Thread: 3-bromopyruvate
View Single Post
Old 10-30-2009, 09:17 PM   #1
Rich66
Senior Member
 
Rich66's Avatar
 
Join Date: Feb 2008
Location: South East Wisconsin
Posts: 3,431
3-bromopyruvate

Curr Pharm Biotechnol. 2010 Aug;11(5):510-7.
3-bromopyruvate: a new targeted antiglycolytic agent and a promise for cancer therapy.

Ganapathy-Kanniappan S, Vali M, Kunjithapatham R, Buijs M, Syed LH, Rao PP, Ota S, Kwak BK, Loffroy R, Geschwind JF.

LINK

Source

Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.

Abstract

The pyruvate analog, 3-bromopyruvate, is an alkylating agent and a potent inhibitor of glycolysis. This antiglycolytic property of 3-bromopyruvate has recently been exploited to target cancer cells, as most tumors depend on glycolysis for their energy requirements. The anticancer effect of 3-bromopyruvate is achieved by depleting intracellular energy (ATP) resulting in tumor cell death. In this review, we will discuss the principal mechanism of action and primary targets of 3-bromopyruvate, and report the impressive antitumor effects of 3-bromopyruvate in multiple animal tumor models. We describe that the primary mechanism of 3-bromopyruvate is via preferential alkylation of GAPDH and that 3-bromopyruvate mediated cell death is linked to generation of free radicals. Research in our laboratory also revealed that 3-bromopyruvate induces endoplasmic reticulum stress, inhibits global protein synthesis further contributing to cancer cell death. Therefore, these and other studies reveal the tremendous potential of 3-bromopyruvate as an anticancer agent.

PMID:
20420565
[PubMed - indexed for MEDLINE]


J Bioenerg Biomembr. 2007 Feb;39(1):1-12.
The cancer cell's "power plants" as promising therapeutic targets: an overview.

Pedersen PL.
Source

Department of Biological Chemistry, Johns Hopkins University, School of Medicine, 725 North Wolfe Street, Baltimore, Maryland 21205-2185, USA. ppederse@jhmi.edu


LINK


Abstract

This introductory article to the review series entitled "The Cancer Cell's Power Plants as Promising Therapeutic Targets" is written while more than 20 million people suffer from cancer. It summarizes strategies to destroy or prevent cancers by targeting their energy production factories, i.e., "power plants." All nucleated animal/human cells have two types of power plants, i.e., systems that make the "high energy" compound ATP from ADP and P( i ). One type is "glycolysis," the other the "mitochondria." In contrast to most normal cells where the mitochondria are the major ATP producers (>90%) in fueling growth, human cancers detected via Positron Emission Tomography (PET) rely on both types of power plants. In such cancers, glycolysis may contribute nearly half the ATP even in the presence of oxygen ("Warburg effect"). Based solely on cell energetics, this presents a challenge to identify curative agents that destroy only cancer cells as they must destroy both of their power plants causing "necrotic cell death" and leave normal cells alone. One such agent, 3-bromopyruvate (3-BrPA), a lactic acid analog, has been shown to inhibit both glycolytic and mitochondrial ATP production in rapidly growing cancers (Ko et al., Cancer Letts., 173, 83-91, 2001), leave normal cells alone, and eradicate advanced cancers (19 of 19) in a rodent model (Ko et al., Biochem. Biophys. Res. Commun., 324, 269-275, 2004). A second approach is to induce only cancer cells to undergo "apoptotic cell death." Here, mitochondria release cell death inducing factors (e.g., cytochrome c). In a third approach, cancer cells are induced to die by both apoptotic and necrotic events. In summary, much effort is being focused on identifying agents that induce "necrotic," "apoptotic" or apoptotic plus necrotic cell death only in cancer cells. Regardless how death is inflicted, every cancer cell must die, be it fast or slow.

PMID:
17404823
[PubMed - indexed for MEDLINE]






Biochem Biophys Res Commun. 2004 Nov 5;324(1):269-75.
Advanced cancers: eradication in all cases using 3-bromopyruvate therapy to deplete ATP.

Ko YH, Smith BL, Wang Y, Pomper MG, Rini DA, Torbenson MS, Hullihen J, Pedersen PL.
The Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205-2185, USA. yko@jhmi.edu
A common feature of many advanced cancers is their enhanced capacity to metabolize glucose to lactic acid. In a challenging study designed to assess whether such cancers can be debilitated, we seeded hepatocellular carcinoma cells expressing the highly glycolytic phenotype into two different locations of young rats. Advanced cancers (2-3cm) developed and were treated with the alkylating agent 3-bromopyruvate, a lactate/pyruvate analog shown here to selectively deplete ATP and induce cell death. In all 19 treated animals advanced cancers were eradicated without apparent toxicity or recurrence. These findings attest to the feasibility of completely destroying advanced, highly glycolytic cancers.

PMID: 15465013 [PubMed - indexed for MEDLINE]




Mol Oncol. 2008 Jun;2(1):94-101. Epub 2008 Jan 13.
3-Bromopyruvate as inhibitor of tumour cell energy metabolism and chemopotentiator of platinum drugs.

Ihrlund LS, Hernlund E, Khan O, Shoshan MC.
Department of Oncology-Pathology, Cancer Centre Karolinska, Karolinska Institute, S-171 76 Stockholm, Sweden.
Tumour cells depend on aerobic glycolysis for adenosine triphosphate (ATP) production, making energy metabolism an interesting therapeutic target. 3-Bromopyruvate (BP) has been shown by others to inhibit hexokinase and eradicate mouse hepatocarcinomas. We report that similar to the glycolysis inhibitor 2-deoxyglucose (DG), BP rapidly decreased cellular ATP within hours, but unlike DG, BP concomitantly induced mitochondrial depolarization without affecting levels of reducing equivalents. Over 24h, and at equitoxic doses, DG reduced glucose consumption more than did BP. The observed BP-induced loss of ATP is therefore largely due to mitochondrial effects. Cell death induced over 24h by BP, but not DG, was blocked by N-acetylcysteine, indicating involvement of reactive oxygen species. BP-induced cytotoxicity was independent of p53. When combined with cisplatin or oxaliplatin, BP led to massive cell death. The anti-proliferative effects of low-dose platinum were strikingly potentiated also in resistant p53-deficient cells. Together with the reported lack of toxicity, this indicates the potential of BP as a clinical chemopotentiating agent.

PMID: 19383331 [PubMed - indexed for MEDLINE]



J Hepatobiliary Pancreat Sci. 2010 Jul;17(4):405-6. Epub 2009 Nov 5.
Interventional oncology: new options for interstitial treatments and intravascular approaches: targeting tumor metabolism via a loco-regional approach: a new therapy against liver cancer.

Liapi E, Geschwind JF. jfg@jhmi.edu
Source

Division of Cardiovascular and Interventional Radiology, The Russell H Morgan Department of Radiology and Radiological Sciences, The Johns Hopkins Hospital, Baltimore, MD 21287, USA.


FREE TEXT


Abstract

Recent research in tumor metabolism has uncovered cancer-cell-specific pathways that cancer cells depend on for energy production. 3-Bromopyruvate (3-BrPA), a specific alkylating agent and potent ATP inhibitor, has been shown both in vitro and in vivo to disrupt some of these cancer-specific metabolic pathways, thereby leading to the demise of the cancer cells through apoptosis. 3-BrPA has been successfully tested in animal models of liver cancer. For optimal results, 3-BrPA can be delivered intra-arterially, with minimal toxicity to the surrounding hepatic parenchyma. In the era of development of drugs with lower toxicity for the treatment of liver cancer, inhibition of cancer metabolism with 3-BrPA appears to be a very attractive potent novel therapeutic option.

PMID:
19890602
[PubMed - indexed for MEDLINE]

PMCID: PMC3063000

Quote:
3-BrPA has proven to be quite effective at killing tumors in various animal models. An initial study conducted in a rabbit model of liver cancer showed that 3-BrPA acted as an irreversible inhibitor of metabolic enzyme(s) associated with cancer glycolysis [6, 8].

Quote:
In this study, direct intra-arterial infusion of 3-BrPA showed complete tumor destruction, without affecting the surrounding normal liver parenchyma. This is an important finding, in view of the fact that most liver cancers arise in the background of underlying liver disease (cirrhosis) and that liver failure is a major risk of treatment-related morbidity and mortality [10]. Furthermore, this method of delivery of 3-BrPA showed a significant survival benefit when compared to other established intra-arterial treatments in the rabbit Vx-2 model of liver cancer [11]. In addition, FDG–PET imaging has proven quite useful in determining adequate delivery of the drug to the target tumor, in assessing tumor response after therapy with 3-BrPa and in detecting possible tumor recurrence.3-BrPa is now being tested in human cancer cell lines transplanted in animals (xenografts). The results have so far matched those demonstrated in the Vx-2 rabbit model. In the era of development of less toxic drugs for treatment of liver cancer, inhibition of cancer metabolism is an appealing therapeutic option. This brand new anticancer approach is extremely promising and perfectly suited for catheter-based delivery. Clinical trials should be under way within 1 year.





Semin Cancer Biol. 2009 Feb;19(1):17-24. Epub 2008 Dec 3.
Hexokinase-2 bound to mitochondria: cancer's stygian link to the "Warburg Effect" and a pivotal target for effective therapy.

Mathupala SP, Ko YH, Pedersen PL.
Department of Neurological Surgery and Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, United States.
The most common metabolic hallmark of malignant tumors, i.e., the "Warburg effect" is their propensity to metabolize glucose to lactic acid at a high rate even in the presence of oxygen. The pivotal player in this frequent cancer phenotype is mitochondrial-bound hexokinase [Bustamante E, Pedersen PL. High aerobic glycolysis of rat hepatoma cells in culture: role of mitochondrial hexokinase. Proc Natl Acad Sci USA 1977;74(9):3735-9; Bustamante E, Morris HP, Pedersen PL. Energy metabolism of tumor cells. Requirement for a form of hexokinase with a propensity for mitochondrial binding. J Biol Chem 1981;256(16):8699-704]. Now, in clinics worldwide this prominent phenotype forms the basis of one of the most common detection systems for cancer, i.e., positron emission tomography (PET). Significantly, HK-2 is the major bound hexokinase isoform expressed in cancers that exhibit a "Warburg effect". This includes most cancers that metastasize and kill their human host. By stationing itself on the outer mitochondrial membrane, HK-2 also helps immortalize cancer cells, escapes product inhibition and gains preferential access to newly synthesized ATP for phosphorylating glucose. The latter event traps this essential nutrient inside the tumor cells as glucose-6-P, some of which is funneled off to serve as carbon precursors to help promote the production of new cancer cells while much is converted to lactic acid that exits the cells. The resultant acidity likely wards off an immune response while preparing surrounding tissues for invasion. With the re-emergence and acceptance of both the "Warburg effect" as a prominent phenotype of most clinical cancers, and "metabolic targeting" as a rational therapeutic strategy, a number of laboratories are focusing on metabolite entry or exit steps. One remarkable success story [Ko YH, Smith BL, Wang Y, Pomper MG, Rini DA, Torbenson MS, et al. Advanced cancers: eradication in all cases using 3-bromopyruvate therapy to deplete ATP. Biochem Biophys Res Commun 2004;324(1):269-75] is the use of the small molecule 3-bromopyruvate (3-BP) that selectively enters and destroys the cells of large tumors in animals by targeting both HK-2 and the mitochondrial ATP synthasome. This leads to very rapid ATP depletion and tumor destruction without harm to the animals. This review focuses on the multiple roles played by HK-2 in cancer and its potential as a metabolic target for complete cancer destruction.

PMID: 19101634 [PubMed - indexed for MEDLINE]





Invest New Drugs. 2009 Apr;27(2):120-3. Epub 2008 Jun 14.
Specificity of the anti-glycolytic activity of 3-bromopyruvate confirmed by FDG uptake in a rat model of breast cancer.

Buijs M, Vossen JA, Geschwind JF, Ishimori T, Engles JM, Acha-Ngwodo O, Wahl RL, Vali M.
Russell H. Morgan Department of Radiology, and Radiological Sciences, Division of Vascular and Interventional Radiology, 600 North Wolfe Street, Blalock 545, Baltimore, MD 21287, USA.
PURPOSE: To evaluate the anti-glycolytic effects of 3-BrPA on rats bearing RMT mammary tumors, by determining FDG uptake after intravenous administration of the therapeutic dose. MATERIALS AND METHODS: Sixteen rats bearing RMT tumors were treated either with 15 mM 3-BrPA in 2.5 ml of PBS or with 2.5 ml of PBS. After treatment, all rats received FDG and were sacrificed 1 h later. Results: 3-BrPA treatment significantly decreased FDG uptake in tumors by 77% (p = 0.002). FDG uptake did not significantly decrease in normal tissues after treatment. CONCLUSION: Our study showed that 3-BrPA exhibits a strong anti-glycolytic effect on RMT cells implanted in rats.





Science. 2009 Sep 18;325(5947):1555-9. Epub 2009 Aug 6.
Glucose deprivation contributes to the development of KRAS pathway mutations in tumor cells.

Yun J, Rago C, Cheong I, Pagliarini R, Angenendt P, Rajagopalan H, Schmidt K, Willson JK, Markowitz S, Zhou S, Diaz LA Jr, Velculescu VE, Lengauer C, Kinzler KW, Vogelstein B, Papadopoulos N.
Ludwig Center for Cancer Genetics and Therapeutics and Howard Hughes Medical Institute, Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21231, USA.
Tumor progression is driven by genetic mutations, but little is known about the environmental conditions that select for these mutations. Studying the transcriptomes of paired colorectal cancer cell lines that differed only in the mutational status of their KRAS or BRAF genes, we found that GLUT1, encoding glucose transporter-1, was one of three genes consistently up-regulated in cells with KRAS or BRAF mutations. The mutant cells exhibited enhanced glucose uptake and glycolysis and survived in low-glucose conditions, phenotypes that all required GLUT1 expression. In contrast, when cells with wild-type KRAS alleles were subjected to a low-glucose environment, very few cells survived. Most surviving cells expressed high levels of GLUT1, and 4% of these survivors had acquired KRAS mutations not present in their parents. The glycolysis inhibitor 3-bromopyruvate preferentially suppressed the growth of cells with KRAS or BRAF mutations. Together, these data suggest that glucose deprivation can drive the acquisition of KRAS pathway mutations in human tumors.

PMID: 19661383 [PubMed - indexed for MEDLINE]





Cancer. 2009 Oct 15;115(20):4655-66.
Transport by SLC5A8 with subsequent inhibition of histone deacetylase 1 (HDAC1) and HDAC3 underlies the antitumor activity of 3-bromopyruvate.

Thangaraju M, Karunakaran SK, Itagaki S, Gopal E, Elangovan S, Prasad PD, Ganapathy V.
Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, Georgia 30912, USA. mthangaraju@mcg.edu
BACKGROUND: 3-bromopyruvate is an alkylating agent with antitumor activity. It is currently believed that blockade of adenosine triphosphate production from glycolysis and mitochondria is the primary mechanism responsible for this antitumor effect. The current studies uncovered a new and novel mechanism for the antitumor activity of 3-bromopyruvate. METHODS: The transport of 3-bromopyruvate by sodium-coupled monocarboxylate transporter SMCT1 (SLC5A8), a tumor suppressor and a sodium (Na+)-coupled, electrogenic transporter for short-chain monocarboxylates, was studied using a mammalian cell expression and the Xenopus laevis oocyte expression systems. The effect of 3-bromopyruvate on histone deacetylases (HDACs) was monitored using the lysate of the human breast cancer cell line MCF7 and human recombinant HDAC isoforms as the enzyme sources. Cell viability was monitored by fluorescence-activated cell-sorting analysis and colony-formation assay. The acetylation status of histone H4 was evaluated by Western blot analysis. RESULTS: 3-Bromopyruvate is a transportable substrate for SLC5A8, and that transport process is Na+-coupled and electrogenic. MCF7 cells did not express SLC5A8 and were not affected by 3-bromopyruvate. However, when transfected with SLC5A8 or treated with inhibitors of DNA methylation, these cells underwent apoptosis in the presence of 3-bromopyruvate. This cell death was associated with the inhibition of HDAC1/HDAC3. Studies with different isoforms of human recombinant HDACs identified HDAC1 and HDAC3 as the targets for 3-bromopyruvate. CONCLUSIONS: 3-Bromopyruvate was transported into cells actively through the tumor suppressor SLC5A8, and the process was energized by an electrochemical Na+ gradient. Ectopic expression of the transporter in MCF7 cells led to apoptosis, and the mechanism involved the inhibition of HDAC1/HDAC3. Copyright (c) 2009 American Cancer Society.

PMID: 19637353 [PubMed - in process]






Oncogene. 2006 Aug 7;25(34):4777-86.
Hexokinase II: cancer's double-edged sword acting as both facilitator and gatekeeper of malignancy when bound to mitochondria.

Mathupala SP, Ko YH, Pedersen PL.
Department of Neurological Surgery and Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
A key hallmark of many cancers, particularly the most aggressive, is the capacity to metabolize glucose at an elevated rate, a phenotype detected clinically using positron emission tomography (PET). This phenotype provides cancer cells, including those that participate in metastasis, a distinct competitive edge over normal cells. Specifically, after rapid entry of glucose into cancer cells on the glucose transporter, the highly glycolytic phenotype is supported by hexokinase (primarily HK II) that is overexpressed and bound to the outer mitochondrial membrane via the porin-like protein voltage-dependent anion channel (VDAC). This protein and the adenine nucleotide transporter move ATP, newly synthesized by the inner membrane located ATP synthase, to active sites on HK II. The abundant amounts of HK II bind both the ATP and the incoming glucose producing the product glucose-6-phosphate, also at an elevated rate. This critical metabolite then serves both as a biosynthetic precursor to support cell proliferation and as a precursor for lactic acid, the latter exiting cancer cells causing an unfavorable environment for normal cells. Although helping facilitate this chemical warfare, HK II via its mitochondrial location also suppresses the death of cancer cells, thus increasing their possibility for metastasis and the ultimate death of the human host. For these reasons, targeting this key enzyme is currently being investigated in several laboratories in a strategy to develop novel therapies that may turn the tide on the continuing struggle to find effective cures for cancer. One such candidate is 3-bromopyruvate that has been shown recently to eradicate advanced stage, PET positive hepatocellular carcinomas in an animal model without apparent harm to the animals.

PMID: 16892090 [PubMed - indexed for MEDLINE]






(The FASEB Journal. 2007;21:890.6)
© 2007 FASEB


890.6

Effects of the Anti-Tumor Agent 3-Bromopyruvate (3BrPA) on Glycolytic Energy Metabolism

R. Brooks Robey1, Richard Hong2, Lihui Zhong1, Lanfei Feng2 and Hongmei Zhang1 1 Dartmouth/WRJVAMC, 215 N Main St, White River Jct, VT, 05009,
2 UIC/JBVAMC, 820 S Wood St, Chicago, IL, 60612


ABSTRACT
BACKGROUND & METHODS: 3BrPA has been reported to eradicate liver cancer in animals without associated systemic toxicity. The molecular basis of this effect is incompletedly characterized but has been attributed to selective hexokinase (HK) inhibition and ATP depletion. We therefore examined 3BrPA and other alkylating agents - 3-fluoropyruvate (3FPA), iodoacetate (IAA), and iodoacetamide (IAM) - for the ability to alter glycolytic HK and GAPDH activities, lactate accumulation, ATP content, and cell viability in non-transformed renal epithelial cells.
RESULTS: 3BrPA inhibited HK activity in cell-free lysates in a concentration-dependent manner, an effect that was less potently mimicked by IAA (IC50 8 vs 0.7 mM), but not by pyruvate, 3FPA, or IAM. Monothioglycerol had no effect on basal activity but markedly decreased sensitivity to 3BrPA and IAA inhibition. 3BrPA was also non-competitive with Glc and ATP. When examined in intact cells, 3BrPA and IAA reduced ATP content and lactate accumulation at µM concentrations that were orders of magnitude lower than those required for HK inhibition in vitro. Cytotoxic LDH release was only observed following profound ATP depletion but was uniformly higher for IAA. Interestingly, 3BrPA inhibited GAPDH more potently than its classic antagonist IAA in cell-free lysates (IC50 2.5 vs 150 µM).
CONCLUSIONS: 3BrPA inhibits HK activity, presumably via selective alkylation of sulfhydryl groups important for enzymatic function but not involved in Glc or ATP binding. However, 3BrPA glycolytic inhibitory potency correlates better with GAPDH inhibition than with HK inhibition in non-tumor epithelial cells. IAA - but not 3FPA or IAM - can mimic these effects, albeit with greater relative ATP depletion and cytotoxicity that suggest actions additional to those shared with 3BrPA.
Rich66 is offline   Reply With Quote