Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 140(21): 2193-2227, 2022 11 24.
Article in English | MEDLINE | ID: mdl-36001803

ABSTRACT

With the introduction of large-scale molecular profiling methods and high-throughput sequencing technologies, the genomic features of most lymphoid neoplasms have been characterized at an unprecedented scale. Although the principles for the classification and diagnosis of these disorders, founded on a multidimensional definition of disease entities, have been consolidated over the past 25 years, novel genomic data have markedly enhanced our understanding of lymphomagenesis and enriched the description of disease entities at the molecular level. Yet, the current diagnosis of lymphoid tumors is largely based on morphological assessment and immunophenotyping, with only few entities being defined by genomic criteria. This paper, which accompanies the International Consensus Classification of mature lymphoid neoplasms, will address how established assays and newly developed technologies for molecular testing already complement clinical diagnoses and provide a novel lens on disease classification. More specifically, their contributions to diagnosis refinement, risk stratification, and therapy prediction will be considered for the main categories of lymphoid neoplasms. The potential of whole-genome sequencing, circulating tumor DNA analyses, single-cell analyses, and epigenetic profiling will be discussed because these will likely become important future tools for implementing precision medicine approaches in clinical decision making for patients with lymphoid malignancies.


Subject(s)
Lymphoma , Neoplasms , Humans , Lymphoma/diagnosis , Lymphoma/genetics , Lymphoma/therapy , Genomics/methods , Precision Medicine , High-Throughput Nucleotide Sequencing , Clinical Decision-Making
2.
Yeast ; 36(4): 211-221, 2019 04.
Article in English | MEDLINE | ID: mdl-30462852

ABSTRACT

3-Bromopyruvate (3BP) is a small, highly reactive molecule formed by bromination of pyruvate. In the year 2000, the antitumor properties of 3BP were discovered. Studies using animal models proved its high efficacy for anticancer therapy with no apparent side effects. This was also found to be the case in a limited number of cancer patients treated with 3BP. Due to the "Warburg effect," most tumor cells exhibit metabolic changes, for example, increased glucose consumption and lactic acid production resulting from mitochondrial-bound overexpressed hexokinase 2. Such alterations promote cell migration, immortality via inhibition of apoptosis, and less dependence on the availability of oxygen. Significantly, these attributes also make cancer cells more sensitive to agents, such as 3BP that inhibits energy production pathways without harming normal cells. This selectivity of 3BP is mainly due to overexpressed monocarboxylate transporters in cancer cells. Furthermore, 3BP is not a substrate for any pumps belonging to the ATP-binding cassette superfamily, which confers resistance to a variety of drugs. Also, 3BP has the capacity to induce multiple forms of cell death, by, for example, ATP depletion resulting from inactivation of both glycolytic and mitochondrial energy production pathways. In addition to its anticancer property, 3BP also exhibits antimicrobial activity. Various species of microorganisms are characterized by different susceptibility to 3BP inhibition. Among tested strains, the most sensitive was found to be the pathogenic yeast-like fungus Cryptococcus neoformans. Significantly, studies carried out in our laboratories have shown that 3BP exhibits a remarkable capacity to eradicate cancer cells, fungi, and algae.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Pyruvates/pharmacology , Pyruvic Acid/metabolism , Animals , Apoptosis/drug effects , Disease Models, Animal , Fungi/drug effects , Glycolysis , Hexokinase/genetics , Humans , Melanoma/drug therapy , Mitochondria , Multiple Myeloma/drug therapy , Pyruvic Acid/analogs & derivatives
3.
World J Urol ; 36(12): 2065-2071, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29802428

ABSTRACT

INTRODUCTION AND OBJECTIVES: Medical expulsive therapy is based on pharmacologic ureteral relaxation. We hypothesized this concept may facilitate the deployment of the large 16 French (F) ureteral access sheath (UAS) when patients are intentionally pre-treated with oral tamsulosin, i.e., medical impulsive therapy. METHODS: We retrospectively analyzed our experience with UAS deployment during endoscopic-guided percutaneous nephrolithotomy in prone position in patients pre-treated for 1 week with oral tamsulosin with a contemporary untreated cohort. Between January 2015 and September 2016, seventy-seven patients without a pre-existing ureteral stent met inclusion criteria. Demographic data, tamsulosin usage, UAS size, deployment failure, ureteral injuries, stone-free rates, and complications were recorded. Univariate and multivariate analysis was conducted to assess the impact of tamsulosin on deployment of the 16F UAS. RESULTS: There was no statistical difference between the tamsulosin (n = 40) group and non-tamsulosin (n = 37) group in regard to demographic data. The tamsulosin group had a significantly higher percentage of 16F UAS deployment, 87 vs. 43% (p < 0.001), and no significant difference in ureteral injuries (p = 0.228). Univariate and multivariate analysis revealed that tamsulosin significantly increased the odds ratio (9.3 and 19.4, respectively) for successful passage of a 16F UAS. Despite a larger stone volume, there was no significant difference in computed tomography scan complete stone-free rates (29 vs. 42%; p = 0.277) at median post-operative time of only 3 days. CONCLUSIONS: In this retrospective study, 1 week of preoperative tamsulosin was associated with an increase in the deployment of a 16F UAS in patients without preoperative ureteral stent placement.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/therapeutic use , Nephrolithotomy, Percutaneous/methods , Preoperative Care/methods , Stents , Tamsulosin/therapeutic use , Ureteral Calculi/surgery , Urinary Catheterization/methods , Urinary Catheters , Case-Control Studies , Female , Humans , Male , Middle Aged , Multivariate Analysis , Odds Ratio , Patient Positioning , Prone Position , Retrospective Studies
4.
Semin Cancer Biol ; 35 Suppl: S129-S150, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26454069

ABSTRACT

Cancer is a disease characterized by unrestrained cellular proliferation. In order to sustain growth, cancer cells undergo a complex metabolic rearrangement characterized by changes in metabolic pathways involved in energy production and biosynthetic processes. The relevance of the metabolic transformation of cancer cells has been recently included in the updated version of the review "Hallmarks of Cancer", where dysregulation of cellular metabolism was included as an emerging hallmark. While several lines of evidence suggest that metabolic rewiring is orchestrated by the concerted action of oncogenes and tumor suppressor genes, in some circumstances altered metabolism can play a primary role in oncogenesis. Recently, mutations of cytosolic and mitochondrial enzymes involved in key metabolic pathways have been associated with hereditary and sporadic forms of cancer. Together, these results demonstrate that aberrant metabolism, once seen just as an epiphenomenon of oncogenic reprogramming, plays a key role in oncogenesis with the power to control both genetic and epigenetic events in cells. In this review, we discuss the relationship between metabolism and cancer, as part of a larger effort to identify a broad-spectrum of therapeutic approaches. We focus on major alterations in nutrient metabolism and the emerging link between metabolism and epigenetics. Finally, we discuss potential strategies to manipulate metabolism in cancer and tradeoffs that should be considered. More research on the suite of metabolic alterations in cancer holds the potential to discover novel approaches to treat it.


Subject(s)
Carcinogenesis/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Carcinogenesis/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Energy Metabolism/genetics , Epigenesis, Genetic , Humans , Metabolic Networks and Pathways/genetics , Mitochondria/genetics , Mitochondria/pathology , Neoplasms/genetics , Neoplasms/pathology
5.
Molecules ; 21(12)2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27983708

ABSTRACT

This review summarizes the current state of knowledge about the metabolism of cancer cells, especially with respect to the "Warburg" and "Crabtree" effects. This work also summarizes two key discoveries, one of which relates to hexokinase-2 (HK2), a major player in both the "Warburg effect" and cancer cell immortalization. The second discovery relates to the finding that cancer cells, unlike normal cells, derive as much as 60% of their ATP from glycolysis via the "Warburg effect", and the remaining 40% is derived from mitochondrial oxidative phosphorylation. Also described are selected anticancer agents which generally act as strong energy blockers inside cancer cells. Among them, much attention has focused on 3-bromopyruvate (3BP). This small alkylating compound targets both the "Warburg effect", i.e., elevated glycolysis even in the presence oxygen, as well as mitochondrial oxidative phosphorylation in cancer cells. Normal cells remain unharmed. 3BP rapidly kills cancer cells growing in tissue culture, eradicates tumors in animals, and prevents metastasis. In addition, properly formulated 3BP shows promise also as an effective anti-liver cancer agent in humans and is effective also toward cancers known as "multiple myeloma". Finally, 3BP has been shown to significantly extend the life of a human patient for which no other options were available. Thus, it can be stated that 3BP is a very promising new anti-cancer agent in the process of undergoing clinical development.


Subject(s)
Antineoplastic Agents/therapeutic use , Energy Metabolism/drug effects , Glycolysis/drug effects , Hexokinase/metabolism , Oxidative Phosphorylation/drug effects , Pyruvates/therapeutic use , Humans , Liver Neoplasms/drug therapy , Mitochondria/metabolism , Multiple Myeloma/drug therapy
6.
Anticancer Drugs ; 25(6): 673-82, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24557015

ABSTRACT

The small molecule 3-bromopyruvate (3-BP), which has emerged recently as the first member of a new class of potent anticancer agents, was tested for its capacity to kill multiple myeloma (MM) cancer cells. Human MM cells (RPMI 8226) begin to lose viability significantly within 8 h of incubation in the presence of 3-BP. The Km (0.3 mmol/l) for intracellular accumulation of 3-BP in MM cells is 24 times lower than that in control cells (7.2 mmol/l). Therefore, the uptake of 3-BP by MM cells is significantly higher than that by peripheral blood mononuclear cells. Further, the IC50 values for human MM cells and control peripheral blood mononuclear cells are 24 and 58 µmol/l, respectively. Therefore, specificity and selectivity of 3-BP toward MM cancer cells are evident on the basis of the above. In MM cells the transcription levels of the gene encoding the monocarboxylate transporter MCT1 is significantly amplified compared with control cells. The level of intracellular ATP in MM cells decreases by over 90% within 1 h after addition of 100 µmol/l 3-BP. The cytotoxicity of 3-BP, exemplified by a marked decrease in viability of MM cells, is potentiated by the inhibitor of glutathione synthesis buthionine sulfoximine. In addition, the lack of mutagenicity and its superior capacity relative to Glivec to kill MM cancer cells are presented in this study.


Subject(s)
Antineoplastic Agents/pharmacology , Multiple Myeloma/pathology , Pyruvates/pharmacology , Adenosine Triphosphate/metabolism , Antineoplastic Agents/metabolism , Benzamides/pharmacology , Buthionine Sulfoximine/pharmacology , Cell Survival/drug effects , Glutathione/antagonists & inhibitors , Glutathione/biosynthesis , Humans , Imatinib Mesylate , Inhibitory Concentration 50 , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Multiple Myeloma/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Pyruvates/metabolism , Symporters/genetics , Symporters/metabolism , Tumor Cells, Cultured/drug effects
7.
Cell Death Discov ; 10(1): 106, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38429272

ABSTRACT

Pancreatic cancer has a five-year survival rate of only 10%, mostly due to late diagnosis and limited treatment options. In patients with unresectable disease, either FOLFIRINOX, a combination of 5-fluorouracil (5-FU), oxaliplatin and irinotecan, or gemcitabine plus nab-paclitaxel combined with radiation are frontline standard regimens. However, chemo-radiation therapy has shown limited success because patients develop resistance to chemotherapy and/or radiation. In this study, we evaluated the role of pancreatic cancer stem cells (CSC) using OCT4 and SOX2, CSC markers in mouse pancreatic tumor organoids. We treated pancreatic tumor organoids with 4 or 8 Gy of radiation, 10 µM of 5-FU (5-Fluorouracil), and 100 µM 3-Bromopyruvate (3BP), a promising anti-cancer drug, as a single treatment modalities, and in combination with RT. Our results showed significant upregulation of, OCT4, and SOX2 expression in pancreatic tumor organoids treated with 4 and 8 Gy of radiation, and downregulation following 5-FU treatment. The expression of CSC markers with increasing treatment dose exhibited elevated upregulation levels to radiation and downregulation to 5-FU chemotherapy drug. Conversely, when tumor organoids were treated with a combination of 5-FU and radiation, there was a significant inhibition in SOX2 and OCT4 expression, indicating CSC self-renewal inhibition. Noticeably, we also observed that human pancreatic tumor tissues exhibited heterogeneous and aberrant OCT4 and SOX2 expression as compared to normal pancreas, indicating their potential role in pancreatic cancer growth and therapy resistance. In addition, the combination of 5-FU and radiation treatment exhibited significant inhibition of the ß-catenin pathway in pancreatic tumor organoids, resulting in sensitization to treatment and organoid death. In conclusion, our study emphasizes the crucial role of CSCs in therapeutic resistance in PC treatment. We recommend using tumor organoids as a model system to explore the impact of CSCs in PC and identify new therapeutic targets.

8.
Pharmaceutics ; 16(4)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38675105

ABSTRACT

Altered glycolytic metabolism has been associated with chemoresistance in acute myeloid leukemia (AML). However, there are still aspects that need clarification, as well as how to explore these metabolic alterations in therapy. In the present study, we aimed to elucidate the role of glucose metabolism in the acquired resistance of AML cells to cytarabine (Ara-C) and to explore it as a therapeutic target. Resistance was induced by stepwise exposure of AML cells to increasing concentrations of Ara-C. Ara-C-resistant cells were characterized for their growth capacity, genetic alterations, metabolic profile, and sensitivity to different metabolic inhibitors. Ara-C-resistant AML cell lines, KG-1 Ara-R, and MOLM13 Ara-R presented different metabolic profiles. KG-1 Ara-R cells exhibited a more pronounced glycolytic phenotype than parental cells, with a weaker acute response to 3-bromopyruvate (3-BP) but higher sensitivity after 48 h. KG-1 Ara-R cells also display increased respiration rates and are more sensitive to phenformin than parental cells. On the other hand, MOLM13 Ara-R cells display a glucose metabolism profile similar to parental cells, as well as sensitivity to glycolytic inhibitors. These results indicate that acquired resistance to Ara-C in AML may involve metabolic adaptations, which can be explored therapeutically in the AML patient setting who developed resistance to therapy.

9.
Biochem Biophys Res Commun ; 434(2): 322-7, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23541578

ABSTRACT

We have investigated the antifungal activity of the pyruvic acid analogue: 3-bromopyruvate (3-BP). Growth inhibition by 3-BP of 110 strains of yeast-like and filamentous fungi was tested by standard spot tests or microdilution method. The human pathogen Cryptococcus neoformans exhibited a low Minimal Inhibitory Concentration (MIC) of 0.12-0.15 mM 3-BP. The high toxicity of 3-BP toward C. neoformans correlated with high intracellular accumulation of 3-BP and also with low levels of intracellular ATP and glutathione. Weak cytotoxicity towards mammalian cells and lack of resistance conferred by the PDR (Pleiotropic Drug Resistance) network in the yeast Saccharomyces cerevisiae, are other properties of 3-BP that makes it a novel promising anticryptococcal drug.


Subject(s)
Antifungal Agents/pharmacology , Cryptococcus neoformans/drug effects , Pyruvates/pharmacology , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Amphotericin B/pharmacology , Biological Transport , Cryptococcus neoformans/pathogenicity , Fluconazole/pharmacology , Glutathione/metabolism , Microbial Sensitivity Tests , Microbial Viability/drug effects , Time Factors
10.
Adv Healthc Mater ; 12(31): e2301815, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37706285

ABSTRACT

Lipid metabolism and glycolysis play crucial roles in the progression and metastasis of cancer, and the use of 3-bromopyruvate (3-BP) as an antiglycolytic agent has shown promise in killing pancreatic cancer cells. However, developing an effective strategy to avoid chemoresistance requires the ability to probe the interaction of cancer drugs with complex tumor-associated microenvironments (TAMs). Unfortunately, no robust and multiplexed molecular imaging technology is currently available to analyze TAMs. In this study, the simultaneous profiling of three protein biomarkers using SERS nanotags and antibody-functionalized nanoparticles in a syngeneic mouse model of pancreatic cancer (PC) is demonstrated. This allows for comprehensive information about biomarkers and TAM alterations before and after treatment. These multimodal imaging techniques include surface-enhanced Raman spectroscopy (SERS), immunohistochemistry (IHC), polarized light microscopy, second harmonic generation (SHG) microscopy, fluorescence lifetime imaging microscopy (FLIM), and untargeted liquid chromatography and mass spectrometry (LC-MS) analysis. The study reveals the efficacy of 3-BP in treating pancreatic cancer and identifies drug treatment-induced lipid species remodeling and associated pathways through bioinformatics analysis.


Subject(s)
Pancreatic Neoplasms , Tumor Microenvironment , Mice , Animals , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/drug therapy , Microscopy, Fluorescence , Biomarkers , Multimodal Imaging , Spectrum Analysis, Raman
11.
Biochemistry ; 51(7): 1532-46, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22243519

ABSTRACT

The mitochondrial ATP synthase (F(o)F(1)) is one of the most abundant, important, and complex enzymes found in animals and humans. In earlier studies, we used the photosensitive phosphate analogue vanadate (V(i)) to study the enzyme's mechanism in the transition state. Significantly, these studies showed that Mg(2+) plays an important role in transition state formation during ATP synthesis. Additionally, in both MgADP·V(i)-F(1) and MgV(i)-F(1) complexes, photoactivation of orthovanadate (V(i)) induced cleavage at the third residue within the P-loop (GGAGVGKT), i.e., ßA158, suggesting its proximity to the γ-phosphate during transition state formation. However, despite our recent release of the F(1)-ATPase structure containing V(i), the structural details regarding the role of Mg(2+) have remained elusive. Therefore, in this study, we sought to improve our understanding of the essential role of Mg(2+) during transition state formation. We utilized Protein Data Bank structural data representing different conformational intermediates of key steps in ATP synthesis to assemble a database of positional relationships between landmark residues of the catalytic site and the bound ligand. Applying novel bioinformatics methods, we combined the resulting interatomic spatial data with an animated model of the catalytic site to visualize the exact nature of the changes in these positional relationships during ATP synthesis. The results of these studies reported here show that the absence of Mg(2+) results in migration of inorganic phosphate (P(i)) from ßA158 to a more medial position in the P-loop binding pocket, thereby disrupting essential placement and orientation of the P(i) needed to form the transition state structure and therefore MgATP.


Subject(s)
Magnesium/chemistry , Mitochondrial Proton-Translocating ATPases/chemistry , Adenosine Triphosphate/chemistry , Alanine/chemistry , Animals , Catalysis , Catalytic Domain , Computer Simulation , Humans , Hydrolysis , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Water/chemistry
12.
J Bioenerg Biomembr ; 44(1): 155-61, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22359102

ABSTRACT

We have investigated the cytotoxicity in Saccharomyces cerevisiae of the novel antitumor agent 3-bromopyruvate (3-BP). 3-BP enters the yeast cells through the lactate/pyruvate H(+) symporter Jen1p and inhibits cell growth at minimal inhibitory concentration of 1.8 mM when grown on non-glucose conditions. It is not submitted to the efflux pumps conferring Pleiotropic Drug Resistance in yeast. Yeast growth is more sensitive to 3-BP than Gleevec (Imatinib methanesulfonate) which in contrast to 3-BP is submitted to the PDR network of efflux pumps. The sensitivity of yeast to 3-BP is increased considerably by mutations or chemical treatment by buthionine sulfoximine that decrease the intracellular concentration of glutathione.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Antineoplastic Agents, Alkylating/toxicity , Pyruvates/pharmacokinetics , Pyruvates/toxicity , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Antineoplastic Agents, Alkylating/metabolism , Buthionine Sulfoximine/pharmacology , Glutathione/metabolism , Microbial Sensitivity Tests , Monocarboxylic Acid Transporters/metabolism , Pyruvates/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/metabolism , Symporters/metabolism
13.
J Bioenerg Biomembr ; 44(1): 141-53, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22350013

ABSTRACT

Most malignant tumors exhibit the Warburg effect, which consists in increased glycolysis rates with production of lactate, even in the presence of oxygen. Monocarboxylate transporters (MCTs), maintain these glycolytic rates, by mediating the influx and/or efflux of lactate and are overexpressed in several cancer cell types. The lactate and pyruvate analogue 3-bromopyruvate (3-BP) is an inhibitor of the energy metabolism, which has been proposed as a specific antitumor agent. In the present study, we aimed at determining the effect of 3-BP in breast cancer cells and evaluated the putative role of MCTs on this effect. Our results showed that the three breast cancer cell lines used presented different sensitivities to 3-BP: ZR-75-1 ER (+)>MCF-7 ER (+)>SK-BR-3 ER (-). We also demonstrated that 3-BP reduced lactate production, induced cell morphological alterations and increased apoptosis. The effect of 3-BP appears to be cytotoxic rather than cytostatic, as a continued decrease in cell viability was observed after removal of 3-BP. We showed that pre-incubation with butyrate enhanced significantly 3-BP cytotoxicity, especially in the most resistant breast cancer cell line, SK-BR-3. We observed that butyrate treatment induced localization of MCT1 in the plasma membrane as well as overexpression of MCT4 and its chaperone CD147. Our results thus indicate that butyrate pre-treatment potentiates the effect of 3-BP, most probably by increasing the rates of 3-BP transport through MCT1/4. This study supports the potential use of butyrate as adjuvant of 3-BP in the treatment of breast cancer resistant cells, namely ER (-).


Subject(s)
Antineoplastic Agents, Alkylating/metabolism , Breast Neoplasms/metabolism , Butyrates/pharmacology , Gene Expression Regulation, Neoplastic/physiology , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Pyruvates/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Butyrates/metabolism , Butyrates/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Chemotherapy, Adjuvant/methods , Female , Gene Expression Regulation, Neoplastic/drug effects , Glycolysis , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Lactic Acid/antagonists & inhibitors , Pyruvates/pharmacology , Pyruvates/therapeutic use , Tetrazolium Salts , Thiazoles
14.
Am J Cancer Res ; 12(11): 4977-4987, 2022.
Article in English | MEDLINE | ID: mdl-36504891

ABSTRACT

Pancreatic cancer (PC) is the fourth-most-deadly cancer in the United States with a 5-year survival rate of only 8%. The majority of patients with locally advanced pancreatic cancer undergo chemotherapy and/or radiation therapy (RT). However, current treatments are inadequate and novel strategies are desperately required. 3-Bromopyruvate (3-BP) is a promising anticancer drug against pancreatic cancer. It exerts potent anticancer effects by inhibiting hexokinase II enzyme (HK2) of the glycolytic pathway in cancer cells while not affecting the normal cells. 3-BP killed 95% of Panc-2 cells at 15 µM concentration and severely inhibited ATP production by disrupting the interaction between HK2 and mitochondrial Voltage Dependent Anion Channel-1 (VDAC1) protein. Electron microscopy data revealed that 3-BP severely damaged mitochondrial membrane in cancer cells. We further examined therapeutic effect of 3-BP in syngeneic mouse pancreatic cancer model by treating animals with 10, 15 and 20 mg/kg dose. 3-BP at 15 & 20 mg/kg dose level significantly reduced tumor growth by approximately 75-80% in C57BL/6 female mice. Immunohistochemistry data showed complete inhibition of hexokinase II (HK2) and TGFß, in animals treated with 3-BP drug. We also observed enhanced expression of active caspase-3 in tumor tissues exhibited apoptotic death. Flow Cytometry analysis showed significant inhibition in MDSC (CD11b) population in treated tumor which may have allowed infiltration of CD8+ T cells and inhibited tumor growth. Notably, metabolomic data also revealed severe inhibition in glycolysis, NADP, ATP and lactic acid production in cancer cells treated with 40 µM 3-BP. Importantly, we also observed inhibition in lactic acid production responsible for tumor aggression. These results provide new evidence that 3-BP severely inhibit glucose metabolism in cancer cells by blocking hexokinase II, and disrupting mitochondria by suppressing BCL2L1 in pancreatic cancer.

15.
Biochim Biophys Acta ; 1797(6-7): 1225-30, 2010.
Article in English | MEDLINE | ID: mdl-20381449

ABSTRACT

Tumors usurp established metabolic steps used by normal tissues for glucose utilization and ATP production that rely heavily on mitochondria and employ a route that, although involving mitochondria, includes a much greater dependency on glycolysis. First described by Otto Warburg almost nine decades ago [1], this aberrant phenotype becomes more pronounced with increased tumor malignancy [2]. Thus, while maintaining their capacity for respiration, tumors "turn more parasitic" by enhancing their ability to scavenge glucose from their surroundings. With excess glucose at hand, tumors shunt their metabolic flux more toward glycolysis than do their normal cells of origin, a strategy that allows for their survival when oxygen is limiting while providing them a mechanism to poison their extra-cellular environment with acid, thus paving the way for invasion and metastasis. Significantly, tumors harness a crucial enzyme to regulate and support this destructive path--to entrap and channel glucose toward glycolysis. This enzyme is an isoform of hexokinase, referred to as hexokinase type II, and also in abbreviated form as HK-2 or HK II. Due to many-faceted molecular features at genetic, epigenetic, transcriptional, and enzymatic levels, including sub-cellular localization to mitochondria, HK-2 facilitates and promotes the high glycolytic tumor phenotype [3]. Thus, HK-2 represents a pivotal model gene or enzyme that tumors "select for" during tumorigenesis in order to facilitate their destructive path. In this review, we examine the roles played by mitochondrial bound HK-2 within the context of the highly choreographed metabolic roulette of malignant tumors. Recent studies that outline how the aberrant glycolytic flux can be subverted toward a more "normal" metabolic phenotype, and how the glycolytic flux affects the tumor microenvironment to facilitate tumor dissemination are also described, including how these very features can be harnessed in new metabolic targeting strategies to selectively debilitate tumors.


Subject(s)
Mitochondria/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Animals , Glucose/metabolism , Glycolysis , Hexokinase/genetics , Hexokinase/metabolism , History, 20th Century , History, 21st Century , Humans , Lactic Acid , Models, Biological , Monocarboxylic Acid Transporters/metabolism , Neoplasms/genetics , Neoplasms/pathology , Proton Pumps/metabolism , Research/history
16.
BJU Int ; 108(6 Pt 2): 1007-17, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21917104

ABSTRACT

• Historically, the ideal outcome of radical prostatectomy (RP) has been measured by achievement of the so-called 'trifecta', or the concurrent attainment of continence and potency with no evidence of biochemical recurrence. However, in the PSA era, younger and healthier men are more frequently diagnosed with prostate cancer. Such patients have higher expectations from the advanced minimally invasive surgical technologies. Mere trifecta is no longer an ideal outcome measure to meet the demands of such patients. • Keeping the limitations of trifecta in mind, we have earlier proposed a new method of outcomes analysis, called the 'pentafecta', which adds early complications and positive surgical margins (PSMs) to trifecta. • We performed a Medline search for articles reporting the complications, PSM rates, continence, potency and biochemical recurrence after robot-assisted RP. Related articles were selected and individual outcomes were reviewed.


Subject(s)
Erectile Dysfunction/etiology , Laparoscopy/adverse effects , Prostatectomy/adverse effects , Prostatic Neoplasms/surgery , Robotics , Urinary Incontinence/etiology , Disease-Free Survival , Humans , Laparoscopy/methods , Male , Middle Aged , Minimally Invasive Surgical Procedures/adverse effects , Minimally Invasive Surgical Procedures/methods , Outcome and Process Assessment, Health Care , Postoperative Complications/etiology , Prostatectomy/methods , Surgery, Computer-Assisted/adverse effects , Surgery, Computer-Assisted/methods , Treatment Outcome
17.
Semin Cancer Biol ; 19(1): 17-24, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19101634

ABSTRACT

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.


Subject(s)
Glucose/metabolism , Hexokinase/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Pyruvates/metabolism , Adenosine Triphosphate/metabolism , Animals , Energy Metabolism/physiology , Gene Expression Regulation, Neoplastic/genetics , Glycolysis/physiology , Humans , Mitochondria/enzymology , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Phosphorylation/physiology , Protein Isoforms/metabolism , Pyruvates/therapeutic use
18.
Circ Res ; 99(7): 706-14, 2006 Sep 29.
Article in English | MEDLINE | ID: mdl-16946135

ABSTRACT

Ischemic preconditioning is characterized by resistance to ischemia reperfusion injury in response to previous short ischemic episodes, a protective effect that can be mimicked pharmacologically. The underlying mechanism of protection remains controversial and requires greater understanding before it can be fully exploited therapeutically. To investigate the overall effect of preconditioning on the myocardial proteome, isolated rabbit ventricular myocytes were treated with drugs known to induce preconditioning, adenosine or diazoxide (each at 100 micromol/L for 60 minutes). Their protein profiles were then compared with vehicle-treated controls (n=4 animals per treatment) using a multitiered 2D gel electrophoresis approach. Of 28 significantly altered protein spots, 19 nonredundant proteins were identified (5 spots remained unidentified). The majority of these proteins are involved in mitochondrial energetics, including subunits of tricarboxylic acid cycle enzymes and oxidative phosphorylation complexes. These changes were not indiscriminate, with only a small number of enzymes or complex subunits altered, indicating a very specific and targeted affect of these 2 preconditioning mimetics. Among the changes were shifts in the extent of posttranslational modification of 4 proteins. One of these, the adenosine-induced phosphorylation of the ATP synthase beta subunit, was fully characterized with the identification of 5 novel phosphorylation sites. This proteomics approach provides an overall assessment of the cellular response to pharmacological treatment with adenosine and diazoxide and identifies a distinct subset of enzymes and protein complex subunit that may underlie the preconditioned phenotype.


Subject(s)
Adenosine/pharmacology , Diazoxide/pharmacology , Ischemic Preconditioning, Myocardial/methods , Myocardium/metabolism , Proteome/metabolism , Animals , Heart Ventricles , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Protein Processing, Post-Translational , Proteomics , Rabbits
19.
Pathol Int ; 58(4): 209-17, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18324913

ABSTRACT

EBV-associated T/natural killer (NK)-cell lymphoproliferative disorder (EBV-T/NK LPD) of children and young adults is generally referred to with the blanket nosological term of severe chronic active EBV infection (CAEBV). This disease is rare, associated with high morbidity and mortality, and appears to be more prevalent in East Asian countries. But because there is no grading or categorization system for CAEBV, pathologists and clinicians often disagree regarding diagnosis and therapy. EBV-T/NK LPD includes polyclonal, oligoclonal, and monoclonal proliferation of cytotoxic T and/or NK cells. Moreover, a unique disease previously described as infantile fulminant EBV-associated T-LPD has been identified and overlaps with EBV-T/NK LPD. In the present review a clinicopathological categorization of EBV-T/NK LPD is proposed, based on pathological evaluation and molecular data, as follows: (i) category A1, polymorphic LPD without clonal proliferation of EBV-infected cells; (ii) category A2, polymorphic LPD with clonality; (iii) category A3, monomorphic LPD (T-cell or NK cell lymphoma/leukemia) with clonality; and (iv) category B, monomorphic LPD (T-cell lymphoma) with clonality and fulminant course. Categories A1, A2, and A3 possibly constitute a continuous spectrum and together are equivalent to CAEBV. Category B is the exact equivalent of infantile fulminant EBV-associated T-LPD. It is expected that this categorization system will provide a guide for the better understanding of this disorder. This proposal was approved at the third meeting of the Asian Hematopathology Association (Nagoya, 2006).


Subject(s)
Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/isolation & purification , Killer Cells, Natural/pathology , Lymphoproliferative Disorders/pathology , T-Lymphocytes/pathology , Adolescent , Adult , Child , Child, Preschool , Epstein-Barr Virus Infections/classification , Epstein-Barr Virus Infections/virology , Female , Herpesvirus 4, Human/genetics , Humans , Infant , Killer Cells, Natural/virology , Lymphoproliferative Disorders/classification , Lymphoproliferative Disorders/virology , Male , T-Lymphocytes/virology
20.
Med Oncol ; 25(4): 447-50, 2008.
Article in English | MEDLINE | ID: mdl-18431552

ABSTRACT

We assessed the quality of life (QOL) at least one year after sequential chemoradiotherapy for the treatment of localized gastric diffuse large B-cell lymphoma (DLBCL). We used the EORTC Quality of Life Questionnaire for Stomach Cancer (EORTC QLQ-STO22). Among the 45 patients available at the one-year follow-up after radiation therapy, 40 patients completed the EORTC QLQ-STO22 questionnaire. Their median age was 54.5 (range, 20-70 years). Social functioning was most adversely affected among the respondents with a score of 59, whereas other functions and the global scales were preserved above a score of 70 by linearly transformed values. Fatigue, the financial impact and specific emotional problems such as "thinking about their illness" (STO-ANX) and "worry about weight loss or future health" (STO-BI) were persistently bothersome for some patients. Other stomach-related symptoms such as dysphagia, pain, or reflux were negligible at 1 year after treatment. Therefore, this organ-preserving combined approach was effective for the maintenance of the QOL and minimization of stomach abnormalities in patients with gastric lymphoma.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/psychology , Lymphoma, Large B-Cell, Diffuse/therapy , Quality of Life , Stomach Neoplasms/psychology , Stomach Neoplasms/therapy , Adolescent , Adult , Aged , Antineoplastic Agents/therapeutic use , Combined Modality Therapy , Female , Humans , Male , Middle Aged , Radiotherapy , Surveys and Questionnaires
SELECTION OF CITATIONS
SEARCH DETAIL