Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Int J Oncol ; 62(2)2023 Feb.
Article in English | MEDLINE | ID: mdl-36579669

ABSTRACT

Genetic alterations drive tumor onset and progression. However, the cross­talk between tumor cells and the benign components of the surrounding stroma can also promote the initiation, progression and metastasis of solid tumors. These cellular and non­cellular stromal components form the tumor microenvironment (TME), which co­evolves with tumor cells. Their dynamic and mutualistic interactions are currently considered to be among the distinctive hallmarks of cancer. Biochemical and physical cues from the TME serve an essential role in regulating tumor onset and progression. They are also associated with resistance to treatment and poor prognosis in patients with cancer. Therefore, a deep understanding of the TME is vital for developing potent anticancer therapeutics and improving patient outcomes. The present review aims to review the biology of both cellular and non­cellular constituents of the TME and novel findings regarding their contribution to core as well as emerging cancer hallmarks. The present review also describes key TME markers that are either targeted in interventional clinical trials or serve as promising potential anticancer therapies. Understanding TME components and their intercellular interactions is key toward identifying the mechanisms of progression and treatment resistance. Such understanding is of utmost significance for personalized and effective cancer therapy strategies.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Tumor Microenvironment/genetics , Neoplasms/pathology
2.
Hum Cell ; 35(1): 23-36, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34761350

ABSTRACT

The tumor microenvironment contributes significantly to tumor initiation, progression, and resistance to chemotherapy. Much of our understanding of the tumor and its microenvironment is developed using various methods of cell culture. Throughout the last two decades, research has increasingly shown that 3D cell culture systems can remarkably recapitulate the complexity of tumor architecture and physiology compared to traditional 2D models. Unlike the flat culture system, these novel models enabled more cell-cell and cell-extracellular matrix interactions. By mimicking in vivo microenvironment, 3D culture systems promise to become accurate tools ready to be used in diagnosis, drug screening, and personalized medicine. In this review, we discussed the importance of 3D culture in simulating the tumor microenvironment and focused on the effects of cancer cell-microenvironment interactions on cancer behavior, resistance, proliferation, and metastasis. Finally, we assessed the role of 3D cell culture systems in the contexts of drug screening. 2D culture system is used to study cancer cell growth, progression, behavior, and drug response. It provides contact between cells and supports paracrine crosstalk between host cells and cancer cells. However, this system fails to simulate the architecture and the physiological aspects of in vivo tumor microenvironment due to the absence of cell-cell/ cell-ECM interactions as well as unlimited access to O2 and nutrients, and the absence of tumor heterogeneity. Recently advanced research has led researchers to generate 3D culture system that can better recapitulate the in vivo environment by providing hypoxic medium, facilitating cell-cell and cell-ECM, interactions, and recapitulating heterogeneity of the tumor. Several approaches are used to maintain and expand cancer cells in 3D culture systems such as tumor spheroids (cell aggregate that mimics the in vivo growth of tumor cells), scaffold-based approaches, bioreactors, microfluidic derives, and organoids. 3D systems are currently used for disease modeling and pre-clinical drug testing.


Subject(s)
Cell Culture Techniques, Three Dimensional/methods , Neoplasms/pathology , Tumor Microenvironment , Antineoplastic Agents/pharmacology , Cell Communication , Cell Proliferation , Disease Progression , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor/methods , Extracellular Matrix , Humans , Neoplasms/diagnosis , Neoplasms/drug therapy , Tumor Cells, Cultured
3.
Pancreas ; 50(8): 1187-1194, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34714283

ABSTRACT

OBJECTIVES: Pancreatic cancer is one of the most aggressive solid cancers and the fourth leading cause of cancer death in men and women. We previously showed that arginine depletion, using arginase I [HuArgI(Co)-PEG5000], selectively triggers cell death by autophagy in PANC-1 pancreatic cancer cells. The mechanism of action of [HuArgI(Co)-PEG5000], however, has remained poorly understood. In this study, we investigated the effects of arginine depletion on PANC-1 cell migration, adhesion, and invasion and determined the main molecular targets, which mediate PANC-1 cell response to treatment with HuArgI(Co)-PEG5000. METHODS: This was done through examining 2-dimensional (2D) cell motility assays (wound healing and time lapse), cell adhesion, and cell invasion assays, as well as immunostaining for focal adhesions and invadopodia in cells without or with the treatment with arginase. RESULTS: We demonstrate that arginine depletion decreases PANC-1 2D cell migration, adhesion, and 3D invasion. Moreover, our data suggest that these effects are mediated by autophagy and subsequent decrease in the activation of members of Ras homolog gene family (Rho) GTPase family. CONCLUSIONS: Altogether, these findings uncover the mechanism of action of [HuArgI(Co)-PEG5000] and highlight the promising and selective anticancer potential for arginine depletion in the treatment of pancreatic cancer cells.


Subject(s)
Arginase/pharmacology , Autophagy/drug effects , Cell Movement/drug effects , Pancreatic Neoplasms/metabolism , Arginine/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Polyethylene Glycols/pharmacology , Recombinant Proteins/pharmacology
4.
Oncol Lett ; 21(2): 163, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33552281

ABSTRACT

Breast cancer is the leading cause of cancer-associated death among women worldwide. Targeting breast cancer cell metastasis is an important therapeutic approach. The MAPK pathway is a key cell signaling pathway that plays a pivotal role in cellular invasion and migration. Numerous studies have identified the MAPK pathway as a way to target cell survival and motility. The present study treated MBA-MD-231 breast cancer cells with anthrax lethal toxin (LeTx), a potent MAPK inhibitor that selectively cleaves and inactivates all MEKs, as a potential therapeutic method to inhibit breast cancer cell migration. LeTx has been demonstrated to affect breast cancer cell migration. Cells treated with LeTx showed a significant decrease in motility, as observed using wound healing and random 2D motility assays. Additionally, cells treated with LeTx showed an increase in adhesion, which would explain the decrease in migration. Pull-down assays examining the activation status of the members of the Rho family of GTPases revealed an increase in RhoA activation accompanied by a decrease in Cdc42 activation following LeTx treatment. Finally, LeTx mediated a decrease in invasion using a Boyden chamber assay, which could be a result of the decrease in Cdc42 activation. The present study reported the effect of LeTx treatment on the migration, adhesion and invasion of breast cancer cells, demonstrating that this effect was associated with the dysregulation of the Rho GTPases, RhoA and Cdc42.

5.
J Ovarian Res ; 14(1): 13, 2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33423701

ABSTRACT

Ovarian carcinoma is the second most common malignancy of the female reproductive system and the leading cause of death from female reproductive system malignancies. Cancer cells have increased proliferation rate and thus require high amounts of amino acids, including arginine. L-arginine is a non-essential amino acid synthesized from L-citrulline by the Arginosuccinate synthetase (ASS1) enzyme. We have previously shown that the ovarian cancer cells, SKOV3, are auxotrophic to arginine, and that arginine deprivation by treatment with the genetically engineered human arginase I (HuArgI (Co)-PEG5000) triggers the death of SKOV3 cells by autophagy. In this study we examine the effect of HuArgI (Co)-PEG5000 on ovarian cancer cell migration and we dissect the mechanism involved. Wound healing assays, 2D random cell migration assays and cell adhesion analysis indicate that arginine deprivation decreases SKOV3 cell migration and adhesion. This effect was mimicked when autophagy was induced through rapamycin and reversed with the autophagy inhibitor chloroquine when autophagy was inhibited. This proved that arginine deprivation leads to the inhibition of cancer cell migration through autophagy, in addition to cell death. In addition, we were able to establish through pull-down assays and reversal experiments, that arginine deprivation-mediated autophagy inhibits cell migration through a direct inhibition of RhoA, member of the Rho family of GTPases. In conclusion, here we identify, for the first time, an autophagy-mediated inhibition of RhoA that plays an important role in regulating ovarian cancer cells motility and adhesion in response to arginine depletion.


Subject(s)
Arginase/metabolism , Arginine/metabolism , Ovarian Neoplasms/genetics , rhoA GTP-Binding Protein/metabolism , Autophagy , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Ovarian Neoplasms/pathology , Transfection
6.
Hum Cell ; 34(1): 152-164, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32979152

ABSTRACT

Deregulating cellular energetics by reprogramming metabolic pathways, including arginine metabolism, is critical for cancer cell onset and survival. Drugs that target the specific metabolic requirements of cancer cells have emerged as promising targeted cancer therapeutics. In this study, we investigate the therapeutic potential of targeting colon cancer cells using arginine deprivation induced by a pegylated cobalt-substituted recombinant human Arginase I [HuArgI (Co)-PEG5000]. Four colon cancer cell lines were tested for their sensitivity to [HuArgI (Co)-PEG5000] as well as for their mechanism of cell death following arginine deprivation. All four cell lines were sensitive to arginine deprivation induced by [HuArgI (Co)-PEG5000]. All cells expressed ASS1 and were rescued from arginine deprivation-induced cytotoxicity by the addition of excess L-citrulline, indicating they are partially auxotrophic for arginine. Mechanistically, cells treated with [HuArgI (Co)-PEG5000] were negative for AnnexinV and lacked caspase activation. Further investigation revealed that arginine deprivation leads to a marked and prolonged activation of autophagy in both Caco-2 and T84 cell lines. Finally, we show that [HuArgI (Co)-PEG5000] causes cell death by sustained activation of autophagy as evidenced by the decrease in cell cytotoxicity upon treatment with chloroquine, an autophagy inhibitor. Altogether, these data demonstrate that colon cancer cells are partially auxotrophic for arginine and sensitive to [HuArgI (Co)-PEG5000]-induced arginine deprivation. They also show that the activation of autophagy does not play protective roles but rather, induces cytotoxicity and leads to cell death.


Subject(s)
Arginase/adverse effects , Arginine/deficiency , Arginine/genetics , Autophagy/genetics , Autophagy/physiology , Cell Death/genetics , Colonic Neoplasms/pathology , Polyethylene Glycols/adverse effects , Arginine/metabolism , Cell Line, Tumor , Humans
7.
Cancer Cell Int ; 20: 150, 2020.
Article in English | MEDLINE | ID: mdl-32390765

ABSTRACT

Arginine is a semi essential amino acid that is used in protein biosynthesis. It can be obtained from daily food intake or synthesized in the body through the urea cycle using l-citrulline as a substrate. Arginine has a versatile role in the body because it helps in cell division, wound healing, ammonia disposal, immune system, and hormone biosynthesis. It is noteworthy that l-arginine is the precursor for the biosynthesis of nitric oxide (NO) and polyamines. In the case of cancer cells, arginine de novo synthesis is not enough to compensate for their high nutritional needs, forcing them to rely on extracellular supply of arginine. In this review, we will go through the importance of arginine deprivation as a novel targeting therapy by discussing the different arginine deprivation agents and their mechanism of action. We will also focus on the factors that affect cell migration and on the influence of arginine on metastases through polyamine and NO.

8.
Invest New Drugs ; 38(1): 10-19, 2020 02.
Article in English | MEDLINE | ID: mdl-30887252

ABSTRACT

In this study, we assess arginine auxotrophy in ovarian cancer cells and attempt to target them using arginine deprivation induced by a pegylated recombinant human Arginase I cobalt [HuArgI (Co)-PEG5000]. Ovarian cancer cells were sensitive to [HuArgI (Co)-PEG5000]-induced arginine deprivation with IC50 values in the low pM range. Addition of excess L-citrulline rescued only one of three cell lines tested, indicating that the majority of cell lines are completely auxotrophic for arginine. The expression pattern of argininosuccinate synthetase (ASS1) confirmed the degree of auxotrophy of ovarian cancer cell lines with completely auxotrophic cells not expressing ASS1 and partially auxotrophic cells expressing the enzyme. Ovarian cancer cell lines were negative for annexinV staining while showing loss of membrane integrity and absence of caspase activation, indicating caspase-independent, non-apoptotic cell death. [HuArgI (Co)-PEG5000]-induced arginine deprivation led to extensive and prolonged activation of autophagy, which proved to be deleterious to cell survival since its inhibition led to a significant decrease in cytotoxicity. This indicates that the activation of autophagy following arginine-deprivation, rather than being protective, mediates cell cytotoxicity leading to death by autophagy.


Subject(s)
Arginase/administration & dosage , Arginine/deficiency , Autophagy , Ovarian Neoplasms/pathology , Polyethylene Glycols/administration & dosage , Apoptosis , Arginase/chemistry , Cell Proliferation , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Polyethylene Glycols/chemistry , Tumor Cells, Cultured
9.
Invest New Drugs ; 38(5): 1236-1246, 2020 10.
Article in English | MEDLINE | ID: mdl-31823161

ABSTRACT

In this study, we examined the sensitivity of pancreatic cancer cells to [HuArgI (Co)-PEG5000]-induced arginine deprivation as well as the mechanisms underlying deprivation-induced cell death. [HuArgI (Co)-PEG5000]-induced arginine deprivation was cytotoxic to all cell lines tested with IC50 values in the pM range at 72 h post-treatment. Three of the five cell lines were rescued by the addition of excess L-citrulline and expressed ASS1, indicating partial arginine auxotrophy. The remaining two cell lines, on the other hand, were not rescued by the addition of L-citrulline and did not express ASS1, indicating complete auxotrophy to arginine. In addition, all cell lines exhibited G0/G1 cell cycle arrest, in the surviving cell fraction, at 72 h following arginine deprivation. Analysis of the type of cell death revealed negative staining for annexin V and a lack of caspase activation in all five cancer cell lines, following treatment, indicating that arginine deprivation leads to caspase-independent, non-apoptotic cell death. Finally, we demonstrated that arginine deprivation leads to a marked activation of autophagy and that inhibition of this autophagy greatly decreases cytotoxicity, indicating that arginine deprivation induces autophagic cell death in pancreatic cancer cells. We have shown that pancreatic cancer cells are auxotrophic for arginine and sensitive to [HuArgI (Co)-PEG5000]-induced arginine deprivation, hence demonstrating that arginine deprivation is a potentially potent and selective treatment for pancreatic cancer. We have also demonstrated that autophagy is activated following arginine-deprivation and that its prolonged activation leads to autophagic cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Arginase/pharmacology , Arginine , Autophagic Cell Death/drug effects , Polyethylene Glycols/pharmacology , Argininosuccinate Synthase/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism
10.
Int J Mol Sci ; 20(23)2019 Nov 29.
Article in English | MEDLINE | ID: mdl-31795337

ABSTRACT

PURPOSE: Colorectal cancer (CRC) is the third most common type of cancer worldwide, and it represents over half of all gastrointestinal cancer deaths. Knowing that cancer cells have a high proliferation rate, they require high amounts of amino acids, including arginine. In addition, several tumor types have been shown to downregulate ASS-1 expression, becoming auxotrophic for arginine. Therefore, Arginine deprivation is one of the promising therapeutic approaches to target cancer cells. This can be achieved through the use of a recombinant human arginase, HuArgI(Co)-PEG5000, an arginine degrading enzyme. METHODS: In this present study, the cytotoxic effect of HuArgI(Co)-PEG5000 on CRC cell lines (HT-29, Caco-2, Sw837) is examined though cytotoxicity assays. Wound healing assays, invasion assays, and adhesion assays were also performed to detect the effect on metastasis. RESULTS: Wound healing and invasion assays revealed a decrease in cell migration and invasion after treatment with arginase. Cells that were treated with arginase also showed a decrease in adhesion, which coincided with a decrease in RhoA activation, demonstrated though the use of a FRET biosensor to detect RhoA activation in a single cell assay, and a decrease in MMP-9 expression. Treating cells with both arginase and L-citrulline, which significantly restores intracellular arginine levels, reversed the effect of HuArgI(Co)-PEG5000 on cell viability, migration, and invasion. CONCLUSION: We can, therefore, conclude that colorectal cancer is partially auxotrophic to arginine and that arginine depletion is a potential selective inhibitory approach for motility and invasion in colon cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Arginase/pharmacology , Arginine/metabolism , Colorectal Neoplasms/drug therapy , Neoplasm Invasiveness/prevention & control , Polyethylene Glycols/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Neoplasm Invasiveness/pathology
11.
Dalton Trans ; 46(35): 11529-11532, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28748239

ABSTRACT

A photochemically dissociating ligand in Ru(bpy)2(dmphen)Cl2 [bpy = 2,2'-bipyridine; dmphen = 2,9-dimethyl-1,10-phenanthroline] was found to be more cytotoxic on the ML-2 Acute Myeloid Leukemia cell line than Ru(bpy)2(H2O)22+ and prototypical cisplatin. Our findings illustrate the potential potency of diimine ligands in photoactivatable Ru(ii) complexes.

12.
Int J Oncol ; 48(5): 1913-20, 2016 May.
Article in English | MEDLINE | ID: mdl-26984023

ABSTRACT

Malignant astrocytomas are highly invasive into adjacent and distant regions of the normal brain. Understanding and targeting cancer cell invasion is an important therapeutic approach. Cell invasion is a complex process that replies on many signaling pathways including the mitogen-activated protein (MAP) kinase (MAPK). In many cell lines, the use of MAPK-targeted drugs proved to be a potential method to inhibit cancer cell motility. In the present study, we use a recombinant anthrax lethal toxin (LeTx), which selectively inhibits the MAPK pathway, in order to target invasion. LeTx proved ineffective on cell survival in astrocytoma (as well as normal cells). However, astrocytoma cells that were treated with LeTx showed a significant decrease in cell motility as seen by wound healing as well as random 2D motility in serum. The cells also showed a decrease in invasion across a collagen matrix. The effect of LeTx on cell migration was mediated though the deregulation of Rho GTPases, which play a role in cell motility. Finally, the effect of LeTx on cell migration and Rho GTPases was mimicked by the inhibition of the MAPK pathway. In this study, we describe for the first time the effect of the LeTx on cancer cell motility and invasion not cell survival making it a potentially selective brain tumor invasion inhibitor.


Subject(s)
Antigens, Bacterial/pharmacology , Astrocytoma/metabolism , Bacterial Toxins/pharmacology , Brain Neoplasms/metabolism , MAP Kinase Signaling System/drug effects , rho GTP-Binding Proteins/metabolism , Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Invasiveness , Recombinant Proteins/pharmacology
13.
Transl Oncol ; 8(5): 347-357, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26500025

ABSTRACT

In this study, we attempt to target both the urokinase plasminogen activator and the mitogen-activated protein kinase pathway in acute myeloid leukemia (AML) cell lines and primary AML blasts using PrAgU2/LF, a urokinase-activated anthrax lethal toxin. PrAgU2/LF was cytotoxic to five out of nine AML cell lines. Cytotoxicity of PrAgU2/LF appeared to be nonapoptotic and was associated with MAPK activation and urokinase activity because all the PrAgU2/LF-sensitive cell lines showed both uPAR expression and high levels of MEK1/2 phosphorylation. Inhibition of uPAR or desensitization of cells to MEK1/2 inhibition blocked toxicity of PrAgU2/LF, indicating requirement for both uPAR expression and MAPK activation for activity. PrAgU2/LF was also cytotoxic to primary blasts from AML patients, with blasts from four out of five patients showing a cytotoxic response to PrAgU2/LF. Cytotoxicity of primary AML blasts was also dependent on uPAR expression and phos-MEK1/2 levels. CD34(+) bone marrow blasts and peripheral blood mononuclear cells lacked uPAR expression and were resistant to PrAgU2/LF, demonstrating the lack of toxicity to normal hematological cells and, therefore, the tumor selectivity of this approach. Dose escalation in mice revealed that the maximal tolerated dose of PrAgU2/LF is at least 5.7-fold higher than that of the wild-type anthrax lethal toxin, PrAg/LF, further demonstrating the increased safety of this molecule. We have shown, in this study, that PrAgU2/LF is a novel, dual-specific molecule for the selective targeting of AML.

14.
Asian Pac J Cancer Prev ; 16(2): 761-7, 2015.
Article in English | MEDLINE | ID: mdl-25684522

ABSTRACT

BACKGROUND: In this study, we used Daucus carota oil extract (DCOE) to target acute myeloid leukemia (AML) cells. All the AML cell lines tested were sensitive to the extract while peripheral mononuclear cells were not. Analysis of mechanism of cell death showed an increase in cells positive for annexinV and for active caspases, indicating that DCOE induces apoptotic cell death in AML. Inhibition of the MAPK pathway decreased sensitivity of AML cells to DCOE, indicating that cytotoxicity may be dependent on its activity. In conclusion, DCOE induces selective apoptosis in AML cells, possibly through a MAPK-dependent mechanism.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Daucus carota/chemistry , Leukemia, Myeloid, Acute/drug therapy , Plant Extracts/pharmacology , Plant Oils/pharmacology , Blotting, Western , Flow Cytometry , Humans , Immunoenzyme Techniques , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Tumor Cells, Cultured
15.
J Neurooncol ; 122(1): 75-85, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25567351

ABSTRACT

In this study, we attempt to target Arginine auxotrophy in glioblastoma multiforme (GBM) cells using a pegylated recombinant human Arginase I cobalt [HuArgI (Co)-PEG5000]. We tested and characterized the activity of HuArgI (Co)-PEG5000 on a panel of 9 GBM cell lines and on human fetal glial cells (SVG-p12). HuArgI (Co)-PEG5000 was cytotoxic to all GBM cells tested. SVG-p12 cells were not sensitive demonstrating the selective cytotoxicity of HuArgI (Co)-PEG5000-induced arginine deprivation. Addition of L-citrulline led to the rescue of 6 GBM cell lines but only at concentrations of 11.4 mM, reflecting the extent of arginine auxotrophy in GBM. The ability of L-citrulline to rescue cells was dependent on the expression of argininosuccinate synthetase-1 (ASS1) with the cells that were not rescued by L-citrulline being negative for ASS1 expression. Knocking-down ASS1 reversed the ability of L-citrulline to rescue GBM cells, further illustrating the dependence of arginine auxotrophy on ASS1 expression. Inhibition of autophagy increased cell sensitivity to HuArgI (Co)-PEG5000 indicating that, following arginine deprivation, autophagy plays a protective role in GBM cells. Analysis of the type of cell death revealed a lack of AnnexinV staining and caspase activation in HuArgI (Co)-PEG5000-treated cells, indicating that arginine deprivation induces caspase-independent, non-apoptotic cell death in GBM. We have shown that GBM cells are auxotrophic for arginine and can be selectively targeted using HuArgI (Co)-PEG5000-induced arginine depletion, thus demonstrating that L-Arginine deprivation is a potent and selective potential treatment for GBM.


Subject(s)
Apoptosis/drug effects , Arginase/pharmacology , Arginine/metabolism , Glioblastoma/pathology , Polyethylene Glycols/pharmacology , Argininosuccinate Synthase/antagonists & inhibitors , Argininosuccinate Synthase/metabolism , Autophagy , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Flow Cytometry , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Recombinant Proteins/metabolism , Tumor Cells, Cultured
16.
Leuk Res ; 37(11): 1565-71, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24018014

ABSTRACT

In this study, we target arginine auxotrophy of AML cell lines using human arginase I cobalt-PEG5000. HuArgI(Co)-PEG5000 was cytotoxic to all AML cell lines tested. Mononuclear cells and CD34(+) blasts were not sensitive demonstrating the selectivity of HuArgI(Co)-PEG5000-induced arginine deprivation. Addition of L-citrulline led to the rescue of five cell lines. The four cell lines that were not rescued by L-citrulline did not express argininosuccinate synthetase-1, indicating complete arginine auxotrophy. Inhibition of autophagy increased cell sensitivity to HuArgI(Co)-PEG5000 demonstrating the protective role of autophagy following arginine deprivation. We have shown that AML can be selectively targeted using HuArgI(Co)-PEG5000-induced arginine depletion.


Subject(s)
Apoptosis/drug effects , Arginase/metabolism , Arginine/metabolism , Autophagy , Leukemia, Myeloid, Acute/pathology , Polyethylene Glycols/chemistry , Recombinant Proteins/metabolism , Argininosuccinate Synthase , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Flow Cytometry , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Tumor Cells, Cultured
17.
Transl Oncol ; 6(1): 25-32, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23418614

ABSTRACT

In this study, we attempt to target the mitogen-activated protein kinase (MAPK) pathway in acute myeloid leukemia (AML) cells using a recombinant anthrax lethal toxin (LeTx). LeTx consists of protective antigen (PrAg) and lethal factor (LF). PrAg binds cells, is cleaved by furin, oligomerizes, binds three to four molecules of LF, and undergoes endocytosis, releasing LF into the cytosol. LF cleaves MAPK kinases, inhibiting the MAPK pathway. We tested potency of LeTx on a panel of 11 human AML cell lines. Seven cell lines showed cytotoxic responses to LeTx. Cytotoxicity of LeTx was mimicked by the specific mitogen-activated protein/extracellular signal-regulated kinase kinase 1/2 (MEK1/2) inhibitor U0126, indicating that LeTx-induced cell death is mediated through the MEK1/2-extracellular signal-regulated kinase (ERK1/2) branch of the MAPK pathway. The four LeTx-resistant cell lines were sensitive to the phosphatidylinositol 3-kinase inhibitor LY294002. Co-treatment of AML cells with both LeTx and LY294002 did not lead to increased sensitivity, showing a lack of additive/synergistic effects when both pathways are inhibited. Flow cytometry analysis of MAPK pathway activation revealed the presence of phospho-ERK1/2 only in LeTx-sensitive cells. Staining for Annexin V/propidium iodide and active caspases showed an increase in double-positive cells and the absence of caspase activation following treatment, indicating that LeTx-induced cell death is caspase-independent and nonapoptotic. We have shown that a majority of AML cell lines are sensitive to the LF-mediated inhibition of the MAPK pathway. Furthermore, we have demonstrated that LeTx-induced cytotoxicity in AML cells is nonapoptotic and dependent on phospho-ERK1/2 levels.

18.
Eur Urol ; 59(5): 747-54, 2011 May.
Article in English | MEDLINE | ID: mdl-21129846

ABSTRACT

BACKGROUND: PRX302 is a prostate specific antigen (PSA)-activated pore-forming protein toxin under development as a targeted approach for improving lower urinary tract symptoms (LUTS) caused by benign prostatic hyperplasia (BPH) without affecting sexual function. OBJECTIVE: To evaluate the safety and efficacy of PRX302 in men with moderate to severe BPH. DESIGN, SETTING, AND PARTICIPANTS: Eligible subjects were refractory, intolerant, or unwilling to undergo medical therapies for BPH and had International Prostate Symptom Score (IPSS) ≥12, a quality of life (QoL) score ≥3, and prostate volumes between 30 and 80 g. Fifteen patients were enrolled in phase 1 studies, and 18 patients entered phase 2 studies. INTERVENTIONS: Subjects received intraprostatic injection of PRX302 into the right and left transition zone via a transperineal approach in an office-based setting. Phase 1 subjects received increasing concentrations of PRX302 at a fixed volume; phase 2 subjects received increasing volumes per deposit at a fixed concentration. MEASUREMENTS: IPSS, QoL, prostate volume, maximum flow rate (Q(max)), International Index of Erectile Function, serum PSA levels, pharmacokinetics, and adverse events were recorded at 30, 60, 90, 180, 270, and 360 d after treatment with PRX302. RESULTS AND LIMITATIONS: Sixty percent of men in the phase 1 study and 64% of men in the phase 2 study treated with PRX302 had ≥30% improvement compared to baseline in IPSS out to day 360. Patients also experienced improvement in QoL and reduction in prostate volume out to day 360. Patients receiving ≥1 ml of PRX302 per deposit had the best response overall. PRX302 had no deleterious effect on erectile function. Adverse events were mild to moderate and transient in nature. The major study limitation was the small sample size. CONCLUSIONS: The promising safety profile and evidence of efficacy in the majority of treated subjects in these phase 1 and 2 studies supports further development of PRX302 as a minimally invasive, targeted treatment for BPH.


Subject(s)
Bacterial Toxins/administration & dosage , Pore Forming Cytotoxic Proteins/administration & dosage , Prostatic Hyperplasia/drug therapy , Urologic Diseases/drug therapy , Aged , Aged, 80 and over , Bacterial Toxins/adverse effects , Bacterial Toxins/pharmacokinetics , Canada , Humans , Injections , Male , Middle Aged , Penile Erection/drug effects , Pore Forming Cytotoxic Proteins/adverse effects , Pore Forming Cytotoxic Proteins/pharmacokinetics , Prostate-Specific Antigen/blood , Prostatic Hyperplasia/complications , Prostatic Hyperplasia/physiopathology , Quality of Life , Severity of Illness Index , Surveys and Questionnaires , Time Factors , Treatment Outcome , Urodynamics/drug effects , Urologic Diseases/etiology , Urologic Diseases/physiopathology
19.
Anticancer Drugs ; 18(7): 809-16, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17581303

ABSTRACT

Native proaerolysin is a channel-forming bacterial protoxin that binds to cell-surface receptors and then is activated by furin or furin-like proteases. We genetically engineered proaerolysin by replacing the furin-cleavage sequence with a prostate-specific antigen-selective sequence. The recombinant modified proaerolysin was expressed and purified from Aeromonas salmonicida in good yields and purity. Recombinant modified proaerolysin had no furin sensitivity and markedly increased prostate-specific antigen sensitivity relative to wild-type proaerolysin. Human prostate cancer cells were significantly more sensitive to recombinant modified proaerolysin in the presence of active prostate-specific antigen when compared with the absence of prostate-specific antigen or the presence of potent prostate-specific antigen inhibitors. Most normal human cells with the exception of prostate and renal epithelial cells showed very low sensitivity to recombinant modified proaerolysin. Our results suggest that recombinant modified proaerolysin is a potent prostate-specific antigen-sensitive protoxin that deserves further development for regional therapy of benign and malignant prostate growths.


Subject(s)
Antineoplastic Agents/pharmacology , Bacterial Toxins/pharmacology , Pore Forming Cytotoxic Proteins/pharmacology , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/drug therapy , Aeromonas salmonicida/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Bacterial Toxins/administration & dosage , Bacterial Toxins/chemistry , Cell Line, Tumor , Chromatography, High Pressure Liquid , Drug Delivery Systems , Drug Screening Assays, Antitumor , Electrophoresis, Polyacrylamide Gel , Genetic Engineering , Humans , Male , Peptide Hydrolases/metabolism , Pore Forming Cytotoxic Proteins/administration & dosage , Pore Forming Cytotoxic Proteins/chemistry , Prodrugs
20.
Clin Cancer Res ; 12(24): 7437-43, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17189417

ABSTRACT

PURPOSE: Anthrax Lethal Toxin (LeTx), composed of protective antigen and lethal factor, catalytically cleaves mitogen-activated protein kinase (MAPK) kinases and inhibits the MAPK signaling pathways. The majority of metastatic melanomas possess the V599E BRAF mutation, which constitutively activates MAPK1/2 signaling. LeTx is cytotoxic to BRAF mutant melanoma cell lines in vitro, whereas most normal cells are resistant to this toxin. In this study, we determine the in vivo potency and safety of systemically administered LeTx. EXPERIMENTAL DESIGN: A s.c. xenograft melanoma model in athymic nude mice was treated with different i.p. doses of LeTx. RESULTS: In this study, we show that in vivo systemic LeTx treatment of s.c. xenograft melanoma tumors in athymic nude mice yields partial and complete tumor regressions with minor toxicity to mice. When animal toxicity was observed, we did not find any histologic evidence of tissue damage. CONCLUSIONS: LeTx is one of the rare targeted agents to produce complete remissions of human melanomas in an animal model and thus warrants further preclinical development.


Subject(s)
Antigens, Bacterial/therapeutic use , Bacterial Toxins/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/adverse effects , Bacterial Toxins/administration & dosage , Bacterial Toxins/adverse effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Subcutaneous , Melanoma/pathology , Mice , Mice, Nude , Skin Neoplasms/pathology , Treatment Outcome , Tumor Burden , Xenograft Model Antitumor Assays/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...