Polysaccharide from Eucheuma Gelatinae Enhance Immunocompetence against Murine H22 Tumor In Vivo

2014 ◽  
Vol 989-994 ◽  
pp. 1056-1059
Author(s):  
Juan Tang ◽  
Zhen Kong ◽  
Dong Yue Liu ◽  
An Jun Liu

New antitumor strategies are underway and play important roles in clinical trials for combating cancer in future. Eucheuma gelatinae contains a certain amount of polysaccharides, which has various biological activities. In this study, the antitumor effect of Eucheuma gelatinae polysaccharide on murine H22 tumor bearing mice has been investigated. Histological stain, flow cytometry and other methods are applied to evaluate the effects of Eucheuma gelatinae polysaccharide on immunocompetence in vivo. The data indicates that Eucheuma gelatinae polysaccharide has antitumor effect in vivo by enhancing immunocompetence of the tumor bearers.

Molecules ◽  
2021 ◽  
Vol 26 (9) ◽  
pp. 2506
Author(s):  
Wamidh H. Talib ◽  
Ahmad Riyad Alsayed ◽  
Alaa Abuawad ◽  
Safa Daoud ◽  
Asma Ismail Mahmod

Melatonin is a pleotropic molecule with numerous biological activities. Epidemiological and experimental studies have documented that melatonin could inhibit different types of cancer in vitro and in vivo. Results showed the involvement of melatonin in different anticancer mechanisms including apoptosis induction, cell proliferation inhibition, reduction in tumor growth and metastases, reduction in the side effects associated with chemotherapy and radiotherapy, decreasing drug resistance in cancer therapy, and augmentation of the therapeutic effects of conventional anticancer therapies. Clinical trials revealed that melatonin is an effective adjuvant drug to all conventional therapies. This review summarized melatonin biosynthesis, availability from natural sources, metabolism, bioavailability, anticancer mechanisms of melatonin, its use in clinical trials, and pharmaceutical formulation. Studies discussed in this review will provide a solid foundation for researchers and physicians to design and develop new therapies to treat and prevent cancer using melatonin.


2021 ◽  
Vol 42 ◽  
pp. e67649
Author(s):  
Marta Sánchez ◽  
Elena González-Burgos ◽  
Irene Iglesias ◽  
M. Pilar Gómez-Serranillos Cuadrado

Valeriana officinalis L. (Caprifoliaceae family) has been traditionally used to treat mild nervous tension and sleep problems. The basis of these activities are mainly attributed to valerenic acid through the modulation of the GABA receptor. Moreover, V. officinalis is claimed to have other biological activities such as cardiovascular benefits, anticancer, antimicrobial and spasmolytic.  The current review aims to update the biological and pharmacological studies (in vitro, in vivo and clinical trials) of V. officinalis and its major secondary metabolites in order to guide future research. Databases PubMed, Science Direct and Scopus were used for literature search including original papers written in English and published between 2014 and 2020. There have been identified 33 articles which met inclusion criteria. Most of these works were performed with V. officinalis extracts and only a few papers (in vitro and in vivo studies) evaluated the activity of isolated compounds (valerenic acid and volvalerenal acid K). In vitro studies focused on studying antioxidant and neuroprotective activity. In vivo studies and clinical trials mainly investigated activities on the nervous system (anticonvulsant activity, antidepressant, cognitive problems, anxiety and sleep disorders). Just few studies were focused on other different activities, highlight effects on symptoms of premenstrual and postmenopausal syndromes. Valeriana officinalis continues to be one of the medicinal plants most used by today's society for its therapeutic properties and whose biological and pharmacological activities continue to arouse great scientific interest as evidenced in recent publications. This review shows scientific evidence on traditional uses of V. officinalis on nervous system.


Author(s):  
Zahra Hashemi ◽  
Mohammad Ali Ebrahimzadeh

Abstract: Inherited beta-thalassemia is a major disease caused by irregular production of hemoglobin through reducing beta-globin chains. It has been observed that increasing fetal hemoglobin (HbF) production improves symptoms in the patients. Therefore, an increase in the level of HbF has been an operative approach for treating patients with beta-thalassemia. This review represents compounds with biological activities and pharmacological properties that can promote the HBF level and therefore used in the β-thalassemia patients' therapy. Various natural products with different mechanisms of action can be helpful in this medication cure. Clinical trials were efficient in improving the signs of patients. Association of in vivo, and in vitro studies of HbF induction and γ-globin mRNA growth displays that in vitro experiments could be an indicator of the in vivo response. The current study shows that; (a) HbF inducers can be grouped in several classes based on their chemical structures and mechanism of actions; b) According to several clinical trials, well-known drugs such as hydroxyurea and decitabine are useful HbF inducers; (c) The cellular biosensor K562 carrying genes under the control of the human γ-globin and β-globin gene promoters were applied during the researches; d) New natural products and lead compounds were found based on various studies as HbF inducers.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 413-413
Author(s):  
Alexander E Perl ◽  
Kasner Margaret ◽  
Shank Doris ◽  
Selina Luger ◽  
Martin Carroll

Abstract Abstract 413 The mammalian target of rapamycin (mTOR) is an emerging molecular target in cancer therapy, however the relationship between target activation in individual tumors as well as inhibition of target and clinical response is poorly defined. This is in large part due to the difficulty of real-time monitoring of mTOR activation in patient samples. mTOR is activated in acute myelogneous leukemia (AML) cells and the mTOR inhibitor rapamycin enhances cytotoxic effects of chemotherapy in vitro and in vivo. We therefore developed a new application of a recently-developed, whole blood fixation/permeabilization technique for intracellular flow cytometry (Chow, et al. Cytometry A 2005). Using this approach, we sought to serially monitor S6 ribosomal protein (S6) phosphorylation in peripheral blood leukemic blasts during clinical trials of mTOR inhibitors. S6 is a known target of mTOR and its phosphorylation is a surrogate marker for mTOR kinase activation. We applied this methodology during a recent pilot trial in which an oral mTOR inhibitor, sirolimus (rapamycin), was administered in sequence with intensive combination chemotherapy (mitoxantrone, etoposide, and cytarabine, or MEC) in patients with relapsed, refractory, or secondary AML. The whole blood fixation process sufficiently preserved surface epitopes and light scatter properties for immunophenotyping, allowing specific signaling analysis of leukemic blasts as well as non-malignant cell populations. Importantly, even leukopenic trial samples containing as few as 20% blasts provided robust signaling data in malignant cells. S6 phosphorylation was readily apparent in leukemic blasts prior to therapy and, consistent with prior reports, occurred only in a subset of blasts. Exposing aliquots of pre-treatment whole blood samples to increasing concentrations of rapamycin ex vivo determined that leukemic blasts from most samples showed inhibition of S6 phosphorylation at clinically achievable concentrations (between 10-20 nM). Notably, some subjects' leukemic blasts showed no inhibition to >50 nM rapamycin, which far exceeded trough concentrations measured on our studies. To examine rapamycin's in vivo biochemical effects, we performed a paired analysis of clinical samples drawn at study entry and after 72 hours of oral sirolimus. 10 subjects provided paired samples, of which 2 did not show baseline S6 phosphorylation, 6 showed baseline S6 phosphorylation that inhibited during therapy, and 2 showed baseline S6 phosphorylation but no inhibition. Trough rapamycin levels were similar among rapamycin responsive and resistant subjects. Considering the 6 subjects with in vivo mTOR inhibition, 3 subjects achieved complete or partial remissions from the regimen. Neither subject with in vivo rapamycin resistance had a clinical response. Overall, we conclude that effective inhibition of mTOR signaling in AML blasts occurs in the majority of subjects during sirolimus treatment at the dose studied. However, cell-intrinsic rapamycin resistance occurs in a minority of patients and requires further study to clarify its mechanism and effects upon concurrent chemotherapy response. These data demonstrate the feasibility of real-time, intra-tumoral pharmacodynamic monitoring of S6 phosphorylation by flow cytometry during clinical trials combining intensive chemotherapy and signal transduction inhibitors for leukemia. Our approach greatly clarifies pharmacokinetic/pharmacodynamic relationships and has broad application to pre-clinical and clinical testing of drugs whose direct or downstream effects disrupt PI3K/AKT/mTOR signaling. Such compounds include inhibitors of FLT3, c-KIT, BCR-ABL, JAK2, and ras/raf/MAPK. Multicenter/cooperative group phase II testing of sirolimus plus MEC in AML has been initiated to establish the regimen's response rate and test the extent to which our pharmacodynamic studies predict clinical response. Disclosures: Off Label Use: The use of sirolimus in the therapy of AML is investigational and off-label. Carroll:Cephalon consultancy: Consultancy; Sanofi Aventis Corporation: Research Funding; Kyowa Hakko Kirin Pharmaceutical: Research Funding.


2020 ◽  
Vol 10 (4) ◽  
pp. 372-383
Author(s):  
Rajani Kurup ◽  
Ajikumaran Nair Sadasivan ◽  
Uthayakumari Kalavathy ◽  
Sabulal Baby

Background: Polyscias guilfoylei, commonly called ‘geranium aralia’, is an erect shrub with dark green leaves. P. guilfoylei has been introduced to tropical countries and is generally cultivated in gardens for ornamental purposes. There are no previous studies on the essential oil of P. guilfoylei and its biological activities. Objective: In this study, we report the chemical profile of P. guilfoylei leaf essential oil and its anticancer activity tested by various in vitro and in vivo assays. Methods: The chemical profile of P. guilfoylei leaf oil was elucidated by Gas Chromatographic analyses (GC-FID, GC-MS). Anticancer activity of P. guilfoylei leaf oil was tested by MTT, morphological observations, DNA ladder, comet, caspase, flow cytometry and in vivo assays. Results: Gas chromatographic profiling of P. guilfoylei leaf oil identified 50 constituents (β-selinene 49.59%, α-selinene 21.68%, (Z)-falcarinol 11.65%). In MTT assay, P. guilfoylei leaf oil at 50, 25, 10, 5 and 1 μg/ml showed 98.6 ± 1.2, 95.3 ± 0.78, 76.8 ± 1.59, 43.6 ± 0.99 and 39.8 ± 1.17% DLA cell death, respectively (CD50 5.96 μg/ml). In flow cytometry, the majority of P. guilfoylei leaf oil (25 μg/ml) treated DLA cells showed an accumulation/cell arrest in G2M phase (61.7 ± 2.6%). In P. guilfoylei leaf oil treated mice (40 days), 5 animals (83.3%, each) were protected in 25, 50 mg/kg groups. Conclusion: P. guilfoylei leaf oil, with minimal toxicity to normal cells, exhibited significant anticancer activity against lymphoma cells enhancing its potential as an anticancer agent.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1015-1015 ◽  
Author(s):  
Elissa Furutani ◽  
Su Su ◽  
Aleah Smith ◽  
Maria Berg ◽  
Richard Childs

Abstract Abstract 1015 Natural killer (NK) cells are a component of the innate immune system that target both tumors and virally infected cells. NK cell killing of tumors is regulated by a delicate balance of activating and inhibitory receptors. These inhibitory receptors bind HLA ligands which prevent NK cell targeting of normal “self” cells. Up regulation of surface expression of HLA molecules has been utilized by tumors as a method to evade NK cell cytotoxicity. Disrupting the function or expression of inhibitory receptors on NK cells could potentially be used as a method to overcome this effect. While most inhibitory receptors are present in only a subset of NK cells, NK cells universally express the HLA-E binding inhibitory receptor NKG2A. We hypothesized that siRNA inactivation of NK cell NKG2A would could be used as a therapeutic approach to enhance NK cell tumor cytotoxicity in vivo. The human natural killer cell line NKL was transduced with lentiviral vectors encoding shRNA targeting various regions of the NKG2A transcript. Following lentiviral transduction, knockdown of receptor expression was confirmed by flow cytometry and RT-qPCR. Compared to wild type (WT) and GFP-transduced NKL controls, NKG2A silenced NKL cells had increased secretion of IFN-gamma and Fas-L by ELISA and increased granzymes A and B and Nkp30 expression by flow cytometry. In contrast, expression of NKG2D, Nkp44, Nkp46, LFA-1, DNAM, and TRAIL was not altered by NKG2A silencing. Chromium-based cytotoxicity assays showed shRNA knockdown of NKG2A significantly enhanced NK cell cytotoxicity of tumor cells: at a 20:1 effector to target ratio, NKG2A knockdown NKLs, WT NKLs and GFP-transduced NKLs induced 68.9%, 8.2% and 8.3% lysis respectively of 721.221 EBV-LCL tumor targets (p=0.001). Remarkably, NKG2A silencing enhanced NKL killing of both HLA-E positive (721.221 EBV-LCL and 526 melanoma cells) and HLA-E negative (K562) tumor cell lines, suggesting NKG2A inactivation increased NK cell cytotoxicity through both HLA-E dependent and independent mechanisms. Using a xenogeneic model, we next explored the in vivo effects of transferring NKG2A silenced NK cells in tumor bearing mice. Immunodeficient NSG mice were injected with 1 million human luciferase transduced 721.221 HLA-E expressing EBV-LCL tumor cells. Twenty-four hours later, tumor-bearing mice were injected with 2–5 million WT NKL cells, GFP-control-transduced NKL, or NKG2A silenced NKL cells, then received IL-2 sq for 10 days to induce in vivo NK cell proliferation. NKL numbers in blood were subsequently analyzed by flow cytometry and tumor burden was assessed by luciferase-based bioluminescence imaging (BLI). At 16 and 21 days following adoptive NK cell transfer, BLI showed that recipients of NKG2A silenced NKL cells had slower tumor growth and significantly smaller tumor burden compared to NKL wt and NKL-GFP transduced controls (figure). Importantly, no toxicity related to infusing NKG2A inactivated NK cells was observed. These in vitro and in vivo data suggest shRNA knockdown of the NKG2A inhibitory receptor could be used as a method to augment NK cell tumor cytotoxicity in patients with hematological malignancies. Figure: Tumor burden in mice Luciferase-tagged 721.221 HLA-E EBV LCLs were injected into mice and imaged using a bioluminescence imager at days 10, 16, and 22 following NKL injection. 5 mice were followed in each group. Figure:. Tumor burden in mice . / Luciferase-tagged 721.221 HLA-E EBV LCLs were injected into mice and imaged using a bioluminescence imager at days 10, 16, and 22 following NKL injection. 5 mice were followed in each group. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Ming Yi ◽  
Mengke Niu ◽  
Jing Zhang ◽  
Shiyu Li ◽  
Shuangli Zhu ◽  
...  

Abstract Background Our previous work showed that the anti-TGF-β/PD-L1 bispecific antibody YM101 effectively overcame anti-PD-L1 resistance in immune-excluded tumor models. However, in immune-desert models, the efficacy of YM101 was limited. Bivalent manganese (Mn2+) is identified as a natural stimulator of interferon genes (STING) agonist, which might enhance cancer antigen presentation and improve the therapeutic effect of YM101. Methods The effect of Mn2+ on STING pathway was validated by western blotting and enzyme-linked immunosorbent assay. Dendritic cell (DC) maturation was measured by flow cytometry. The synergistic effect between Mn2+ and YM101 in vitro was determined by one-way mixed lymphocyte reaction, CFSE dilution assay, and cytokine detection. The in vivo antitumor effect of Mn2+ plus YM101 therapy was assessed in CT26, EMT-6, H22, and B16 tumor models. Flow cytometry, RNA-seq, and immunofluorescent staining were adopted to investigate the alterations in the tumor microenvironment. Results Mn2+ could activate STING pathway and promote the maturation of human and murine DC. The results of one-way mixed lymphocyte reaction showed that Mn2+ synergized YM101 in T cell activation. Moreover, in multiple syngeneic murine tumor models, Mn2+ plus YM101 therapy exhibited a durable antitumor effect and prolonged the survival of tumor-bearing mice. Relative to YM101 monotherapy and Mn2+ plus anti-PD-L1 therapy, Mn2+ plus YM101 treatment had a more powerful antitumor effect and a broader antitumor spectrum. Mechanistically, Mn2+ plus YM101 strategy simultaneously regulated multiple components in the antitumor immunity and drove the shift from immune-excluded or immune-desert to immune-inflamed tumors. The investigation in the TME indicated Mn2+ plus YM101 strategy activated innate and adaptive immunity, enhanced cancer antigen presentation, and upregulated the density and function of tumor-infiltrating lymphocytes. This normalized TME and reinvigorated antitumor immunity contributed to the superior antitumor effect of the combination therapy. Conclusion Combining Mn2+ with YM101 has a synergistic antitumor effect, effectively controlling tumor growth and prolonging the survival of tumor-bearing mice. This novel cocktail strategy has the potential to be a universal regimen for inflamed and non-inflamed tumors.


Molecules ◽  
2020 ◽  
Vol 25 (9) ◽  
pp. 2218
Author(s):  
Yilin Sun ◽  
Jiankun Wang ◽  
Kun Hao

Gemcitabine (dFdC) demonstrates significant effectiveness against solid tumors in vitro and in vivo; however, its clinical application is limited because it tends to easily undergo deamination metabolism. Therefore, we synthesized 4-N-carbobenzoxy-gemcitabine (Cbz-dFdC) as a lead prodrug and conducted a detailed pharmacokinetic, metabolic, and pharmacodynamic evaluation. After intragastric Cbz-dFdC administration, the Cmax of Cbz-dFdC and dFdC was 451.1 ± 106.7 and 1656.3 ± 431.5 ng/mL, respectively. The Tmax of Cbz-dFdC and dFdC was 2 and 4 h, respectively. After intragastric administration of Cbz-dFdC, this compound was mainly distributed in the intestine due to low carboxylesterase-1 (CES1) activity. Cbz-dFdC is activated by CES1 in both humans and rats. The enzyme kinetic curves were well fitted by the Michaelis–Menten equation in rats’ blood, plasma, and tissue homogenates and S9 of the liver and kidney, as well as human liver S9 and CES1 recombinase. The pharmacodynamic results showed that the Cbz-dFdC have a good antitumor effect in the HepG2 cell and in tumor-bearing mice, respectively. In general, Cbz-dFdC has good pharmaceutical characteristics and is therefore a good candidate for a potential prodrug.


2021 ◽  
Vol 9 (7) ◽  
pp. e002644
Author(s):  
Montserrat Puigdelloses ◽  
Marc Garcia-Moure ◽  
Sara Labiano ◽  
Virginia Laspidea ◽  
Marisol Gonzalez-Huarriz ◽  
...  

BackgroundGlioblastoma (GBM) is a devastating primary brain tumor with a highly immunosuppressive tumor microenvironment, and treatment with oncolytic viruses (OVs) has emerged as a promising strategy for these tumors. Our group constructed a new OV named Delta-24-ACT, which was based on the Delta-24-RGD platform armed with 4-1BB ligand (4-1BBL). In this study, we evaluated the antitumor effect of Delta-24-ACT alone or in combination with an immune checkpoint inhibitor (ICI) in preclinical models of glioma.MethodsThe in vitro effect of Delta-24-ACT was characterized through analyses of its infectivity, replication and cytotoxicity by flow cytometry, immunofluorescence (IF) and MTS assays, respectively. The antitumor effect and therapeutic mechanism were evaluated in vivo using several immunocompetent murine glioma models. The tumor microenvironment was studied by flow cytometry, immunohistochemistry and IF.ResultsDelta-24-ACT was able to infect and exert a cytotoxic effect on murine and human glioma cell lines. Moreover, Delta-24-ACT expressed functional 4-1BBL that was able to costimulate T lymphocytes in vitro and in vivo. Delta-24-ACT elicited a more potent antitumor effect in GBM murine models than Delta-24-RGD, as demonstrated by significant increases in median survival and the percentage of long-term survivors. Furthermore, Delta-24-ACT modulated the tumor microenvironment, which led to lymphocyte infiltration and alteration of their immune phenotype, as characterized by increases in the expression of Programmed Death 1 (PD-1) on T cells and Programmed Death-ligand 1 (PD-L1) on different myeloid cell populations. Because Delta-24-ACT did not induce an immune memory response in long-term survivors, as indicated by rechallenge experiments, we combined Delta-24-ACT with an anti-PD-L1 antibody. In GL261 tumor-bearing mice, this combination showed superior efficacy compared with either monotherapy. Specifically, this combination not only increased the median survival but also generated immune memory, which allowed long-term survival and thus tumor rejection on rechallenge.ConclusionsIn summary, our data demonstrated the efficacy of Delta-24-ACT combined with a PD-L1 inhibitor in murine glioma models. Moreover, the data underscore the potential to combine local immunovirotherapy with ICIs as an effective therapy for poorly infiltrated tumors.


2021 ◽  
Vol 12 ◽  
Author(s):  
Min Ye ◽  
Yongkang Gai ◽  
Hao Ji ◽  
Yaqun Jiang ◽  
Pengxin Qiao ◽  
...  

Sphingosine-1-phosphate (S1P) is a phospholipid that regulates pleiotropic biological activities and exerts extracellular functions by binding to five specific G-protein-coupled receptors, S1P receptors (S1PR) 1–5. When activated by S1P, S1PR promote the proliferation and invasion of tumor cells by inducing the formation of new blood vessels. We developed and assessed a new monoclonal antibody imaging probe 99mTc-HYNIC-S1PR1mAb, to explore the feasibility of targeting the S1PR1 in vitro and in vivo. S1PR1mAb was prepared and followed by technetium-99m labeling with succinimidyl 6-hydraziniumnicotinate hydrochloride. Cell uptake and blocking studies were performed to investigate the binding specificity of 99mTc-HYNIC-S1PR1mAb in vitro. 99mTc-HYNIC-S1P1mAb was also tested in vivo in mice xenografted with SK-HEP-1 (high-expression of S1PR1) and MCF-7 (low-expression of S1PR1) using single-photon emission-computed tomography (SPECT). Ex vivo gamma counting of tissues from tumor-bearing mice was used to evaluate 99mTc-HYNIC-S1PR1mAb biodistribution. The biodistribution study results showed significantly higher uptake in SK-HEP-1 tumors than in MCF-7 tumors (P < 0.001). Reduced uptake of 99mTc-HYNIC-S1PR1mAb in SK-HEP-1 was observed in tumor-bearing nude mice pretreated with fingolimod, which binds competitively to the receptors, especially S1PR1. 99mTc-HYNIC-S1PR1mAb can be synthesized and specifically targeted to S1PR1 in vitro and in vivo, allowing S1PR1 expression assessment with SPECT imaging.


Sign in / Sign up

Export Citation Format

Share Document