Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Cancers (Basel) ; 16(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38254853

ABSTRACT

Dabrafenib therapy for metastatic melanoma focuses on blocking growth-promoting signals produced by a hyperactive BRAF protein. We report the metabolic differences of four human melanoma cell lines with diverse responses to dabrafenib therapy (30 mg/kg; oral): WM3918 < WM9838BR < WM983B < DB-1. Our goal was to determine if metabolic changes produced by the altered signaling pathway due to BRAF mutations differ in the melanoma models and whether these differences correlate with response to treatment. We assessed metabolic changes in isolated cells using high-resolution proton magnetic resonance spectroscopy (1H MRS) and supplementary biochemical assays. We also noninvasively studied mouse xenografts using proton and phosphorus (1H/31P) MRS. We found consistent changes in lactate and alanine, either in isolated cells or mouse xenografts, correlating with their relative dabrafenib responsiveness. In xenografts, we also observed that a more significant response to dabrafenib correlated with higher bioenergetics (i.e., increased ßNTP/Pi). Notably, our noninvasive assessment of the metabolic status of the human melanoma xenografts by 1H/31P MRS demonstrated early metabolite changes preceding therapy response (i.e., tumor shrinkage). Therefore, this noninvasive methodology could be translated to assess in vivo predictive metabolic biomarkers of response in melanoma patients under dabrafenib and probably other signaling inhibition therapies.

2.
Leukemia ; 37(12): 2436-2447, 2023 12.
Article in English | MEDLINE | ID: mdl-37773266

ABSTRACT

As we show in this study, NAMPT, the key rate-limiting enzyme in the salvage pathway, one of the three known pathways involved in NAD synthesis, is selectively over-expressed in anaplastic T-cell lymphoma carrying oncogenic kinase NPM1::ALK (ALK + ALCL). NPM1::ALK induces expression of the NAMPT-encoding gene with STAT3 acting as transcriptional activator of the gene. Inhibition of NAMPT affects ALK + ALCL cells expression of numerous genes, many from the cell-signaling, metabolic, and apoptotic pathways. NAMPT inhibition also functionally impairs the key metabolic and signaling pathways, strikingly including enzymatic activity and, hence, oncogenic function of NPM1::ALK itself. Consequently, NAMPT inhibition induces cell death in vitro and suppresses ALK + ALCL tumor growth in vivo. These results indicate that NAMPT is a novel therapeutic target in ALK + ALCL and, possibly, other similar malignancies. Targeting metabolic pathways selectively activated by oncogenic kinases to which malignant cells become "addicted" may become a novel therapeutic approach to cancer, alternative or, more likely, complementary to direct inhibition of the kinase enzymatic domain. This potential therapy to simultaneously inhibit and metabolically "starve" oncogenic kinases may not only lead to higher response rates but also delay, or even prevent, development of drug resistance, frequently seen when kinase inhibitors are used as single agents.


Subject(s)
Lymphoma, Large-Cell, Anaplastic , Receptor Protein-Tyrosine Kinases , Humans , Receptor Protein-Tyrosine Kinases/metabolism , Anaplastic Lymphoma Kinase/metabolism , Lymphoma, Large-Cell, Anaplastic/genetics , Signal Transduction , Nuclear Proteins/genetics , Cell Line, Tumor
3.
RSC Adv ; 13(29): 19813-19816, 2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37404315

ABSTRACT

Lonidamine (LND) is an anti-cancer drug with great potential as a metabolic modulator of chemotherapy, radiotherapy, hyperthermia, and photodynamic therapy in cancer treatment. LND affects several important aspects of cancer cell metabolism: it inhibits Complex I and II of the electron transport chain (ETC) and pyruvate carriers (mitochondrial), and monocarboxylate transporters in the plasma membrane of the cell. Cancer cells are affected by changes in pH on the molecular level, and so are the drugs used to treat cancer, thus it is important to understand how pH affects their structures and LND is no exception. LND dissolves at a pH of 8.3 in tris-glycine buffer but has limited solubility at pH 7. To understand how pH affects the structure of LND, and its effect as a metabolic modulator on cancer therapy, we made up samples of LND at pH 2, pH 7, and pH 13, and analyzed these samples using 1H and 13C NMR. We looked for ionization sites to explain the behavior of LND in solution. Our results showed considerable chemical shifts between the extremes of our experimental pH range. LND was ionized at its indazole α-nitrogen, however, we did not directly observe the protonation of the carboxyl group oxygen that is expected at pH 2, which may be the result of a chemical-exchange phenomenon.

4.
NMR Biomed ; 36(4): e4716, 2023 04.
Article in English | MEDLINE | ID: mdl-35196744

ABSTRACT

Bonded cumomers are sets of isotopomers of 13 C-labeled metabolites containing a particular sequence of contiguously or singly labeled carbon atoms. Only these isotopomers contribute to multiplet structure in the 13 C NMR spectrum. We discuss the application of this technique to the study of quantitative tumor metabolism, bioenergetics, and the Warburg effect. The advantages and sensitivity of bonded cumomer analysis over positional enrichment analysis are discussed. When sensitivity requirements are met, bonded cumomer analysis enables the extraction of fluxes through specific metabolic pathways with higher precision. In conjunction with isotopomer control analysis, we evaluate the sensitivity of experimentally measurable metabolite multiplets to determine the robustness of flux analysis in 13 C spectra of tumors. This review examines the role of glycolytic and tricarboxylic acid cycle metabolism with special emphasis on flux through the pentose phosphate pathway (PPP). The impact of reversibility of the nonoxidative branch of the PPP with various 13 C glucose tracers on fine-structure multiplets is analyzed.


Subject(s)
Models, Biological , Neoplasms , Humans , Magnetic Resonance Spectroscopy/methods , Energy Metabolism , Citric Acid Cycle , Glucose/metabolism , Carbon Isotopes/metabolism
5.
Magn Reson Imaging ; 94: 127-143, 2022 12.
Article in English | MEDLINE | ID: mdl-36089181

ABSTRACT

1H magnetic resonance spectroscopy (MRS) with the multiple quantum coherence (MQC) technique allows for the detection of lactate, an end product of glycolysis, in the environment of lipids. The method can also be used to detect alanine, a byproduct of glutaminolysis. An issue is that when both lactate and alanine are detected together by the MQC technique, a phase mismatch arises between lactate and alanine signals due to off-resonance rotations and the difference in double quantum coherence frequencies between the two molecules. Such phase mismatch can cause errors in spectral fitting and metabolite quantification. In this study, we designed two pulse sequences that eliminate such phase differences of lactate and alanine while suppressing lipid signals by modifications of the Selective Multiple Quantum Coherence (Sel-MQC) sequence, a well-known MQC technique. Using the product operator formalism and the off-resonance rotation matrices, the phase evolutions of lactate and alanine during the spectrally selective pulses and the free precession times of the sequence at the single quantum, double quantum and zero quantum coherence states of these molecules were calculated. The multiple quantum (MQ) evolution time t1 that can remove the phase difference of lactate and alanine at the echo was calculated and fine-tuned with experiments. The lactate and alanine signal intensities and the editing efficiencies from the two modified Sel-MQC sequences were theoretically predicted by using the product operator evolutions and compared with the experimental data. The J-coupled lipid signals were successfully suppressed by both sequences. One of the two developed sequences was applied to a human body with a phantom of lactate and alanine, which resulted in successful in-phase editing of lactate and alanine and suppression of the lipid signals from the body. The study sets an important foundation for the noninvasive detection of lactate and alanine from tumors of cancer patients.


Subject(s)
Alanine , Lactic Acid , Humans , Magnetic Resonance Spectroscopy/methods , Magnetic Resonance Imaging/methods , Lipids/chemistry
6.
Biomed Phys Eng Express ; 8(3)2022 03 08.
Article in English | MEDLINE | ID: mdl-35193126

ABSTRACT

Objective.The selective multiple quantum coherence (Sel-MQC) sequence is a magnetic resonance spectroscopy (MRS) technique used to detect lactate and suppress co-resonant lipid signalsin vivo. The coherence pathways of J-coupled lipids upon the sequence, however, have not been studied, hindering a logical design of the sequence to fully attenuate lipid signals. The objective of this study is to elucidate the coherence pathways of J-coupled lipids upon the Sel-MQC sequence and find a strategy to effectively suppress lipid signals from these pathways while keeping the lactate signal.Approach.The product operator formalism was used to express the evolutions of the J-coupled spins of lipids and lactate. The transformations of the product operators by the spectrally selective pulses of the sequence were calculated by using the off-resonance rotation matrices. The coherence pathways and the conversion rates of the individual pathways were derived from them. Experiments were performed on phantoms and two human subjects at 3 T.Main results.The coherence pathways contributing to the various lipid resonance signals by the Sel-MQC sequence depending on the gradient ratios and RF pulse lengths were identified. Theoretical calculations of the signals from the determined coherence pathways and signal attenuations by gradients matched the experimental data very well. Lipid signals from fatty tissues of the subjects were successfully suppressed to the noise level by using the gradient ratio -0.8:-1:2 or 1:0.8:2. The new gradient ratios kept the lactate signal the same as with the previously used gradient ratio 0:-1:2.Significance.The study has elucidated the coherence pathways of J-coupled lipids upon the Sel-MQC sequence and demonstrated how lipid signals can be effectively suppressed while keeping lactate signals by using information from the coherence pathway analysis. The findings enable applying the Sel-MQC sequence to lactate detection in an environment of high concentrations of lipids.


Subject(s)
Lactic Acid , Magnetic Resonance Imaging , Humans , Lactic Acid/analysis , Lactic Acid/chemistry , Lactic Acid/metabolism , Lipids/chemistry , Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy/methods , Phantoms, Imaging
7.
Front Oncol ; 9: 568, 2019.
Article in English | MEDLINE | ID: mdl-31334109

ABSTRACT

While studies have identified a number of mutations in mantle cell lymphoma (MCL), the list may still be incomplete and contribution to the pathogenesis remains unclear. We analyzed the mutational landscape of four mantle cell lymphoma biopsies obtained during an 8-year period from the same patient with his normal cells serving as control; we also established a cell line from the final stage of the disease. Numerous mutations with high allelic burden have been identified in all four biopsies. While a large subset of mutations was seen only in individual biopsies, the core of 21 mutations persisted throughout the disease. This mutational core is also maintained in the cell line that also displays DNA-methylation and cytokine secretion profiles of the primary mantle cell lymphoma cells. This cell line is uniquely sensitive to clinically relevant inhibitors of Bruton's Tyrosine Kinase. The response to Bruton Tyrosine Kinase's inhibition is enhanced by inhibitors of CDK4/6 and mTOR. Among the mutations seen in the primary and cultured MCL cells, mutations of three genes are involved in the control of H3K4 methylation: demethylase KDM5C, present already in the early disease, and methyltransferase KMT2D and cofactor BCOR, both of which are seen late in the disease and are novel and predicted to be pathogenic. The presence of these mutations was associated with hypermethylation of H3K4. Restoration of KDM5C expression affected expression of numerous genes involved in cell proliferation, adherence/movement, and invasiveness.

8.
Mol Cancer Res ; 17(6): 1365-1377, 2019 06.
Article in English | MEDLINE | ID: mdl-30862686

ABSTRACT

Current methods to evaluate effects of kinase inhibitors in cancer are suboptimal. Analysis of changes in cancer metabolism in response to the inhibitors creates an opportunity for better understanding of the interplay between cell signaling and metabolism and, from the translational perspective, potential early evaluation of response to the inhibitors as well as treatment optimization. We performed genomic, metabolomic, and fluxomic analyses to evaluate the mechanism of action of the Bruton's tyrosine kinase (BTK) inhibitor ibrutinib (IBR) in mantle cell lymphoma (MCL) cells. Our comprehensive analysis of the data generated by these diverse technologies revealed that IBR profoundly affected key metabolic pathways in IBR-sensitive cells including glycolysis, pentose phosphate pathway, TCA cycle, and glutaminolysis while having much less effects on IBR-poorly responsive cells. Changes in 1H magnetic resonance spectroscopy (MRS)-detectable lactate and alanine concentrations emerged as promising biomarkers of response and resistance to IBR as demonstrated from experiments on various MCL cell lines. The metabolic network analysis on the 13C MRS and 13C LC/MS experimental data provided quantitative estimates of various intracellular fluxes and energy contributions. Glutaminolysis contributed over 50% of mitochondrial ATP production. Administration of the glutaminase inhibitor CB-839 induced growth suppression of the IBR-poorly responsive cells. IMPLICATIONS: Our study demonstrates application of the advanced metabolomic/fluxomic techniques for comprehensive, precise, and prompt evaluations of the effects of kinase inhibition in MCL cells and has strong translational implications by potentially permitting early evaluation of cancer patient response versus resistance to kinase inhibitors and on design of novel therapies for overcoming the resistance.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/metabolism , Metabolic Networks and Pathways/physiology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Adenine/analogs & derivatives , Benzeneacetamides/pharmacology , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Glutaminase/metabolism , Humans , Metabolic Networks and Pathways/drug effects , Piperidines , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Thiadiazoles/pharmacology
9.
Sci Rep ; 8(1): 14654, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30279592

ABSTRACT

Lonidamine (LND), a metabolic modulator, sensitizes DB-1 human melanoma to doxorubicin (DOX) chemotherapy by acidifying and de-energizing the tumor. This report compares the effects of LND on two human melanoma lines, DB-1 and WM983B, which exhibit different metabolic properties. Using liquid chromatography mass spectrometry and Seahorse analysis, we show that DB-1 was more glycolytic than WM983B in vitro. 31P magnetic resonance spectroscopy (MRS) indicates that LND (100 mg/kg, i.p.) induces similar selective acidification and de-energization of WM983B xenografts in immunosuppressed mice. Over three hours, intracellular pH (pHi) of WM983B decreased from 6.91 ± 0.03 to 6.59 ± 0.10 (p = 0.03), whereas extracellular pH (pHe) of this tumor changed from 7.03 ± 0.05 to 6.89 ± 0.06 (p = 0.19). A decline in bioenergetics (ß-NTP/Pi) of 55 ± 5.0% (p = 0.03) accompanied the decline in pHi of WM983B. Using 1H MRS with a selective multiquantum pulse sequence and Hadamard localization, we show that LND induced a significant increase in tumor lactate levels (p < 0.01). LND pre-treatment followed by DOX (10 mg/kg, i.v.) produced a growth delay of 13.7 days in WM983B (p < 0.01 versus control), a growth delay significantly smaller than the 25.4 days that occurred with DB-1 (p = 0.03 versus WM983B). Differences in relative levels of glycolysis may produce differential therapeutic responses of DB-1 and WM983B melanomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Doxorubicin/pharmacology , Energy Metabolism/drug effects , Indazoles/pharmacology , Melanoma/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Doxorubicin/therapeutic use , Drug Synergism , Glucose/analysis , Glucose/metabolism , Glycolysis/drug effects , Humans , Hydrogen-Ion Concentration , Indazoles/therapeutic use , Lactic Acid/analysis , Lactic Acid/metabolism , Magnetic Resonance Spectroscopy , Male , Melanoma/pathology , Mice , Mice, Nude , Oxygen/analysis , Oxygen/metabolism , Oxygen Consumption/drug effects , Treatment Outcome , Xenograft Model Antitumor Assays
10.
J Exp Clin Cancer Res ; 36(1): 169, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29179732

ABSTRACT

BACKGROUND: Glioblastoma Multiforme is the deadliest type of brain tumor and is characterized by very poor prognosis with a limited overall survival. Current optimal therapeutic approach has essentially remained unchanged for more than a decade, consisting in maximal surgical resection followed by radiotherapy plus temozolomide. MAIN BODY: Such a dismal patient outcome represents a compelling need for innovative and effective therapeutic approaches. Given the development of new drugs is a process presently characterized by an immense increase in costs and development time, drug repositioning, finding new uses for existing approved drugs or drug repurposing, re-use of old drugs when novel molecular findings make them attractive again, are gaining significance in clinical pharmacology, since it allows faster and less expensive delivery of potentially useful drugs from the bench to the bedside. This is quite evident in glioblastoma, where a number of old drugs is now considered for clinical use, often in association with the first-line therapeutic intervention. Interestingly, most of these medications are, or have been, widely employed for decades in non-neoplastic pathologies without relevant side effects. Now, the refinement of their molecular mechanism(s) of action through up-to-date technologies is paving the way for their use in the therapeutic approach of glioblastoma as well as other cancer types. SHORT CONCLUSION: The spiraling costs of new antineoplastic drugs and the long time required for them to reach the market demands a profoundly different approach to keep lifesaving therapies affordable for cancer patients. In this context, repurposing can represent a relatively inexpensive, safe and fast approach to glioblastoma treatment. To this end, pros and cons must be accurately considered.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Clinical Trials as Topic , Drug Repositioning , Energy Metabolism/drug effects , Gene Regulatory Networks/drug effects , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Structure-Activity Relationship
11.
Anticancer Res ; 37(7): 3413-3421, 2017 07.
Article in English | MEDLINE | ID: mdl-28668829

ABSTRACT

BACKGROUND/AIM: Since temozolomide (TMZ) is activated under alkaline conditions, we expected lonidamine (LND) to have no effect or perhaps diminish its activity, but initial results suggest it may actually enhance either or both short- and long-term activity of TMZ in melanoma xenografts. MATERIALS AND METHODS: Cohorts of 5 mice with subcutaneous xenografts ~5 mm in diameter were treated with saline (control (CTRL)), LND only, TMZ only or LND followed by TMZ at t=40 min (time required for maximal tumor acidification). RESULTS: Mean tumor volume for LND+TMZ for the period between 6 and 26 days was reduced compared to TMZ alone (repeated measures ANOVA F (1, 8), p=0.006), suggesting a pronounced impact of LND on this phenomenon. TMZ and LND+TMZ produced median growth delays of 82 and 106 days, respectively. CONCLUSION: The use of TMZ alone and in combination with LND deserves further investigation in treatment of melanoma and other malignancies.


Subject(s)
Dacarbazine/analogs & derivatives , Indazoles/administration & dosage , Melanoma/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/administration & dosage , Drug Synergism , Heterografts/drug effects , Humans , Mice , Mice, Nude , Temozolomide , Transplantation, Heterologous/methods
12.
Rapid Commun Mass Spectrom ; 30(16): 1835-45, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27476658

ABSTRACT

RATIONALE: Mass spectrometric (MS) analysis of low molecular weight polar metabolites can be challenging because of poor chromatographic resolution of isomers and insufficient ionization efficiency. These metabolites include intermediates in key metabolic pathways, such as glycolysis, the pentose phosphate pathway, and the Krebs cycle. Therefore, sensitive, specific, and comprehensive quantitative analysis of these metabolites in biological fluids or cell culture models can provide insight into multiple disease states where perturbed metabolism plays a role. METHODS: An ion-pairing reversed-phase ultra-high-performance liquid chromatography (IP-RP-UHPLC)/MS approach to separate and analyze biochemically relevant phosphate- and carboxylic acid-containing metabolites was developed. Diisopropylethylamine (DIPEA) was used as an IP reagent in combination with reversed-phase liquid chromatography (RP-LC) and a triple quadrupole mass spectrometer using selected reaction monitoring (SRM) and negative electrospray ionization (NESI). An additional reagent, hexafluoroisopropanol (HFIP), which has been previously used to improve sensitivity of nucleotide analysis by UHPLC/MS, was used to enhance sensitivity. RESULTS: HFIP versus acetic acid, when added with the IP base, increased the sensitivity of IP-RP-UHPLC/NESI-MS up to 10-fold for certain analytes including fructose-1,6-bisphosphate, phosphoenolpyruvate, and 6-phosphogluconate. It also improved the retention of the metabolites on a C18 reversed-phase column, and allowed the chromatographic separation of important isomeric metabolites. This methodology was amenable to quantification of key metabolites in cell culture experiments. The applicability of the method was demonstrated by monitoring the metabolic adaptations resulting from rapamycin treatment of DB-1 human melanoma cells. CONCLUSIONS: A rapid, sensitive, and specific IP-RP-UHPLC/NESI-MS method was used to quantify metabolites from several biochemical pathways. IP with DIPEA and HFIP increased the sensitivity and improved chromatographic separation when used with reversed-phase UHPLC.


Subject(s)
Carboxylic Acids/metabolism , Chromatography, High Pressure Liquid/methods , Ethylamines/chemistry , Phosphates/metabolism , Propanols/chemistry , Carboxylic Acids/analysis , Cell Line, Tumor , Cytological Techniques , Humans , Metabolic Networks and Pathways , Phosphates/analysis
13.
Biochim Biophys Acta ; 1866(2): 151-162, 2016 12.
Article in English | MEDLINE | ID: mdl-27497601

ABSTRACT

Lonidamine (LND) was initially introduced as an antispermatogenic agent. It was later found to have anticancer activity sensitizing tumors to chemo-, radio-, and photodynamic-therapy and hyperthermia. Although the mechanism of action remained unclear, LND treatment has been known to target metabolic pathways in cancer cells. It has been reported to alter the bioenergetics of tumor cells by inhibiting glycolysis and mitochondrial respiration, while indirect evidence suggested that it also inhibited l-lactic acid efflux from cells mediated by members of the proton-linked monocarboxylate transporter (MCT) family and also pyruvate uptake into the mitochondria by the mitochondrial pyruvate carrier (MPC). Recent studies have demonstrated that LND potently inhibits MPC activity in isolated rat liver mitochondria (Ki 2.5µM) and cooperatively inhibits l-lactate transport by MCT1, MCT2 and MCT4 expressed in Xenopus laevis oocytes with K0.5 and Hill coefficient values of 36-40µM and 1.65-1.85, respectively. In rat heart mitochondria LND inhibited the MPC with similar potency and uncoupled oxidation of pyruvate was inhibited more effectively (IC50~7µM) than other substrates including glutamate (IC50~20µM). LND inhibits the succinate-ubiquinone reductase activity of respiratory Complex II without fully blocking succinate dehydrogenase activity. LND also induces cellular reactive oxygen species through Complex II and has been reported to promote cell death by suppression of the pentose phosphate pathway, which resulted in inhibition of NADPH and glutathione generation. We conclude that MPC inhibition is the most sensitive anti-tumour target for LND, with additional inhibitory effects on MCT-mediated l-lactic acid efflux, Complex II and glutamine/glutamate oxidation.


Subject(s)
Antineoplastic Agents/pharmacology , Indazoles/pharmacology , Animals , Hexokinase/antagonists & inhibitors , Humans , Hydrogen-Ion Concentration , Indazoles/toxicity , Membrane Transport Proteins/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins , Monocarboxylic Acid Transporters/metabolism , Pyruvic Acid/metabolism
14.
Front Oncol ; 6: 135, 2016.
Article in English | MEDLINE | ID: mdl-27379200

ABSTRACT

We present the first validated metabolic network model for analysis of flux through key pathways of tumor intermediary metabolism, including glycolysis, the oxidative and non-oxidative arms of the pentose pyrophosphate shunt, the TCA cycle as well as its anaplerotic pathways, pyruvate-malate shuttling, glutaminolysis, and fatty acid biosynthesis and oxidation. The model that is called Bonded Cumomer Analysis for application to (13)C magnetic resonance spectroscopy ((13)C MRS) data and Fragmented Cumomer Analysis for mass spectrometric data is a refined and efficient form of isotopomer analysis that can readily be expanded to incorporate glycogen, phospholipid, and other pathways thereby encompassing all the key pathways of tumor intermediary metabolism. Validation was achieved by demonstrating agreement of experimental measurements of the metabolic rates of oxygen consumption, glucose consumption, lactate production, and glutamate pool size with independent measurements of these parameters in cultured human DB-1 melanoma cells. These cumomer models have been applied to studies of DB-1 melanoma and DLCL2 human diffuse large B-cell lymphoma cells in culture and as xenografts in nude mice at 9.4 T. The latter studies demonstrate the potential translation of these methods to in situ studies of human tumor metabolism by MRS with stable (13)C isotopically labeled substrates on instruments operating at high magnetic fields (≥7 T). The melanoma studies indicate that this tumor line obtains 51% of its ATP by mitochondrial metabolism and 49% by glycolytic metabolism under both euglycemic (5 mM glucose) and hyperglycemic conditions (26 mM glucose). While a high level of glutamine uptake is detected corresponding to ~50% of TCA cycle flux under hyperglycemic conditions, and ~100% of TCA cycle flux under euglycemic conditions, glutaminolysis flux and its contributions to ATP synthesis were very small. Studies of human lymphoma cells demonstrated that inhibition of mammalian target of rapamycin (mTOR) signaling produced changes in flux through the glycolytic, pentose shunt, and TCA cycle pathways that were evident within 8 h of treatment and increased at 24 and 48 h. Lactate was demonstrated to be a suitable biomarker of mTOR inhibition that could readily be monitored by (1)H MRS and perhaps also by FDG-PET and hyperpolarized (13)C MRS methods.

15.
PLoS One ; 11(6): e0157125, 2016.
Article in English | MEDLINE | ID: mdl-27285585

ABSTRACT

Previous NMR studies demonstrated that lonidamine (LND) selectively diminishes the intracellular pH (pHi) of DB-1 melanoma and mouse xenografts of a variety of other prevalent human cancers while decreasing their bioenergetic status (tumor ßNTP/Pi ratio) and enhancing the activities of melphalan and doxorubicin in these cancer models. Since melphalan and doxorubicin are highly toxic agents, we have examined three other nitrogen (N)-mustards, chlorambucil, cyclophosphamide and bendamustine, to determine if they exhibit similar potentiation by LND. As single agents LND, melphalan and these N-mustards exhibited the following activities in DB-1 melanoma xenografts; LND: 100% tumor surviving fraction (SF); chlorambucil: 100% SF; cyclophosphamide: 100% SF; bendamustine: 79% SF; melphalan: 41% SF. When combined with LND administered 40 min prior to administration of the N-mustard (to maximize intracellular acidification) the following responses were obtained; chlorambucil: 62% SF; cyclophosphamide: 42% SF; bendamustine: 36% SF; melphalan: 10% SF. The effect of LND on the activities of these N-mustards is generally attributed to acid stabilization of the aziridinium active intermediate, acid inhibition of glutathione-S-transferase, which acts as a scavenger of aziridinium, and acid inhibition of DNA repair by O6-alkyltransferase. Depletion of ATP by LND may also decrease multidrug resistance and increase tumor response. At similar maximum tolerated doses, our data indicate that melphalan is the most effective N-mustard in combination with LND when treating DB-1 melanoma in mice, but the choice of N-mustard for coadministration with LND will also depend on the relative toxicities of these agents, and remains to be determined.


Subject(s)
Alkylating Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Indazoles/pharmacology , Mechlorethamine/pharmacology , Melanoma/pathology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Chlorambucil/pharmacology , Cyclophosphamide/pharmacology , Doxorubicin/pharmacology , Drug Synergism , Humans , Male , Melanoma/drug therapy , Melphalan/pharmacology , Mice , Mice, Nude , Mice, SCID , Xenograft Model Antitumor Assays
16.
Biochem J ; 473(7): 929-36, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26831515

ABSTRACT

Lonidamine (LND) is an anti-tumour drug particularly effective at selectively sensitizing tumours to chemotherapy, hyperthermia and radiotherapy, although its precise mode of action remains unclear. It has been reported to perturb the bioenergetics of cells by inhibiting glycolysis and mitochondrial respiration, whereas indirect evidence suggests it may also inhibit L-lactic acid efflux from cells mediated by members of the proton-linked monocarboxylate transporter (MCT) family and also pyruvate uptake into the mitochondria by the mitochondrial pyruvate carrier (MPC). In the present study, we test these possibilities directly. We demonstrate that LND potently inhibits MPC activity in isolated rat liver mitochondria (Ki2.5 µM) and co-operatively inhibits L-lactate transport by MCT1, MCT2 and MCT4 expressed in Xenopus laevisoocytes with K0.5 and Hill coefficient values of 36-40 µM and 1.65-1.85 respectively. In rat heart mitochondria LND inhibited the MPC with similar potency and uncoupled oxidation of pyruvate was inhibited more effectively (IC50~ 7 µM) than other substrates including glutamate (IC50~ 20 µM). In isolated DB-1 melanoma cells 1-10 µM LND increased L-lactate output, consistent with MPC inhibition, but higher concentrations (150 µM) decreased L-lactate output whereas increasing intracellular [L-lactate] > 5-fold, consistent with MCT inhibition. We conclude that MPC inhibition is the most sensitive anti-tumour target for LND, with additional inhibitory effects on MCT-mediated L-lactic acid efflux and glutamine/glutamate oxidation. Together these actions can account for published data on the selective tumour effects of LND onL-lactate, intracellular pH (pHi) and ATP levels that can be partially mimicked by the established MPC and MCT inhibitor α-cyano-4-hydroxycinnamate (CHC).


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/antagonists & inhibitors , Cell Membrane/metabolism , Indazoles/pharmacology , Membrane Transport Proteins , Mitochondrial Proteins/antagonists & inhibitors , Monocarboxylic Acid Transporters/antagonists & inhibitors , Symporters/antagonists & inhibitors , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/genetics , Ion Transport/drug effects , Ion Transport/genetics , Lactic Acid/metabolism , Male , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Pyruvic Acid/metabolism , Rats , Rats, Wistar , Solute Carrier Proteins , Symporters/genetics , Symporters/metabolism
17.
J Biol Chem ; 291(1): 42-57, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26521302

ABSTRACT

The antitumor agent lonidamine (LND; 1-(2,4-dichlorobenzyl)-1H-indazole-3-carboxylic acid) is known to interfere with energy-yielding processes in cancer cells. However, the effect of LND on central energy metabolism has never been fully characterized. In this study, we report that a significant amount of succinate is accumulated in LND-treated cells. LND inhibits the formation of fumarate and malate and suppresses succinate-induced respiration of isolated mitochondria. Utilizing biochemical assays, we determined that LND inhibits the succinate-ubiquinone reductase activity of respiratory complex II without fully blocking succinate dehydrogenase activity. LND also induces cellular reactive oxygen species through complex II, which reduced the viability of the DB-1 melanoma cell line. The ability of LND to promote cell death was potentiated by its suppression of the pentose phosphate pathway, which resulted in inhibition of NADPH and glutathione generation. Using stable isotope tracers in combination with isotopologue analysis, we showed that LND increased glutaminolysis but decreased reductive carboxylation of glutamine-derived α-ketoglutarate. Our findings on the previously uncharacterized effects of LND may provide potential combinational therapeutic approaches for targeting cancer metabolism.


Subject(s)
Antineoplastic Agents/pharmacology , Electron Transport Complex II/antagonists & inhibitors , Indazoles/pharmacology , Mitochondria/metabolism , Cell Death/drug effects , Cell Line, Tumor , Citric Acid Cycle/drug effects , Diacetyl/analogs & derivatives , Diacetyl/pharmacology , Electron Transport Complex II/metabolism , Fumarates/metabolism , Glutamine/metabolism , Glutathione/metabolism , Humans , Malates/metabolism , Melanoma/metabolism , Melanoma/pathology , Metabolic Flux Analysis , Mitochondria/drug effects , Models, Biological , NADP/metabolism , Naphthalenes/pharmacology , Oxidation-Reduction/drug effects , Pentose Phosphate Pathway/drug effects , Reactive Oxygen Species/metabolism , Succinic Acid/metabolism
18.
J Biol Chem ; 291(10): 5157-71, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26703469

ABSTRACT

A network model for the determination of tumor metabolic fluxes from (13)C NMR kinetic isotopomer data has been developed and validated with perfused human DB-1 melanoma cells carrying the BRAF V600E mutation, which promotes oxidative metabolism. The model generated in the bonded cumomer formalism describes key pathways of tumor intermediary metabolism and yields dynamic curves for positional isotopic enrichment and spin-spin multiplets. Cells attached to microcarrier beads were perfused with 26 mm [1,6-(13)C2]glucose under normoxic conditions at 37 °C and monitored by (13)C NMR spectroscopy. Excellent agreement between model-predicted and experimentally measured values of the rates of oxygen and glucose consumption, lactate production, and glutamate pool size validated the model. ATP production by glycolytic and oxidative metabolism were compared under hyperglycemic normoxic conditions; 51% of the energy came from oxidative phosphorylation and 49% came from glycolysis. Even though the rate of glutamine uptake was ∼ 50% of the tricarboxylic acid cycle flux, the rate of ATP production from glutamine was essentially zero (no glutaminolysis). De novo fatty acid production was ∼ 6% of the tricarboxylic acid cycle flux. The oxidative pentose phosphate pathway flux was 3.6% of glycolysis, and three non-oxidative pentose phosphate pathway exchange fluxes were calculated. Mass spectrometry was then used to compare fluxes through various pathways under hyperglycemic (26 mm) and euglycemic (5 mm) conditions. Under euglycemic conditions glutamine uptake doubled, but ATP production from glutamine did not significantly change. A new parameter measuring the Warburg effect (the ratio of lactate production flux to pyruvate influx through the mitochondrial pyruvate carrier) was calculated to be 21, close to upper limit of oxidative metabolism.


Subject(s)
Melanoma/metabolism , Oxidative Phosphorylation , Adenosine Triphosphate/metabolism , Carbon-13 Magnetic Resonance Spectroscopy , Cell Line, Tumor , Citric Acid Cycle , Glucose/metabolism , Glutamine/metabolism , Humans , Models, Theoretical , Mutation, Missense , Oxygen/metabolism , Proto-Oncogene Proteins B-raf/genetics
19.
NMR Biomed ; 28(3): 395-403, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25702942

ABSTRACT

We seek to exploit the natural tendency of melanomas and other tumors to convert glucose to lactate as a method for the selective intracellular acidification of cancer cells and for the potentiation of the activity of nitrogen-mustard antineoplastic agents. We performed this study to evaluate whether the induction of hyperglycemia (26 mM) could enhance the effects of lonidamine (LND, 100 mg/kg; intraperitoneally) on the induction of intracellular acidification, bioenergetic decline and potentiation of the activity of melphalan (LPAM) against DB-1 melanoma xenografts in mice. Intracellular pH (pHi ), extracellular pH (pHe ) and bioenergetics (ß-nucleoside triphosphate to inorganic phosphate ratio, ß-NTP/Pi) were reduced by 0.7 units (p < 0.001), 0.3 units (p > 0.05) and 51.4% (p < 0.05), respectively. The therapeutic response to LPAM (7.5 mg/kg; intravenously) + LND (100 mg/kg; intraperitoneally) was reduced by about a factor of three under hyperglycemic conditions relative to normoglycemia, producing a growth delay of 7.76 days (tumor doubling time, 5.31 days; cell kill, 64%) compared with LND alone of 1.70 days and LPAM alone of 0.29 days. Under normoglycemic conditions, LND plus LPAM produced a growth delay of 17.75 days, corresponding to a cell kill of 90% at the same dose for each of these agents. The decrease in tumor cell kill under hyperglycemic conditions correlates with an increase in tumor ATP levels resulting from increased glycolytic activity. However, hyperglycemia substantially increases lactic acid production in tumors by a factor of approximately six (p < 0.05), but hyperglycemia did not increase the effects of LND on acidification of the tumor, most probably because of the strong buffering action of carbon dioxide (the pKa of carbonic acid is 6.4). Therefore, this study demonstrates that the addition of glucose during treatment with LND diminishes the activity of this agent.


Subject(s)
Acids/metabolism , Energy Metabolism/drug effects , Hyperglycemia/complications , Indazoles/pharmacology , Melanoma/metabolism , Melphalan/pharmacology , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Hydrogen-Ion Concentration , Intracellular Space/metabolism , Magnetic Resonance Spectroscopy , Male , Melanoma/complications , Melanoma/pathology , Mice, Nude , Organ Specificity
20.
NMR Biomed ; 28(3): 281-90, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25504852

ABSTRACT

We demonstrate that the effects of lonidamine (LND, 100 mg/kg, i.p.) are similar for a number of xenograft models of human cancer including DB-1 melanoma and HCC1806 breast, BT-474 breast, LNCaP prostate and A2870 ovarian carcinomas. Following treatment with LND, each of these tumors exhibits a rapid decrease in intracellular pH, a small decrease in extracellular pH, a concomitant monotonic decrease in nucleoside triphosphate and an increase in inorganic phosphate over a 2-3 h period. We have previously demonstrated that selective intracellular tumor acidification potentiates response of this melanoma model to melphalan (7.5 mg/kg, i.v.), producing an estimated 89% cell kill based on tumor growth delay analysis. We now show that, in both DB-1 melanoma and HCC1806 breast carcinoma, LND potentiates response to doxorubicin, producing 95% cell kill in DB-1 melanoma at 7.5 mg/kg, i.v. doxorubicin and 98% cell kill at 10.0 mg/kg doxorubicin, and producing a 95% cell kill in HCC1806 breast carcinoma at 12.0 mg/kg doxorubicin. Potentiation of doxorubicin may result from cation trapping of the weakly basic anthracycline. Recent experience with the clinical treatment of melanoma and other forms of human cancer suggests that these diseases will probably not be cured by a single therapeutic procedure other than surgery. A multimodality therapeutic approach will be required. As a potent modulator of tumor response to N-mustards and anthracyclines as well as tumor thermo- and radiosensitivity, LND promises to play an important clinical role in the management and possible complete local control of a number of prevalent forms of human cancer.


Subject(s)
Adenosine Triphosphate/deficiency , Breast Neoplasms/metabolism , Doxorubicin/pharmacology , Indazoles/pharmacology , Ovarian Neoplasms/metabolism , Prostatic Neoplasms/metabolism , Xenograft Model Antitumor Assays , Acids/metabolism , Animals , Blotting, Western , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Energy Metabolism/drug effects , Female , Humans , Hydrogen-Ion Concentration , Intracellular Space/metabolism , Magnetic Resonance Spectroscopy , Male , Mice, Nude , Monocarboxylic Acid Transporters/metabolism , Ovarian Neoplasms/pathology , Prostatic Neoplasms/pathology , Symporters/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...