study identified the MDM2-p53 axis as a novel therapeutic vulnerability, or an Achilles' heel, which is unique to glioma stem cells. Our results, which suggest that non-stem, bulk tumor cells are less sensitive to MDM2 inhibitors, may help guide the selection of glioblastoma patients suitable for MDM2 inhibitor therapy.


Subject(s)
Glioblastoma , Glioma , Humans , Tumor Suppressor Protein p53/genetics , Glioma/drug therapy , Glioma/genetics , Apoptosis , Neoplastic Stem Cells , Proto-Oncogene Proteins c-mdm2/genetics
2.
Cancers (Basel) ; 15(17)2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37686602

ABSTRACT

The development of MDM4 inhibitors as an approach to reactivating p53 in human cancer is attracting increasing attention; however, whether they affect the function of MDM2 and how they interact with MDM2 inhibitors remain unknown. We addressed this question in the present study using CEP-1347, an inhibitor of MDM4 protein expression. The effects of CEP-1347, the genetic and/or pharmacological inhibition of MDM2, and their combination on the p53 pathway in malignant brain tumor cell lines expressing wild-type p53 were investigated by RT-PCR and Western blot analyses. The growth inhibitory effects of CEP-1347 alone or in combination with MDM2 on inhibition were examined by dye exclusion and/or colony formation assays. The treatment of malignant brain tumor cell lines with CEP-1347 markedly increased MDM2 protein expression, while blocking CEP-1347-induced MDM2 overexpression by genetic knockdown augmented the effects of CEP-1347 on the p53 pathway and cell growth. Blocking the MDM2-p53 interaction using the small molecule MDM2 inhibitor RG7112, but not MDM2 knockdown, reduced MDM4 expression. Consequently, RG7112 effectively cooperated with CEP-1347 to reduce MDM4 expression, activate the p53 pathway, and inhibit cell growth. The present results suggest the combination of CEP-1347-induced MDM2 overexpression with the selective inhibition of MDM2's interaction with p53, while preserving its ability to inhibit MDM4 expression, as a novel and rational strategy to effectively reactivate p53 in wild-type p53 cancer cells.

3.
Biomedicines ; 11(7)2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37509605

ABSTRACT

A significant proportion of meningiomas are clinically aggressive, but there is currently no effective chemotherapy for meningiomas. An increasing number of studies have been conducted to develop targeted therapies, yet none have focused on the p53 pathway as a potential target. In this study, we aimed to determine the in vitro and in vivo effects of CEP-1347, a small-molecule inhibitor of MDM4 with known safety in humans. The effects of CEP-1347 and MDM4 knockdown on the p53 pathway in human meningioma cell lines with and without p53 mutation were examined by RT-PCR and Western blot analyses. The growth inhibitory effects of CEP-1347 were examined in vitro and in a mouse xenograft model of meningioma. In vitro, CEP-1347 at clinically relevant concentrations inhibited MDM4 expression, activated the p53 pathway in malignant meningioma cells with wild-type p53, and exhibited preferential growth inhibitory effects on cells expressing wild-type p53, which was mostly mimicked by MDM4 knockdown. CEP-1347 effectively inhibited the growth of malignant meningioma xenografts at a dose that was far lower than the maximum dose that could be safely given to humans. Our findings suggest targeting the p53 pathway with CEP-1347 represents a novel and viable approach to treating aggressive meningiomas.

4.
Int J Mol Sci ; 24(13)2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37445993

ABSTRACT

The deregulation of the FOXM1 transcription factor is a key molecular alteration in ovarian cancer, contributing to the development and progression of ovarian cancer via activation of the target genes. As such, FOXM1 is a highly attractive therapeutic target in the treatment of ovarian cancer, but there has been no clinically tested FOXM1 inhibitor to date. We investigated in this study the effects of domatinostat, a class I-selective HDAC inhibitor currently in the clinical stage of development as a cancer therapeutic, on the expression of FOXM1 and viability of ovarian cancer cells. Cell viability, as well as protein and mRNA expression of FOXM1 and its transcriptional target survivin, was examined after domatinostat treatment of TOV21G and SKOV3 ovarian cancer cell lines in the absence or presence of cisplatin and paclitaxel. The effect of FOXM1 knockdown on survivin expression and those of genetic and pharmacological inhibition of survivin alone or in combination with the chemotherapeutic agents on cell viability were also examined. Domatinostat reduced the protein and mRNA expression of FOXM1 and survivin and also the viability of ovarian cancer cells alone and in combination with cisplatin or paclitaxel at clinically relevant concentrations. Knockdown experiments showed survivin expression was dependent on FOXM1 in ovarian cancer cells. Survivin inhibition was sufficient to reduce the viability of ovarian cancer cells alone and in combination with the chemotherapeutic agents. Our findings suggest that domatinostat, which effectively targets the FOXM1-survivin axis required for the viability of ovarian cancer cells, is a promising option for the treatment of ovarian cancer.


Subject(s)
Cisplatin , Ovarian Neoplasms , Humans , Female , Survivin/genetics , Survivin/metabolism , Cisplatin/therapeutic use , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Cell Line, Tumor , RNA, Messenger/genetics , Apoptosis , Cell Proliferation , Gene Expression Regulation, Neoplastic , Drug Resistance, Neoplasm , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism
5.
J Craniofac Surg ; 34(7): 2092-2094, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37271863

ABSTRACT

Some cases of moderate or severe cryptotia are accompanied by a shortage of the helix. Although various operative techniques for correcting cryptotia have been reported, elongation of the helix is not considered in most of those techniques. In cases of a shortage of the helix, a drooped wide helix like a constricted ear or a cranially and posteriorly hypoplastic ear, which is characteristic of cryptotia, can appear after surgery if the helix has not been elongated. We previously reported a large Z-plasty technique that has become one of the popular techniques for correcting cryptotia. However, satisfactory results are not always achieved by using this technique in cases with a shortage of the helix. We developed a new technique (double Z-plasty) in which a small Z-plasty in the helical rim is added to the usual large Z-plasty technique. An improved helical shape and enlargement of the ear can be achieved by using this technique. Almost all types of cryptotia can be treated by appropriately using the large Z-plasty and double Z-plasty techniques.

6.
Anticancer Res ; 43(3): 1131-1138, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36854532

ABSTRACT

BACKGROUND/AIM: Givinostat is a pan-histone deacetylase (HDAC) inhibitor that has demonstrated excellent tolerability as well as efficacy in patients with polycythemia vera. Accumulating in vitro and in vivo evidence suggests givinostat is also promising as a therapeutic agent targeting glioma stem cells (GSCs), the cancer stem cells of glioblastoma (GBM) considered responsible for its intractable nature. However, it remains to be shown how givinostat impacts the therapeutic effects of temozolomide, a DNA-alkylating agent and the key component of GBM treatment given not only during postoperative radiotherapy but also thereafter as maintenance chemotherapy. MATERIALS AND METHODS: The effects of givinostat and knockdown of O6-methylguanine-DNA methyltransferase (MGMT) or Sp1 on the mRNA and protein expression of relevant genes in human GSC lines were examined by RT-PCR and western blot analyses. The dye exclusion method was used to evaluate cell viability. RESULTS: Givinostat enhanced the cytotoxic activity of temozolomide in GSC lines expressing MGMT, in which the MGMT expression was shown to contribute to their temozolomide resistance. Givinostat inhibited MGMT expression in GSCs and, in parallel, the expression of Sp1, a transcription factor involved in the control of MGMT promoter activity. Knockdown experiments demonstrated Sp1 expression was indeed required for MGMT expression in GSCs. CONCLUSION: Givinostat, in addition to its own cytotoxic activity, sensitizes GSCs to temozolomide by inhibiting Sp1-dependent MGMT expression in GSCs. Combining givinostat with temozolomide could therefore be a rational therapeutic strategy to effectively eliminate GSCs and thus help overcome the therapy resistance of GBM.


Subject(s)
Glioblastoma , Glioma , Neoplastic Stem Cells , O(6)-Methylguanine-DNA Methyltransferase , Temozolomide , Humans , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Glioblastoma/metabolism , Glioma/drug therapy , Glioma/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/metabolism , Temozolomide/pharmacology , Tumor Suppressor Proteins/genetics
7.
Cancers (Basel) ; 16(1)2023 Dec 25.
Article in English | MEDLINE | ID: mdl-38201546

ABSTRACT

Uveal melanoma (UM) is among the most common primary intraocular neoplasms in adults, with limited therapeutic options for advanced/metastatic disease. Since UM is characterized by infrequent p53 mutation coupled with the overexpression of MDM4, a major negative regulator of p53, we aimed to investigate in this study the effects on UM cells of CEP-1347, a novel MDM4 inhibitor with a known safety profile in humans. We also examined the impact of CEP-1347 on the protein kinase C (PKC) pathway, known to play a pivotal role in UM cell growth. High-grade UM cell lines were used to analyze the effects of genetic and pharmacological inhibition of MDM4 and PKC, respectively, as well as those of CEP-1347 treatment, on p53 expression and cell viability. The results showed that, at its clinically relevant concentrations, CEP-1347 reduced not only MDM4 expression but also PKC activity, activated the p53 pathway, and effectively inhibited the growth of UM cells. Importantly, whereas inhibition of either MDM4 expression or PKC activity alone failed to efficiently activate p53 and inhibit cell growth, inhibition of both resulted in effective activation of p53 and inhibition of cell growth. These data suggest that there exists a hitherto unrecognized interaction between MDM4 and PKC to inactivate the p53-dependent growth control in UM cells. CEP-1347, which dually targets MDM4 and PKC, could therefore be a promising therapeutic candidate in the treatment of UM.

8.
Anticancer Res ; 42(10): 4727-4733, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36192008

ABSTRACT

BACKGROUND/AIM: The development of pharmacological inhibitors targeting negative regulators of p53, such as murine double minute (MDM) 2 and, more recently, MDM4, has been actively pursued as a potential strategy to treat cancers with wild-type p53. We previously showed that CEP-1347, a small molecule kinase inhibitor originally developed for the treatment of Parkinson's disease, suppressed MDM4 expression and activated wild-type p53 in retinoblastoma cells. However, it remains unknown whether CEP-1347 acts as an MDM4 inhibitor and as such activates p53 in other types of human cancer cells. MATERIALS AND METHODS: The effects of CEP-1347 and MDM4 knockdown on the mRNA and protein expression of components of the p53 pathway, including MDM4, in human glioma cell lines with and without p53 mutation were examined by RT-PCR and western blot analyses. Trypan blue dye exclusion was used to examine the effect of CEP-1347 on cell growth. RESULTS: CEP-1347 decreased the expression of MDM4, increase that of p53, and activated the p53 pathway in glioma cells with wild-type p53. Knockdown-mediated inhibition of MDM4 expression in a glioma cell line with wild-type p53 that overexpresses MDM4 resulted in increased p53 expression and activation of the p53 pathway. CEP-1347 preferentially inhibited the growth of glioma cells with wild-type p53 without showing toxicity to normal cells at clinically relevant concentrations. CONCLUSION: Our findings suggest CEP-1347 is a novel inhibitor of MDM4 protein expression and as such activates p53 to inhibit the growth of cancer cells with wild-type p53, including retinoblastoma and glioblastoma.


Subject(s)
Glioma , Retinal Neoplasms , Retinoblastoma , Carbazoles , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Gene Expression Regulation, Neoplastic , Glioma/drug therapy , Glioma/genetics , Humans , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Messenger/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Int J Mol Sci ; 23(15)2022 Aug 07.
Article in English | MEDLINE | ID: mdl-35955917

ABSTRACT

Glioma stem cells (GSCs), the cancer stem cells of glioblastoma multiforme (GBM), contribute to the malignancy of GBM due to their resistance to therapy and tumorigenic potential; therefore, the development of GSC-targeted therapies is urgently needed to improve the poor prognosis of GBM patients. The molecular mechanisms maintaining GSCs need to be elucidated in more detail for the development of GSC-targeted therapy. In comparison with patient-derived GSCs and their differentiated counterparts, we herein demonstrated for the first time that phospholipase C (PLC)ε was highly expressed in GSCs, in contrast to other PLC isoforms. A broad-spectrum PLC inhibitor suppressed the viability of GSCs, but not their stemness. Nevertheless, the knockdown of PLCε suppressed the survival of GSCs and induced cell death. The stem cell capacity of residual viable cells was also suppressed. Moreover, the survival of mice that were transplanted with PLCε knockdown-GSCs was longer than the control group. PLCε maintained the stemness of GSCs via the activation of JNK. The present study demonstrated for the first time that PLCε plays a critical role in maintaining the survival, stemness, and tumor initiation capacity of GSCs. Our study suggested that PLCε is a promising anti-GSC therapeutic target.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Glioblastoma/metabolism , Glioma/drug therapy , Glioma/genetics , Glioma/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Neoplastic Stem Cells/metabolism , Phosphoinositide Phospholipase C , Type C Phospholipases/metabolism
10.
Int J Mol Sci ; 23(15)2022 Jul 22.
Article in English | MEDLINE | ID: mdl-35897656

ABSTRACT

Cancer stem cells (CSCs) are in general characterized by higher resistance to cell death and cancer therapies than non-stem differentiated cancer cells. However, we and others have recently revealed using glioma stem cells (GSCs) as a model that, unexpectedly, CSCs have specific vulnerabilities that make them more sensitive to certain drugs compared with their differentiated counterparts. We aimed in this study to discover novel drugs targeting such Achilles' heels of GSCs as anti-GSC drug candidates to be used for the treatment of glioblastoma, the most therapy-resistant form of brain tumors. Here we report that domatinostat (4SC-202), a class I HDAC inhibitor, is one such candidate. At concentrations where it showed no or minimal growth inhibitory effect on differentiated GSCs and normal cells, domatinostat effectively inhibited the growth of GSCs mainly by inducing apoptosis. Furthermore, GSCs that survived domatinostat treatment lost their self-renewal capacity. These results suggested that domatinostat is a unique drug that selectively eliminates GSCs not only physically by inducing cell death but also functionally by inhibiting their self-renewal. Our findings also imply that class I HDACs and/or LSD1, another target of domatinostat, may possibly have a specific role in the maintenance of GSCs and therefore could be an attractive target in the development of anti-GSC therapies.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Benzamides , Brain Neoplasms/metabolism , Cell Line, Tumor , Glioblastoma/metabolism , Glioma/metabolism , Histone Deacetylase Inhibitors/therapeutic use , Humans , Neoplastic Stem Cells/metabolism
11.
Cancers (Basel) ; 14(7)2022 Mar 27.
Article in English | MEDLINE | ID: mdl-35406478

ABSTRACT

Despite several clinical trials with encouraging findings, effective standard systemic therapies have yet to be established for malignant meningioma and the prognosis of these patients remains poor. Accumulating preclinical and clinical evidence suggests that gemcitabine is effective against malignant meningioma. To identify drugs with therapeutic effects that may be enhanced in combination with gemcitabine, we screened drugs that have been tested in preclinical and clinical trials for meningioma. In IOMM-Lee and HKBMM malignant meningioma cells, gemcitabine enhanced the growth inhibitory effects of the mTOR inhibitor everolimus, the clinical benefits of which have been demonstrated in patients with meningioma. The synergistic growth inhibitory effects of this combination were accompanied by cellular senescence characterized by an increase in senescence-associated ß-galactosidase activity. To enhance the effects of this combination, we screened senolytic drugs that selectively kill senescent cells, and found that navitoclax, an inhibitor of anti-apoptotic BCL-2 family proteins, effectively reduced the number of viable malignant meningioma cells in combination with everolimus and gemcitabine by inducing apoptotic cell death. The suppression of tumor growth in vivo by the combination of everolimus with gemcitabine was significantly stronger than that by either treatment alone. Moreover, navitoclax, in combination with everolimus and gemcitabine, significantly reduced tumor sizes with an increase in the number of cleaved caspase-3-positive apoptotic cells. The present results suggest that the addition of gemcitabine with or without navitoclax to everolimus is a promising strategy that warrants further evaluation in future clinical trials for malignant meningioma.

12.
Neurooncol Adv ; 3(1): vdab148, 2021.
Article in English | MEDLINE | ID: mdl-34765973

ABSTRACT

BACKGROUND: Malignant meningioma is an aggressive tumor that requires adjuvant radiotherapy after surgery, yet there has been no standard systemic therapy established so far. We recently reported that malignant meningioma cells are highly sensitive to gemcitabine; however, it remains unknown whether or how gemcitabine interacts with ionizing radiation (IR) in malignant meningioma cells. METHODS: We examined the radiosensitization effects of gemcitabine using malignant meningioma cell lines and xenografts and explored the underlying mechanisms. RESULTS: Gemcitabine sensitized malignant meningioma cells to IR through the induction of senescence both in vitro and in vivo. Gemcitabine augmented the intracellular production of reactive oxygen species (ROS) by IR, which, together with cell growth suppression/senescence induced by this combination, was inhibited by N-acetyl-cysteine, suggesting a pivotal role for ROS in these combinatorial effects. Navitoclax, a senolytic drug that inhibits Bcl-2 proteins, further enhanced the effects of the combination of gemcitabine and IR by strongly inducing apoptotic cell death in senescent cells. CONCLUSION: These results not only indicate the potential of gemcitabine as a candidate radiosensitizer for malignant meningioma, but also reveal a novel role for gemcitabine radiosensitization as a means to create a therapeutic vulnerability of senescent meningioma cells to senolytics.

13.
Int J Mol Sci ; 22(21)2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34769063

ABSTRACT

Glioblastoma (GBM) is one of the deadliest of all human cancers. Developing therapies targeting GBM cancer stem cells or glioma stem cells (GSCs), which are deemed responsible for the malignancy of GBM due to their therapy resistance and tumor-initiating capacity, is considered key to improving the dismal prognosis of GBM patients. In this study, we found that folate antagonists, such as methotrexate (MTX) and pemetrexed, are selectively cytotoxic to GSCs, but not to their differentiated counterparts, normal fibroblasts, or neural stem cells in vitro, and that the high sensitivity of GCSs to anti-folates may be due to the increased expression of RFC-1/SLC19A1, the reduced folate carrier that transports MTX into cells, in GSCs. Of note, in an in vivo serial transplantation model, MTX alone failed to exhibit anti-GSC effects but promoted the anti-GSC effects of CEP1347, an inducer of GSC differentiation. This suggests that folate metabolism, which plays an essential role specifically in GSCs, is a promising target of anti-GSC therapy, and that the combination of cytotoxic and differentiation therapies may be a novel and promising approach to effectively eliminate cancer stem cells.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cell Differentiation/drug effects , Folic Acid/metabolism , Glioma/drug therapy , Neoplastic Stem Cells/drug effects , Neural Stem Cells/drug effects , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioma/metabolism , Heterografts/drug effects , Heterografts/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism
14.
Life (Basel) ; 11(9)2021 Aug 27.
Article in English | MEDLINE | ID: mdl-34575034

ABSTRACT

Cancer stem cells (CSCs) have high tumor-initiating capacity and are resistant to chemotherapeutic reagents; thus eliminating CSCs is essential to improving the prognosis. Recently, we reported that dexamethasone increases the effects of gemcitabine on pancreatic CSCs; however, the mechanism involved remains to be fully elucidated. In this study, we explored the role of reactive oxygen species (ROS) in the dexamethasone-induced chemosensitization of CSCs. Dexamethasone increased the growth-inhibitory effects of gemcitabine and 5-fluorouracil, whereas N-acetyl-cysteine, a ROS scavenger, abolished this effect. Although dexamethasone alone did not increase ROS levels, dexamethasone promoted the increase in ROS levels induced by gemcitabine and 5-fluorouracil. Dexamethasone treatment reduced the expression of NRF2, a key regulator of antioxidant responses, which was attenuated by siRNA-mediated knockdown of the glucocorticoid receptor. Furthermore, brusatol, a suppressor of NRF2, sensitized pancreatic CSCs to gemcitabine and 5-fluorouracil. Of note, essentially, the same mechanism was functional in ovarian and colon CSCs treated by the combination of dexamethasone and chemotherapeutic agents. Our study suggests that dexamethasone can sensitize CSCs to chemotherapeutic agents by promoting chemotherapy-induced ROS production through suppressing NRF2 expression.

15.
Anticancer Res ; 41(9): 4321-4331, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34475052

ABSTRACT

BACKGROUND/AIM: Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are key drugs in cancer treatment due to their minor adverse effects and outstanding anticancer effects. However, drugs for overcoming EGFR-TKI resistance are not in clinical use so far. Therefore, to overcome resistance, we focused on lurasidone, a new antipsychotic drug, due to its mild adverse effect profile from the viewpoint of drug repositioning. MATERIALS AND METHODS: We explored the effects of lurasidone alone or in combination with EGFR-TKI on the growth of osimertinib-resistant cancer cells the anti-apoptotic marker expression such as survivin, and autophagy levels by LC-3B expression. RESULTS: Within a non-toxic concentration range in normal cells, lurasidone and osimertinib combination therapy showed a growth-inhibitory effect in osimertinib-resistant cancer cells in vitro and in vivo. Furthermore, lurasidone decreased survivin expression and mildly induced autophagy. CONCLUSION: Lurasidone may increase the sensitivity to osimertinib in osimertinib-resistant cancer cells in drug repurposing.


Subject(s)
Acrylamides/administration & dosage , Aniline Compounds/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/drug therapy , Lurasidone Hydrochloride/administration & dosage , Survivin/metabolism , A549 Cells , Acrylamides/pharmacology , Aniline Compounds/pharmacology , Animals , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/metabolism , Lurasidone Hydrochloride/pharmacology , Mice , Microtubule-Associated Proteins/metabolism , Xenograft Model Antitumor Assays
16.
Plast Reconstr Surg Glob Open ; 9(2): e3058, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33680629

ABSTRACT

Damage and loss of ear cartilage can easily occur in a burned ear accompanied by severe extensive burns due to exposure of the cartilage or chondritis. Deformity can also occur due to later development of scar contracture despite minimal damage to the ear cartilage when the injury occurred. We have developed a new technique for treatment of a deep burn in the whole ear. METHODS: In the new technique, the ear is amputated, and soft tissues are denuded. The cartilage is banked in a subcutaneous pocket in the chest and later regrafted in combination with costal cartilage. RESULTS: Although techniques for regrafting banked ear cartilage have been reported, the reconstructed ear cannot acquire a good shape because of absorption of the cartilage or lack of intensity to sustain the outline of the ear. Meanwhile, when we tried to reconstruct an ear by only using costal cartilage, we found it difficult to fabricate a frame because most patients are adults in whom the costal cartilage is too rigid and fragile to be shaved or combined. In our technique, the frame has both the advantages of sufficient intensity in costal cartilage and a smooth curved surface together with elasticity in the ear cartilage. Ears reconstructed by our technique have a natural appearance. CONCLUSION: Our technique can be used for cases in which treatment for another large area of the body surface needs to be performed first to save the patient's life.

17.
Neuro Oncol ; 23(6): 945-954, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33556172

ABSTRACT

BACKGROUND: High-grade meningiomas are aggressive tumors with high morbidity and mortality rates that frequently recur even after surgery and adjuvant radiotherapy. However, limited information is currently available on the biology of these tumors, and no alternative adjuvant treatment options exist. Although we previously demonstrated that high-grade meningioma cells were highly sensitive to gemcitabine in vitro and in vivo, the underlying molecular mechanisms remain unknown. METHODS: We examined the roles of hENT1 (human equilibrative nucleoside transporter 1) and dCK (deoxycytidine kinase) in the gemcitabine sensitivity and growth of meningioma cells in vitro. Tissue samples from meningiomas (26 WHO grade I and 21 WHO grade II/III meningiomas) were immunohistochemically analyzed for hENT1 and dCK as well as for Ki-67 as a marker of proliferative activity. RESULTS: hENT1 and dCK, which play critical roles in the intracellular transport and activation of gemcitabine, respectively, were responsible for the high gemcitabine sensitivity of high-grade meningioma cells and were strongly expressed in high-grade meningiomas. hENT1 expression was required for the proliferation and survival of high-grade meningioma cells and dCK expression. Furthermore, high hENT1 and dCK expression levels correlated with stronger tumor cell proliferative activity and shorter survival in meningioma patients. CONCLUSIONS: The present results suggest that hENT1 is a key molecular factor influencing the growth capacity and gemcitabine sensitivity of meningioma cells and also that hENT1, together with dCK, may be a viable prognostic marker for meningioma patients as well as a predictive marker of their responses to gemcitabine.


Subject(s)
Meningeal Neoplasms , Meningioma , Pancreatic Neoplasms , Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Deoxycytidine Kinase/metabolism , Deoxycytidine Kinase/therapeutic use , Equilibrative Nucleoside Transporter 1 , Humans , Meningeal Neoplasms/drug therapy , Meningioma/drug therapy , Gemcitabine
18.
Biomedicines ; 8(8)2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32764319

ABSTRACT

Osimertinib, which is a third-generation epidermal growth factor receptor tyrosine kinase inhibitor, is an important anticancer drug because of its high efficacy and excellent safety profile. However, resistance against osimertinib is inevitable; therefore, therapeutic strategies to overcome the resistance are needed. Doxazosin, a classic quinazoline-based alpha 1-adrenoceptor antagonist is used to treat hypertension and benign prostatic hyperplasia with a known safety profile. The anticancer effects of doxazosin have been examined in various types of malignancies from the viewpoint of drug repositioning or repurposing. However, it currently remains unclear whether doxazosin sensitizes cancer cells to osimertinib. Herein, we demonstrated that doxazosin induced autophagy and enhanced the anticancer effects of osimertinib on the cancer cells and cancer stem cells of non-small cell lung cancer, pancreatic cancer, and glioblastoma at a concentration at which the growth of non-tumor cells was not affected. The osimertinib-sensitizing effects of doxazosin were suppressed by 3-methyladenine, an inhibitor of autophagy, which suggested that the effects of doxazosin were mediated by autophagy. The present study provides evidence for the efficacy of doxazosin as a combination therapy with osimertinib to overcome resistance against osimertinib.

19.
Plast Reconstr Surg Glob Open ; 8(6): e2871, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32766039

ABSTRACT

BACKGROUND: Problems with poor circulation often occur when a large defect or a distant region, such as the apex of the nose, is covered with a paramedian forehead flap. Delay technique increases the safety of reconstruction procedures, but it has been used less frequently because a 2-stage surgery is necessary, and various other flaps and techniques have been developed. METHOD: We performed the delay technique of paramedian forehead flap at the same time as tumor resection. For the flap, a narrow pedicle of about 1-cm was prepared on the supratrochlear artery and vein, and the incision was extended toward the lateral side conforming to the defect morphology, and a paramedian forehead flap with a design consistent with the esthetic unit containing the defect was prepared. The region below the flap was dissected to create the flap bipedicle, and surgery was completed. RESULT: This procedure was used in 4 patients with malignant tumor of the external nose, and the flap survived perfectly in all patients. The postoperative esthetic outcome was also found to be good. CONCLUSIONS: This procedure does not increase the frequency of surgery, circulation in the flap is maintained, the flap pedicle on the supratrochlear artery can be made narrow, and flap thinning can be performed from the beginning. Coverage of an extensive defect is possible because a large flap can be excised, and satisfactory esthetic appearance can be obtained by matching with the esthetic unit. The delay technique for various flaps (not limited to forehead flap alone) should be considered an effective technique for the current treatment of malignant tumors.

20.
J Plast Reconstr Aesthet Surg ; 73(7): 1312-1317, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32205053

ABSTRACT

BACKGROUND: Reconstruction of the upper eyelid with the same eyelid tissue is desirable because of the ability to achieve eye opening/closing and corneal protection, and a lid switch flap is a useful method. For total defects, almost all of the tissues of the lower eyelid should be used; however, the reconstruction of the lower eyelid donor site has often been undervalued. Reconstruction with an insufficient amount of soft tissue often results in complications such as lagophthalmos and ectropion. Here, we report our method of management of total upper eyelid defects and secondary reconstruction of the lower eyelid donor site. METHOD: A lid switch flap is designed on the lower eyelid as the first operation. As important points, the height of the flap of the anterior lamina should be the same but the conjunctiva as the posterior lamina should be harvested up to the conjunctival fornix to obtain sufficient tissue. After switching the flap, the lower eyelid donor site is reconstructed with sufficient tissue: cheek mucosa, conchal cartilage, and a reverse superficial temporal artery flap as a three-layered structure. RESULTS: Three patients were treated using our method, and we achieved favorable results with a sufficient amount of soft tissue for the reconstruction of the lower eyelid. CONCLUSION: Reconstruction of the upper eyelid with sufficient tissue from the lower eyelid is important for eyelid function.


Subject(s)
Blepharoplasty/methods , Eyelids/surgery , Surgical Flaps , Temporal Arteries/surgery , Aged , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...