scholarly journals Anti-tumor efficacy of CKD-516 in combination with radiation in xenograft lung cancer mouse model

2019 ◽  
Author(s):  
Min-Young Kim ◽  
Jung-Young Shin ◽  
Jeong-Oh Kim ◽  
Kyoung-Hwa Son ◽  
Chan-Kwon Jung ◽  
...  

Abstract Background: To evaluate the anti-tumor efficacy of CKD-516 in combined with irradiation (IR) and examine tumor necrosis, delayed tumor growth, and expression of molecules involved in hypoxia and angiogenesis. Methods : A xenograft mouse model of lung cancer was established. The tumor was exposed to irradiation (IR) for 5 days per week. CKD-516 was administered with ​​two treatment schedules (day 1 or days 1 and 5) at one hour after IR. After the administration, tumor tissues were stained with hematoxylin and eosin and pimonidazole. HIF1 , Glut-1, VEGF, CD31 and Ki-67 expression were evaluated by Immunohistochemical staining. Results: With short-term administration, IR and CKD-516+IR (d1) significantly reduced tumor size ( p = 0.0062 and p = 0.0051, respectively). CKD-516+IR groups were remarkably reduced blood vessels ( p < 0.005). In particular, CKD-516+IR (d1) resulted in the most extensive tumor necrosis, which was significantly increased with large hypoxic area ( p = 0.02) and decreased HIF1 , Glut-1, VEGF, and Ki-67 expressions. Long-term administration of CKD-516+IR reduced tumor size and delayed tumor growth. This combination also greatly reduced the number of blood vessels ( p = 0.0006) and significantly enhanced tumor necrosis ( p = 0.004). CKD-516+IR notably increased HIF1 expression ( p = 0.0047), but significantly diminished VEGF expression ( p = 0.0046). Conclusion: Taken together, our results demonstrate that CKD-516 in combination with IR can significantly enhance the anti-tumor efficacy compared to CKD-516 or IR alone in lung cancer xenograft mice. Keywords: Irradiation, Tumor necrosis, Hypoxia, Squamous cell carcinoma of lung, Xenograft mice

2020 ◽  
Author(s):  
Min-Young Kim ◽  
Jung-Young Shin ◽  
Jeong-Oh Kim ◽  
Kyoung-Hwa Son ◽  
Yeon Sil Kim ◽  
...  

Abstract Background: To evaluate the anti-tumor efficacy of CKD-516 in combined with irradiation (IR) and examine tumor necrosis, delayed tumor growth, and expression of molecules involved in hypoxia and angiogenesis. Methods : A xenograft mouse model of lung cancer was established. The tumor was exposed to irradiation (IR) for 5 days per week. CKD-516 was administered with ​​two treatment schedules (day 1 or days 1 and 5) at one hour after IR. After the administration, tumor tissues were stained with hematoxylin and eosin and pimonidazole. HIF1 , Glut-1, VEGF, CD31 and Ki-67 expression were evaluated by Immunohistochemical staining. Results: With short-term administration, IR and CKD-516+IR (d1) significantly reduced tumor size ( p = 0.0062 and p = 0.0051, respectively). CKD-516+IR groups were remarkably reduced blood vessels ( p < 0.005). In particular, CKD-516+IR (d1) resulted in the most extensive tumor necrosis, which was significantly increased with large hypoxic area ( p = 0.02) and decreased HIF1 , Glut-1, VEGF, and Ki-67 expressions. Long-term administration of CKD-516+IR reduced tumor size and delayed tumor growth. This combination also greatly reduced the number of blood vessels ( p = 0.0006) and significantly enhanced tumor necrosis ( p = 0.004). CKD-516+IR notably increased HIF1 expression ( p = 0.0047), but significantly diminished VEGF expression ( p = 0.0046). Conclusions : Taken together, our results demonstrate that CKD-516 in combination with IR can significantly enhance the anti-tumor efficacy compared to CKD-516 or IR alone in lung cancer xenograft mice.


2020 ◽  
Author(s):  
Min-Young Kim ◽  
Jung-Young Shin ◽  
Jeong-Oh Kim ◽  
Kyoung-Hwa Son ◽  
Yeon Sil Kim ◽  
...  

Abstract Background: Hypoxic tumors are known to be highly resistant to radiotherapy and cause poor prognosis in non-small-cell lung cancer (NSCLC) patients. Vascular disrupting agents (VDAs) mainly affect blood vessels located in the central area of the tumor and lead to a rapid decrease in blood, resulting in massive apoptotic tumor cell death. Thus, we evaluate the anti-tumor efficacy of CKD-516 in combination with irradiation (IR) and examine tumor necrosis, delayed tumor growth, and expression of proteins involved in hypoxia and angiogenesis in this study.Methods: A xenograft mouse model of lung squamous cell carcinoma was established, and the tumor was exposed to IR 5 days per week. CKD-516 was administered with ​​two treatment schedules (day 1 or days 1 and 5) 1 h after IR. After the treatment, tumor tissues were stained with hematoxylin and eosin and pimonidazole. HIF-1α, Glut-1, VEGF, CD31, and Ki-67 expression levels were evaluated by immunohistochemical staining.Results: Short-term treatment with IR alone and CKD-516+IR (d1) significantly reduced tumor volume (p = 0.006 and p = 0.048, respectively). Treatment with CKD-516+IR (d1 and d1, 5) resulted in a marked reduction in the number of blood vessels (p < 0.005). More specifically, CKD-516+IR (d1) caused the most extensive tumor necrosis, which resulted in a significantly large hypoxic area (p = 0.02) and decreased HIF-1α, Glut-1, VEGF, and Ki-67 expression. Long-term administration of CKD-516+IR reduced tumor volume and delayed tumor growth. This combination also greatly reduced the number of blood vessels (p = 0.0006) and significantly enhanced tumor necrosis (p = 0.004). CKD-516+IR significantly increased HIF-1α expression (p = 0.0047), but significantly reduced VEGF expression (p = 0.0046).Conclusions: Taken together, our data show that when used in combination, CKD-516 and IR can significantly enhance anti-tumor efficacy compared to monotherapy in lung cancer xenograft mice.


2020 ◽  
Author(s):  
Min-Young Kim ◽  
Jung-Young Shin ◽  
Jeong-Oh Kim ◽  
Kyoung-Hwa Son ◽  
Yeon Sil Kim ◽  
...  

Abstract Background: Hypoxic tumors are known to be highly resistant to radiotherapy and cause poor prognosis in non-small cell lung cancer (NSCLC) patients. CKD-516, a novel vascular disrupting agent (VDA), mainly affects blood vessels in the central area of the tumor and blocks tubulin polymerization, thereby destroying the aberrant tumor vasculature with a rapid decrease in blood, resulting in massive apoptotic tumor cell death. Therefore, we evaluated the anti-tumor efficacy of CKD-516 in combination with irradiation (IR) and examined tumor necrosis, delayed tumor growth, and expression of proteins involved in hypoxia and angiogenesis in this study.Methods: A xenograft mouse model of lung squamous cell carcinoma was established, and the tumor was exposed to IR 5 days per week. CKD-516 was administered with two treatment schedules (day 1 or days 1 and 5) 1 h after IR. After treatment, tumor tissues were stained with hematoxylin and eosin, and pimonidazole. HIF-1α, Glut-1, VEGF, CD31, and Ki-67 expression levels were evaluated using immunohistochemical staining.Results: Short-term treatment with IR alone and CKD-516+IR (d1) significantly reduced tumor volume (p = 0.006 and p = 0.048, respectively). Treatment with CKD-516+IR (d1 and d1, 5) resulted in a marked reduction in the number of blood vessels (p < 0.005). More specifically, CKD-516+IR (d1) caused the most extensive tumor necrosis, which resulted in a significantly large hypoxic area (p = 0.02) and decreased HIF-1α, Glut-1, VEGF, and Ki-67 expression. Long-term administration of CKD-516+IR reduced tumor volume and delayed tumor growth. This combination also greatly reduced the number of blood vessels (p = 0.0006) and significantly enhanced tumor necrosis (p = 0.004). CKD-516+IR significantly increased HIF-1α expression (p = 0.0047), but significantly reduced VEGF expression (p = 0.0046).Conclusions: Taken together, our data show that when used in combination, CKD-516 and IR can significantly enhance anti-tumor efficacy compared to monotherapy in lung cancer xenograft mice.


BMC Cancer ◽  
2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Min-Young Kim ◽  
Jung-Young Shin ◽  
Jeong-Oh Kim ◽  
Kyoung-Hwa Son ◽  
Yeon Sil Kim ◽  
...  

Abstract Background Hypoxic tumors are known to be highly resistant to radiotherapy and cause poor prognosis in non-small cell lung cancer (NSCLC) patients. CKD-516, a novel vascular disrupting agent (VDA), mainly affects blood vessels in the central area of the tumor and blocks tubulin polymerization, thereby destroying the aberrant tumor vasculature with a rapid decrease in blood, resulting in rapid tumor cell death. Therefore, we evaluated the anti-tumor efficacy of CKD-516 in combination with irradiation (IR) and examined tumor necrosis, delayed tumor growth, and expression of proteins involved in hypoxia and angiogenesis in this study. Methods A xenograft mouse model of lung squamous cell carcinoma was established, and the tumor was exposed to IR 5 days per week. CKD-516 was administered with two treatment schedules (day 1 or days 1 and 5) 1 h after IR. After treatment, tumor tissues were stained with hematoxylin and eosin, and pimonidazole. HIF-1α, Glut-1, VEGF, CD31, and Ki-67 expression levels were evaluated using immunohistochemical staining. Results Short-term treatment with IR alone and CKD-516 + IR (d1) significantly reduced tumor volume (p = 0.006 and p = 0.048, respectively). Treatment with CKD-516 + IR (d1 and d1, 5) resulted in a marked reduction in the number of blood vessels (p < 0.005). More specifically, CKD-516 + IR (d1) caused the most extensive tumor necrosis, which resulted in a significantly large hypoxic area (p = 0.02) and decreased HIF-1α, Glut-1, VEGF, and Ki-67 expression. Long-term administration of CKD-516 + IR reduced tumor volume and delayed tumor growth. This combination also greatly reduced the number of blood vessels (p = 0.0006) and significantly enhanced tumor necrosis (p = 0.004). CKD-516 + IR significantly increased HIF-1α expression (p = 0.0047), but significantly reduced VEGF expression (p = 0.0046). Conclusions Taken together, our data show that when used in combination, CKD-516 and IR can significantly enhance anti-tumor efficacy compared to monotherapy in lung cancer xenograft mice.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3276-3276
Author(s):  
Yen T. M. Nguyen ◽  
Manabu Fujisawa ◽  
Tran B. Nguyen ◽  
Yasuhito Suehara ◽  
Tatsuhiro Sakamoto ◽  
...  

Abstract Introductions: Loss-of-function TET2 mutations are frequent in clonal hematopoiesis in patients with solid cancers as well as that in healthy individuals. It remains to be elucidated whether and how TET2-mutated immune cells affect cancer progression in patients with TET2-mutated clonal hematopoiesis. Here, we assessed activity of Tet2-deficient immune cells using a mouse lung cancer model. Methods: Lewis Lung Carcinoma (LLC) cells were subcutaneously transplanted into blood-specific Mx-Cre or myeloid-specific LysM-Cre x Tet2 f/f mice (Tet2 -/- or Tet2 mye-) or control mice (CT). Single-cell RNA sequencing (scRNA-seq) was performed to determine the immune-cell profiles and mediators in tumors of Tet2 -/- mice (Tet2 -/- tumors). Whole transcriptome analysis (WTA) was also performed for granulocytic myeloid-derived cells (GMD), monocytic myeloid-derived cells (MMD), and tumor associated macrophages (TAM), as well as LLC cells sorted from Tet2 -/- tumors and CT tumors. Results: We found that tumor growth was enhanced in both Tet2 -/- and Tet2 mye- comparing to CT. Unsupervised clustering of scRNA-seq data identified 14 cell clusters: GMD into 3 (GMD1, GMD2, and GMD3), MMD into 5 (MMD1, MMD2, MMD3, MMD4, and MMD5), TAMs into 4 (TAM1, TAM2, TAM3, and TAM4), and DCs into 2 (DC1 and DC2). Notably, among all subclusters, the proportions of GMD1, GMD3, TAM3 and TAM4 were markedly expanded in Tet2 -/- tumors comparing to CT. Differentially expressed gene (DEG) analysis of scRNA-seq data found that S100a8 and S100a9 were highly expressed in Tet2-deficient GMD1 compared to CT. Furthermore, S100a8/S100a9 proteins were elevated in plasmas of Tet2 -/- comparing to those of CT. Pathway analysis using DEGs (p &lt; 0.05) from WTA of GMD determined interleukin 1b (Il1b) signaling as upstream of S100a8/S100a9 activity. Gene set enrichment analysis (GSEA) also showed that 6 pathways related to Il1b were enriched in Tet2-deficient group compared to CT group. Gene ontology analysis (GO) for DEGs of GMD, MMD, and TAMs by WTA as well as 13 subclusters by scRNA-seq revealed that the "cellular response to IL-1" pathway was enriched in Tet2-deficient group compared to CT group. To define the downstream effectors in LLC cells, we performed WTA for LLC cells sorted from Tet2 -/- and CT tumors. We found that Vegfa, encoding a mediator for angiogenesis was highly upregulated in LLC cells sorted from Tet2 -/- tumors comparing to CT tumors. GSEA for WTA further identified that multiple Vegfa-related pathways as well as MAPK cascade were enriched in LLC cells from Tet2 -/- tumors comparing to those from CT tumors . Furthermore, S100a8/S100a9 induced Vegfa secretion from LLC cells in vitro. Remarkably, the area of blood vessels was increased in Tet2 -/- tumors comparing to CT tumors. Immunostaining exhibited that the number of Ly6g +GMD foci (&gt;1000 px 2) expressing S100a8/S100a9 was increased in Tet2 -/- tumors comparing to CT tumors. Furthermore, LLC cells surrounding GMD foci highly expressed Vegfa in Tet2 -/- tumors. Finally, administration of an antibody against Emmprin, a receptor for S100a8/S100a9 inhibited the tumor growth in Tet2 -/-. Notably, the area of blood vessels in Tet2 -/- tumors with anti-Emmprin group was decreased at 2-fold compared to that seen in isotype group (p &lt; 0.05). Consistently, S100A8/S100A9 induced VEGFA production in human lung cancer cells in vitro. Conclusions: Tet2-deificient immune cells promote lung cancer progression through S100a8/S100a9-Emmprin-Vegfa axis. Our study suggests a novel role of TET2-mutated clonal hematopoiesis in cancer progression and even provides a novel therapeutic target. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Author(s):  
Juan Yang ◽  
Zhouxue Wu ◽  
Yao Chen ◽  
Chuanfei Hu ◽  
Dong Li ◽  
...  

Abstract Background:The hypoxic microenvironment of solid tumors reduces the susceptibility of cancer cells to radiotherapy. Current treatments are focused on the development of anti-cancer agents that selectively target tumor cells with no toxicity to healthy tissue. Bacteria colonize and destroy tumors and have emerged as biological vectors that can survive in the tumor microenvironment. Methods:In this study, a Lewis lung carcinoma transplant mouse model was established and treated with a combination of bifidobacterium infantis (Bi), its specific monoclonal antibodies (Ab) and radiotherapy (RT). 18F-FDG PET/CT and 18F-FMISO PET/CT imaging were performed to monitor tumor growth and hypoxia in the tumor tissue. Phosphorylated histone (γ-H2AX), the proliferation index (Ki-67), platelet endothelial cell adhesion molecules (CD31), tumor necrosis factor-α (TNF-α), hypoxia inducible factor-1α (HIF-1α) and glucose transporter 1 (Glut-1) levels were assessed through immunohistochemistry. Results:The results showed that the combined treatment group (Ab+ Bi+ RT) displayed delayed tumor growth and prolonged the survival of mice. The combined treatment group also had lower levels of HIF-1α, Glut-1, and CD31 expression, and a lower uptake of FDG and FMISO. The tumors treated with the combination therapy also had lower levels of hypoxia, and increased γ-H2AX and TNF-α expression. Conclusion:Taken together, these data suggest that the combination of bifidobacterium infantis and its specific monoclonal antibodies can markedly improve the efficacy of radiotherapy for the treatment of lung cancer.


2019 ◽  
Vol 26 (6) ◽  
pp. 565-574 ◽  
Author(s):  
S Latteyer ◽  
S Christoph ◽  
S Theurer ◽  
G S Hönes ◽  
K W Schmid ◽  
...  

Thyroid hormones are important for physiology and homeostasis. In addition to nuclear thyroid hormone receptors, the plasma membrane protein integrin αvβ3 has been recognized as a receptor for both thyroxine (T4) and triiodothyronine (T3). Here, we studied whether thyroid hormone promotes growth of murine lung cancer via αvβ3 in vivo. Murine Lewis lung carcinoma cells (3LL), stably transfected with luciferase, were injected into mouse lungs. Tumor growth in untreated mice was compared to hypothyroid mice and hypothyroid mice treated with T3 or T4 with or without the αvβ3 inhibitor 3,5,3′,5′-tetraiodothyroacetic acid (Tetrac). Tumor progression was determined by serial in vivo imaging of bioluminescence emitted from the tumor. Tumor weight was recorded at the end of the experiment. Neoangiogenesis was determined by immunohistochemistry for CD31. Tumor growth was reduced in hypothyroidism and increased by T4 treatment. Strikingly, only T4 but not T3 treatment promoted tumor growth. This T4 effect was abrogated by the αvβ3 inhibitor Tetrac. Tumor weight and neoangiogenesis were also significantly increased only in T4-treated mice. The T4 effect on tumor weight and neoangiogenesis was abolished by Tetrac. In vitro, T4 did not stimulate 3LL cell proliferation or signaling pathway activation. We conclude that T4 promotes lung cancer growth in this orthotopic mouse model. The tumor-promoting effect is mediated via the plasma membrane integrin αvβ3 and increased neoangiogenesis rather than direct stimulation of 3LL cells. These data suggest that such effects of levothyroxine may need to be considered in cancer patients on T4 substitution.


2013 ◽  
Author(s):  
Hiroshi Kitagawa ◽  
Matthew Dallos ◽  
Brendan Quinn ◽  
Ajaikumar Kunnumakkara ◽  
Irem Dogan ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document