Therapeutic Potential for Toll-Like Receptor 2 Agonists In Pediatric Acute Lymphoblastic Leukemia (pre-B ALL): Induction of Apoptosis and Anti-ALL Immunogenicity

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 870-870
Author(s):  
Nina Rolf ◽  
Amina Kariminia ◽  
Sabine Ivison ◽  
Kirk R. Schultz

Abstract Abstract 870 Introduction: Despite high cure-rates achieved by an intense chemotherapy regimen in pediatric pre-B ALL, 20% of children suffer relapse after which they face limited therapy options and poor prognosis. New treatment options are urgently needed. Background: Hematopoietic cell transplantation (HCT) has become an important therapy for ALL-relapse, both by eradicating leukemic blasts through myeloablative chemotherapy and by inducing a graft-versus-leukemia effect (GVL). Inadequate post-HCT immune responses can increase risk of relapse, while a GVL effect often comes at the expense of graft-versus-host disease leading to significant morbidity and mortality. Stimulation of Toll-like receptors (TLRs), which detect conserved pathogen- and stressed-self-derived ligands, represents a possible strategy for augmenting anti-ALL immunogenicity. TLR stimulation of antigen-presenting cells induces the expression of co-stimulatory molecules and cytokines, resulting in Th1-polarized immune responses important for anti-tumor responses. Furthermore, it has been shown that TLR stimulation can increase the sensitivity of hematological malignancies to conventional chemotherapeutics (Spaner et al, Leukemia 2010). We have previously used agonists of endosome-localized TLR9 (synthetic CpG-ODN) to enhance anti-ALL immunity (Reid et al, Blood 2005). This approach, which was successful in eliminating leukemia and extending survival in a syngeneic mouse model of minimal residual disease (Seif et al, Blood 2009), is now entering a phase I clinical trial (TACL group). However, our recent research demonstrated that TLR2 receptors, localized on the surface and thus independent of endocytosis, are more abundantly and consistently expressed on pre-B ALL cells. Unlike most other TLRs, which are functionally active as homodimers, TLR2s can form heterodimers with TLR1 or TLR6 suggesting that TLR2 ligands can modulate multiple downstream signaling pathways. Altogether, TLR2 agonists may have better efficacy in generating anti-B-ALL immunity, apoptosis and chemosensitivity. Objective: We tested the hypothesis that i) TLR2 agonists increase pre-B ALL blast sensitivity to doxorubicin (DOX) and asparaginase (ASP) in vitro. We further investigated if ii) distinctive synthetic TLR2 agonists (TLR2/6: Pam2CSK4=Pam2, TLR2/1: Pam3CSK4=Pam3 and FSL-1) differ in their ability to 1) transduce specific signaling pathways, 2) induce apoptosis, and 3) augment pre B-ALL cell immunogenicity. Methods: Pre-B ALL cell lines pre-treated with TLR2 agonists were compared to untreated samples in vitro using the following methods: 1) NFkB phosphorylation (pNFkB) and IkB degradation was detected by flow cytometry in a time and dose-dependent manner; 2) Induction of apoptosis/necrosis of blasts by TLR2 stimulation was examined by flow cytometric detection of AnnexinV/7AAD. 3)In vitro augmentation of immunogenicity was investigated by measuring induction of co-stimulatory molecules and increment of allogeneic T-cell proliferation. Results: 1) Pam2 rapidly and potently induced NFkB signaling (pNFkB 23–42% at 1ug/ml after 15–30min), while Pam3 (10ug/ml) displayed a slow and continuous increment at 60min, thus underlining the differences in signaling kinetics. 2) Pam3 stimulation induced significant, dose-dependent cell death: 36% (6hr), 66% (24hr), 81% (48hr) at 10ug/ml. Unexpectedly, both TLR2/6 agonists, Pam2 and FSL-1, did not induce comparable degree of cell death. Thus, Pam3 killed pre-B ALL cells more potently than therapeutic levels of Doxorubicin (0.005ug/ml) at the earliest time point of 6hr (8.5%) with equal cytotoxicity at 24hr (44%) and 48hr (93%). 3) All TLR2 agonists induced a comparably high expression of CD40/80/86 after 48hr (83-93%/63-76%/84-86%). However, only Pam3 induced a dose-dependent, early CD40 expression at 6hr (32%) and 24hr (60%). Furthermore, blast pre-treatment with Pam3 (but not Pam2) increased the sensitivity to ASP (49%/73% live cells with/without Pam3, respectively). Finally, there was a 30% increase in immunogenicity of pre B-ALL blasts by Pam3 in T-cell alloreactivity studies when compared to medium. Conclusion: TLR2 agonists increased anti-ALL immunogenicity in vitro. Pam3 also had strong cell-death inducing qualities and sensitized pre-B ALL blasts to chemotherapy. This supports that TLR2 agonists have promise for improving relapsed pre-B ALL cure-rates. Disclosures: No relevant conflicts of interest to declare.

Nutrients ◽  
2021 ◽  
Vol 13 (7) ◽  
pp. 2178
Author(s):  
Fabio Morandi ◽  
Veronica Bensa ◽  
Enzo Calarco ◽  
Fabio Pastorino ◽  
Patrizia Perri ◽  
...  

Neuroblastoma (NB) is the most common extra-cranial solid tumor of pediatric age. The prognosis for high-risk NB patients remains poor, and new treatment strategies are desirable. The olive leaf extract (OLE) is constituted by phenolic compounds, whose health beneficial effects were reported. Here, the anti-tumor effects of OLE were investigated in vitro on a panel of NB cell lines in terms of (i) reduction of cell viability; (ii) inhibition of cell proliferation through cell cycle arrest; (iii) induction of apoptosis; and (iv) inhibition of cell migration. Furthermore, cytotoxicity experiments, by combining OLE with the chemotherapeutic topotecan, were also performed. OLE reduced the cell viability of NB cells in a time- and dose-dependent manner in 2D and 3D models. NB cells exposed to OLE underwent inhibition of cell proliferation, which was characterized by an arrest of the cell cycle progression in G0/G1 phase and by the accumulation of cells in the sub-G0 phase, which is peculiar of apoptotic death. This was confirmed by a dose-dependent increase of Annexin V+ cells (peculiar of apoptosis) and upregulation of caspases 3 and 7 protein levels. Moreover, OLE inhibited the migration of NB cells. Finally, the anti-tumor efficacy of the chemotherapeutic topotecan, in terms of cell viability reduction, was greatly enhanced by its combination with OLE. In conclusion, OLE has anti-tumor activity against NB by inhibiting cell proliferation and migration and by inducing apoptosis.


1996 ◽  
Vol 63 (2) ◽  
pp. 257-267 ◽  
Author(s):  
Chun W. Wong ◽  
Geoffrey O. Regester ◽  
Geoffrey L. Francis ◽  
Dennis L. Watson

SummaryStudies on the immunomodulatory activities of ruminant milk and colostral whey fractions were undertaken. By comparing with boiled colostral whey in a preliminary experiment, a putative heat-labile immunostimulatory factor for antibody responses was found to be present in ovine colostral whey. Studies were then undertaken in sheep in which the efferent prefemoral lymphatic ducts were cannulated bilaterally, and immune responses in the node were measured following subcutaneous injection in the flank fold of whey protein preparations of various purities. A significant sustained decline of efferent lymphocyte output was observed following injection with autologous crude milk whey or colostral whey preparations, but no changes were observed in interferon-gamma levels in lymph plasma. Two bovine milk whey fractions (lactoperoxidase and lactoferrin) of high purity were compared in bilaterally cannulated sheep. A transient decline over the first 6 h was seen in the efferent lymphocyte output and lymph flow rate after injection of both fractions. A significant difference was seen between the two fractions in interferongamma levels in lymph at 6 h after injection. However, no significant changes in the proportion of the various efferent lymphocyte phenotypes were seen following either treatment. Whereas both fractions showed a significant inhibitory effect in a dose-dependent manner on the proliferative response of T lymphocytes, but not B lymphocytes, to mitogenic stimulation in vitro, no similar changes were seen following in vivo stimulation with these two fractions.


2021 ◽  
Vol 3 (Supplement_6) ◽  
pp. vi6-vi6
Author(s):  
Takashi Fujii ◽  
Shun Yamamuro ◽  
Masamichi Takahashi ◽  
Akihide Kondo ◽  
Yoshitaka Narita ◽  
...  

Abstract The therapeutic outcome of glioblastomas (GBMs) is still very poor. Therefore, invention of novel therapeutic methods against GBM cases is considered urgent. The antitumor effects of naturally-derived compounds are attracting attention recently, and therapeutic efficacy of curcumin, a plant-derived compound previously used for multiple purpose, has been indicated in many cancer systems; however, clinical application of curcumin is considered difficult because of its poor bioavailability (under 1 %). Curcumin monoglucuronide (CMG), a water-soluble prodrug of curcumin recently developed for overcoming this weakness, has been demonstrated excellent antitumor effects for several malignancies in vitro and in vivo; therefore, we investigated the effects of CMG against GBM cells. CMG induced cell death of human GBM cells lines (T98G, U251MG, and U87MG) by dose dependent manner by triggering multiple forms of cell death such as apoptosis and perthanatos. Immunoblotting of CMG-treated GBM cell lysates demonstrated activation of multiple cell death signaling. Furthermore, immunodeficiency mice harboring intracerebral U87MG cell xenografts systemically treated by CMG showed significantly prolonged survival compared with control mice. These results suggest CMG would be a novel therapeutic agent against GBM cases.


1983 ◽  
Vol 245 (4) ◽  
pp. G463-G469
Author(s):  
B. Richelsen ◽  
J. F. Rehfeld ◽  
L. I. Larsson

A technique for studying in vitro release of gastric hormones has been developed. The system utilizes nonenzymatically isolated antropyloric glands from humans or rats, which are perifused in a Bio-Gel P-2 column. The system permits the study of kinetics and dose-response characteristics using the glands as their own control. The glands were stimulated with carbachol and bombesin, and the antral peptides gastrin and somatostatin were measured. Bombesin and carbachol both evoked a dose-dependent stimulation of gastrin release, beginning at below 10(-10) M (bombesin) and 10(-7) M (carbachol). Carbachol inhibited the release of somatostatin in a dose-dependent manner, being maximally effective at 10(-6) M and then producing 60% inhibition of somatostatin release. Bombesin was without effect on antropyloric somatostatin release. These data suggest that the gastrin-stimulating effect of carbachol is partially or totally due to inhibition of somatostatin release, whereas bombesinergic stimulation of gastrin release must work in an independent manner. In addition, data on the effects of these substances on the release of gastrin and ACTH-like peptides from human antropyloric glands are presented. Due to the absence of local neural reflexes, this system is a useful supplement to the isolated perfused stomach model.


1990 ◽  
Vol 5 (1) ◽  
pp. 55-60 ◽  
Author(s):  
L. B. O'Toole ◽  
K.J. Armour ◽  
C. Decourt ◽  
N. Hazon ◽  
B. Lahlou ◽  
...  

ABSTRACT An isolated in-vitro perifused interrenal gland preparation from the dogfish Scyliorhinus canicula was used to study production of quantitatively the major corticosteroid 1α-hydroxycorticosterone (1α-OH-B), measured by radioimmunoassay. Basal secretory rates were 877·1 ± 145 (s.e.m.) fmol/mg per 15 min (n=14) and the preparation remained viable for up to 22 h, as reflected in a brisk response to 10 μm cyclic AMP (cAMP) after this time. Steroid production responded in a dose-dependent manner to porcine ACTH, with 10 μm producing a maximum stimulation of 225% above the basal secretory rate. cAMP (10 μm) produced an increase of 278% above basal, while 1 μm forskolin increased basal secretory rates by 127%. [Val5]- and [Ile5]-angiotensin II (0·1 μm) increased 1α-OH-B production by 120 and 372% respectively over basal secretory rates. Increasing the concentration of K+ in the perfusate from 8 mm to 12, 18, 28 and 40 mm produced a significant rise only at 28 mm. Alterations in the concentration of Na+ and osmolarity of the perifusion medium had inconsistent effects on steroid production. Increased concentrations of urea (from 360 to 720 mm) increased the basal secretory rate by 121%, whilst reducing the concentration of urea (from 360 to 90 mm) had no effect.


Cancers ◽  
2020 ◽  
Vol 12 (12) ◽  
pp. 3641
Author(s):  
Tatsuya Kobayashi ◽  
Makoto Miyazaki ◽  
Nobuyoshi Sasaki ◽  
Shun Yamamuro ◽  
Eita Uchida ◽  
...  

To manage refractory and invasive glioblastomas (GBM)s, photodynamic therapy (PDT) using talaporfin sodium (NPe6) (NPe6-PDT) was recently approved in clinical practice. However, the molecular machineries regulating resistance against NPe6-PDT in GBMs and mechanisms underlying the changes in GBM phenotypes following NPe6-PDT remain unknown. Herein, we established an in vitro NPe6-mediated PDT model using human GBM cell lines. NPe6-PDT induced GBM cell death in a NPe6 dose-dependent manner. However, this NPe6-PDT-induced GBM cell death was not completely blocked by the pan-caspase inhibitor, suggesting NPe6-PDT induces both caspase-dependent and -independent cell death. Moreover, treatment with poly (ADP-ribose) polymerase inhibitor blocked NPe6-PDT-triggered caspase-independent GBM cell death. Next, it was also revealed resistance to re-NPe6-PDT of GBM cells and GBM stem cells survived following NPe6-PDT (NPe6-PDT-R cells), as well as migration and invasion of NPe6-PDT-R cells were enhanced. Immunoblotting of NPe6-PDT-R cells to assess the behavior of the proteins that are known to be stress-induced revealed that only ERK1/2 activation exhibited the same trend as migration. Importantly, treatment with the MEK1/2 inhibitor trametinib reversed resistance against re-NPe6-PDT and suppressed the enhanced migration and invasion of NPe6-PDT-R cells. Overall, enhanced ERK1/2 activation is suggested as a key regulator of elevated malignant phenotypes of GBM cells surviving NPe6-PDT and is therefore considered as a potential therapeutic target against GBM.


Author(s):  
Hongfang Wang ◽  
Jinlian Fu ◽  
Aiguo Wang

Obesity has become a global health problem. Research suggests that leptin, a hormone that responds to fat deposition, may be involved in mammalian reproduction; however, its precise role in embryo implantation is poorly understood. Here, primary porcine endometrium epithelium cells (PEECs) were cultured in vitro and used to evaluate the regulatory role of different leptin levels on β3-integrin, MMP9, HB-EGF, and IL-1β, which are, respectively, involved in four critical steps of embryo implantation. Results showed that only 0.01 nM leptin significantly improved β3-integrin mRNA expression (p < 0.05). MMP9 and HB-EGF mRNA expressions were upregulated by 0.10–10.00 nM leptin (p < 0.05). The IL-1β expression level was only increased by 10.00 nM leptin (p < 0.05). β3-integrin, MMP9, HB-EGF, and IL-1β mRNA and protein have a similar fluctuant response to increased leptin. Leptin’s influence on β3-integrin, MMP9, HB-EGF, and IL-1β disappeared when the JAK2, PI(3)K, or MAPK signaling pathways were blocked, respectively. In conclusion, leptin affected porcine implantation by regulating the expression of β3-integrin, MMP9, HB-EGF, and IL-1β in a dose-dependent manner. The signaling pathways of JAK2, PI(3)K, and MAPK may participate in this regulatory process. These findings will contribute to further understanding the mechanisms of reproductive disorders in obesity.


2020 ◽  
Vol 2020 ◽  
pp. 1-15 ◽  
Author(s):  
Xinyi Lu ◽  
Xingli Wu ◽  
Lin Jing ◽  
Lingjia Tao ◽  
Yingxuan Zhang ◽  
...  

Objective. To analyze the active compounds, potential targets, and diseases of JianPi Fu Recipe (JPFR) based on network pharmacology and bioinformatics and verify the potential biological function and mechanism of JPFR in vitro and in vivo. Methods. Network pharmacology databases including TCMSP, TCM-PTD, TCMID, and DrugBank were used to screen the active compounds and potential drug targets of JPFR. Cytoscape 3.7 software was applied to construct the interaction network between active compounds and potential targets. The DAVID online database analysis was performed to investigate the potential effective diseases and involved signaling pathways according to the results of the GO function and KEGG pathways enrichment analysis. To ensure standardization and maintain interbatch reliability of JPFR, High Performance Liquid Chromatography (HPLC) was used to establish a “chemical fingerprint.” For biological function validation, the effect of JPFR on the proliferation and migration of CRC cells in vitro was investigated by CCK-8 and transwell and wound healing assay, and the effect of JPFR on the growth and metastasis of CRC cells in vivo was detected by building a lung metastasis model in nude mice and in vivo imaging. For the potential mechanism validation, the expressions of MALAT1, PTBP-2, and β-catenin in CRC cells and transplanted CRC tumors were detected by real-time PCR, western blot, and immunohistochemical staining analysis. Results. According to the rules of oral bioavailability (OB) > 30% and drug-likeness (DL) > 0.18, 244 effective compounds in JPFR were screened out, as well as the corresponding 132 potential drug targets. By the analysis of DAVID database, all these key targets were associated closely with the cancer diseases such as prostate cancer, colorectal cancer, bladder cancer, small cell lung cancer, pancreatic cancer, and hepatocellular carcinoma. In addition, multiple signaling pathways were closely related to JPFR, including p53, Wnt, PI3K-Akt, IL-17, HIF-1, p38-MAPK, NF-κB, PD-L1 expression and PD-1 checkpoint pathway, VEGF, JAK-STAT, and Hippo. The systematical analysis showed that various active compounds of JPFR were closely connected with Wnt/β-catenin, EGFR, HIF-1, TGFβ/Smads, and IL6-STAT3 signaling pathway, including kaempferol, isorhamnetin, calycosin, quercetin, medicarpin, phaseol, spinasterol, hederagenin, beta-sitosterol, wighteone, luteolin, and isotrifoliol. For in vitro experiments, the migration and growth of human CRC cells were inhibited by the JPFR extract in a dose-dependent way, and the expression of MALAT1, PTBP-2, β-catenin, MMP7, c-Myc, and Cyclin D1 in CRC cells were downregulated by the JPFR extract in a dose-dependent way. For in vivo metastasis experiments, the numbers of lung metastasis were found to be decreased by the JPFR extract in a dose-dependent manner, and the expressions of metastasis-associated genes including MALAT1, PTBP-2, β-catenin, and MMP7 in the lung metastases were downregulated dose dependently by the JPFR extract. For the orthotopic transplanted tumor experiments, the JPFR extract could inhibit the growth of orthotopic transplanted tumors and downregulate the expression of c-Myc and Cyclin D1 in a dose-dependent manner. Moreover, the JPFR extract could prolong the survival time of tumor-bearing mice in a dose-dependent manner. Conclusions. Through effective network pharmacology analysis, we found that JPFR contains many effective compounds which may directly target cancer-associated signaling pathways. The in vitro and in vivo experiments further confirmed that JPFR could inhibit the growth and metastasis of CRC cells by regulating β-catenin signaling-associated genes or proteins.


Drug Research ◽  
2017 ◽  
Vol 68 (03) ◽  
pp. 153-158 ◽  
Author(s):  
Rahmaniah Rahmaniah ◽  
Yuyuntia Yuyuntia ◽  
Vivian Soetikno ◽  
Wawaimuli Arozal ◽  
Radiana Antarianto ◽  
...  

Abstract Background Alpha mangostin has been reported to have activity for the treatment of liver fibrosis in the rats. However, the mechanisms of action are poorly understood. This study was aimed to investigate the effect of alpha mangostin on hepatic stellate cells (HSC) activation and proliferation through TGF-β/Smad and Akt signaling pathways. Methods Immortalized HSC, LX2 cells, were incubated with TGF-β with or without alpha mangostin (5 or 10 μM). Sorafenib 10 µM was used as positive control. LX2 viability was counted using trypan blue exclusion method. The effect of alpha mangostin on TGF-β concentrations, and the expressions of proliferation and fibrogenic markers were evaluated. Results Alpha mangostin treatment resulted in a reduced proliferation of HSC, decreased Ki-67 and p-Akt expressions. These findings were followed with decreased concentrations of TGF-β in the medium of cells treated with alpha mangostin, decreased expressions of COL1A1, TIMP1, PAI1, α-SMA, and p-Smad3 as fibrogenic markers. These effects were shown to be dose-dependent. Conclusions Alpha mangostin inhibits hepatic stellate cells proliferation and activation through TGF-β/Smad and Akt signaling pathways in dose dependent manner.


Sign in / Sign up

Export Citation Format

Share Document