scholarly journals Antitumor activity of melinjo ( Gnetum gnemon L.) seed extract in human and murine tumor models in vitro and in a colon‐26 tumor‐bearing mouse model in vivo

2015 ◽  
Vol 4 (11) ◽  
pp. 1767-1780 ◽  
Author(s):  
Narayanan K. Narayanan ◽  
Kazuhiro Kunimasa ◽  
Yukio Yamori ◽  
Mari Mori ◽  
Hideki Mori ◽  
...  
2014 ◽  
Vol 2 (23) ◽  
pp. 3659-3666 ◽  
Author(s):  
Guoyong Xu ◽  
Jiafeng Wang ◽  
Tao Liu ◽  
Mahong Wang ◽  
Shuangsheng Zhou ◽  
...  

The results of TPEF imaging in a tumor-bearing mouse model demonstrated the potential of the obtained complex for in vivo tumor diagnosis.


2014 ◽  
Vol 7 (4) ◽  
pp. 1147-1150 ◽  
Author(s):  
MASAKI ONOYAMA ◽  
KAZUO AZUMA ◽  
TAKESHI TSUKA ◽  
TOMOHIRO IMAGAWA ◽  
TOMOHIRO OSAKI ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3835-3835 ◽  
Author(s):  
Michael Fitzgerald ◽  
Yueying Cao ◽  
Bret Bannerman ◽  
Zhi Li ◽  
Olga Tayber ◽  
...  

Abstract Abstract 3835 Poster Board III-771 Introduction The first generation proteasome inhibitor VELCADE® (bortezomib) is indicated for the treatment of patients with multiple myeloma (MM), a form of plasma cell malignancy (PCM). MLN9708 is our novel proteasome inhibitor that selectively and reversibly binds to, and potently inhibits the b5 site of the 20s proteasome in preclinical studies. We have recently demonstrated that MLN9708 significantly prolongs tumor-free survival of double transgenic iMycCa/Bcl-XL mice, a genetically-engineered mouse model of de novo PCM. Here we describe the in vivo evaluation of cell lines derived from double transgenic iMycCa/Bcl-XL mice and the antitumor activity of MLN9708 in a disseminated mouse model of iMycCa/Bcl-XL PCM. Materials MLN9708 immediately hydrolyzes to MLN2238, the biologically active form, upon exposure to aqueous solutions or plasma. MLN2238 was used for all preclinical studies described below. Double transgenic iMycCa/Bcl-XL mice develop de novo PCM, in which neoplastic plasma cell development is driven by the targeted expression of the oncoprotein Myc and anti-apoptotic Bcl-XL (J. Clin. Invest. 113:1763-1773, 2004). DP54 and DP42 are plasma cell tumor cell lines isolated from the bone marrow and lymph nodes, respectively, of syngeneic mice previously inoculated with iMycCa/Bcl-XL tumors (Cancer Res. 67:4069-4078, 2007). In vitro, DP54 and DP42 cells express both the Myc and Bcl-XL transgenes, various plasma cell and B-cell markers including CD38, CD138 and B220, and have gene expression profiles very similar to human MM. Methods Cell viability studies were performed to determine the antiproliferative effects of MLN2238 in DP54 and DP42 cells in vitro. To evaluate DP54 and DP42 cells in vivo, these cells were aseptically inoculated into the tail vein of NOD-SCID mice. Progressions of the resultant PCM were monitored and tumor burdens were evaluated by magnetic resonance imaging (MRI), ex vivo mCT imaging, and histopathology. Mouse plasma samples were collected at the end of the studies and levels of immunoglobulin were assessed. To establish a preclinical disseminated mouse model of iMycCa/Bcl-XL PCM, freshly dissociated DP54-Luc cells (constitutively expressing firefly luciferase under a mouse Ig-k promoter) were aseptically inoculated into the tail vein of NOD-SCID mice. Once tumor growth has been established, mice were randomized into treatment groups and then treated with vehicle, bortezomib (at 0.7mg/kg intravenously [IV] twice weekly [BIW]) or MLN2238 (at 11 mg/kg IV BIW) for 3 consecutive weeks. Tumor burden was measured by bioluminescent imaging. Results In vitro, both DP54 and DP42 cells were sensitive to MLN2238 treatment (LD50 values of 14 and 25 nM, respectively). In vivo, NOD-SCID mice rapidly succumbed to PCM after being inoculated with DP54 and DP42 cells (25 and 14 days post-inoculation, respectively), where the disease was accompanied by marked elevation of plasma immunoglobulins. MRI scans revealed the presence of multiple lesions and several abnormalities were found including: cranial deformation, bowel distortion, splenomegaly and renal edema. Tumor infiltrates, ranging from minor to extensive, were identified in multiple organ compartments (brain<kidney<liver<lymph nodes<spleen<bone marrow) by histopathological analysis. Ex vivo mCT imaging has also revealed signs of bone erosion in the cranial sagittal sutures. Dissemination of DP54-Luc cells after tail vein inoculations was detected by in vivo bioluminescent and confirmed by ex vivo imaging where luminescent tumor nodules were identified in the spleen, kidneys, liver, intestine, lymph nodes, spinal bone and cranium. To assess the antitumor activity of MLN2238, an efficacy study was performed using the DP54-Luc disseminated model. Tumor burden (bioluminescence), skeletal malformation (mCT) and overall survival after treatment with bortezomib and MLN2238 will be presented. Conclusion The DP54-Luc disseminated mouse model of double transgenic iMycCa/Bcl-XL PCM recapitulated several key features of human MM and provided real-time assessment of novel MM therapy preclinically. MLN9708 is currently in human clinical development for both hematological and solid tumor indications. Disclosures: Cao: Milllennium: Employment, Equity Ownership. Bannerman:Milllennium: Employment. Li:Milllennium: Employment. Bradley:Milllennium: Employment, Equity Ownership, Research Funding. Silverman:Milllennium: Employment. Janz:Milllennium: Research Funding. Van Ness:Milllennium: Research Funding. Kupperman:Milllennium: Employment. Manfredi:Milllennium: Employment. Lee:Milllennium: Employment, Equity Ownership.


2012 ◽  
Vol 72 (20) ◽  
pp. 5348-5362 ◽  
Author(s):  
Pedram Kharaziha ◽  
Hendrik De Raeve ◽  
Charlotte Fristedt ◽  
Qiao Li ◽  
Astrid Gruber ◽  
...  

2020 ◽  
Vol 15 (4) ◽  
pp. 341-359
Author(s):  
Moloud Kazemi ◽  
Jaber Emami ◽  
Farshid Hasanzadeh ◽  
Mohsen Minaiyan ◽  
Mina Mirian ◽  
...  

Background: The development of biocompatible tumor-targeting delivery systems for anticancer agents is essential for efficacious cancer chemotherapy. Nanoparticles, as drug delivery cargoes for cancer therapy, are rapidly improving to overcome the limitations of conventional chemotherapeutic agents. Heparin–modified nanoparticles are currently being considered as one of the favorable carriers for the delivery of chemotherapeutics to cancer tissues. Objective: This study was aimed at evaluating the in vitro and in vivo antitumor activity of a novel targeted, pH-sensitive, heparin-based polymeric micelle loaded with the poorly water-soluble anticancer drug, docetaxel (DTX). The micelles could overcome the limited water solubility, non-specific distribution, and insufficient drug concentration in tumor tissues. Methods: DTX-loaded folate targeted micelles were prepared and evaluated for physicochemical properties, drug release, in vitro cellular uptake and cytotoxicity in folate receptor-positive and folate receptor-negative cells. Furthermore, the antitumor activity of DTX-loaded micelles was evaluated in the tumor-bearing mice. Some related patents were also studied in this research. Results: The heparin-based targeted micelles exhibited higher in vitro cellular uptake and cytotoxicity against folate receptor over-expressed cells due to the specific receptor-mediated endocytosis. DTX-loaded micelles displayed greater antitumor activity, higher anti-angiogenesis effects, and lower systemic toxicity compared with free DTX in a tumor-induced mice model as confirmed by tumor growth monitoring, immunohistochemical evaluation, and body weight shift. DTX-loaded targeting micelles demonstrated no considerable toxicity on major organs of tumor-bearing mice compared with free DTX. Conclusion: Our results indicated that DTX-loaded multifunctional heparin-based micelles with desirable antitumor activity and low toxicity possess great potential as a targeted drug delivery system in the treatment of cancer.


Talanta ◽  
2017 ◽  
Vol 175 ◽  
pp. 421-426 ◽  
Author(s):  
Hong-Wen Liu ◽  
Xiao-Xiao Hu ◽  
Longmin Zhu ◽  
Ke Li ◽  
Qiming Rong ◽  
...  

2021 ◽  
Vol 8 ◽  
Author(s):  
Ramazan Rezaei ◽  
Kaveh Baghaei ◽  
Seyed Mahmoud Hashemi ◽  
Mohammad Reza Zali ◽  
Hossein Ghanbarian ◽  
...  

Exosomes have been introduced as a new alternative delivery system for the transmission of small molecules. Tumor-derived exosomes (TEXs) not only contain tumor-associated antigens to stimulate antitumor immune responses but also act as natural carriers of microRNAs. The aim of the current study was to evaluate the efficacy of miR-124-3p-enriched TEX (TEXomiR) as cell-free vaccine in the induction of antitumor immune responses in a mouse model of colorectal cancer. Briefly, the exosomes were isolated from cultured CT-26 cell line, and modified calcium chloride method was used to deliver miR-124-3p mimic into the exosomes. We used a CT-26-induced BALB/c mouse model of colorectal cancer and analyzed the effect of TEXomiR on survival, tumor size, spleen and tumor-infiltrated lymphocytes, and splenocyte proliferation. Furthermore, intra-tumor regulatory T cells, cytotoxic activity of the splenocytes, and cytokine secretion was also evaluated to describe the anti-tumor immune response. When the tumor size reached 100 mm3, the mice were injected with TEXomiR, TEX, and/or phosphate-buffered saline (PBS) subcutaneously three times with 3-day interval, and then tumor size was monitored every 2 days. The in vitro results indicated that TEXs could efficiently deliver functional miR-124-3p mimic. The in vivo evaluation in tumor-bearing mice showed that treatment with TEXomiR can elicit a stronger anti-tumor immune response than unloaded TEX and PBS. Significant tumor growth inhibition and increased median survival time was achieved in tumor-bearing mice treated with TEXomiR. A significant decrease in CD4/CD8 and Treg/CD8 ratio in tumor tissue was demonstrated. Moreover, increased cytotoxicity and proliferation of splenocytes in the TEXomiR group compared to the TEX and PBS groups were identified. Taken together, our data demonstrated that tumor-derived exosomes efficiently deliver miR-124-3p mimic, and TEXomiR promotes anti-tumor immune responses.


Nanomedicine ◽  
2021 ◽  
Vol 16 (29) ◽  
pp. 2587-2604
Author(s):  
Chaoqi Li ◽  
Xiangbo Gou ◽  
Hui Gao

Aim: The authors aimed to develop Dox@Rg1 nanoparticles with decreased cardiotoxicity to expand their application in cancer. Materials & methods: Dox@Rg1 nanoparticles were developed by encapsulating doxorubicin (Dox) in a self-assembled Rg1. The antitumor effect of the nanoparticles was estimated using 4T1 tumor-bearing mice and the protective effect on the heart was investigated in vitro and in vivo. Results: Different from Dox, the Dox@Rg1 nanoparticles induced increased cytotoxicity to tumor cells, which was decreased in cardiomyocytes by the inhibition of apoptosis. The study in vivo revealed that the Dox@Rg1 nanoparticles presented a perfect tumor-targeting ability and improved antitumor effects. Conclusion: Dox@Rg1 nanoparticles could enhance the antitumor effects and decrease the cardiotoxicity of Dox.


2020 ◽  
Author(s):  
Yaojun Yu ◽  
jing zhang ◽  
Leyi Ni ◽  
Yuesheng Zhu ◽  
Hejie Yu ◽  
...  

Abstract Background: The role of neoantigens in cancer immunotherapy is crucial. However, the effectiveness and safety of personalized neoantigen vaccines in colorectal cancer (CRC), especially in Chinese population, has not been well studied. This paper mainly explores the feasibility and effectiveness of personalized neoantigen vaccines in CRC treatment. Methods: Whole-exome sequencing and transcriptome sequencing were used to identify somatic mutations, RNA expression and human leukocyte antigen (HLA) alleles. Neoantigens were predicted, and the immunogenicity of neoantigen candidates was evaluated by ELISPOT in vitro. To verify the immunogenicity in vivo, neoantigen candidates from HLA-A0201+PW11 were used to immunized female 6-8-week-old HLA-A2.1/Kb-transgenic (Tg) mice. Neoantigen-reactive T cells (NRTs) were induced by immunogenic peptides from autologous HLA-A2.1/Kb to adoptive transfer transgenic mice, and C57BL/6nu/nu mice were used for in vivo antitumor response assays.Results: Compared to medium alone (no peptide) or the unrelated peptide VSV-NP43-69, the neoantigens TSHZ3-L523P, RARA-R83H, TP53-R248W, EYA2-V333I and NRAS-G12D from Patient 4 (PW4); HAVCR2-F39V, SEC11A-R11L, TASP1-P161L, RAP1GAP-S215R, MOSPD1-V63I and NAV2-D1973N from Patient 10 (PW10); and SMPDL3B-T452M, LRFN3-R118Q and ULK1-S248L from Patient 11 (PW11) induced notable peptide-specific T cell responses. The results indicated that about half of the predicted neoantigens for all 3 patients can stimulate T cell responses and antitumor effects in CRC. In addition, predicted neoantigens from PW11 (HLA-A0201) showed promising antitumor efficacy in HLA-A2.1/Kb-Tg mice and tumor-bearing mouse models.Conclusion: With the application of next-generation sequencing (NGS) sequencing of patient specimens, neoantigen prediction and a rapid immunoassay system, an evaluation system utilizing in vitro studies and in vivo transgenic and tumor-bearing mouse models can be used to screen strong immunogenic neoantigens in CRC patients. Accurate identification of neoantigens with strong immunogenicity would promote personalized cancer vaccine development.


Sign in / Sign up

Export Citation Format

Share Document