orthotopic model
Recently Published Documents


TOTAL DOCUMENTS

391
(FIVE YEARS 88)

H-INDEX

39
(FIVE YEARS 4)

Biomedicines ◽  
2021 ◽  
Vol 9 (12) ◽  
pp. 1873
Author(s):  
Chiara Tobia ◽  
Daniela Coltrini ◽  
Roberto Ronca ◽  
Alessandra Loda ◽  
Jessica Guerra ◽  
...  

Uveal melanoma is a highly metastatic tumor, representing the most common primary intraocular malignancy in adults. Tumor cell xenografts in zebrafish embryos may provide the opportunity to study in vivo different aspects of the neoplastic disease and its response to therapy. Here, we established an orthotopic model of uveal melanoma in zebrafish by injecting highly metastatic murine B16-BL6 and B16-LS9 melanoma cells, human A375M melanoma cells, and human 92.1 uveal melanoma cells into the eye of zebrafish embryos in the proximity of the developing choroidal vasculature. Immunohistochemical and immunofluorescence analyses showed that melanoma cells proliferate during the first four days after injection and move towards the eye surface. Moreover, bioluminescence analysis of luciferase-expressing human 92.1 uveal melanoma cells allowed the quantitative assessment of the antitumor activity exerted by the canonical chemotherapeutic drugs paclitaxel, panobinostat, and everolimus after their injection into the grafted eye. Altogether, our data demonstrate that the zebrafish embryo eye is a permissive environment for the growth of invasive cutaneous and uveal melanoma cells. In addition, we have established a new luciferase-based in vivo orthotopic model that allows the quantification of human uveal melanoma cells engrafted in the zebrafish embryo eye, and which may represent a suitable tool for the screening of novel drug candidates for uveal melanoma therapy.


Cancers ◽  
2021 ◽  
Vol 13 (22) ◽  
pp. 5775
Author(s):  
Hae Hyun Hwang ◽  
Hee Jeong Jeong ◽  
Sangwu Yun ◽  
Youngro Byun ◽  
Teruo Okano ◽  
...  

Pancreatic cancers are classified based on where they occur, and are grouped into those derived from exocrine and those derived from neuroendocrine tumors, thereby experiencing different anticancer effects under medication. Therefore, it is necessary to develop anticancer drugs that can inhibit both types. To this end, we developed a heparin–taurocholate conjugate, i.e., LHT, to suppress tumor growth via its antiangiogenic activity. Here, we conducted a study to determine the anticancer efficacy of LHT on pancreatic ductal adenocarcinoma (PDAC) and pancreatic neuroendocrine tumor (PNET), in an orthotopic animal model. LHT reduced not only proliferation of cancer cells, but also attenuated the production of VEGF through ERK dephosphorylation. LHT effectively reduced the migration, invasion and tube formation of endothelial cells via dephosphorylation of VEGFR, ERK1/2, and FAK protein. Especially, these effects of LHT were much stronger on PNET (RINm cells) than PDAC (PANC1 and MIA PaCa-2 cells). Eventually, LHT reduced ~50% of the tumor weights and tumor volumes of all three cancer cells in the orthotopic model, via antiproliferation of cancer cells and antiangiogenesis of endothelial cells. Interestingly, LHT had a more dominant effect in the PNET-induced tumor model than in PDAC in vivo. Collectively, these findings demonstrated that LHT could be a potential antipancreatic cancer medication, regardless of pancreatic cancer types.


2021 ◽  
Vol 12 ◽  
Author(s):  
Garcia-Lazaro Rocio Soledad ◽  
Caligiuri Lorena Gisel ◽  
Lorenzo Norailys ◽  
Lamdan Humberto ◽  
Alonso Daniel Fernando ◽  
...  

Breast cancer (BC) is the most frequent cancer in women and tumor metastasis is a major cause of cancer-related deaths. Our aim was to evaluate anti-metastatic properties of yerba mate extract (YMe) in BC models. 4T1, F3II, MCF-7, and MDA-MB231 cell lines were used to perform in vitro assays. The F3II syngeneic mammary carcinoma model in BALB/c mice was used to evaluate tumor progression, BC metastasis and survival. Cells were inoculated subcutaneously into the flank for the heterotopic model and into the mammary fat pad for the orthotopic model. YMe was administered p.o. in a dose of 1.6 g/kg/day. In vitro YMe inhibited cell proliferation and reduced tumor cell adhesion, migration and invasion. These biological effects were cell-line dependent. In vivo YMe reduced tumor metastasis and increased mice survival in both models. Our preclinical results suggest that YMe could modulate tumor progression and metastasis in BC models.


2021 ◽  
Author(s):  
Yunzhi Dang ◽  
Jiao Yu ◽  
Shuhong Zhao ◽  
Ximing Cao ◽  
Qing Wang

Abstract Background: KRAS mutation accounts for 30-50% of human colorectal cancer (CRC). Due to paucity of effective treatment options, KRAS mutant CRC is difficult to treat in clinic. Metastasis is still the major reason for the high mortality of KRAS mutant CRC, but the exact mechanism remains unclear. Here, we report a novel role of Homeobox 7 (HOXA7) in promoting KRAS mutant CRC metastasis and probed therapy strategies for these subpopulation patients.Methods: The expression of HOXA7 was detected in human CRC cohort by immunohistochemistry. The function of HOXA7 in KRAS mutant CRC metastasis was analyzed by cecum orthotopic model. Results: The elevated expression of HOXA7 was positively correlated with lymph node metastasis, distant metastasis, poorer tumor differentiation, higher TNM stage, and poor prognosis in CRC patients. Furthermore, HOXA7 is an independent prognostic marker of KRAS mutant CRC patients (P<0.001), while not for KRAS wild-type CRC patients (P=0.575). HOXA7 overexpression increased the metastasis ability of KRAS mutant CT26 cell, and promoted the infiltration of MDSCs at the same time. When MDSCs infiltration was depleted by CXCR2 inhibitor, it can markedly suppress the metastasis rate in CT26 cell. The combination of CXCR2 inhibitor SB265610 and anti-programmed death-ligand 1 (anti-PD-L1) can largely inhibit metastasis in KRAS mutant CRC. Conclusions: HOXA7 overexpression upregulated CXCL1, which promoted MDSCs infiltration. Interruption of this loop might provide a promising treatment strategy for HOXA7-mediated KRAS mutant CRC metastasis.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi23-vi23
Author(s):  
Miranda Tallman ◽  
Abby Zalenski ◽  
Amanda Deighen ◽  
Treg Grubb ◽  
Morgan Schrock ◽  
...  

Abstract Glioblastoma (GBM) is a fatal and incurable brain tumor, with an average life expectancy after diagnosis of only 12-15 months. A main reason for the lethality of GBM is inevitable recurrence, caused by a small population of the tumor cells, called cancer stem cells (CSCs). These cells are aggressive, infiltrative, and resistant to current GBM treatments of chemotherapy and radiotherapy. We use a small molecule drug, CBL0137, which inhibits the FACT (facilitates chromatin transcription) complex leading to cancer cell specific cytotoxicity. Here, we show that CBL0137 sensitized GBM CSCs to radiotherapy and hence lead to increased CSC death and prolonged survival in preclinical models. Clonogenic assays were used to show that CSCs were radiosensitized after CBL0137 treatment. We saw increased DNA damage when GBM CSCs were treated with CBL0137, as well as a decrease in foci resolution over time, when CBL0137 was combined with irradiation. In order to elucidate if the increase in DNA damage was directly due to the inhibition of the FACT complex, we depleted the level of FACT in our GBM CSCs. FACT depletion also led to increased DNA damage, and even more so when combined with irradiation. To validate whether combination therapy sensitized CSCs to radiotherapy in vivo, we used a subcutaneous mouse model and showed combination treatment decreased CSCs frequency in these tumors as well as decreased tumor volume. With an orthotopic model of GBM, we showed that CBL0137 treatment followed by radiotherapy significantly increased survival of mice bearing tumors over either treatment alone. Together, this work establishes a new treatment paradigm for GBM, which sensitizes radio-resistant GBM CSCs to irradiation, a critical component of patient care. Radio-sensitizing agents, including CBL0137, pose an exciting new therapeutic capable of increasing the efficacy of irradiation, by inclusively targeting CSCs.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi81-vi81
Author(s):  
Dimpy Koul ◽  
Veerakumar Balasubramaniyan ◽  
Xiaolong Li ◽  
Sabbir Khan ◽  
Davide Guggi ◽  
...  

Abstract Glioblastoma (GBM) remains an incurable tumor with median overall survival of 15 months despite radiation and alkylating temozolomide (TMZ) chemotherapy. DNA damage response (DDR) pathways are among the most important key players of oncogenic mutations associated with resistance to both chemotherapy and radiation in GBM. The high frequency of alterations in DDR pathways in GBM suggests that its inhibition by DDR inhibitors may render GBM cells more susceptible to DNA damaging interventions. Here, we report the preclinical in vitro and in vivo activity of a novel, orally bioavailable Ataxia-telangiectasia mutated serine/threonine protein kinase and Rad3-related (ATR) inhibitor LR02 (Laevoroc Oncology) in a panel of 15 well-characterized glioma stem-like cells (GSCs). Effects on cell proliferation, survival and tumor formation were analyzed following treatment with LR02. Growth inhibition was time- and dose-dependent with a 3-day exposure resulting in a growth inhibitory IC50 (gIC50) in the low nM range in all the glioblastoma cell lines tested. LR02 inhibited growth of GSCs at IC50 values ranging from 500nmol/L to-~2umol/L. Additional studies showed that temozolomide sensitized GSC to LR02. Importantly, we demonstrate that MGMT promotor methylation status was associated with cellular response to LR02 treatment with preferential inhibition of cell growth in MGMT promotor methylated (MGMT deficient) cell lines. LR02 showed efficacy and survival benefit in a GSC262 (MGMT methylated) orthotopic model of GBM. Further administration of LR02 further enhanced the in vivo antitumor efficacy of temozolomide (TMZ) against GBM using the GSC262 model demonstrating that ATR inhibitor LR02 may enhance alkylating agent-mediated cytotoxicity and provide a novel treatment combination for GBM patients. Our present findings establish that the ATR inhibitor LR02 can specifically be used in tumors with MGMT deficiency when combined with alkylating chemotherapy. Further studies are ongoing to evaluate the potential of LR02 to overcome radiation and chemotherapy resistance in glioblastoma.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi181-vi181
Author(s):  
Rafal Zielinski ◽  
Roberto Cardenas-Zuniga ◽  
Maria Poimenidou ◽  
Magdalena Remiszewski ◽  
Edd Felix ◽  
...  

Abstract As part of our drug discovery program, we have developed structure-based modular designs of unique DNA-binding agents. The approach combines DNA intercalating and DNA “minor-groove-binding” modules. We have discovered compound WP1244 that potentially binds up to 10 bp long sequences of DNA. The unique and intriguing feature of WP1244 is its high CNS uptake combined with the picomolar to low nanomolar cytotoxicity against ependymoma and glioblastoma multiforme (GBM) cell lines and demonstrated in vivo activity in the orthotopic model of GBM. To improve water solubility and develop an IV formulation, we have synthesized WP1874, a mesylate salt of WP1244, and initiated its preclinical characterization. WP1874, similarly to its parental compound, shows high cytotoxicity in ependymoma, GBM, and medulloblastoma cell lines with IC50 in low nanomolar range and it was up to 100 to 200 times more potent than doxorubicin. Interestingly, WP1874 does not appear to be cytotoxic against normal kidney cells (VeroC1008) with IC50 &gt; 10 μM. Preliminary pharmacokinetic and biodistribution studies performed in CD-1 mice with intact brains revealed enhanced penetration of WP1874 to the brain with Cmax 1.5-fold greater than in plasma. Respectively, WP1874 Cmax in the brain was 2.3 ug/g (~2.0 μM) vs. 1.5 μg/ml (1.3 μM) in plasma. Acute toxicity in intravenously administered WP1874 was LD50 &gt;15mg/kg. No mortalities or any apparent toxicity symptoms were recorded for six intravenous weekly doses of WP1874 at 2.5 or 5 mg/kg in CD-1, Balb/c, or nude athymic mice. Intraperitoneal administration was well-tolerated up to 5 mg/kg given three times a week for four cycles. High CNS uptake, excellent cytotoxicity against different brain cancer cell lines, and low toxicity in vivo and in vitro against normal cells warrant further investigation of WP1874 as a mechanically unique potential anticancer agent against CNS malignancies.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi22-vi22
Author(s):  
Jack Korleski ◽  
Sweta Shudir ◽  
Christopher Caputo ◽  
Bachchu Lal ◽  
Yuan Rui ◽  
...  

Abstract Multi-potent stem-like cells (i.e. cancer stem cells, CSCs) are critical determinants of tumor propagation, therapeutic resistance, and recurrence in glioblastoma (GBM). Modifications in chromatin architecture play a fundamental role in the tumor cell phenotype of GBM. The polycomb repressor complex 2 (PRC2) is a key histone modifier that supports multi-potency and oncogenesis via H3K27 trimethylation (H3K27me3). Understanding how these epigenetic modifications cooperatively drive cancer cell stemness should unveil new targets for therapeutic development in GBM. Using a combination of next-generation sequencing, bioinformatics, and molecular approaches we identified EZH2, the catalytic domain of the PRC2 complex, as a critical mediator of reprograming events in GBM cells. We found that EZH2 is highly induced in response to transgenic Oct4/Sox2 with global increases in H3K27me3. Pharmacological inhibition of EZH2 diminishes self-renewal capacity of GBM neurospheres concurrent with a reduction in gene expression levels of markers and drivers of stemness. Furthermore, we identified and validated a set of 6 putative tumor suppressor genes repressed by Oct4 and Sox2 in a PRC2-dependent manner. We identified miR-217 as an EZH2 regulator in GBM cells and miR-217 reconstitution using advanced nanoparticle formulations re-activates the PRC2-repressed tumor suppressors, inhibited tumor growth and enhanced the effects of ionizing radiation in an orthotopic model of GBM. Taken together, these data show that PRC2-mediated chromatin changes in H3K27me3 help regulate the stem-cell phenotype induced by Oct4 and Sox2 in GBM cells and predict that targeting EZH2 could have therapeutic benefit in GBM.


Biomedicines ◽  
2021 ◽  
Vol 9 (11) ◽  
pp. 1592
Author(s):  
Natalia Yudintceva ◽  
Ekaterina Lomert ◽  
Natalia Mikhailova ◽  
Elena Tolkunova ◽  
Nikol Agadzhanian ◽  
...  

Despite multimodal approaches for the treatment of multiforme glioblastoma (GBM) advances in outcome have been very modest indicating the necessity of novel diagnostic and therapeutic strategies. Currently, mesenchymal stem cells (MSCs) represent a promising platform for cell-based cancer therapies because of their tumor-tropism, low immunogenicity, easy accessibility, isolation procedure, and culturing. In the present study, we assessed the tumor-tropism and biodistribution of the superparamagnetic iron oxide nanoparticle (SPION)-labeled MSCs in the orthotopic model of C6 glioblastoma in Wistar rats. As shown in in vitro studies employing confocal microscopy, high-content quantitative image cytometer, and xCelligence system MSCs exhibit a high migratory capacity towards C6 glioblastoma cells. Intravenous administration of SPION-labeled MSCs in vivo resulted in intratumoral accumulation of the tagged cells in the tumor tissues that in turn significantly enhanced the contrast of the tumor when high-field magnetic resonance imaging was performed. Subsequent biodistribution studies employing highly sensitive nonlinear magnetic response measurements (NLR-M2) supported by histological analysis confirm the retention of MSCs in the glioblastoma. In conclusion, MSCs due to their tumor-tropism could be employed as a drug-delivery platform for future theranostic approaches.


Sign in / Sign up

Export Citation Format

Share Document