Co-treatment of dichloroacetate, omeprazole and tamoxifen exhibited synergistically antiproliferative effect on malignant tumors: in vivo experiments and a case report

Tatsuaki Ishiguro 1, Ryumei Ishiguro, Miyu Ishiguro, Sayuri Iwai 1Kamui Medical CO., Ltd., Tokyo, Japan. [email protected]: Received: Accepted: Published: Abstract Background/aims: Omeprazole (OPZ) and tamoxifen (TAM) strengthen the effects of anticancer drugs and dichloroacetate (DCA) inhibits tumor growth. This study assesses the synergistic effects of these drugs. Methodology: HT1080 human fibrosarcoma cells and WI-38 human fibroblasts were used as test and control cells, respectively. DCA, OPZ and TAM alone or in combination were applied and cells were counted after a one week culture. The combination of these drugs was prescribed to a cholangiocarcinoma patient and serum CA19-9 was monitored. Results: DCA combined with OPZ and TAM exhibited more potent antitumor activity than DCA alone in HT1080 fibrosarcoma cells, but did not influence proliferation of WI-38 human fibroblasts. All these drugs induce caspase-dependent cell growth inhibition through superoxide production. Since they can be taken orally and have been used clinically without major side effects, it was thought that this combination therapy would be a readily translated strategy to treat malignant tumors. Under the patient's consent these three drugs were prescribed to a 51-year old female cholangiocarcinoma patient to whom neither gemcitabine+S-1 nor adoptive immunotherapy with natural killer cells was effective. Disease progression was successfully blocked (the rise of serum CA19-9 value) for three months, also confirmed by CT. Conclusions: Although findings are preliminary, this study is a sample of translational research. Since there is no consensus regarding treatment strategy of cholangiocarcinoma and chemotherapy has only limited efficacy, it is expected that it might open a new possibility of treatment. Keywords: © INTRODUCTION Breast canc REFERENCES 1 World ]

Therapeutic Modulation of Cancer Metabolism with Dichloroacetate and Metformin

Nathan Patrick Ward Approved: 7 November 2016 Abstract The robust glycolytic metabolism of glioblastoma multiforme (GBM) has proven them susceptible to increases in oxidative metabolism induced by the pyruvate mimetic dichloroacetate (DCA). Recent reports demonstrate that the anti-diabetic drug metformin enhances the damaging oxidative stress associated with DCA treatment in cancer cells. We sought to elucidate the role of metformin’s reported activity as a mitochondrial complex I inhibitor in the enhancement of DCA cytotoxicity in the VM-M3 model of GBM. We demonstrated that metformin potentiated DCA-induced superoxide production and that this was required for enhanced cytotoxicity towards VM-M3 cells with the combination. Similarly, rotenone enhanced oxidative stress resultant from DCA treatment and this too was required for the noted augmentation of cytotoxicity. Adenosine monophosphate kinase (AMPK) activation was not observed with the concentration of metformin required to enhance DCA activity. Moreover, addition of an activator of AMPK did not enhance DCA cytotoxicity, whereas an inhibitor of AMPK heightened the cytotoxicity of the combination. We also show that DCA and metformin reduce tumor burden and prolong survival in VM-M3 tumor-burdened mice as individual therapies. In contrast to our in vitro work, we did not observe synergy between DCA and metformin in vivo. Our data indicate that metformin enhancement of DCA cytotoxicity is dependent on complex I inhibition. Particularly, that complex I inhibition cooperates with DCA-induction of glucose oxidation to enhance cytotoxic oxidative stress in VM-M3 GBM cells. This work supports further investigation and optimization of a DCA/metformin combination as a potential pro-oxidant combinatorial therapy for GBM. Keywords: Cancer metabolism, mitochondrial glucose oxidation, complex I inhibition, oxidative stress, DCA, metformin Copyright © 2017, Nathan P. Ward CANCER METABOLISM Chapter Synopsis Herein we provide a review of the metabolic programs employed by tumors to meet the biosynthetic requirements of tumorigenesis. The metabolism of tumors is intricately linked to the hallmarks of the disease and provides cancer cells with a survival advantage in response to the stresses imposed by the tumor microenvironment. An understanding of the metabolic characteristics of tumors provides a basis for rational targeting of these metabolic dependencies as a therapeutic strategy. Current approaches in targeting cancer metabolism are also discussed in this chapter. Altered Energy Metabolism Cancer is traditionally considered a genetic disease, characterized by genomic instability and frequent mutation that cooperate to promote a distinct cellular environment that permits unbridled proliferation (1). Genomic sequencing of tumors has identified a multitude of drug targetable mutations that have driven research and pharmaceutical development. Unfortunately, the promise of encouraging pre-clinical findings has not often translated to clinical efficacy. This has driven the field to consider additional hallmarks of tumor development and disease progression and devise alternative strategies for cancer management (1). Resultant from this initiative was a renewed appreciation for the distinct metabolic activity of tumors (2). Beyond the dysregulation of the cell cycle and loss of deoxyribonucleic acid (DNA) quality control that accompany cancer cell proliferation is a fundamental demand for biomass. An intricate network of metabolic pathways converges to generate…

Targeting metabolic flexibility by simultaneously inhibiting respiratory complex I and lactate generation retards melanoma progression

Balkrishna Chaube1, Parmanand Malvi1, Shivendra Vikram Singh1, Naoshad Mohammad1, Avtar Singh Meena1,2 and Manoj Kumar Bhat1 1 National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India2 Current address: Department of Physiology, University of Tennessee Health Science Center, Memphis, USA Correspondence: Manoj Kumar Bhat, email: [email protected]: 16 June 2015 Accepted: 23 September 2015Published: 15 October 2015 Abstract Melanoma is a largely incurable skin malignancy owing to the underlying molecular and metabolic heterogeneity confounded by the development of resistance. Cancer cells have metabolic flexibility in choosing either oxidative phosphorylation (OXPHOS) or glycolysis for ATP generation depending upon the nutrient availability in tumor microenvironment. In this study, we investigated the involvement of respiratory complex I and lactate dehydrogenase (LDH) in melanoma progression. We show that inhibition of complex I by metformin promotes melanoma growth in mice via elevating lactate and VEGF levels. In contrast, it leads to the growth arrest in vitro because of enhanced extracellular acidification as a result of increased glycolysis. Inhibition of LDH or lactate generation causes decrease in glycolysis with concomitant growth arrest both in vitro and in vivo. Blocking lactate generation in metformin-treated melanoma cells results in diminished cell proliferation and tumor progression in mice. Interestingly, inhibition of either LDH or complex I alone does not induce apoptosis, whereas inhibiting both together causes depletion in cellular ATP pool resulting in metabolic catastrophe induced apoptosis. Overall, our study suggests that LDH and complex I play distinct roles in regulating glycolysis and cell proliferation. Inhibition of these two augments synthetic lethality in melanoma. Keywords: melanoma; complex I; LDH; metabolic catastrophe; synthetic lethality INTRODUCTION Malignant melanoma is one of the most aggressive forms of skin cancer with high metastatic potential and resistance to many cytotoxic agents [1, 2]. Despite extensive research and partial successes gained by the use of currently available drugs there is no effective treatment against malignant melanoma [1-3]. Melanoma cases are increasing every year and account for about 75% of skin cancer-related deaths worldwide [2, 3]. Poor response to currently available therapeutic options and development of resistance to therapy warrant exploration of new strategies to treat melanoma. Enhanced aerobic glycolysis is a characteristic feature of many cancers [3-5]. It has been reported that melanoma cells, owing to the BRAF mutation, depend majorly on glycolysis for ATP generation, and exhibit dysfunctional oxidative phosphorylation [6, 7]. Cancer cells derive ATP, biosynthetic intermediates, and reducing equivalents by unusually engaging in biochemical pathways such as glycolysis, glutaminolysis, and the pentose phosphate pathway [5]. Normal (non-cancerous) cells derive ATP primarily through mitochondrial OXPHOS; while cancer cells rely mainly on aerobic glycolysis to generate ATP and glycolytic intermediates those facilitate rapid growth [4, 5]. Enhanced lactate generation has been correlated with aggressiveness of cancer. Numerous studies have identified lactate dehydrogenase (LDH), which catalyzes the conversion of pyruvate to lactate, as the most consistent marker of the aggressive and rapidly growing cancers [8-11]. LDH plays an important role in regulating glycolysis, maintaining cellular redox state, mitochondrial physiology and tumor maintenance [12]. Altered metabolism of cancer cell can be associated with mitochondrial dysfunction which…

Bioenergetic modulation with dichloroacetate reduces the growth of melanoma cells and potentiates their response to BRAFV600E inhibition

Cecilie Abildgaard1 , Christina Dahl1 , Astrid L Basse2 , Tao Ma2 and Per Guldberg1* 1 Danish Cancer Society Research Center, Copenhagen, Denmark2 Department of Biology, University of Copenhagen, Copenhagen, Denmark.Correspondence: [email protected]: 14 September 2020Accepted: 4 December 2020Published: 9 December 2020 Abstract Background: Advances in melanoma treatment through targeted inhibition of oncogenic BRAF are limited owing to the development of acquired resistance. The involvement of BRAFV600E in metabolic reprogramming of melanoma cells provides a rationale for co-targeting metabolism as a therapeutic approach. Methods: We examined the effects of dichloroacetate (DCA), an inhibitor of pyruvate dehydrogenase kinase, on the growth and metabolic activity of human melanoma cell lines. The combined effect of DCA and the BRAF inhibitor vemurafenib was investigated in BRAFV600E -mutated melanoma cell lines. Vemurafenib-resistant cell lines were established in vitro and their sensitivity to DCA was tested. Results: DCA induced a reduction in glycolytic activity and intracellular ATP levels, and inhibited cellular growth. Co-treatment of BRAFV600E-mutant melanoma cells with DCA and vemurafenib induced a greater reduction in intracellular ATP levels and cellular growth than either compound alone. In addition, melanoma cells with in vitro acquired resistance to vemurafenib retained their sensitivity to DCA. Conclusions: These results suggest that DCA potentiates the effect of vemurafenib through a cooperative attenuation of energy production. Furthermore, the demonstration of retained sensitivity to DCA in melanoma cells with acquired resistance to vemurafenib could have implications for melanoma treatment. Keywords: Dichloroacetate, Melanoma, BRAF, Bioenergetics, Metabolism, ATPAbbreviations: (Acetyl-CoA): Acetyl coenzyme A, (AMPK): AMP activated protein kinase, (DCA): Dichloroacetate, (ECAR): Extracellular acidification rate, (HEMn-LP): Human epidermal melanocytes(IC50): half maximal inhibitory concentration, (LKB1): Liver kinase B1, (MITF): Microphthalmia-associated transcription factor, (OCR): Oxygen consumption rate, (PDH): Pyruvate dehydrogenase, (PDK): Pyruvate dehydrogenase kinase © 2014 Abildgaard et al.; licensee BioMed Central Ltd. Background A hallmark of cancer is the reprogramming of cellular metabolism towards aerobic glycolysis. This metabolic pattern is characterized by increased glucose uptake and highly up-regulated glycolytic activity with fermentation of glucose into lactic acid instead of complete aerobic decomposition in the mitochondria. Aerobic glycolysis, also referred to as the Warburg effect, resembles the anaerobic metabolism of normal cells, but occurs in the context of an adequate oxygen supply [1]. The reprogramming of metabolism in cancer cells is a highly complex and heterogeneous process, which is driven by a wide variety of genetic and non-genetic strategies to overcome energy restriction [2]-[4]. The BRAF V600E oncogene, present in more than 50% of melanomas [5], has been directly implicated in the reprogramming of cellular metabolism. The constitutive activity of mutant BRAF reduces the expression of oxidative enzymes and the number of mitochondria, while increasing the expression of glycolytic enzymes and lactic acid production [6],[7]. Furthermore, a molecular link was recognized between the RAS-RAF-MEK-ERK-MAPK pathway and the energetic-stress check-point mediated by the liver kinase B1 (LKB1)-AMP activated protein kinase (AMPK) pathway, suggesting a role of BRAFV600E in mediating resistance to energetic stress [8],[9]. BRAF affects oxidative metabolism through microphthalmia-associated transcription factor (MITF)-dependent control of the mitochondrial master regulator PGC1α [7]. Previous studies have shown that melanomas expressing…

In Vivo Evaluation of the Antitumor and Immunogenic Properties of Silver and Sodium Dichloroacetate Combination against Melanoma

Zaida Torres-Cavazos1, Moisés Armides Franco-Molina2, Silvia Elena Santana-Krímskaya2, Cristina Rodríguez-Padilla2, Jorge Ramsy Kawas-Garza1, Gustavo Hernández-Vidal1, Gustavo Moreno-Degollado1, and Diana Elisa Zamora-Ávila1 1 Posgrado Conjunto de las Facultades de Agronomía y Medicina Veterinaria y Zootecnia, Universidad Autónoma de Nuevo León,Ave. Universidad S/N, Cd. Universitaria, San Nicolás de los Garza, N. L., CP 66455, Mexico2 Laboratorio de Inmunología y Virología, Unidad C, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León,Ave. Universidad S/N, Cd. Universitaria, San Nicolás de los Garza, N. L., CP 66455, Mexico Correspondence: Moisés Armides Franco-Molina; [email protected]: 13 July 2020Revised: 28 September 2020Accepted: 14 October 2020Published: 7 November 2020 Abstract Our main focus was to evaluate the efficacy of silver and sodium dichloroacetate as dual-function agents to be used in melanoma treatment. This strategy is designed to increase the activity of these two compounds that affect DNA integrity and the mitochondria at different levels. Furthermore, we evaluated if the cell death mechanism induced by our treatments was immunogenic cell death. To evaluate antitumor efficacy, we assessed tumor volume and production of tumor necrosis factor-α, nuclear factor κ B (both by ELISA), and nitric oxide levels (Nitrate/Nitrite colorimetric assay kit); for immunogenic cell death, we evaluated the release of danger-associated molecular patterns using immunohistochemistry and flow cytometry, as well as an in vivo challenge. Our results showed that the combination of colloidal silver and sodium dichloroacetate is more effective than each treatment alone and that the antitumor mechanism is not through immunogenic cell death. Furthermore, this study can broadly contribute to the development of dichloroacetate-loaded silver nanoparticles and to the design targeted pharmacological formulations to fight melanoma as well as other types of cancer. Academic Editor: Yanis Toledaño Magaña Copyright © 2020 Zaida Torres-Cavazos et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. INTRODUCTION Targeted therapies have increased the chances of survival for people with melanoma [1]; however, cancer cells present within the tumor favor different metabolic pathways [2]; as a consequence, the tumor eventually becomes resistant to targeted therapies, especially the ones designed against a single target [3]. The development of silver-based therapies is a promising tool in cancer treatment. Silver ions and silver nanoparticles induce oxidative stress, mitochondrial membrane dysfunction, DNA damage, and cytokines upregulation [4]. The exact action mechanism varies depending on the physical and chemical properties of the nanoparticle and the type of cancer [5]. Furthermore, the clinical use of colloidal silver for bactericidal and antiviral purposes proves that this treatment is safe [6],  7]. Sodium dichloroacetate (DCA) is a pyruvate analog which interferes with tumor-associated glycolysis (Warburg effect), decreases cancer malignancy, and reduces lactate production by altering cancer cell metabolic pathways [8]. A decrease in lactate counteracts the acidic state of tumoral microenvironment, reducing tumor growth and metastasis [8]. WZB117, a bis-hydroxybenzoate, 2-deoxy-d-glucose, metformin, and DCA reduce glycolysis and block glucose uptake in cancer cells. Under low intracellular glucose levels, biosynthetic pathways, such as nucleotides…

Combination of Sulindac and Dichloroacetate Kills Cancer Cells via Oxidative Damage

Kasirajan Ayyanathan1,2,*,#, Shailaja Kesaraju1,#, Ken Dawson-Scully1,2, Herbert Weissbach1 1 Center for Molecular Biology and Biotechnology, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, Florida, United States of America,2 Department of Biological Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, Florida, United States of America*E-mail: [email protected]#These authors contributed equally to this work. Received: 12 August 2011Accepted: 4 June 2012Published: 1 July 2012 Abstract Sulindac is an FDA-approved non-steroidal anti-inflammatory drug with documented anticancer activities. Our recent studies showed that sulindac selectively enhanced the killing of cancer cells exposed to oxidizing agents via production of reactive oxygen species (ROS) resulting in mitochondrial dysfunction. This effect of sulindac and oxidative stress on cancer cells could be related to the defect in respiration in cancer cells, first described by Warburg 50 years ago, known as the Warburg effect. We postulated that sulindac might enhance the selective killing of cancer cells when combined with any compound that alters mitochondrial respiration. To test this hypothesis we have used dichloroacetate (DCA), which is known to shift pyruvate metabolism away from lactic acid formation to respiration. One might expect that DCA, since it stimulates aerobic metabolism, could stress mitochondrial respiration in cancer cells, which would result in enhanced killing in the presence of sulindac. In this study, we have shown that the combination of sulindac and DCA enhances the selective killing of A549 and SCC25 cancer cells under the conditions used. As predicted, the mechanism of killing involves ROS production, mitochondrial dysfunction, JNK signaling and death by apoptosis. Our results suggest that the sulindac-DCA drug combination may provide an effective cancer therapy. Citation: Ayyanathan K, Kesaraju S, Dawson-Scully K, Weissbach H (2012) Combination of Sulindac and Dichloroacetate Kills Cancer Cells via OxidativeDamage. PLoS ONE 7(7): e39949. doi:10.1371/journal.pone.0039949Editor: Joseph Alan Bauer, Bauer Research Foundation, United States of America © 2012 Ayyanathan et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permitsunrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Funding: Funding assistance from the National Institutes of Health to KA (grant 5K01CA95620) and HW (grant R15 CA122001) and from the State of Florida to HW (SURECAG grant R94007) to carry out this work is gratefully acknowledged. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.Competing Interests: The authors have declared that no competing interests exist. INTRODUCTION Sulindac is an FDA-approved non-steroidal anti-inflammatory drug (NSAID), which has also been shown to have anti-cancer activity [1–6]. Recent studies from our laboratory have demonstrated that RKO, A549 and SCC25 cancer cell lines exhibited sensitivity towards a combination of sulindac and an oxidizing agent, such as TBHP or H2O2 [7]. The data indicated that the sulindac effect was not related to its NSAID activity but that sulindac made cancer cells more sensitive to oxidative stress resulting in mitochondrial dysfunction and loss of viability. In contrast, normal cells did not show enhanced…

Inhibition of retinoic acid receptor β signaling confers glycolytic dependence and sensitization to dichloroacetate in melanoma cells

Cecilie Abildgaard1, Christina Dahl1, Ahmad Abdul-Al1, Annette Christensen1 and Per Guldberg1 1 Danish Cancer Society Research Center, Copenhagen, Denmark Correspondence: Per Guldberg, email: [email protected]: 19 April 2017 Accepted: 19 July 2017Published: 24 August 2017 Abstract Dysregulation of metabolism during melanoma progression is tightly associated with the acquisition of genetic and epigenetic alterations in regulators of metabolic pathways. Retinoic acid receptor beta (RARβ) is epigenetically silenced in a large proportion of melanomas, but a link between RARβ and metabolic rewiring of melanoma has not been established. Here, we show that in primary human melanocytes, all-trans retinoic acid (a RARβ agonist) induced growth inhibition accompanied by a decrease in both glycolytic and oxidative metabolism, whereas selective inhibition of RARβ led to an increase in the basal glycolytic rate and increased sensitivity to inhibition of glycolysis. In melanoma cells, inhibition of RARβ promoted lower mitochondrial respiration and higher glycolytic activity, which led to energetic stress and activation of the energy sensor AMP-activated protein kinase. This metabolic shift increased the sensitivity to both glycolytic inhibition and stimulation of mitochondrial metabolism with dichloroacetate, an inhibitor of pyruvate dehydrogenase kinase. In melanoma cells harboring the BRAFV600E mutation, RARβ activation antagonized the effect of the BRAF inhibitor PLX4032 (vemurafenib). Collectively, these data suggest that RARβ signaling is involved in regulating cellular metabolism in melanoma and may provide a potential target in combination treatment strategies. Keywords: melanoma, cancer metabolism, retinoic acid receptor β, mitochondrial respiration, dichloroacetateAbbreviations: ATRA: all-trans retinoic acid; DCA: dichloroacetate; ECAR: extracellular acidification rate; OCR: oxygen consumption rate; ROS: reactive oxygen species © Abildgaard et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. INTRODUCTION Melanoma, the most lethal form of skin cancer, causes 50,000 deaths annually with the incidence continuing to increase worldwide. While primary cutaneous melanoma is curable by surgery, the most advanced form of the disease (stage IV) is associated with a 10-year survival of 10-15% [1], reflecting its notorious resistance to conventional anti-cancer therapy. Recent therapeutic advances include immune checkpoint inhibitors and therapies targeting oncogenes or downstream effectors of the MAPK pathway (e.g., BRAF and MEK inhibitors). However, the development of acquired drug resistance eventually leads to relapse in the majority of cases [2, 3]. Melanoma develops from melanin-producing cells, called melanocytes, through the acquisition of multiple genomic alterations. The most common melanoma drivers include activating mutations in BRAF and NRAS and inactivating mutations or deletions in CDKN2A (encoding p16INK4A and p14ARF), PTEN and TP53 [4]. Recent evidence suggests that a common function shared among some of these genes is to control cellular metabolism [5, 6]. During the progression of melanoma, cellular metabolism is reprogrammed, implying a shift from mitochondrial respiration toward aerobic glycolysis, leading to increased glucose consumption and lactic acid production (the Warburg effect) [7]. Several reports based on in vitro and in vivo models of melanoma and clinical studies of melanoma patients have demonstrated a link between activating mutations at codon V600 of BRAF (most commonly BRAFV600E) and aerobic…

In vitro cytotoxicity of novel platinum-based drugs and dichloroacetate against lung carcinoid cell lines

Wolfgang Fiebiger1, Ulrike Olszewski2, Ernst Ulsperger2, Klaus Geissler2, Gerhard Hamilton2,3 1 Department of Internal Medicine I, Division of Oncology, St. Poelten Hospital, St. Poelten, Austria2 Ludwig Boltzmann Cluster of Translational Oncology, C/o Balderichgasse, 26/13, 1170 Vienna, Austriae-mail: [email protected]. Hamilton3 Department of Surgery, Medical University Vienna, Vienna, AustriaReceived: 1 April 2010 Accepted: 6 June 2010 Abstract Introduction: Chemotherapy for advanced well-differentiated carcinoids is characterised by low response rates and short duration of responses. The present study aimed to assess the in vitro activity of novel platinum-based chemotherapeutic drugs in combination with dichloroacetate (DCA), a sensitiser to apoptosis, against lung carcinoid cell lines. Methods: Three permanent cell lines (UMC-11, H727 and H835) were exposed to 14 different established cytotoxic drugs and the novel platinum-based compounds as satraplatin, JM118 and picoplatin in combination with DCA, and viability of the cells was measured using a tetrazolium based dye assay. Results: With exception of the highly chemoresistant UMC- 11 line, the carcinoid cell lines (H727, H835) were sensitive to the majority of chemotherapeutics in vitro. Among the platinum-based drugs, carboplatin and oxaliplatin showed highest efficacy. H835 cells growing as multicellular spheroids were 2.7-8.7-fold more resistant to picoplatin, satraplatin and its metabolite compared to single cell suspensions. DCA (10 mM) inhibited the growth of UMC- 11 cells by 22% and sensitised these highly resistant cells to carboplatin, satraplatin and JM118 1.4-2.4-fold. Conclusion: The highly resistant UMC-11 lung carcinoid cells are sensitive to carboplatin, oxaliplatin and the satraplatin metabolite JM118, but multicellular spheroidal growth, as observed in the H835 cell line and pulmonary tumourlets, seems to increase chemoresistance markedly. The activity of carboplatin and JM118 is significantly and specifically increased in combination with the apoptosis sensitiser DCA that promotes mitochondrial respiration over aerobic glycolysis. In summary, among the novel platinum drugs satraplatin has the potential for treatment of lung carcinoids and DCA potentiates the cytotoxicity of selected platinum drugs. Keywords: Carcinoid; Chemosensitivity; Drug resistance; Platinum complex; Picoplatin; Satraplatin; Dichloroacetate INTRODUCTION Carcinoids derive from enterochromaffin cells of the neuroendocrine cell system that is widely distributed in the body [1–3]. These rather rare tumours have an incidence of 2.0–2.5 cases per 100,000 people for clinically conspicuous carcinoids [4]. Foregut carcinoids comprise 20% of all carcinoid tumours and include thymic and lung (representing 2% of primary lung cancers) in addition to gastric and duodenal carcinoids [5]. According to their histologic characteristics, these heterogenous tumours are classifi ed either as well-differentiated neuroendocrine tumours with small cells and regular nuclei (typical carcinoid) or well differentiated neuroendocrine carcinomas with nuclear atypia and necrotic areas (atypical carcinoid) [6, 7]. The “carcinoid syndrome”, which is induced by secretion of vasoactive substances, occurs in less than 5% of pulmonary carcinoids [8]. From 13 to 22% of patients have distant metastases at time of presentation [4]. Primary diagnostic procedures include biochemical testing, particularly analysis of serotonin and urinary 5-hydroxyindoleacetic acid (5-HIAA). The most sensitive technique for localisation of the tumours is somatostatin receptor scintigraphy supplemented by CT for visualisation of liver metastases and concomitant biopsy for histopathological verification [9]. Since…

EFFECT OF DICHLOROACETATE ON LEWIS LUNG CARCINOMA GROWTH AND METASTASIS

D.L. Kolesnik1, O.N. Pyaskovskaya1, I.V. Boychuk1, O.I. Dasyukevich1, O.R. Melnikov1, A.S. Tarasov1, G.I. Solyanik1 1R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, Kyiv 03022, Ukraine.Correspondence: D.L. Kolesnik, E-mail: [email protected]: 20 March 2015 Abstract A hallmark of malignancy is excessive tumor glycolysis, even in the presence of oxygen, which causes lactacidosis in the tumor microenvironment and favors tumor cell proliferation and survival. For this reason antimetabolic agents which target tumor cell metabolism are being researched extensively as promising anticancer drugs. Aim: To study the effect of lactacidosis on survival of Lewis lung carcinoma (LLC) cells at the conditions of nutritional substrate deficiency in vitro and evaluate antitumor and antimetastatic activity against LLC/R9 in vivo. Materials and Methods: LLC variant LLC/R9 was used as experimental tumor model. Tumor cell viability was determined using trypan blue staining. Apoptosis level was counted with the use of Hoechst 33258 dye. Lactate content in the tumor tissue was evaluated by enzyme method with the use of lactate dehydrogenase. Reactive oxygen species was determined using 2.7-dichlorofluorescein diacetate. Effects of dichloroacetate (DCA) on the growth and metastasis of LLC/R9 were analyzed by routine procedures. Evaluation of DCA effect toward electron-transport chain (ETC) components was performed using EPR. Results: It has been shown that at the conditions of lactacidosis and glucose deficiency, LLC/R9 cell viability in vitro was higher by 30% (р < 0.05) and apoptosis level was triply lower (р < 0.05) than these indices at the conditions of glucose deficiency only. In mice with transplanted LLC/R9 tumors treated for 3 weeks per os with DCA at the total dose of 1.5 g/kg of body weight starting from the next day after tumor transplantation, the primary tumor volume was just by 30% lower than that in control group. At the same time, the number and volume of lung metastases in animals treated with DCA were by 59% (р < 0.05) and 94% (р < 0.05) lower, respectively, than these indices in the control group. DCA treatment resulted in nearly 30% increase (р < 0.05) of lactate content in tumor tissue compared to that in the control, but did not affect significantly the levels of heme iron complexes with NO (at gmed = 2.007) in mitochondrial ETC proteins and Fe-S cluster proteins (at g = 1.94) in tumor cells. Conclusions: It has been shown that lactacidosis significantly promoted LLC/R9 cell survival at the conditions of glucose deficiency in vitro. If LLC/R9 developed in vivo, DCA as the compound with antilactacidosis activity did not suppress significantly the primary tumor growth but exerted significant antimetastatic activity. Keywords: dichloroacetate; hypoxic radiosensitivity; breast cancer; reactive oxygen speciesAbbreviations: DCA — dichloroacetate, ETC — electron-transport chain; LLC/R9 — Lewis lung carcinoma variant; PDH — pyruvate dehydrogenase; PDK — pyruvate dehydrogenase kinase. INTRODUCTION It is well known that lactacidosis, large accumulation of lactate and decrease of рН, is the main characteristics of metabolic tumor cell microenvironment in vitro and in vivo. Earlier lactacidosis has been considered as a ballast product of tumor cell metabolism. However, recently it has…

Anti-cancer synergy of dichloroacetate and EGFR tyrosine kinase inhibitors in NSCLC cell lines

Zheng Yang1, Kin Y. Tam1 1 Faculty of Health Sciences, University of Macau, Taipa, Macau, China.Correspondence: Kin Y. TamFaculty of Health Sciences, University of Macau, Taipa, Macau, China Tel.: +853-88224988 Fax: +853-88222314. Email: [email protected] Received: 15 April 2016Revised: 27 July 2016Accepted: 2 August 2016 Abstract Glycolysis has been observed as a predominant process for most cancer cells to utilize glucose, which was referred to as "Warburg Effect". Targeting critical enzymes, such as pyruvate dehydrogenase kinase (PDK) that inversely regulating the process of glycolysis could be a promising approach to work alone or in combination with other treatments for cancer therapy. EGFR inhibitors for Non-Small-Cell Lung Cancer (NSCLC) treatment have been applied for decades in clinical practices with great success, but also their clinical benefits were somewhat hampered by the rising acquired-resistance. Combination drug therapy is an effective strategy to cope with the challenge. In this study, we utilized Dichloroacetate (DCA), a widely regarded PDK inhibitor, together with Erlotinib and Gefitinib, two well-known EGFR inhibitors, and demonstrated that the applications of DCA in combination with either Erlotinib or Gefitinib significantly attenuated the viability of EGFR mutant NSCLC cells (NCI-H1975 and NCI-H1650) in a synergistic manner. This synergistic outcome appears to be a combination effect in promoting apoptosis, rather than co-suppression of either EGFR or PDK signaling pathways. Moreover, we have shown that the combination treatment did not exhibit synergistic effect in other NSCLC cell lines without EGFR mutations (A549 or NCI-H460). Together, these observations suggested that combined targeting of EGFR and PDK in NSCLC cells exerted synergistic effects in an EGFR mutation-dependent fashion. Keywords: Pyruvate Dehydrogenase Kinase; Dichloroacetate; Epidermal Growth Factor Receptor; Erlotinib; Gefitinib; Drug Combination Copyright © 2016 Elsevier B.V. All rights reserved. INTRODUCTION Lung cancer ranked the first in males and top five in females of newly diagnosed cancer cases and the cancer-related deaths worldwide according to the latest statistics (Jemal et al., 2011), with over 80% of the patients falling into the category of Non-Small-Cell Lung Cancer, or NSCLC (Ke et al., 2015). Traditional strategies for NSCLC treatment often resorted to chemotherapies with either mono-application or combo-administration of platinum-based or other cytotoxic chemicals. However, the objective response rates of these strategies were usually unsatisfactory, with median overall survival usually less than 1 year (Schiller et al., 2002, Pao and Chmielecki, 2010). EGFR mutation was found in approximate 30% of the NSCLC patients who often responded well to the targeted therapy (Pao and Chmielecki, 2010). This enabled wide applications of small molecular EGFR tyrosine kinase inhibitors (EGFR-TKis), which showed tremendous success in past decades (Hanahan and Weinberg, 2011), as exemplified by Erlotinib and Gefitinib (Dutta and Maity, 2007). However, acquired-resistance usually occurred when patients were treated with EGFR inhibitors, with several identified mechanisms such as original or induced T790M hot spot mutation, activated secondary signaling like MET amplification or PI3K mutation, or conferred epithelial to mesenchymal transition (EMT) (Maione et al., 2015). Disappointingly, combined applications of EGFR inhibitors with chemotherapies brought about higher occurrence of adverse effects, rather than the expected benefit of extending the overall survival of…

Dichloroacetate enhances the antitumor efficacy of chemotherapeutic agents via inhibiting autophagy in non-small-cell lung cancer

Xiao Lu,1,* Dong Zhou,1,* Bing Hou,1 Quan-Xing Liu,1 Qian Chen,2 Xu-Feng Deng,1,3 Zu-Bin Yu,1 Ji-Gang Dai,1 Hong Zheng1 1 Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, People’s Republic of China 2 Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, People’s Republic of China 3 Department of Cardiothoracic Surgery, First People’s Hospital of Zunyi, Guizhou, People’s Republic of China*These authors contributed equally to this work Correspondence: Hong Zheng; Ji-Gang DaiDepartment of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Street, Shapingba District, Chongqing 400037, People’s Republic of ChinaTel.: +86 23 6877 4724Fax: +86 23 6877 4724Email: [email protected]; [email protected]: 14 September 2020Accepted: 4 December 2020Published: 9 December 2020 Abstract Background: Chemotherapy is still the primary adjuvant strategy of cancer therapy; however, the emergence of multi-drug resistance has been a cause for concern. Autophagy has been demonstrated to have a protective role against chemotherapeutic drugs in cancer cells, and autophagy inhibition is generally considered to be a promising therapeutic strategy. However, the paucity of effective and specific autophagy inhibitors limits its application.Purpose: The objective of this study was to explore the effect of DCA, small molecular antitumor agent, on the autophagy regulation and chemosensitization in NSCLC cells.Methods: We investigated the autophagy regulation of dichloroacetate (DCA) by laser confocal microscopy and western blotting in A549 and H1975 cell lines. The MTT assay and flow cytometry was performed for explore the chemosensitization effectiveness of DCA. The results were verified with subcutaneous tumor model in nude mice and the immunohistochemistry was applied for assessing the level of cell apoptosis and autophagy in vivo post treatment.Results: We found that DCA, which exhibited antitumor properties in various carcinoma models, induced apoptosis of non-small cell lung cancer cells (NSCLC) by inhibiting cancer cell autophagy. Furthermore, Perifosine, an AKT inhibitor, can greatly weaken the capacity of inducing apoptosis by DCA. The results indicate that the AKT-mTOR pathway, a main negative regulator of autophagy, is involved in the DCA-induced inhibition of autophagy. Then, we detected the effectiveness of autophagy inhibition by DCA. When used in co-treatment with the chemotherapeutic drug paclitaxel (PTX), DCA markedly decreased cell autophagy, enhanced apoptosis and inhibited proliferation in A549 and H1975 cells. The results of the xenograft experiment demonstrate that co-treatment of PTX and DCA can significantly decrease cell proliferation in vivo and prolong the survival of mice.Conclusion: Our results suggest that DCA can inhibit cell autophagy induced by chemotherapeutics, providing a new avenue for cancer chemotherapy sensitization. Keywords: DCA, autophagy, multi-drug resistance, non-small-cell lung cancer, paclitaxel, xenograft nude mice, chemosensitization INTRODUCTION Non-small-cell lung cancer (NSCLC) is one of the leading causes of cancer mortality worldwide. It is the most commonly occurring cancer in men and women, with an incidence greater than the combined incidence of breast, cervical, and colorectal cancers.[1,2] Although chemotherapy is still the most important means of adjuvant therapy for inoperable cancer patients and patients undergoing surgery, the clinical benefits of platinum- and paclitaxel-based postoperative chemotherapies are modest, especially in advanced NSCLC. At the same time, the adverse drug reactions have become more…

EFFECTIVENESS OF SODIUM DICHLOROACETATE AGAINST GLIOMA С6 DEPENDS ON ADMINISTRATION SCHEDULE AND DOSAGE

A.G. Fedorchuk, O.N. Pyaskovskaya, G.V. Gorbik, I.V. Prokhorova, D.L. Kolesnik, G.I. Solyanik 1R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, Kyiv 03022, Ukraine. Correspondence: [email protected]: 17 May 2016 Abstract Background: Anticancer action of sodium dichloroacetate (DCA) could be related to its ability to activate oxidative phosphorylation leading to enhanced generation of reactive oxygen species and induction of apoptosis. On the other hand, activation of oxidative phosphorylation could promote tumor cell survival, in particular, via increased ATP synthesis. Such ambiguous effects of DCA could influence its anticancer effectiveness, depending on biological properties of a tumor, schedule of DCA administration and its dosage. The aim of the study was to analyze anticancer effect of DCA against glioma С6 in rats under conditions of different schedules of its administration and various dosages. Materials and methods: The study was carried out in Wistar rats with intracerebrally transplanted glioma С6 cells. Therapy with DCA was performed as follows: daily for 6 days starting from the second day after tumor cell transplantation (schedule І) or 7(th) day (schedule ІІ) at a dose of 1.0 g/kg, or daily for 13 days starting from the second day at doses of 1.0; 1.5 or 4.5 g/kg (schedule ІІІ). An influence of hypoxia on anticancer effect of DCA was studied using hypoxic chambers where oxygen content was maintained at a level of 12.5-13% for 3 h after DCA administration to glioma С6 bearing rats. The state of mitochondrial electron transport chain components in tumor cells was studied using electron paramagnetic resonance. Results: It has been shown that therapy with DCA using schedule I resulted in 15% decrease of animals life span (LS; < 0.05), while the use of schedule II had no effect on this index. Prolonged administration of DCA (schedule ІІІ) resulted in significant antitumor effect and increased LS of rats by 25.5% (p < 0.05). Under hypoxic conditions, treatment with DCA resulted in a significant increase of animal LS by 15-22%. Dosage of DCA had a moderate effect of its anticancer action. Maximal effect, an increase of LS by 34.5% (p < 0.05) was detected at a dose of 1.5 g/kg. It has been shown that anticancer activity of DCA under all studied conditions is not related to its influence on a functional state of tumor cell mitochondria. Conclusion: Anticancer effect of DCA significantly depends on a schedule of its administration; being administered at equal total dose, but dependent on the schedule DCA could cause ambiguous effects varying from tumor growth stimulation to significant anticancer activity. Under hypoxic conditions, anticancer efficacy of DCA against glioma С6 is significantly enhanced. Keywords: sodium dichloroacetate, glioma С6, mitochondrial electron transport chain.Abbreviations used: DCA — sodium dichloroacetate; EPR — electron paramagnetic resonance; LS — life span; MtETC — mitochondrial electron transport chain; PDH — pyruvate dehydrogenase kinase; ROS — reactive oxygen species. INTRODUCTION According to statistics of World Health Organization, an average rate of brain tumors incidence is 10.9–12.8 per 100 000 of population [1]. Nearly 60% of all tumors of central nervous…

Dichloroacetate reverses the hypoxic adaptation to bevacizumab and enhances its antitumor effects in mouse xenografts

Krishan Kumar1, Simon Wigfield2, Harriet E. Gee2,5, Cecilia M. Devlin1, Dean Singleton2, Ji-Liang Li2, Francesca Buffa2, Melanie Huffman1, Anthony L. Sinn3, Jayne Silver3, Helen Turley2, Russell Leek2, Adrian L. Harris2 & Mircea Ivan4 1 Department of Medicine, Indiana University, Indianapolis, IN 46202, USA2 Department of Oncology, Weatherall Institute of MolecularMedicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UKe-mail: [email protected] In Vivo Therapeutics Core, Indiana University, Indianapolis, IN 46202, USA4 Department of Medicine, Immunology and Microbiology, Indiana University, 980W. Walnut Street, Room C225, Indianapolis, IN 46202, USAe-mail: [email protected] Department of Radiation Oncology, Sydney Cancer Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales 2050, Australia Received: 7 November 2012 Revised: 20 December 2012Accepted: 2 January 2013Published Online: 30 January 2013 Abstract Inhibition of vascular endothelial growth factor increases response rates to chemotherapy and progression-free survival in glioblastoma. However, resistance invariably occurs, prompting the urgent need for identification of synergizing agents. One possible strategy is to understand tumor adaptation to microenvironmental changes induced by antiangiogenic drugs and test agents that exploit this process. We used an in vivo glioblastoma-derived xenograft model of tumor escape in presence of continuous treatment with bevacizumab. U87-MG or U118-MG cells were subcutaneously implanted into either BALB/c SCID or athymic nude mice. Bevacizumab was given by intraperitoneal injection every 3 days (2.5 mg/kg/dose) and/or dichloroacetate (DCA) was administered by oral gavage twice daily (50 mg/kg/dose) when tumor volumes reached 0.3 cm3 and continued until tumors reached approximately 1.5–2.0 cm3. Microarray analysis of resistant U87 tumors revealed coordinated changes at the level of metabolic genes, in particular, a widening gap between glycolysis and mitochondrial respiration. There was a highly significant difference between U87-MG-implanted athymic nude mice 1 week after drug treatment. By 2 weeks of treatment, bevacizumab and DCA together dramatically blocked tumor growth compared to either drug alone. Similar results were seen in athymic nude mice implanted with U118-MG cells. We demonstrate for the first time that reversal of the bevacizumab-induced shift in metabolism using DCA is detrimental to neoplastic growth in vivo. As DCA is viewed as a promising agent targeting tumor metabolism, our data establish the timely proof of concept that combining it with antiangiogenic therapy represents a potent antineoplastic strategy. Keywords: Dichloroacetate; Hypoxia; Bevacizumab; Oxidative phosphorylation; Glycolysis © Springer-Verlag Berlin Heidelberg 2013 INTRODUCTION Molecular therapies targeting neo-angiogenesis and, in particular, vascular endothelial growth factor (VEGF) have shown antitumor activity in a variety of clinical contexts [1]. Glioblastoma (GBM) is a highly vascularized and lethal primary brain tumor, with median survival of approximately 12–14 months, and therefore, represents an important target for antiangiogenic drugs [2]. Bevacizumab, a humanized anti-VEGF antibody, is currently approved by the Food and Drug Administration as a second-line treatment of GBM, and ongoing clinical trials aim at assessing its potential as a first-line agent [3]. However, as VEGF blockade prolongs progression-free survival but not overall survival, it is imperative to identify strategies that increase its impact and delay the onset of resistance [4]. For example, limited progress has been achieved in combination with irinotecan; however, no impact on…

Overexpression of pyruvate dehydrogenase kinase supports dichloroacetate as a candidate for cutaneous melanoma therapy

Helena Populo1,2, Regina Caldas1,2,3, Jose Manuel Lopes1,2,4,5, Joana Pardal5, Valdemar Maximo1,2,4 & Paula Soares†,2,4 † Institute for Research and Innovation in Health (Instituto de Investigacao e Inovacao em Saude), University of Porto, Porto, Portugal1 Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), University of Porto, Porto, Portugal Tel: +22 557 0700; Fax: +22 557 0799; E-mail: [email protected] Institute for Research and Innovation in Health (Instituto de Investigacao e Inovac¸aoem Saude), University of Porto, Porto, Portugal 3 Faculty of Medicine, University of Porto, Porto, Portugal 4 Department of Pathology and Oncology, Faculty of Medicine, University of Porto, Porto, Portugal 5 Service of Anatomic Pathology Hospital Sao Joao, Porto, Portugal Published Online: 14 May 2015 Abstract Objective: We aimed to verify if there is evidence to consider dichloroacetate (DCA), which inhibits the pyruvate dehydrogenase kinase (PDK) and reverts the metabolic shift of cancer cells from glycolysis to oxidative phosphorylation, as a promising drug for therapy of cutaneous melanoma (CM) patients. Research design and methods: We assessed the expression profile of PDK 1, 2 and 3 in a series of melanoma samples, to verify if melanoma tumors express the DCA targets, if this expression correlates with the activation of important signaling cascades for melanomagenesis and also with the prognosis of melanoma patients. We also established the sensitivity of melanoma cell lines to DCA treatment, by assessing their metabolic alterations, proliferation and survival. Results: We observed that both PDK 1 and 2 isoforms are overexpressed in CM compared to nevi, this expression being associated with the expression of the mTOR pathway effectors and independent of the BRAF mutational status. Melanoma cell lines treated with DCA showed a shift in metabolism, that is, a decrease in glucose consumption and lactate production, downregulation of proliferation, an increase of apoptosis and a decrease in mTOR pathway activation. Conclusion: Our results suggest that PDK expression may play a role in melanoma development and that DCA can be useful for CM therapy, alone or in combination with mTOR inhibitors. Keywords: dichloroacetate, melanoma, metabolism, mTOR, pyruvate dehydrogenase kinase © 2015 Informa UK, Ltd. ISSN 1472-8222, e-ISSN 1744-7631 INTRODUCTION Cutaneous melanoma (CM) is a very aggressive malignancy, and despite being the least common type of skin cancer, it is responsible for the majority of skin cancerrelated deaths. As the incidence of CM is rising, it is at present the most probable invasive cancer to develop before the age of 50 years in male gender [1,2]. Exposure to UV radiation is considered the main risk factor for melanomagenesis [2]. CM can be classified in different histological subtypes, being the most common superficial spreading melanoma (SSM), followed by nodular melanoma (NM), lentigo maligna melanoma (LMM) and acral lentiginous melanoma (ALM). SSM and NM arise in the skin with intermittent sun exposure, whereas LMM occurs in chronic sundamaged skin and ALM is restricted to skin with no sun exposure. This histological classification has no prognostic value [3,4]. Staging of CM takes into account tumor thickness, ulceration, mitotic rate, node involvement and the presence…

Dichloroacetate (DCA) Sensitizes BothWild-Type and Over Expressing Bcl-2Prostate Cancer Cells InVitroto Radiation

Wengang Cao,1,3 Saif Yacoub,1,3 Kathleen T. Shiverick,2,3 Kazunori Namiki,1,3 Yoshihisa Sakai,1,3 Stacy Porvasnik,1,3 Cydney Urbanek,1,3 and Charles J. Rosser1,2,3* 1 Department of Urology, University of Florida, Gainesville, Florida2 Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida3 Prostate CancerTranslationalWorking Group, University of Florida, Gainesville, Florida Correspondence: Dr. Charles J. Rosser, MD, Department of Urology, University of Florida College of Medicine, Suite N215, PO Box 100247, Gainesville, FL 3210. E-mail: [email protected]: 28 February 2008Accepted: 1 April 2008Published: 8 May 2008 Abstract Background: Bcl-2 protects cells from apoptosis and provides a survival advantage to cells over-expressing this oncogene. In addition, over expression of Bcl-2 renders cell resistant to radiation therapy. Recently, dichloroacetate (DCA) was proven to potentiate the apoptotic machinery by interacting with Bcl-2. In this study, we investigated whether treating human prostate cancer cells with DCA could modulate Bcl-2 expression and if the modulation in Bcl-2 expression could render the Bcl-2 over expressing cells more susceptible to cytotoxicity effects of radiation. Methods: PC-3-Bcl-2 and PC-3-Neo human prostate cancer cells treated with DCA in addition to irradiation were analyzed in vitro for changes in proliferation, clonogenic survival, apoptosis, cell cycle phase distribution, mitochondrial membrane potential, and expression of Bcl-2, Bcl-xL, Bax, or Bak proteins. Results: DCA alone produced significant cytotoxic effects and was associated with G1 cell cycle arrest. Furthermore, DCA was associated with an increased rate of apoptosis. The combination of DCA with irradiation sensitized both cell lines to radiation's killing effects. Treatment of PC-3-Bcl-2 or PC-3-Neo with DCA and irradiation resulted in marked changes in various members of the Bcl-2 family. In addition, DCA therapy resulted in a significant change in mitochondria membrane potential, thus supporting the notion that DCAs effect is on the mitochondria. Conclusions: This is the first study to demonstrate DCA can effectively sensitize wild-type and over expressing Bcl-2 human prostate cancer cells to radiation by modulating the expression of key members of the Bcl-2 family. Together, these findings warrant further evaluation of the combination of DCA and irradiation. Keywords: dichloracetate; radiation; prostate cancer; Bcl-2 Prostate 68: 1223–1231, 2008.© 2008 Wiley-Liss, Inc. INTRODUCTION Recurrence after definitive radiation therapy for localized prostate cancer is a common phenomenon occurring in 33–56% of men [1]. Recently, radiation dose escalation has resulted in improved prostate cancer control outcomes [2,3]. Since there is an increased risk of complications in nearby critical structures, the amount of radiation that can be delivered is limited and thus dose escalation is likely not the ultimate solution to overcome radiation resistance. Instead, investigators have turned to strategies for sensitizing prostate tumors to the effects of irradiation [4–7]. However, all such strategies tested over the past 20 years have involved systemic administration of agents whose own unique side effect profiles almost always limit pharmacologic doses to levels below those needed to actually sensitize tumors to irradiation. Moreover, none of the sensitizing strategies tested to date are available for widespread use. Multiple studies have consistently implicated two genes related to apoptosis, p53 and Bcl-2, as being important in post radiation therapy prostate cancer recurrence [8–13]. Specifically, aberration of these genes can induce faulty mitochondria and apoptotic pathways [14]. Recently, researchers…

The effect of dichloroacetate in canine prostate adenocarcinomas and transitional cell carcinomas in vitro

Tatjana Harting1, Mandy Stubbendorff 2, Saskia Willenbrock1, Siegfried Wagner1, Patrik Schadzek3, Anaclet Ngezahayo3, Hugo Murua Escobar1, Ingo Nolte1 1 Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, D-30559 Hannover2 Division of Medicine Clinic III, Hematology, Oncology and Palliative Medicine, University of Rostock, D-18057 Rostock3 Evotec AG, D-22419 Hamburg4 Institute of Biophysics, Leibniz University, D-30419 Hannover, Germany Correspondence: Professor Ingo Nolte, Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, Bünteweg 9, D-30559 Hannover, Germany; E-mail: [email protected]: 12 July 2016Accepted: 5 September 2016Published Online: 5 October 2016 Abstract The Warburg effect describes the ability of cancer cells to produce energy via aerobic glycolysis instead of oxidative phosphorylation of pyruvate. This deviation in mitochondrial metabolism inhibits apoptosis, allowing increased proliferation under conditions of reduced oxygen levels. Dichloroacetate (DCA) was successfully used in several human cancer cell lines to reactivate oxidative phosphorylation in mitochondria. The aim of this study was the characterization and response of canine cancer cell lines after DCA exposure. The effect of 10 mM DCA was characterized in vitro on a set of six canine prostate adenocarcinoma and transitional cell carcinoma (TCC) derived cell lines. Cell counts, lactate levels, apoptosis, expression of apoptotic proteins, survival factors and different miRNAs were analyzed. Additionally, metabolic activity, mitochondrial activity and proliferation were investigated. DCA significantly decreased cell number of all but one utilized cell lines and leads to a significant reduction of lactate release. Decreased survivin levels were found in all cell lines, two of which presented a significant reduction in metabolic activity. Increased miR-375 levels were measured in all TCC cell lines. Reactivation of pyruvate dehydrogenase and an elevated mitochondrial activity appear to induce the transition from aerobic glycolysis back to oxidative phosphorylation. Further, these results display that DCA treatment has a suppressant effect on proliferation of canine cancer cells. Keywords: dichloroacetate, canine prostate adenocarcinoma, canine transitional cell carcinoma, Warburg effectDOI: 10.3892/ijo.2016.3720 INTRODUCTION In the last few years cancer treatments such as chemotherapy, radiation therapy and surgery, as they are used in human cancer treatment, became more important in veterinary medicine. Conventional chemotherapeutic agents target dividing cells, cancerous as well as non-neoplastic cells, causing several side effects as myelosuppression, diarrhea, vomitus and anorexia [1]. Further, due to advanced disease stage and resistance of prostate and bladder cancer, treatment is difficult and often associated with poor prognosis [2,3]. Therefore, new alternatives which are more effective have to be investigated. Dichloroacetate (DCA), a small and cost-efficient molecule, affects different metabolic pathways by inhibiting pyruvate dehydrogenase kinase (PDK) [4,5]. This implicates that pyruvate dehydrogenase (PDH) is potentially indirectly activated by DCA which yields a metabolic shift to favor oxidation of pyruvate to acetyl-co-enzyme-A in mitochondria [5]. Despite this fact in the past decades DCA has been used in the treatment of a multitude of disorders like congenital lactate acidosis [6,7], hypercholesterolemia [8], hyperglycemia [9], congestive heart failure [10] and only recently in cancer research [11–16]. DCA was tested in different in vitro approaches in the field of human oncology including colorectal cancer [17,18], endometrial cancer [14], oral squamous cell carcinoma [19] and breast…

Inhibition of the pentose phosphate pathway by dichloroacetate unravels a missing link between aerobic glycolysis and cancer cell proliferation

Géraldine De Preter1, Marie-Aline Neveu1, Pierre Danhier1, Lucie Brisson2, Valéry L. Payen2, Paolo E. Porporato2, Bénédicte F. Jordan1, Pierre Sonveaux2 and Bernard Gallez1 1 Louvain Drug Research Institute (LDRI), Biomedical Magnetic Resonance Research Group, Université Catholique de Louvain (UCL), Brussels, Belgium2 Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology, Université Catholique de Louvain (UCL), Brussels, Belgium Correspondence: Bernard Gallez, email: [email protected]: 1 June 2015Accepted: 26 December 2020Published: 9 December 2020 Abstract Glucose fermentation through glycolysis even in the presence of oxygen (Warburg effect) is a common feature of cancer cells increasingly considered as an enticing target in clinical development. This study aimed to analyze the link between metabolism, energy stores and proliferation rates in cancer cells. We found that cell proliferation, evaluated by DNA synthesis quantification, is correlated to glycolytic efficiency in six cancer cell lines as well as in isogenic cancer cell lines. To further investigate the link between glycolysis and proliferation, a pharmacological inhibitior of the pentose phosphate pathway (PPP) was used. We demonstrated that reduction of PPP activity decreases cancer cells proliferation, with a profound effect in Warburg-phenotype cancer cells. The crucial role of the PPP in sustaining cancer cells proliferation was confirmed using siRNAs against glucose-6-phosphate dehydrogenase, the first and rate-limiting enzyme of the PPP. In addition, we found that dichloroacetate (DCA), a new clinically tested compound, induced a switch of glycolytic cancer cells to a more oxidative phenotype and decreased proliferation. By demonstrating that DCA decreased the activity of the PPP, we provide a new mechanism by which DCA controls cancer cells proliferation. Keywords: bioenergetics, glycolysis, dichloroacetate, pentose phosphate pathway, proliferation © 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/). INTRODUCTION These last few years, metabolism has generated tremendous interest in the field of cancer research. Many studies focused on the various metabolic profiles of different tumors [1-3] because metabolic plasticity is involved in cancer progression, drug resistance and metastasis [4-6]. In normal cells, glycolysis is coupled to oxidative phosphorylation (OXPHOS) to optimally synthesize intracellular ATP from glucose [7]. However, many cancer cells undergo a fundamental metabolic transformation, the “glycolytic switch”, by which glycolysis is uncoupled from respiration and rewired to lactic fermentation, thus becoming the primary source of cell ATP production. Switching to a glycolytic metabolism primarily occurs under hypoxia as a rescue mechanism for energy production. However, some cancer cells further adopt a particular glycolytic phenotype, first described by Warburg [8] and coined ‘aerobic glycolysis’ [9]. The biological rationale behind the Warburg phenotype remains controversial, but it has been recently proposed that proliferating cancer cells enhance glycolysis because it benefits both bioenergetics and biosynthesis [4,10]. Indeed, a glycolytic metabolism potentially allows fast ATP production and provides carbon intermediates that can be directed to branched biosynthetic pathways, enabling a faster expansion of the cellular biomass. Convincingly, mutations occurring in signaling pathways regulating both cellular biosynthesis and aerobic glycolysis, such as the PI3K/Akt/mTOR…

Dichloroacetate enhances apoptotic cell death via oxidative damage and attenuates lactate production in metformin-treated breast cancer cells

Allison B. Haugrud, Yongxian Zhuang, Joseph D. Coppock, W. Keith Miskimins Cancer Biology Research Center, Sanford Research, 2301 E. 60th St North, Sioux Falls, SD 57104, USAe-mail: [email protected] Received: 24 April 2014 Accepted: 5 September 2014Published: 12 September 2014 Abstract The unique metabolism of breast cancer cells provides interest in exploiting this phenomenon therapeutically. Metformin, a promising breast cancer therapeutic, targets complex I of the electron transport chain leading to an accumulation of reactive oxygen species (ROS) that eventually lead to cell death. Inhibition of complex I leads to lactate production, a metabolic byproduct already highly produced by reprogrammed cancer cells and associated with a poor prognosis. While metformin remains a promising cancer therapeutic, we sought a complementary agent to increase apoptotic promoting effects of metformin while attenuating lactate production possibly leading to greatly improved efficacy. Dichloroacetate (DCA) is a well-established drug used in the treatment of lactic acidosis which functions through inhibition of pyruvate dehydrogenase kinase (PDK) promoting mitochondrial metabolism. Our purpose was to examine the synergy and mechanisms by which these two drugs kill breast cancer cells. Cell lines were subjected to the indicated treatments and analyzed for cell death and various aspects of metabolism. Cell death and ROS production were analyzed using flow cytometry, Western blot analysis, and cell counting methods. Images of cells were taken with phase contrast microscopy or confocal microscopy. Metabolism of cells was analyzed using the Seahorse XF24 analyzer, lactate assays, and pH analysis. We show that when DCA and metformin are used in combination, synergistic induction of apoptosis of breast cancer cells occurs. Metformin-induced oxidative damage is enhanced by DCA through PDK1 inhibition which also diminishes metformin promoted lactate production. We demonstrate that DCA and metformin combine to synergistically induce caspase-dependent apoptosis involving oxidative damage with simultaneous attenuation of metformin promoted lactate production. Innovative combinations such as metformin and DCA show promise in expanding breast cancer therapies. Keywords: Metformin; Dichloroacetate; Breast cancer; Lactate; Apoptosis © Springer Science+Business Media New York 2014 INTRODUCTION Cancer metabolism is a developing into a promising area for development of new therapeutic approaches. Compared to the normal cells from which they derive, cancer cells are metabolically reprogrammed, preferentially utilizing glycolysis even under conditions of sufficient oxygen, a phenomenon known as the Warburg effect [1]. To compensate for lost ATP as a result of preferential glycolysis (vs. progression through oxidative phosphorylation), cancer cells up-regulate genes encoding glucose transporters and glycolytic enzymes such as pyruvate dehydrogenase kinase (PDK) and lactate dehydrogenase (LDH). This high rate of glucose uptake and altered metabolism not only provides ATP and allows cells to survive under hypoxic conditions; it also provides biosynthetic building blocks such as intermediates and substrates for the production of amino acids, NADPH, and ribose-5-phosphate which are essential for nucleotide, protein, and membrane synthesis necessary in rapidly dividing cells. This also means the mitochondrial TCA cycle generates a lower percentage of ATP, allowing citrate to be used in fatty acid and lipid biosynthesis for the production of new membranes [2]. Much…

Sensitization of metformin-cytotoxicity by dichloroacetate via 4 reprogramming glucose metabolism in cancer cells

Yong Won Choi1, In Kyoung Lim * 1 Department of Biochemistry and Molecular Biology, BK21 Cell Transformation and Restoration Project, Ajou University School of Medicine, Suwon 443-721, Republic of Korea* Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, San 5 Woncheon-dong, Yeongtonggu, Suwon 443-721, Republic of Korea; Tel.: +82 31 219 5051; fax: +82 31 219 5059. E-mail address: [email protected] (I.K. Lim).Received: 29 October 2013Accepted: 20 January 2014Revised: in revised form 8 January 2014Published/Available online: 27 January 2014 Abstract To investigate sensitization of metformin-cytotoxicity, cancer cells were treated with dichloroacetate (DCA), an inhibitor of pyruvate dehydrogenase kinase (PDK). Metformin-cytotoxicity was mainly dependent on glucose availability and reducing power generated by pentose phosphate pathway, whereas DCA cotreatment enhanced metformin-cytotoxicity via reprogramming glucose metabolism by inhibiting PDK and increasing mitochondrial respiration. DCA cotreatment elicited cell death rather than cell survival despite high glucose and high GSH condition. In conclusion, DCA sensitized metformin-cytotoxicity by reprogramming glucose metabolism in part from aerobic glycolysis to mitochondrial oxidation, evidenced by measurements of glucose consumption, lactate release, and the ratio of oxygen consumption rate/extracellular acidification rate. Keywords: Metformin Dichloroacetate (DCA), Oxidative stress, Glucose deprivation, Glutathione contents © 2014 Published by Elsevier Ireland Ltd. INTRODUCTION Growing evidences indicate that metabolic perturbations of cancer cells such as aerobic glycolysis or glutamine addiction are inevitable for carcinogenesis beyond the epiphenomenon [13], [31], and enormous amount of efforts have been exhausted to find out drug-able targets and candidate chemicals on cancer metabolism [5], [30]. Based on the epidemiological, preclinical and clinical data, metformin, a biguanide used for treatment of diabetes mellitus, became one of the most attractive and promising drugs targeting for cancer metabolism. Although the mechanism of metformin is not fully elucidated, intracellular function of metformin has been known to inhibit respiratory chain complex I [8], [21]. At present, many evidences indicate that AMPK activation is a master node of anti-tumor effects of metformin [3], [16],[23], therefore, LKB1−/− cancer cells are more resistant to metformin-induced cytotoxicity in the in vitro culture system [22], [34]. In contrast, AMPK activation has been shown to protect cancer cells from energy stress via regulation of NADPH homeostasis [12], therefore, LKB1−/− cancer cells are more sensitive to phenformin-induced metabolic stress in mouse lung cancer model [24]. In the confusing contexts, there are reports that anti-tumor effects metformin are dependent on glucose concentration in culture medium [11], [17], [25]; Glucose-deprivation significantly enhances metformin-cytotoxicity [17], whereas metformin-induced AMPK activation is inefficient under high glucose (25 mM) condition [25]. Therefore, in-depth understanding of biochemical mechanism of metformin-cytotoxicity needs to be clarified before using metformin as an anticancer agent focusing on glucose metabolism. Warburg effect frequently observed in cancer cells is important not only for energy generation but also for maintaining reduced status under hostile tumor microenvironment. Glucose is the main source of reducing power, NADPH, via pentose phosphate pathway [1], [9], [22]. Indeed, 2-deoxyglucose (2DG)-mediated cancer cell death is dependent mainly on the intolerable oxidative stress in addition to energy crisis [15]. Based on the concept that increased glycolysis protects cancer cells from oxidative stress, metformin-increased glycolysis has been thought to be capable of protecting cells from mitochondrial oxidative stress resulting from the inhibition of respiratory chain complex I [35]. Therefore, we investigated whether metformin-induced mitochondrial stress can be augmented by oxidative stress due to GSH depletion under glucose deprivation or H2O2 treatment under glucose-sufficient condition. Based…

Targeting Cancer Cell Metabolism with Metformin, Dichloroacetate and Memantine in Glioblastoma (GBM)

Gulsah ALBAYRAK1 , Ece KONAC1 , Umit Akin DERE2 , Hakan EMMEZ2 1 Gazi University, Faculty of Medicine, Department of Medical Biology and Genetics, Ankara, Turkey 2 Gazi University, Faculty of Medicine, Department of Neurosurgery, Ankara, Turkey Correspondence: Ece KONAC; E-mail: [email protected]: 16 January 2020Accepted: 3 July 2020Published: 29 December 2020 Abstract AIM: To investigate the effects of metformin, dichloroacetate (DCA), and memantine on T98G and U87-MG human glioblastoma (GBM) cells to target tumor cell metabolism in a multi-directional manner. MATERIAL and METHODS: IC50 levels for metformin, DCA, metformin+DCA and memantine were determined by MTT assay in T98G and U87-MG cells in vitro. Casp3, Bcl-2, Bax, c-Myc and GSK-3B protein expressions were investigated post treatments. Fifteen GBM+ tumor tissues were assessed for Casp-3, Bcl-2, Bad, Bax for apoptotic protein expression patterns. RESULTS: Cancer cell metabolism targeting drugs metformin, DCA, metformin+DCA and memantine induced cytotoxicity in a dose-dependent manner in T98G and U87-MG cells. IC50 for memantine is found as 0.5 mM (p<0.01) which is nearly 10 times lower concentration than that of metformin. Fifteen GBM+ tumor tissues had differential apoptotic protein expressions. CONCLUSION: Memantine exerted anti-cancer mechanism of action in T98G and U87-MG cells, however, such a mechanism requires deeper investigation for GBM treatment. Keywords: Glioblastoma, Cancer cell, Metabolism, Metformin, Dichloroacetate, MemantineDOI: 10.5137/1019-5149.JTN.29176-20.3 INTRODUCTION Glioblastoma (GBM) is one of the most aggressive tumors of the central nervous system, and represents approximately 50% of all glial tumor types [12,27]. The median survival rate for GBM patients does not change signicantly using the current standard of care treatments, which involve tumor resection followed by radiotherapy and Temozolomide treatment. The median survival is approxi-mately 12-14 months despite the combined use of surgery, radiotherapy and chemotherapy [7,23]. GBM tumors have a wide range of genetic variations leading to dierent therapeutic responses [5,9,21]. Intratumor hetero-geneity might be the key to identify the cause of treatment failure [22]. Chemotherapy drugs such as Temozolamide in-creases the mutational load within the cancer genome when compared to the untreated GBM cells [17]. Alternative therapy approaches, therefore, are urgently needed in GBM treatment. Metformin is a common anti-diabetic drug used in Type 2 diabetes treatment [18]. Metformin treatment is found to be associated with a lower risk of several cancers, however, its effects on GBM has not been well characterized [4,24]. Metformin treatment decreases Temozolomide resistance in GBM cells [26]. Metformin, however, has safety concerns in clinical setting as most of the preclinical works used supraphysiological doses of metformin [25]. In this study we aimed to overcome this challenge by investigating the effect of metformin by combining it with dichloroacetate (DCA) that targets cancer cell metabolism via pyruvate dehydrogenase kinase inhibition [11]. Targeting cancer cell metabolism might have implications for the treatment of aggressive GBM. Furthermore, we aimed to investigate the apoptotic protein expression profiles to better understand GBM at a molecular level. We also aimed to interfere cancer cell metabolism using metformin, DCA and memantine in T98G and U87-MG GBM cell lines. Materials and methods Cell Culture and…

Dichloroacetate induces apoptosis and cell-cycle arrest in colorectal cancer cells

BM Madhok*,1, S Yeluri1 , SL Perry1 , TA Hughes2 and DG Jayne1 1 Section of Translational Anaesthesia & Surgery, University of Leeds, Level 7 Clinical Sciences Building, St. James’s University Hospital, Leeds, UK2 Leeds Institute of Molecular Medicine, University of Leeds, St. James’s University Hospital, Leeds, UKCorrespondence: Dr BM Madhok; E-mail: [email protected]: 23 March 2010Accepted: 26 April 2010Published: 18 May 2010 Abstract BackgroundCancer cells are highly dependent on glycolysis. Our aim was to determine if switching metabolism from glycolysis towards mitochondrial respiration would reduce growth preferentially in colorectal cancer cells over normal cells, and to examine the underlying mechanisms. MethodsRepresentative colorectal cancer and non-cancerous cell lines were treated with dichloroacetate (DCA), an inhibitor of pyruvate dehydrogenase kinase. ResultsDichloroacetate (20 mM) did not reduce growth of non-cancerous cells but caused significant decrease in cancer cell proliferation (P=0.009), which was associated with apoptosis and G2 phase cell-cycle arrest. The largest apoptotic effect was evident in metastatic LoVo cells, in which DCA induced up to a ten-fold increase in apoptotic cell counts after 48 h. The most striking G2 arrest was evident in well-differentiated HT29 cells, in which DCA caused an eight-fold increase in cells in G2 phase after 48 h. Dichloroacetate reduced lactate levels in growth media and induced dephosphorylation of E1α subunit of pyruvate dehydrogenase complex in all cell lines, but the intrinsic mitochondrial membrane potential was reduced in only cancer cells (P=0.04). ConclusionsPyruvate dehydrogenase kinase inhibition attenuates glycolysis and facilitates mitochondrial oxidative phosphorylation, leading to reduced growth of colorectal cancer cells but not of non-cancerous cells. Keywords: dichloroacetate, colorectal cancer, pyruvate dehydrogenase, pyruvate dehydrogenase kinaseBritish Journal of Cancer (2010) 102, 1746 – 1752, www.bjcancer.comdoi: 10.1038/sj.bjc.6605701 © 2010 Cancer Research UK INTRODUCTION Colorectal cancer is the third most common cancer in the world and the fourth leading cause of cancer-related death (Shike et al, 1990). In 2007 colorectal cancer accounted for 17.1 deaths per 100 000 persons in the United Kingdom (UK Bowel Cancer Statistics, 2009). Despite recent advances, the prognosis of patients with advanced and metastatic colorectal cancer remains poor. Targeting tumour metabolism for cancer therapy is a rapidly developing area (Pan and Mak, 2007). Early observations concerning the metabolic differences between cancer and normal cells were made by Otto Warburg, who showed that cancer cells are inherently dependent on glycolysis for production of chemical energy (Warburg, 1956). There is now mounting evidence that this increased glycolysis results from the influence of multiple molecular pathways, including adaptive responses to the hypoxic tumour microenvironment, oncogenic signalling, and mitochondrial dysfunction (Gatenby and Gillies, 2004; Gillies and Gatenby, 2007; Wu et al, 2007). The glycolytic phenotype offers growth advantages to cancer cells by resisting apoptosis, and facilitating tumour spread and metastasis (Yeluri et al, 2009). A key regulator of cellular metabolism is pyruvate dehydrogenase (PDH). Pyruvate dehydrogenase converts pyruvate, produced from glycolysis, to acetyl-CoA, which is oxidised in the tricarboxylic acid cycle within mitochondria. Pyruvate dehydrogenase activity is tightly regulated by inhibitory phosphorylation by pyruvate dehydrogenase kinase (PDK). Phosphorylation occurs on the E1α sub-unit of PDH (PDHE1α) at three sites: Ser232, Ser293,…

Reversal of the glycolytic phenotype by dichloroacetate inhibits metastatic breast cancer cell growth in vitro and in vivo

Ramon C. Sun, Mitali Fadia, Jane E. Dahlstrom, Christopher R. Parish, Philip G. Board, Anneke C. Blackburn R. C. Sun, P. G. Board, A. C. Blackburn Molecular Genetics Group, John Curtin School of Medical Research, Australian National University, P.O. Box 334, Canberra 2601, Australia e-mail: [email protected]. Fadia, J. E. Dahlstrom Department of Anatomical Pathology, Canberra Hospital and Australian National University Medical School, Woden ACT 2606, AustraliaC. R. Parish Cancer and Vascular Biology Group, John Curtin School of Medical Research, Australian National University, Canberra ACT 0200, AustraliaReceived: 17 April 2009 Accepted: 2 June 2009Published: 19 June 2009 Abstract The glycolytic phenotype is a widespread phenomenon in solid cancer forms, including breast cancer. Dichloroacetate (DCA) has recently been proposed as a novel and relatively non-toxic anti-cancer agent that can reverse the glycolytic phenotype in cancer cells through the inhibition of pyruvate dehydrogenase kinase. We have examined the effect of DCA against breast cancer cells, including in a highly metastatic in vivo model. The growth of several breast cancer cell lines was found to be inhibited by DCA in vitro. Further examination of 13762 MAT rat mammary adenocarcinoma cells found that reversal of the glycolytic phenotype by DCA correlated with the inhibition of proliferation without any increase in cell death. This was despite a small but significant increase in caspase 3/7 activity, which may sensitize cancer cells to other apoptotic triggers. In vivo, DCA caused a 58% reduction in the number of lung metastases observed macroscopically after injection of 13762 MAT cells into the tail vein of rats (P = 0.0001, n ≥ 9 per group). These results demonstrate that DCA has anti-proliferative properties in addition to pro-apoptotic properties, and can be effective against highly metastatic disease in vivo, highlighting its potential for clinical use. Keywords: Dichloroacetate, Breast cancer, Glycolysis, Metastasis, Animal model © Springer Science+Business Media, LLC. 2009 INTRODUCTION The glycolytic phenotype, often referred to as the Warburg effect, is a widespread phenomenon in the majority of cancer forms where high rates of glucose uptake and glycolysis occur while mitochondrial respiration is repressed, despite the presence of oxygen. This metabolic characteristic is believed to be acquired for the production of ATP during anaerobic tumor evolution; however, evidence is increasingly indicating that the glycolytic phenotype is accompanied by gene expression changes that are intimately linked to tumorigenic processes, such as resistance to apoptosis and increased metastatic potential [1,2]. The existence of the glycolytic phenotype in breast cancer has been well described. An altered bioenergetic cellular index (BEC) and a profound shift toward an enhanced glycolytic phenotype has been reported in breast cancers compared to paired normal breast tissue biopsies, and correlated with overall and disease-free survival of the patients [3,4]. The invasiveness of several breast cancer cell lines has been correlated with a higher constitutive level of the transcription factor HIF-1α in normoxic conditions and decreased induction of HIF-1α in hypoxia, as well as higher lactate production [5]. Immunohistochemically, upregulation of two markers of the glycolytic phenotype (glucose transporter GLUT1 and Na+/H+ exchanger NHE-1) was observed in microinvasive…

Activation of mitochondrial oxidation by PDK2 inhibition reverses cisplatin resistance in head and neck cancer

Jong-Lyel Roh a, *, Jin Young Park a , Eun Hye Kim a , Hye Jin Jang a , Minsu Kwon b a Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea b Department of Otolaryngology, Gyeongsang National University Hospital School of Medicine, Jinju, Republic of KoreaCorrespondence author: Tel.: +82 2 3010 3965; fax: +82 2 489 2773. E-mail address: [email protected] (J.-L. Roh). Received: 9 September 2015Revised: revised form 13 November 2015Accepted: 14 November 2015 Abstract Dichloroacetate (DCA), an orphan drug that promotes a shift from glycolysis to oxidative phosphorylation, has been repurposed for cancer therapy. The present study investigated whether DCA may overcome cisplatin resistance in head and neck cancer (HNC). Two cisplatin-resistant HNC cell lines (AMC-HN4R and -HN9R), their parental lines, and other human HNC lines were used. The effect of DCA, alone and in combination with cisplatin, was assessed by measuring cell cycle, viability, death, reactive oxygen species (ROS) production, mitochondrial membrane potential (ΔΨm), and protein expression in preclinical mouse tumor xenograft models. Increased glycolysis correlated with decreased sensitivity to cisplatin and was reduced by DCA. Cisplatin-resistant cells overexpressed pyruvate dehydrogenase kinase 2 (PDK2). DCA induced HNC cell death by decreasing ΔΨm and promoting mitochondrial ROS production. This effect was decreased by the antioxidant N-acetyl-l-cysteine or by inhibition of caspase-mediated apoptosis. Activation of mitochondrial glucose oxidation by DCA eventually activated downstream mitochondrial apoptotic signaling, leading to the death of chemoresistant cancer cells. Therefore, DCA significantly sensitized resistant HNC cells to cisplatin in vitro and in vivo. High glycolysis and PDK2 overexpression are closely linked to cisplatin resistance in HNC cells; the latter can be overcome by DCA. Keywords: Head and neck cancer, Cisplatin resistance, PDK2, Dichloroacetate, Mitochondrial remodelingAbbreviations: HNC, head and neck cancer; DCA, dichloroacetate; CDDP, cisplatin; OXPHOS, oxidative phosphorylation; PDK2, pyruvate dehydrogenase kinase 2; PDHE1α, pyruvate dehydrogenase isoform E1α; ROS, reactive oxygen species; ΔΨm, mitochondrial membrane potential; NAC, N-acetyl-l-cysteine; DCF-DA, 2′,7′- dichlorofluorescein diacetate; PARP, poly(ADP-ribose) polymerase; siRNA, short interfering RNA; 18F-FDG, 18F-fluorodeoxyglucose; PET, positron emission tomography; SUV, standardized uptake value; MTV, metabolic tumor volume; TUNEL, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling. © 2015 Elsevier Ireland Ltd. All rights reserved. INTRODUCTION Head and neck cancer (HNC) is the eighth most common cancer worldwide, with more than half a million new cases diagnosed each year [1] . The tumors arise in the upper aerodigestive tract including the oral or nasal cavity, pharynx, and larynx, and metastasize to regional lymph nodes and distant sites. The current standard of care in HNC involves a multimodal approach including surgery, chemotherapy, and radiotherapy, particularly in advanced HNC. Along with the recent interest in organ preservation strategies, non-surgical modalities, such as radiotherapy combined with chemotherapy, have been increasingly used as first-line therapy in HNC [2]. In the management of patients with advanced HNC, systemic chemotherapy is now a central component of several curative approaches, including combination with definitive radiotherapy or induction treatment, exceeding reservation just for palliation [3]. Cisplatin, the platinum derivative cis-diamminedichloroplatinum (II) (CDDP), remains a first-line chemotherapeutic agent in non-surgical modalities against HNC, despite recent advances in targeted therapy [4]. Over the last three decades, however, despite diagnostic and therapeutic advances, the overall survival rate in HNC has not changed substantially, a result of the…

Mitochondrial induction as a potential radio-sensitizer in lung cancer cells – a short report

Ronen Shavit1, Maya Ilouze, Tali Feinberg, Yaacov Richard Lawrence, Yossi Tzur, Nir Peled 1 Thoracic Cancer Research and Detection Center, Sheba Medical Center Tel Hashomer, Ramat-Gan, 52621, POB 244, Israel. e-mail: [email protected] URL: http://medicine.mytau.org/peled/ Y. R. Lawrence Center for Translational Research in Radiation Oncology, Sheba Medical Center, Ramat-Gan, Israel M. Ilouze : N. Peled Thoracic Cancer Service, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, IsraelCorrespondence: [email protected]: 10 December 2014Published: 7 January 2015 Abstract Introduction: Lung cancer is the leading cause of cancer death. Radiation therapy plays a key role in its treatment. Ionizing radiation induces cell death through chromosomal aberrations, which trigger mitotic catastrophe and apoptosis. However, many lung cancer patients show resistance to radiation. Dichloroacetate (DCA) is a small molecule that can promote mitochondrial activation by increasing the influx of pyruvate. Here, we tested whether DCA may increase the sensitivity of non-small cell lung cancer (NSCLC) cells to radiation through this mechanism. Methods: Two representative NSCLC cell lines (A549 and H1299) were tested for their sensitivity to radiation with and without pre-exposure to DCA. The treatment efficacy was evaluated using a clonogenic survival assay. An extracellular flux analyzer was used to assess the effect of DCA on cellular oxygen consumption as a surrogate marker for mitochondrial activity. Results: We found that DCA increases the oxygen consumption rate in both A549 and H1299 cells by 60% (p = 0.0037) and 20% (p = 0.0039), respectively. Pre-exposure to DCA one hour before radiation increased the cytotoxic death rate 4-fold in A549 cells (55 to 13%, p = 0.004) and 2-fold in H1299 cells (35 to 17%, p = 0.28) respectively, compared to radiation alone. Conclusion: Mitochondrial induction by DCA may serve as a radio-sensitizer in non-small cell lung cancer. Keywords: NSCLC; DCA; Mitochondria; Radiation; Radio-sensitizer; Warburg effect © International Society for Cellular Oncology 2015 INTRODUCTION Lung cancer is the leading cause of cancer-related deaths in the United States with an overall 5-year survival rate for all stages of ~17 % [1–4]. Radiation therapy (RT) plays an important role in the clinical management of lung cancer patients, particularly those with stage IIIB disease who are candidates for definitive chemo-radiotherapy. In addition, RT may be applied as a neo-adjuvant or adjuvant therapy in stage IIIA, or in an ablative manner when stereotactic body radiotherapy (SBRT) is applied. Radiation induced pneumonitis (RIP) is the limiting factor when treating patients with RT. In order to minimize RIP, oncologists aim to keep the V20 (i.e., the percentage of lung volume receiving a radiation dose of ≥20 Gy) below 22 % [5]. Radio-sensitizers may increase the cytotoxic efficacy of radiation, thereby potentially improving cure rates without increasing the V20. RT kills cells by causing DNA double-strand breaks. Unrepaired DNA breaks result in chromosomal aberrations which, in turn, lead to “mitotic catastrophe” - a mode of cell death that results from the premature or inappropriate entry of cells into mitosis [6,7]. Furthermore, radiation may directly affect cell membranes and organelles. Although these latter changes are as yet poorly understood, they may lead to…

DCA increases the antitumor effects of capecitabine in a mouse B16 melanoma allograft and a human non‑small cell lung cancer A549 xenograft

Mao‑fa Zheng1, Si‑yu Shen, Wei‑da Huang 1Department of Biochemistry, School of Life Science, Fudan University, Handan Road 220, Shanghai, 200433, China.Email: e-mail: [email protected]: 16 May 2013Accepted: 27 August 2013Published: 17 September 2013 Abstract Purpose: Capecitabine is one of the few chemotherapy drugs with high oral availability. Recently, sodium dichloroacetate (DCA) has shown great potential as an anticancer agent. In the present study, we assessed the anticancer effect of DCA in combination with capecitabine for cancers that modestly expressed TP. Methods: A mouse B16 melanoma allograft and a human non-small cell lung cancer A549 xenograft were used to assess the effect of DCA and capecitabine combined treatment. Histology and immunohistochemistry were used to detect the apoptosis and proliferation of cancer cells. Real-time PCR and Western blot were carried out to detect the expression of TP and caspases, respectively. Results: For the first time, we report that DCA increased the antitumor effects of capecitabine in a mouse B16 allograft and a human A549 xenograft by promoting apoptosis of tumor cells. DCA has little effect on the expression of TP. Conclusions: Our finding suggests that DCA in combination with capecitabine might be potential as a new therapeutic regimen against some cancers.Keywords: DCA, Capecitabine, Combination, Antitumor effect © Springer-Verlag Berlin Heidelberg 2013Cancer Chemother Pharmacol (2013) 72:1031–1041DOI: 10.1007/s00280-013-2281-z INTRODUCTION Sodium dichloroacetate (DCA) is a small molecular salt of dichloroacetic acid with a molecular weight of 150 Da. DCA inhibits the activity of pyruvate dehydrogenase kinase, thus activating the mitochondrial enzyme complex pyruvate dehydrogenase [1] and converting the glycolytic metabolic pathway to oxidative phosphorylation. In the past 40 years, DCA has been used as an orphan drug in the treatment for children congenital lactic acidosis and other lactic acidosis complicated by other diseases [2] and has shown high efficacy and low toxicity in both preclinical and clinical trials [3]. Recently, DCA has shown great potential as an anticancer agent because of the similarities in metabolic remodeling of some tumor cells to that occurring during lactic acidosis [4]. Cancer cells, especially cancer stem cells (CSCs), resist apoptosis by producing energy through glycolysis and lactic acid fermentation, rather than oxidative phosphorylation, because of the hypoxic nature of the tumor microenvironment, a phenomenon known as the Warburg effect [5,6]. After oral administration, DCA has been shown to restore mitochondrial function and selectively promote tumor cell apoptosis in a mitochondrial-dependent pathway [7,8]. The therapeutic activities of DCA against glioblastoma have been tested in clinical trials (NCT00540176) and shown some positive results [9]. But phase II trial NCT01029925 to determine the response rate of oral dichloroacetate in patients with recurrent and/or metastatic and pretreated breast and non-small cell lung cancer was terminated due to higher than expected risk and safety concerns. So, the clinical utility of DCA for cancer control needs more careful estimation. As an apoptosis sensitizer, DCA has also been used in combination with other cancer therapies. Cao et al. [10] reported that DCA sensitized prostate cancer cells to radiation in vitro. Xiao et al. [11] determined that DCA enhanced tumor cell death when…

Dichloroacetate Induces Apoptosis in Endometrial Cancer Cells

Jason Y.Y. Wong1, Gordon S. Huggins2, Marcella Debidda4, Nikhil C. Munshi4, and Immaculata De Vivo1,3 1 Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston Massachusetts. 2 Molecular Cardiology Research Institute, Tufts-New England Medical Center, Boston Massachusetts. 3 Program in Molecular and Genetic Epidemiology, Harvard School of Public Health, Boston Massachusetts. 4 The Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts.Correspondence: Immaculata De Vivo, Brigham and Women’s Hospital, Department of Medicine, Channing Laboratory, 181 Longwood Ave, Boston, MA, 02115, USA. Phone: 617-525-2094. Fax: 617-525-2008. [email protected]: 14 September 2020Accepted: 4 December 2020Published: 9 December 2020 Abstract Purpose: A recent landmark study demonstrated that Dichloroacetate (DCA) treatment promoted apoptosis in lung, breast, and glioblastoma cancer cell lines by shifting metabolism from aerobic glycolysis to glucose oxidation coupled with NFAT-Kv1.5 axis remodeling. The objective of this study was to determine whether DCA induces apoptosis in endometrial cancer cells and to assess apoptotic mechanism. Methods: A panel of endometrial cancer cell lines with varying degrees of differentiation was treated with DCA and analyzed for apoptosis via flow cytometry. Biological correlates such as gene expression, intracellular Ca2+, and mitochondrial membrane potential were examined to assess apoptotic mechanism. Results: Initiation of apoptosis was observed in five low to moderately invasive cancer cell lines including Ishikawa, RL95-2, KLE, AN3CA, and SKUT1B while treatment had no effect on non-cancerous 293T cells. Two highly invasive endometrial adenocarcinoma cell lines, HEC1A and HEC1B, were found to be resistant to DCA-induced apoptosis. Apoptotic responding cell lines had a significant increase in early and late apoptotis, a decrease in mitochondrial membrane potential, and decreased Survivin transcript abundance, which are consistent with a mitochondrial-regulated mechanism. DCA treatment decreased intracellular calcium levels in most apoptotic responding cell lines which suggests a contribution from the NFAT-Kv1.5-mediated pathway. DCA treatment increased p53 upregulated modulator of apoptosis (PUMA) transcripts in cell lines with an apoptotic response, suggesting involvement of a p53-PUMA-mediated mechanism. Keywords: Dichloroacetate; Endometrial; Cancer; Apoptosis; Mitochondria Conflict of Interest Statement: The authors declare there are no conflicts of interest.Conclusions: Dichloroacetate effectively sensitizes most endometrial cancer cell lines to apoptosis via mitochondrial, NFAT-Kv1.5, and PUMA-mediated mechanisms. Further investigation of the cancer therapeutic potential of DCA is warranted.MMP is further confirmed via TMRM fluorescent staining in a DCA dose-response experiment. Error bars represent standard error from 2 independent experiments performed in triplicate wells. INTRODUCTION Endometrial cancer (EC) is a neoplasia of the epithelial lining of the uterine corpus. It is the most common gynecologic malignancy in the United States and the fourth leading cause of cancer death in the country among women [1]. There are few therapeutic options without serious drawbacks for those with recurrent or metastatic endometrial cancer. Chemotherapy for metastatic disease has high rates of toxicity, neuralgia, and cardiac complications[2, 3]. The impetus in future cancer therapy development will be to reduce serious adverse effects while demonstrating comparable or improved efficacy to existing treatments. Aerobic glycolysis, also known as the ‘Warburg Effect’, is a unique…

Dichloroacetate and metformin synergistically suppress the growth of ovarian cancer cells

Bo Li1,*, Xinzhe Li1,*, Zhenhong Ni1, Yan Zhang1, Yijun Zeng1, Xiaohuan Yan2, Yan Huang3, Jintao He4, Xilin Lyu1, Yaran Wu1, Yuting Wang1, Yingru Zheng2, Fengtian He1 1 Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University,Chongqing 400038, China2 Department of Obstetrics and Gynecology, Daping Hospital and Research Institute of Surgery, Third Military MedicalUniversity, Chongqing 400042, China3 Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042,China4 Battalion 17 of Students, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China* These authors have contributed equally to this work Correspondence: Yingru Zheng, email: [email protected] He, email: [email protected] Received: January 24, 2016Accepted: July 09, 2016Published: July 19, 2016 Abstract Both dichloroacetate (DCA) and metformin (Met) have shown promising antitumor efficacy by regulating cancer cell metabolism. However, the DCA-mediated protective autophagy and Met-induced lactate accumulation limit their tumor-killing potential respectively. So overcoming the corresponding shortages will improve their therapeutic effects. In the present study, we found that DCA and Met synergistically inhibited the growth and enhanced the apoptosis of ovarian cancer cells. Interestingly, we for the first time revealed that Met sensitized DCA via dramatically attenuating DCA-induced Mcl-1 protein and protective autophagy, while DCA sensitized Met through markedly alleviating Met-induced excessive lactate accumulation and glucose consumption. The in vivo experiments in nude mice also showed that DCA and Met synergistically suppressed the growth of xenograft ovarian tumors. These results may pave a way for developing novel strategies for the treatment of ovarian cancer based on the combined use of DCA and Met. Keywords: dichloroacetate, metformin, Mcl-1, cancer metabolism, ovarian cancer INTRODUCTION The mortality of ovarian cancer ranks top among several types of gynecological cancers. At present, platinum and taxol-based chemotherapies are still standard paradigm in addition to surgery, however, their side effects are severe and the chemoresistance has also emerged [1–2]. Therefore, it is urgent to explore novel strategies as alternatives of traditional chemotherapy. In recent years, the growing evidences have shown that cancer is a kind of metabolic abnormalities, which pushes it to the forefront by regulating cancer metabolism to inhibit tumor growth [3]. Targeting key metabolic pathways significantly kill numerous cancer cells including ovarian cancer cells [4–5]. Among various metabolic drugs, dichloroacetate (DCA) and metformin (Met) have shown charming prospects because of their positive functions in cancer therapy. As a mitochondria-targeting agent, DCA can inhibit the activity of pyruvate dehydrogenase kinase (PDK) and subsequently increase the activity of pyruvate dehydrogenase (PDH), which promotes the flux of carbohydrates into mitochondria and thereby enhances aerobic oxidation of glucose. This effect reverses mitochondrial dysfunction and reactivates mitochondria-dependent apoptosis in several tumor cells [6–9]. Simultaneously, DCA inhibits glycolysis and reduces lactate accumulation, which destroys the acidified tumor microenvironment (The acidified microenvironment is generally in favor of tumor survival) [10]. Although DCA has shown promising prospect in fighting against cancers, it has been reported that DCA induces protective autophagy in colon cancer cells which in turn hinders its apoptotic capacity [11]. So far,…

Dichloroacetate is an antimetabolite that antagonizes acetate and deprives cancer cells from its benefits: a novel evidence-based medical hypothesis

Salah Mohamed El Sayed1, 2 , Hussam Baghdadi1 , Nagwa Sayed Ahmed2 , Hamdi H. Almaramhy3 , Ahmed Al-Amir Mahmoud2 , Samer El-Sawy2 , Mongi Ayat1 , Momen Elshazley4,5, Wafaa Abdel-Aziz6 , Haitham Mahmoud Abdel-Latif6 , Walaa Ibrahim6 . 1 Department of Clinical Biochemistry and Molecular Medicine, Taibah College of Medicine, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia. 2 Department of Medical Biochemistry, Sohag Faculty of Medicine, Sohag University, Egypt. 3 Department of surgery, Taibah College of Medicine, Taibah University, AlMadinah Al-Munawwarah, Saudi Arabia. 4 Department of Medicine, Taibah College of Medicine, Taibah University, AlMadinah Al-Munawwarah, Saudi Arabia. 5 Department of occupational diseases and toxigenomics, Sohag Faculty of Medicine, Sohag University, Egypt. 6 Department of Medical Pharmacology, Sohag Faculty of Medicine, Sohag University, Egypt.Corresponding Author: Assist. Professor/ Salah Mohamed El Sayed *Department of Clinical Biochemistry and Molecular Medicine, Taibah College of Medicine, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia. * Department of Medical Biochemistry, Sohag Faculty of Medicine, Sohag University, Egypt Tel.: 00966503551588, 00966542927804. *Department of Medical Biochemistry, Sohag Faculty of Medicine, Sohag University, Egypt. Email: [email protected], [email protected]: 18 July 2018Accepted: 20 November 2018Available online: 22 November 2018 Abstract Dichloroacetate (DCA) is a promising safe anticancer drug that cured a patient with chemoresistant non-Hodgkin's lymphoma and treated lactic acidosis effectively. The well-known mechanism of DCA action is through stimulating Krebs cycle (stimulating pyruvate dehydrogenase via inhibiting pyruvate dehydrogenase kinase). This prevents lactate formation (Warburg effect) depriving cancer cells of lactatebased benefits e.g. angiogenesis, chemoresistance and radioresistance. Here, we introduce novel evidence-based hypotheses to explain DCA-induced anticancer effects. On pharmacological and biochemical bases, we hypothesize that DCA is a structural antagonist of acetate competing with it for target enzymes and biological reactions. We hypothesize that DCA exerts its anticancer effects via depriving cancer of acetate benefits. We hypothesize also that acetate is an antidote of DCA capable of treating DCA toxicity. Many reports support our hypotheses. Acetate is vital for cancer cells (tumors depend on acetate) and DCA is structurally similar to acetate. DCA exerts opposite effects to acetate. Acetate caused a decrease in serum potassium, phosphorus and glucose, and an increase in serum lactate, citrate, free fatty acids and ketone bodies (serum acetoacetate and beta-hydroxybutyrate levels). Acetate decreased the proportion of active (dephosphorylated) pyruvate dehydrogenase in perfused rat heart. DCA produced quite opposite effects. Intravenous infusion of acetate produced metabolic alkalemia while DCA caused minimal effects on acidbase status. Acetate is important for cancer cells metabolism and survival as elevated acetate can drive resistance to targeted cancer treatments. Acetate is required for epidermal growth factor receptor vIII mutation in lethal brain tumors. Experimentally, DCA inhibited acetate oxidation in hearts of normal rats and reversed inhibitory effects of acetate on the oxidation of glucose. During presence of DCA with no glucose in heart perfusions with [1-14C]acetate, DCA decreased the specific radioactivity of acetyl CoA and its product citrate. This proves our hypotheses that DCA is an antimetabolite that antagonizes acetate for vital reactions in cancer cells. Acetate may be used as an…

Pharmacogenetic considerations with dichloroacetate dosing

Margaret O James*,1 & Peter W Stacpoole2,3 1 Department of Medicinal Chemistry, University of Florida, Gainesville, FL 32610-0485, USA 2 Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610-0485, USA 3 Department of Biochemistry & Molecular Biology, University of Florida, Gainesville, FL 32610-0485, USA Correspondence: Tel: +1 352 273 7707 Email: [email protected]: 16 December 2015Accepted: 17 February 2016Published: 4 May 2016 Abstract The investigational drug dichloroacetate (DCA) is a metabolic regulator that has been successfully used to treat acquired and congenital metabolic diseases and, recently, solid tumors. Its clinical use has revealed challenges in selecting appropriate doses. Chronic administration of DCA leads to inhibition of DCA metabolism and potential accumulation to levels that result in side effects. This is because conversion of DCA to glyoxylate is catalyzed by one enzyme, glutathione transferase zeta 1 (GSTZ1-1), which is inactivated by DCA. SNPs in the GSTZ1 gene result in expression of polymorphic variants of the enzyme that differ in activity and rates of inactivation by DCA under physiological conditions: these properties lead to considerable variation between people in the pharmacokinetics of DCA. Keywords: dichloroacetate; GSTZ1; pharmacogenetics Clinical use of dichloroacetate For being such a simple molecule, DCA has a remarkably rich and diverse pharmacological portfolio, dating almost a century [1,2]. However, its modern use as an investigational drug began in 1970, when its selective ability to lower blood glucose levels in diabetic, but not in nondiabetic, animals was discovered [3] and subsequently confirmed in humans [4]. Many metabolic properties of DCA involving glucose and lipid metabolism were identified during the 1970s and 1980s. However, most of its pharmacological effects can be distilled into a few basic sites and mechanisms of action. First, DCA is a noncompetitive inhibitor of the endoplasmic reticulum enzyme HMG CoA reductase, which catalyzes the rate-limiting step in cholesterol biosynthesis. DCA's inhibitory effect is observed in both rodent liver [5] and human leukocytes [6]; it likely accounts for the drug-associated reduction in total and low-density lipoprotein (LDL) cholesterol in patients with LDL receptor-negative homozygous familial hypercholesterolemia [7] and for its designation as the first orphan product for this rare disease. Second, DCA inhibits de novo hepatic triglyceride synthesis in nondiabetic rodents [5] and decreases circulating triglyceride and very low density lipoprotein levels in patients with Type 2 diabetes mellitus [4]. It also decreases blood ketone bodies in rats with experimentally induced diabetic ketoacidosis [8,9]. The precise mechanisms underlying these effects on lipid synthesis and oxidation are unknown. Third, DCA stimulates the mitochondrial PDC, which irreversibly oxidizes pyruvate to acetyl coenzyme A (acetyl CoA) [10], a property shared by certain other halogenated short chain fatty acids [11]. It is the ability of DCA to alter PDC activity that has generated by far the greatest experimental and clinical research on this unusual molecule. PDC is regulated post-translationally mainly by reversible phosphorylation of one or more of three serine residues on the E1α subunit of the first enzyme PDH of PDC that decarboxylates pyruvate [12]. Four PDH kinases (PDK1–4) and two PDH phosphatases (PDP 1 and…

Sensitization of breast cancer cells to paclitaxel by dichloroacetate through inhibiting autophagy

Minghao Wang a, Cuiwei Liao a,b, Ying Hu a , Wenqin Pan a , Jun Jiang a,# a Center of Breast Disease, Southwest Hospital, The Third Military Medical University, Chongqing, 400038, Chinab Department of Radiology, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400038, China Received: 14 May 2017Accepted: Accepted 17 May 2017Available online: 19 May 2017 Abstract Chemotherapy is still the main adjuvant strategy in the treatment of cancer, however, chemoresistance is also frequently encountered. Autophagy inhibition has been widely accepted as a promising therapeutic strategy in cancer, while the lack of effective and specific autophagy inhibitors hinders its application. Here we found that dichloroacetate (DCA), a small molecule compound, could significantly inhibit the autophagy induced by Doxorubicin in breast cancer cells. And DCA markedly enhances Doxorubicin-induced breast cancer cell death and anti-proliferation in vitro. But the sensitization to Dox of DCA was significantly reduced through induction of autophagy by rapamycin. Moreover, the combined therapy of Dox and DCA could significantly inhibit tumor growth in vivo and prolong mouse survival time. Taken together, we demonstrate that DCA could inhibit doxorubicin-inducing autophagy and provide a novel strategy for improving the anti-cancer efficacy of chemotherapy. Keywords: Dichloroacetate; Breast cancer; Autophagy; Chemotherapy Highlights:•Doxorubicin (Dox) induces autophagy in breast cancer cell.•DCA sensibilizes MDA-MB-231 cells to Dox through inhibiting autophagy.•Induction autophagy by rapamycin impaired the sensitizing effects of DCA to Dox.•Synergistic inhibition of tumor growth in mice treated with Dox and DCA. INTRODUCTION Breast cancer is a major public health problem worldwide and its universalism and occurrence rate have markedly risen over the past decades. Currently, chemotherapy is still the main aid to breast cancer treatment. Meanwhile, breast cancer is insensitive to regular chemotherapy and radiationtherapy [1], [2]. Autophagy involves the sequestration of portions of the cytoplasmin double-membraned vesicles, the autophagosomes, which then fuse with lysosomes to generate autolysosomes, in which the autophagic cargo is degraded by catabolichydrolases. Autophagy allows cells to degrade their own proteins and organelles to maintain cellular homoeostasis required for normal growth and development, the short-term adaptation to stress as well as for the long-term survival of optimally fit cells [3], [4], [5]. Deregulations of autophagy have been involved in multiple degenerative diseases, aging, including cancer [6], [7], [8], [9]. Notably, autophagy is often a pro-survival response to chemotherapeutic treatment in cancer cells, and suppression of autophagy during chemotherapy has been proposed as anovel therapeutic strategy [10]. Dichloroacetate (DCA) is a small inhibitor of pyruvate dehydrogenase kinase (PDK), which activates pyruvate dehydrogenase (PDH), and increases glucose oxidation by promoting influx of pyruvate into the Krebs cycle. It has been recently demonstrated as a promising nontoxic antineoplastic agent to promote apoptosis in carcinoma cell [11], [12]. But the correlation of DCA and autophagy in breast cancer cell was unknown. In the present study, we investigate the effect of DCA on modulation of autophagy in human breast cancer cells. We found for the first time that DCA potently inhibited autophagy in MDB-231 cell. Furthermore, we investigated the effects of autophagy inhibition by DCA on anticancer potency of chemotherapeutic drugs. Co-treatment of DCA markedly decreased the viability and increased apoptosis in cells treated with Dox, and the sensitizing efficacy of DCA was significantly…

Dichloroacetate Affects Mitochondrial Function and Stemness-Associated Properties in Pancreatic Cancer Cell Lines

Tiziana Tataranni 1 , Francesca Agriesti 1 , Consiglia Pacelli 2 , Vitalba Ruggieri 1 , Ilaria Laurenzana 1 , Carmela Mazzoccoli 1 , Gerardo Della Sala 1 , Concetta Panebianco 3 , Valerio Pazienza 3 , Nazzareno Capitanio 2 and Claudia Piccoli 1,2,* 1 Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy; [email protected] (T.T.); [email protected] (F.A.); [email protected] (V.R.); [email protected] (I.L.); [email protected] (C.M.); [email protected] (G.D.S.) 2 Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; [email protected] (C.P.); [email protected] (N.C.) 3 Division of Gastroenterology, IRCCS “Casa Sollievo della Sofferenza” Hospital, 71013 San Giovanni Rotondo, Italy; [email protected] (C.P.); [email protected] (V.P.) * Correspondence: [email protected]; Tel.: +39-0881-588-060Received: 21 February 2019Accepted: 15 May 2019Published: 18 May 2019 Abstract Targeting metabolism represents a possible successful approach to treat cancer. Dichloroacetate (DCA) is a drug known to divert metabolism from anaerobic glycolysis to mitochondrial oxidative phosphorylation by stimulation of PDH. In this study, we investigated the response of two pancreatic cancer cell lines to DCA, in two-dimensional and three-dimension cell cultures, as well as in a mouse model. PANC-1 and BXPC-3 treated with DCA showed a marked decrease in cell proliferation and migration which did not correlate with enhanced apoptosis indicating a cytostatic rather than a cytotoxic effect. Despite PDH activation, DCA treatment resulted in reduced mitochondrial oxygen consumption without affecting glycolysis. Moreover, DCA caused enhancement of ROS production, mtDNA, and of the mitophagy-marker LC3B-II in both cell lines but reduced mitochondrial fusion markers only in BXPC-3. Notably, DCA downregulated the expression of the cancer stem cells markers CD24/CD44/EPCAM only in PANC-1 but inhibited spheroid formation/viability in both cell lines. In a xenograft pancreatic cancer mouse-model DCA treatment resulted in retarding cancer progression. Collectively, our results clearly indicate that the efficacy of DCA in inhibiting cancer growth mechanistically depends on the cell phenotype and on multiple off-target pathways. In this context, the novelty that DCA might affect the cancer stem cell compartment is therapeutically relevant. Keywords: metabolism; mitochondria; cancer stem cells © 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/). INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a very aggressive cancer, with a low percentage of affected patients eligible for surgical resection and highly refractory to conventional therapies [1,2]. Therefore, more effective drugs are urged to improve current treatment regimens. Apart from cell growth, DNA repair, invasiveness, and angiogenesis, PDAC cells are hallmarked by mutations in genes involved in metabolism [1,3]. New therapeutic strategies targeting metabolism are emerging as promising approaches to overcome chemoresistance [4]. However, inter- and intra-tumor heterogeneity, often result in different metabolic phenotypes also as a consequence of multiple interactions with the tumor microenvironment [5]. This poses therapeutic limitations and highlights the importance of preliminary metabolic characterizations of the tumor lineages, preparatory to the administration of effective drugs. We recently demonstrated that two pancreatic cancer cell lines, characterized by…

Dichloroacetate Radiosensitizes Hypoxic Breast Cancer Cells

Sven de Mey 1 , Inès Dufait 1 , Heng Jiang 1 , Cyril Corbet 2 , Hui Wang 1 , Melissa Van De Gucht 1 , Lisa Kerkhove 1 , Ka Lun Law 1 , Hugo Vandenplas 3 , Thierry Gevaert 1 , Olivier Feron 2 and Mark De Ridder 1,* 1 Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; [email protected] (S.d.M.); [email protected] (I.D.); [email protected] (H.J.); [email protected] (H.W.); [email protected] (M.V.D.G.); [email protected] (L.K.); [email protected] (K.L.L.); [email protected] (T.G.) 2 Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 1200 Brussels, Belgium; [email protected] (C.C.); [email protected] (O.F.) 3 Department of Medical Oncology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; [email protected]: [email protected]: 14 September 2020Accepted: 4 December 2020Published: 9 December 2020 Abstract Mitochondrial metabolism is an attractive target for cancer therapy. Reprogramming metabolic pathways can potentially sensitize tumors with limited treatment options, such as triple-negative breast cancer (TNBC), to chemo- and/or radiotherapy. Dichloroacetate (DCA) is a specific inhibitor of the pyruvate dehydrogenase kinase (PDK), which leads to enhanced reactive oxygen species (ROS) production. ROS are the primary effector molecules of radiation and an increase hereof will enhance the radioresponse. In this study, we evaluated the effects of DCA and radiotherapy on two TNBC cell lines, namely EMT6 and 4T1, under aerobic and hypoxic conditions. As expected, DCA treatment decreased phosphorylated pyruvate dehydrogenase (PDH) and lowered both extracellular acidification rate (ECAR) and lactate production. Remarkably, DCA treatment led to a significant increase in ROS production (up to 15-fold) in hypoxic cancer cells but not in aerobic cells. Consistently, DCA radiosensitized hypoxic tumor cells and 3D spheroids while leaving the intrinsic radiosensitivity of the tumor cells unchanged. Our results suggest that although described as an oxidative phosphorylation (OXPHOS)-promoting drug, DCA can also increase hypoxic radioresponses. This study therefore paves the way for the targeting of mitochondrial metabolism of hypoxic cancer cells, in particular to combat radioresistance. Keywords: dichloroacetate; hypoxic radiosensitivity; breast cancer; reactive oxygen species © 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/). INTRODUCTION Breast cancer is the most common cancer in women globally and results annually in 627,000 deaths [1]. In the last decades, significant progress has been made in the treatment of breast cancer. However, only limited therapies are available for patients with triple-negative/basal-like breast cancers [2,3,4]. The standard of care for the treatment of high-risk breast cancers consists of neoadjuvant chemotherapy and surgery, followed by postoperative whole-breast/chest wall irradiation. Nowadays, researchers are focusing either on hypofractionation of the adjuvant radiotherapy (FAST-Forward trial [5] or the combination of chemotherapy with preoperative radiotherapy. The preoperative radiotherapy approach could result in improved disease-free survival and quality of life [6,7,8,9,10,11]. The main effect of radiation, particularly for low linear energy transfer radiation, is the induction of reactive oxygen species (ROS). During radiotherapy, ROS are created by the radiolysis of water in extracellular…

Dichloroacetate restores colorectal cancer chemosensitivity through the p53/miR-149-3p/PDK2-mediated glucose metabolic pathway

Yu Liang1, Lidan Hou1, Linjing Li1, Lei Li1, Liming Zhu1, Yu Wang1, Xin Huang1, Yichao Hou1, Danxi Zhu1, Huimin Zou1, Yan Gu2, Xiaoling Weng3,4, Yingying Wang5, Yue Li6, Tianqi Wu3, Mengfei Yao3, Isabelle Gross7,8, Christian Gaiddon9,10, Meng Luo2, Jianhua Wang3, Xiangjun Meng1 1 Department of Gastroenterology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China2 Department of General Sufergery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China3 Cancer institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China 4 Ningbo Aitagene Technology Co. LTD, Shanghai, China 5 Department of Biochemistry and Molecular & Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China 6 Pathology Center, Shanghai First People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China 7 INSERM UMR_S1113, Strasbourg F-67200, France 8 FMTS, Universite de Strasbourg Strasbourg, Strasbourg F-67000, France 9Universite de Strasbourg, Inserm IRFAC UMR_S1113, Laboratory Stress Response and Innovative Therapy “Streinth”, Strasbourg 67200, France 10 CLCC Paul Strauss, Strasbourg, FranceMeng Luo [email protected] Wang [email protected] Meng [email protected] authors contributed equally: Yu Liang, Lidan HouReceived: 16 March 2019Revised: 17 September 2019Accepted: 19 September 2019Published: 9 October 20199 October 2019 Abstract The development of chemoresistance remains a major challenge that accounts for colorectal cancer (CRC) lethality. Dichloroacetate (DCA) was originally used as a metabolic regulator in the treatment of metabolic diseases; here, DCA was assayed to identify the mechanisms underlying the chemoresistance of CRC. We found that DCA markedly enhanced chemosensitivity of CRC cells to fluorouracil (5-FU), and reduced the colony formation due to high levels of apoptosis. Using the microarray assay, we noted that miR-149-3p was involved in the chemoresistance of CRC, which was modulated by wild-type p53 after DCA treatment. In addition, PDK2 was identified as a direct target of miR-149-3p. Mechanistic analyses showed that overexpression of miR-149-3p enhanced 5-FU-induced apoptosis and reduced glucose metabolism, similar to the effects of PDK2 knockdown. In addition, overexpression of PDK2 partially reversed the inhibitory effect of miR-149-3p on glucose metabolism. Finally, both DCA treatment and miR-149-3p overexpression in 5-FU-resistant CRC cells were found to markedly sensitize the chemotherapeutic effect of 5-FU in vivo, and this effect was also validated in a small retrospective cohort of CRC patients. Taken together, we determined that the p53/miR-149-3p/PDK2 signaling pathway can potentially be targeted with DCA treatment to overcome chemoresistant CRC. Supplementary information: The online version of this article (https:// doi.org/10.1038/s41388-019-1035-8) contains supplementary material, which is available to authorized users. Introduction Colorectal cancer (CRC) is the fourth leading cause of cancer-related death in China [1] and is the second leading cause of cancer-related mortality in the United States [2], which mainly attributes to metastasis and chemotherapy failure due to drug resistance, leading to ~50,000 deaths annually [3]. Recently, while the emerging star PD1/PDL1 attracted great interest, and more biotherapeutic agents were showing encouraging results in cancer treatment, the limited efficacy rate and inevitable adverse effects restrain its use in the clinic [4,5]. Currently, chemotherapy is still a major choice in the…

Cotreatment with dichloroacetate and omeprazole exhibits a synergistic antiproliferative effect on malignant tumors

TATSUAKI ISHIGURO1,2, MIYU ISHIGURO1, RYUMEI ISHIGURO1 and SAYURI IWAI1* 1 Department of Experimental Therapeutics, Kamui Medical Co., Ltd., Tokyo 11200022 Hibiya Uchisaiwaicho Clinic, Tokyo 1050004, Japan. Correspondence: Dr Tatsuaki Ishiguro, Kamui Medical Co., Ltd., 2-20-13 Koishikawa Bunkyo-ku, Tokyo 1120002, JapanE-mail: [email protected]: 14 November 2011 Accepted: 28 December 2011DOI: 10.3892/ol.2012.552 Abstract It has been reported that treating cancer cells with dichloroacetate (DCA), an approved treatment for congenital lactic acidosis, reverses the Warburg effect and inhibits tumor growth). Furthermore, omeprazole (OMP) is a well-known agent that enhances the effects of anticancer drugs. The aim of this study was to find clinically-used drugs that enhance the effects of DCA. The combination of DCA and OMP exhibited a more potent antitumor activity than DCA alone in HT1080 fibrosarcoma cells and RKO colon cancer cells, while the drugs did not affect the proliferation of WI-38 human fibroblasts. The inhibitory effect of DCA combined with OMP was reversed with vitamin E and Z-VAD-FMK; therefore conventional caspase-dependent cell growth inhibition through superoxide production was suggested as the mechanism for inhibition. The combination of these drugs also had an effect on HT1080 fibrosarcoma cells inoculated into mice. Since OMP and DCA may be administered orally and have been used clinically for several years without major side effects, we believe that this combination therapy could be readily translated to treat malignant tumors. Keywords: fibrosarcoma, colon cancer, dichloroacetate, omeprazoleAbbreviations: DCA, dichloroacetate; PPI, proton pump inhibitor; OMP, omeprazole; SOD, superoxide; ROS, reactive oxygen species Introduction Warburg first observed that even in the presence of sufficient oxygen, cancer cells prefer to metabolize glucose and produce lactic acid [1-4]. The concomitant increase in glucose uptake may be exploited clinically for the detection of most solid malignant tumors by fluorodeoxyglucose positron emission tomography (FDG-PET). One possible reason for cancers adopting this less efficient pathway for producing adenosine triphosphate (ATP) compared with oxidative phosphorylation is its advantage for survival and proliferation in the unique hypoxic tumor environment [5]. This preference for anaerobic respiration is also considered to be the reason for the resistance cancer cells exhibit to anticancer drugs which induce apoptosis via the mitochondrial pathway. Bonnet et al have reported that treating cancer cells with dichloroacetate (DCA), an approved treatment for congenital lactic acidosis, reverses the Warburg effect and inhibits tumor growth (3,4,6-8). DCA increases the flux of pyruvate into the mitochondria by inhibiting the pyruvate dehydrogenase kinase and promotes glucose oxidation over glycolysis. As a result, DCA decreases the production of lactic acid by the tumor and increases the intracellular pH. DCA induces apoptosis via two pathways, one in the mitochondria, where depolarization and superoxide (SOD) production activates mitochondria-dependent apoptosis, and the other at the plasmalemmal level, where activation/upregulation of Kv1.5 channels decreases the (K+)i, activating caspases [6]. DCA is a promising anticancer drug due to the convenience of its oral administration, low cost, few side effects and long experience of clinical use [7,8]. Although it appeared to be a promising treatment for malignant tumors, its effect is limited in an ongoing…

Synergistic Antitumor Effect of Dichloroacetate in Combination with 5-Fluorouracil in Colorectal Cancer

Jingtao Tong, Ganfeng Xie, Jinxia He, Jianjun Li, Feng Pan, and Houjie Liang Department of Oncology, Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Chongqing 400038, China Correspondence should be addressed to Houjie Liang, [email protected] Received 27 May 2010; Revised 29 December 2010; Accepted 13 January 2011 Academic Editor: Miguel A. Andrade Copyright © 2011 Jingtao Tong et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium provided the original work is properly cited. Dichloroacetate (DCA), an inhibitor of pyruvate dehydrogenase kinase (PDK), has been recently demonstrated as a promising nontoxic antineoplastic agent that promotes apoptosis of cancer cells. In the present study, we aimed to investigate the antitumor effect of DCA combined with 5-Fluorouracil (5-FU) on colorectal cancer (CRC) cells. Four human CRC cell lines were treated with DCA or 5-FU, or a combination of DCA and 5-FU. The cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)- 2,5-diphenyltetrazolium bromide assay. The interaction between DCA and 5-FU was evaluated by the median effect principle. Immunocytochemistry with bromodeoxyuridine (BrdU) was carried out to determine the proliferation of CRC cells. Cell cycle and apoptosis were measured by flow cytometry, and the expression of apoptosis-related molecules was assessed by western blot. Our results demonstrated that DCA inhibited the viability of CRC cells and had synergistic antiproliferation in combination with 5-FU. Moreover, compared with 5-FU alone, the apoptosis of CRC cells treated with DCA and 5-FU was enhanced and demonstrated with the changes of Bcl-2, Bax, and caspase-3 proteins. Our results suggest that DCA has a synergistic antitumor effect with 5-FU on CRC cell lines in vitro. 1.  Introduction Colorectal cancer is one of the most common malignancies worldwide [1]. Other than surgery, treatment for CRC patients relies primarily on chemotherapy, especially the patients with advanced CRC. Among the chemotherapeutic agents for CRC, 5-Fluorouracil (5-FU), which is a classical chemotherapy agent, has been the first line regimen for treating CRC over several decades [2, 3]. However, 5-FU alone is poorly selective to tumor as well as highly toxic to bone marrow, gastrointestinal tract, and skin when used at the therapeutic dose [4]. Metabolic abnormity is one of the critical hallmarks of cancer [5]. As early as the 1920s, Otto Warburg observed that cancer cells generally use glycolysis rather than oxidative phosphorylation for energy [6]. Thus, the metabolic switch to anaerobic respiration through glycolysis from pyruvate, rather than pyruvate conversion to acetyl-CoA by the action of pyruvate dehydrogenase (PDH) in aerobic glucose metabolism, becomes a preferential phenotype of cancer progress. PDH can be inactivated by pyruvate dehydrogenase kinase (PDK) in many glycolytic phenotypes including cancer, while inhibition of PDK switches metabolism to aerobic oxidation which is proved to be disadvantageous to tumour growth [7]. Dichloroacetate (DCA) is a prototypical inhibitor of mitochondrial PDK. By blocking the enzyme, DCA decreases lactate production by shifting the metabolism of pyruvate from glycolysis towards oxidation in the mitochondria. This property has led to trials…

Role of SLC5A8, a plasma membrane transporter and a tumor suppressor, in the antitumor activity of dichloroacetate

Ellappan Babu, Ph. D, Sabarish Ramachandran, Ph. D, Veena CoothanKandaswamy, Ph. D., Selvakumar Elangovan, Ph. D, Puttur D. Prasad, Ph. D, Vadivel Ganapathy, Ph. D, and Muthusamy Thangaraju, Ph. D. Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia, USA Users may view, print, copy, download and text and data- mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms Correspondence: M. Thangaraju, Ph. D., Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA., [email protected] of interest: The authors declare that there is no competing financial interest in relation to the work described in this manuscript.Published in final edited form as: Oncogene. 2011 September 22; 30(38): 4026–4037. doi:10.1038/onc.2011.113.Keywords: SLC5A8; dichloroacetate; anticancer drug; Warburg effect; pyruvate dehydrogenase kinase; mitochondrial oxidation in cancer Abstract There has been growing interest among the public and scientists in dichloroacetate as a potential anticancer drug. Credible evidence exists for the antitumor activity of this compound, but high concentrations are needed for significant therapeutic effect. Unfortunately, these high concentrations produce detrimental side effects involving nervous system, thereby precluding its use for cancer treatment. The mechanistic basis of the compound’s antitumor activity is its ability to activate pyruvate dehydrogenase complex through inhibition of pyruvate dehydrogenase kinase. Since the compound inhibits the kinase at micromolar concentrations, it is not known why therapeutically prohibitive high doses are needed for suppression of tumor growth. We hypothesized that lack of effective mechanisms for the entry of dichloroacetate into tumor cells may underlie this phenomenon. Here we show that SLC5A8 transports dichloroacetate very effectively with high affinity. This transporter is expressed in normal cells, but the expression is silenced in tumor cells via epigenetic mechanisms. The lack of the transporter makes tumor cells resistant to the antitumor activity of dichloroacetate. However, if the transporter is expressed in tumor cells ectopically, the cells become sensitive to the drug at low concentrations. This is evident in breast cancer cells, colon cancer cells, and prostate cancer cells. Normal cells, which constitutively express the transporter, are however not affected by the compound, indicating the tumor cell-selective therapeutic activity. The mechanism of the antitumor activity of the compound is still its ability to inhibit pyruvate dehydrogenase kinase and force mitochondrial oxidation of pyruvate. Since the silencing of SLC5A8 in tumors involves DNA methylation and its expression can be induced by treatment with DNA methylation inhibitors, our findings suggest that combining dichloroacetate with a DNA methylation inhibitor would offer a means to reduce the doses of dichloroacetate to avoid detrimental effects associated with high doses but without compromising antitumor activity. INTRODUCTION Dichloroacetate is currently used for the treatment of congenital lactic acidosis (Stacpoole et al., 2003, 2008). The therapeutic efficacy of this drug is due to its ability to activate pyruvate dehydrogenase complex (PDC) in mitochondrial matrix. However, the enzyme complex is not the direct target for the drug. Dichloroacetate is an inhibitor of pyruvate…

Case Report: Sodium dichloroacetate (DCA) inhibition of the “Warburg Effect” in a human cancer patient: complete response in non-Hodgkin’s lymphoma after disease progression with rituximab-CHOP

Stephen B. Strum & Örn Adalsteinsson & Richard R. Black & Dmitri Segal & Nancy L. Peress & James Waldenfels S. B. StrumInternational Strategic Cancer Alliance, 538 Granite Street, Ashland, OR 97520, USAe-mail: [email protected]. B. StrumAmerican Society of Clinical Oncology, 538 Granite Street, Ashland, OR 97520, USAS. B. StrumAmerican Urological Association, 538 Granite Street, Ashland, OR 97520, USAÖ. AdalsteinssonInternational Strategic Cancer Alliance, 873 E. Baltimore Pike #333, Kennett Square, PA 19348, USAe-mail: [email protected]. R. BlackNuclear Medicine and PET Imaging, Radisphere Teleradiology Group, Beachwood, OH 44022, USAe-mail: [email protected]. SegalValley Radiology Consultants, Poway, CA 92064, USAe-mail: [email protected]. L. PeressLife Extension Foundation, 5760 S. Scenic Drive, Sault Sainte Marie, MI 49783, USAe-mail: [email protected]. WaldenfelsLife Extension Foundation, 9114 Steeplebush Court, Annandale, VA 22003-4051, USAe-mail: [email protected] Keywords: Dichloroacetate . DCA . non-Hodgkin’s lymphoma . NHL . PET . PET/CT . Glycolysis . Metabolomics . WarburgAbbreviations: DCA (dichloroacetate)NHL (non-Hodgkin’s lymphoma)PET (Positron Emission Tomography)CT (computerized tomography)FDG (fluorodeoxyglucose)SUV (standardized uptake value)mg (milligrams)kg (kilograms)R-CHOP (rituximab-Cytoxan,Hydroxydaunomycin, Oncovin, Prednisone) Received: 27 October 2012 Accepted: 23 November 2012Springer Science+Business Media New York 2012 Abstract The uptake of fluorodeoxyglucose Positron Emission Tomography in the tumors of various cancer types demonstrates the key role of glucose in the proliferation of cancer. Dichloroacetate is a 2-carbon molecule having crucial biologic activity in altering the metabolic breakdown of glucose to lactic acid. Human cell line studies show that dichloroacetate switches alter the metabolomics of the cancer cell from one of glycolysis to oxidative phosphorylation, and in doing so restore mitochondrial functions that trigger apoptosis of the cancer cell. Reports of dichloroacetate in human subjects are rare. The authors contacted individuals from Internet forums who had reported outstanding anti-cancer responses to selfmedication with dichloroacetate. With informed consent, complete medical records were requested to document response to dichloroacetate, emphasizing the context of monotherapy with dichloroacetate. Of ten patients agreeing to such an evaluation, only one met the criteria of having comprehensive clinic records as well as pathology, imaging and laboratory reports, along with single agent therapy with dichloroacetate. That individual is the focus of this report. In this case report of a man with documented relapse after state-of-the-art chemotherapy for non-Hodgkin’s lymphoma, a significant response to dichloroacetate is documented with a complete remission, which remains ongoing after 4 years. Dichloroacetate appears to be a novel therapy warranting further investigation in the treatment of cancer. Background The metabolic profile of malignancy has been characterized as one associated with metabolic adaptations directed to preferentially utilize pathways involved with glycolysis (Warburg et al. 1927), which in the recent literature has been termed the glycolytic phenotype of cancer. (Bui and Thompson 2006; Fang et al. 2008; Gatenby and Gawlinski 2003) In essence, this glycolytic phenotype is a Darwinian adaptation in that the cancer cell diminishes and undermines the metabolic pathways of glucose oxidation used by normal cells for energy production, and also for tumor cell elimination (Fang et al. 2008). One crucial normal cell function compromised in the battle with cancer involves mitochondrial programmed cell death or apoptosis. On the basis of the…

Anticancer drugs that target metabolism: is dichloroacetate the new paradigm?

Ioanna Papandreou, Tereza Goliasova, and Nicholas C. Denko Department of Radiation Oncology, Division of Radiation and Cancer Biology, Stanford University School of Medicine, Stanford, CA Keywords: tumor metabolism, pyruvate dehydrogenase, Warburg effect, metabolic inhibitorsAbbreviations: DCA: dichloroacetate; HIF1: hypoxia-inducible factor 1; LDH: lactate dehydrogenase; PDC: pyruvate dehydrogenase complex; PDH: pyruvate dehydrogenase; PDK: pyruvate dehydrogenase kinase; PDP: pyruvate dehydrogenase phosphatase Correspondence to: Nicholas C. Denko, Department of Radiation Oncology, Division of Radiation and Cancer Biology, Stanford University School of Medicine, Stanford, CA 94305, USATel.: 650-724-5066, Fax: 650-723-7382,E-mail: [email protected] Received: 22 Jul 2010Accepted: Accepted 30 Sep 2010Online: 18 Oct 2010DOI: 10.1002/ijc.25728 In the past 20 years, the number of articles containing ‘‘tumor metabolism’’ has increased from 3 to 28 per year, and the number of times these articles have been cited has increased from 23 to 929 per year (ISI, Thompson Reuters statistics). The renewed interest in understanding the mechanisms and consequences of altered tumor metabolism has clearly captured the imagination of the scientific community. The idea that tumors have altered metabolism was first recognized by Nobel Prizewinning biochemist Otto Warburg when describing glucose metabolism.1 More recently, the concept that tumors are metabolically different has grown to encompass other characteristics, such as glutaminolysis, fatty acid oxidation and lipid biogenesis. There is clearly a different metabolic demand that drives these changes in cells that are continuously dividing when compared with terminally differentiated cells. The discovery of these alterations has raised the possibility that they may be therapeutically targeted because of their unique importance to cancer cells.2 The concept that metabolic changes are a response to unique demands within the tumor has been proposed,3 even when it is hard to quantitate those demands. There is an interplay between oncogenic changes in the tumor cell with the unique aspects of the tumor microenvironment that impact on cellular metabolism and vice versa (Fig. 1). It is therefore difficult to establish the exact metabolic demands within the tumor by studying the cells from the tumor grown ex vivo. The environmental conditions used to grow cells in culture are very different from the environmental conditions in vivo. High glucose Dulbecco’s modified Eagles media and an atmosphere of 21% oxygen is very different from the hypoxic and/or hypoglycemic conditions found in the tumor.4,5 The glucose concentration of 25 mM is approximately five times that of normal blood levels, and the oxygen tension is at least four times greater than that found in vivo. The fact that the cells are bathed in these metabolic substrates significantly alters their inherent metabolic programs.4,6 Elevated glucose concentrations favors glycolysis (the Crabtree effect7 ) while elevated oxygenation produces increased oxygen byproducts and shortens cellular lifespan.8 Glucose metabolism illustrates the interplay of these three factors in the tumor. Oncogenic transformation drives tumor cell proliferation more than vascular capacity, generating hypoxia. Hypoxia within the tumor microenvironment enhances glycolytic metabolism, largely through the activation of the hypoxiainducible factor 1 (HIF1) transcription factor.9 Increased glycolysis leads to increased production of lactate, which contributes to an acidic extracellular pH and further…

Metabolic Modulation of Glioblastoma with Dichloroacetate

E. D. Michelakis,1 * G. Sutendra,1 P. Dromparis,1 L. Webster,1 A. Haromy,1 E. Niven,2 C. Maguire,2 T.-L. Gammer,1 J. R. Mackey,3 D. Fulton,3 B. Abdulkarim,3 M. S. McMurtry,1 K. C. Petruk4 1Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2B7. 2Department of Biomedical Engineering and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada T6G 2B7. 3Department of Oncology, University of Alberta, Edmonton, Alberta, Canada T6G 2B7. 4Department of Neurosurgery, University of Alberta, Edmonton, Alberta, Canada T6G 2B7. *To whom correspondence should be addressed. E-mail: [email protected] Volume 2 Issue 31 31ra34  Submitted: 11 November 2009Accepted: 23 April 2010Published: 12 May 2010 Solid tumors, including the aggressive primary brain cancer glioblastoma multiforme, develop resistance to cell death, in part as a result of a switch from mitochondrial oxidative phosphorylation to cytoplasmic glycolysis. This metabolic remodeling is accompanied by mitochondrial hyperpolarization. We tested whether the small-molecule and orphan drug dichloroacetate (DCA) can reverse this cancer-specific metabolic and mitochondrial remodeling in glioblastoma. Freshly isolated glioblastomas from 49 patients showed mitochondrial hyperpolarization, which was rapidly reversed by DCA. In a separate experiment with five patients who had glioblastoma, we prospectively secured baseline and serial tumor tissue, developed patient-specific cell lines of glioblastoma and putative glioblastoma stem cells (CD133+ , nestin+ cells), and treated each patient with oral DCA for up to 15 months. DCA depolarized mitochondria, increased mitochondrial reactive oxygen species, and induced apoptosis in GBM cells, as well as in putative GBM stem cells, both in vitro and in vivo. DCA therapy also inhibited the hypoxia-inducible factor–1α, promoted p53 activation, and suppressed angiogenesis both in vivo and in vitro. The dose-limiting toxicity was a dose-dependent, reversible peripheral neuropathy, and there was no hematologic, hepatic, renal, or cardiac toxicity. Indications of clinical efficacy were present at a dose that did not cause peripheral neuropathy and at serum concentrations of DCA sufficient to inhibit the target enzyme of DCA, pyruvate dehydrogenase kinase II, which was highly expressed in all glioblastomas. Metabolic modulation may be a viable therapeutic approach in the treatment of glioblastoma. INTRODUCTION Glioblastoma multiforme (GBM) is an aggressive primary brain tumor that exhibits extremely poor responses to approved therapies (1). Chemotherapy with temozolomide (TMZ) plus radiation therapy (RT), administered after debulking surgery, increases median survival from 12.1 months with RT alone to 14.6 months (1). The median time to progression of the tumor after RT and TMZ is only 6.9 months (1). In recurrent gliomas, the progression-free survival and the response to TMZ are much worse (2). GBMs are very vascular tumors with remarkable molecular and genetic heterogeneity (1). An ideal therapy should increase GBM apoptosis, overcome the molecular heterogeneity, inhibit angiogenesis, and cross the blood-brain barrier while having minimal systemic toxicity. On the basis of our recent findings in animal models (3, 4), we hypothesized that the orphan small-molecule dichloroacetate (DCA) fulfills these criteria and may be effective in the treatment of GBM in humans. DCA inhibits the mitochondrial enzyme pyruvate dehydrogenase kinase (PDK) (5). By inhibiting PDK, DCA activates pyruvate dehydrogenase…

Dichloroacetate (DCA) and Cancer: An Overview towards Clinical Applications

Tiziana Tataranni 1 and Claudia Piccoli 1,2 1Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (Pz), 85028, Italy 2Department of Clinical and Experimental Medicine, University of Foggia, Foggia 71121, Italy Correspondence should be addressed to Tiziana Tataranni; [email protected] Guest Editor: Kanhaiya Singh Copyright © 2019 Tiziana Tataranni and Claudia Piccoli. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Received: 24 July 2019Revised: 12 September 2019Accepted: 11 October 2019Published online: 14 November 2019 An extensive body of literature describes anticancer property of dichloroacetate (DCA), but its effective clinical administration in cancer therapy is still limited to clinical trials. The occurrence of side effects such as neurotoxicity as well as the suspicion of DCA carcinogenicity still restricts the clinical use of DCA. However, in the last years, the number of reports supporting DCA employment against cancer increased also because of the great interest in targeting metabolism of tumour cells. Dissecting DCA mechanism of action helped to understand the bases of its selective efficacy against cancer cells. A successful coadministration of DCA with conventional chemotherapy, radiotherapy, other drugs, or natural compounds has been tested in several cancer models. New drug delivery systems and multiaction compounds containing DCA and other drugs seem to ameliorate bioavailability and appear more efficient thanks to a synergistic action of multiple agents. The spread of reports supporting the efficiency of DCA in cancer therapy has prompted additional studies that let to find other potential molecular targets of DCA. Interestingly, DCA could significantly affect cancer stem cell fraction and contribute to cancer eradication. Collectively, these findings provide a strong rationale towards novel clinical translational studies of DCA in cancer therapy. INTRODUCTION Cancer is one of the leading causes of death worldwide. Despite the significant progression in diagnostic and therapeutic approaches, its eradication still represents a challenge. Too many factors are responsible for therapy failure or relapse, so there is an urgent need to find new approaches to treat it. Apart from the typical well-known properties featuring malignant cells, including abnormal proliferation, deregulation of apoptosis, and cell cycle [1, 2], cancer cells also display a peculiar metabolic machine that offers a further promising approach for cancer therapy [3–5]. Our group had already suggested the importance of a metabolic characterization of cancer cells to predict the efficacy of a metabolic treatment [6]. Drugs able to affect cancer metabolism are already under consideration, showing encouraging results in terms of efficacy and tolerability [7]. In the last decade, the small molecule DCA, already used to treat acute and chronic lactic acidosis, inborn errors of mitochondrial metabolism, and diabetes [8], has been largely purposed as an anticancer drug. DCA is a 150 Da water-soluble acid molecule, analog of acetic acid in which two of the three hydrogen atoms of the methyl group have been replaced by chlorine atoms (Figure 1(a)) [9]. DCA administration in doses ranging from 50 to…

METFORMIN ENHANCES ANTITUMOR ACTION OF SODIUM DICHLOROACETATE AGAINST GLIOMA C6

D.L. Kolesnik*, O.N. Pyaskovskaya, O.V. Yurchenko, G.I. SolyanikR.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, Kyiv 03022, UkraineSubmitted: May 06, 2019. *Correspondence: E-mail: [email protected] Abbreviations used: DCA — sodium dichloroacetate; MTF — metformin; Δψm — mitochondrial membrane potential; IC50 — half maximal inhibitory concentration Abstract It is known that the arsenal of chemotherapeutic agents for the treatment of malignant brain tumors is quite limited, which causes the high relevance of research aimed at finding new effective antitumor regimens, including the use of energy metabolism modifiers. Aim: To investigate the anti-glioma activity of sodium dichloroacetate (DCA) and metformin (MTF) used in combination in vitro and in vivo. Materials and Methods: Cell survival, cell cycle, apoptosis, mitochondrial membrane potential (Δψm), ATP level, the glucose consumption rate, and lactate production rate were determined in vitro in cultured glioma C6 cells. The antitumor action of agents in vivo was evaluated routinely by the prolongation of the life span of rats with transplanted intracerebral glioma C6 and was confirmed by histological examination of tumor tissue. Results: The half maximal inhibitory concentration (IC50) for DCA and MTF used separately was 79.2 ± 2.1 mM and 78.4 ± 4.0 mM, respectively, whereas IC50 for DCA used in combination with 7.8 mM MTF was 3.3 fold lower (24.0 ± 1.2 mM, p < 0.05). The 1-day incubation of cells with DCA at a concentration close to IC50 (25 mM), in combination with MTF at a concentration by order lower than IC50 (7.8 mM), in contrast to their separate use, resulted in a decrease in the number of viable cells by 40% (p < 0.05); redistribution of the cells by the cell cycle phases toward decreased proportion of cells in the S-phase by 46% (p < 0.05) and an increased percentage of cells in the G0/G1 phase by 24% (p < 0.05) compared to similar indices in the control. High proapoptotic activity of DCA in combination with MTF was supported by a significantly higher percentage of apoptotic cells in vitro than in the control (18.9 ± 4.4% vs 5.7 ± 1.3%, p < 0.05) and a high number of tumor cells with signs of apoptosis revealed during the histological examination of tumor pathomorphosis. The combined effect of DCA and MTF resulted in almost 4-fold decrease of the glucose consumption rate by glioma C6 cells (0.23 ± 0.05 μmol/106 cells/h vs 0.91 ± 0.12 μmol/106 cells/h, p<0.05) compared to the corresponding parameters in the control, and 2-fold increased rate of lactate production (1.06 ± 0.03 μmol/106 cells/h vs 0.53 ± 0.03 μmol/106 cells/h, p < 0.05). At the same time, both Δψm and the level of intracellular ATP in the glioma C6 cells treated with DCA and MTF, both separately and in combination, did not differ significantly from those indices in the control. In in vivo studies, the average life span of rats with intracranial transplanted glioma C6, treated with DCA in combination with MTF in a total dose of 1.1 and 2.6 g/kg body…

Dichloroacetate potentiates tamoxifen-induced cell death in breast cancer cells via downregulation of the epidermal growth factor receptor

Sang Hyeok Woo1,*, Sung-Keum Seo1,*, Yoonhwa Park1,2, Eun-Kyu Kim3, Min-Ki Seong4, Hyun-Ah Kim4, Jie-Young Song1, Sang-Gu Hwang1, Jin Kyung Lee5, Woo Chul Noh4, In-Chul Park1 1Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea 2School of Life Science and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea 3Department of Surgery, Breast Cancer Center, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Bundang-gu, Seongnam, 13620, Republic of Korea 4Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea 5KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, 01812, Republic of Korea *These authors contributed equally to this work Correspondence to: In-Chul Park, email: [email protected] Keywords: tamoxifen, breast cancer, dichloroacetate, epidermal growth factor receptor, pyruvate dehydrogenase kinase  Received: May 18, 2016 Accepted: July 22, 2016 Published online: August 01, 2016 Abstract Metabolic reprogramming in cancer cells has recently been recognized as an essential hallmark of neoplasia. In this context, metabolic alterations represent an attractive therapeutic target, and encouraging results with drugs targeting various metabolic processes have been obtained in preclinical studies. Recently, several studies have suggested that dichloroacetate (DCA), a specific pyruvate dehydrogenase kinase inhibitor, may be a potential anticancer drug in a large number of diverse tumors. However, the precise mechanism is not fully understood, which is important for the use of DCA in cancer treatment. In the present study, we found that DCA sensitized MCF7 breast cancer cells to tamoxifen-induced cell death by decreasing epidermal growth factor receptor (EGFR) expression. The downregulation of EGFR was caused by degradation of the protein. Furthermore, p38 mitogen-activated protein kinase played an important role in DCA/tamoxifen-induced EGFR degradation. Finally, DCA also promoted comparable tamoxifen-induced cell death in tamoxifen-resistant MCF7 cells, which were established by long-term treatment with tamoxifen. In summary, our results suggest that DCA is an attractive potential drug that sensitizes cells to tamoxifen-induced cell death and overcome tamoxifen resistance via downregulation of EGFR expression in breast cancer cells. INTRODUCTION Proliferating cancer cells have considerably different metabolic requirements compared to most normal differentiated cells. For example, to support rapid cell growth and proliferation, cancer cells differentially alter metabolic flux compared to the surrounding tissue to provide sufficient bioenergetics and biosynthetic intermediates. A well-known phenomenon observed in most cancer cells is a shift to aerobic glycolysis, regardless of oxygen supply, which is termed the “Warburg effect”, in which pyruvate is directly converted to lactic acid instead of entering the citric acid cycle [1]. As all cancer cells are dependent on this change in metabolism, these altered pathways represent attractive therapeutic targets [2]. Efforts have been made to target reprogrammed metabolism alone or in combination with cancer chemotherapy both in preclinical and clinical studies [3]. Interestingly, this cancer-specific metabolic remodeling is reversed by dichloroacetate (DCA), a mitochondria-targeting small molecule that can penetrate most tissues after oral administration [4]. It specifically inhibits pyruvate dehydrogenase kinase (PDK), a member of the kinase family, leading to reactivation…

Medullary thyroid carcinoma relapse reversed with dichloroacetate: A case report

Dana Flavin Foundation for Collaborative Medicine and Research, Greenwich, CT, USA and AMC/co Klinik im Alpenpark, Ringsee, Germany Correspondence to: Dr Dana Flavin, Foundation for Collaborative Medicine and Research, 24 Midwood Drive, Greenwich, CT, USA E-mail: [email protected] Received: April 7, 2010Accepted: June 15, 2010DOI: 10.3892/ol_00000158 Abstract A 51-year-old male patient diagnosed with medullary thyroid carcinoma (MTC) in 2001, with progression to lung metastases following adriamycin therapy, was then successfully treated with dimethyltriazenoimidazole carboximide. He remained in partial remission for 7 years following numerous chemotherapy attempts to induce partial remission. In October 2008, the patient, then 58 years old, relapsed with numerous tumors throughout his central body. On December 1, 2008, the tumor marker for MTC, calcitonin, was at 38,611 pg/ ml, i.e., much higher than the norm of <20 pg/ml. Since all other chemotherapy attempts had failed, he was ineligible for any new studies. Subsequently, the patient was immediately started on 10 mg/kg of dichloroacetate (DCA). By April 2009, the calcitonin level was reduced to 2,000 pg/ml. In May 2009, a new positron emission tomography showed a dramatic reduction in all tumor locations. The patient presently remains in remission and continues receiving the same dosage of DCA, with his tumor marker remaining stable in laboratory data since November 2009. Key words: dichloroacetate, medullary thyroid carcinoma INTRODUCTION Medullary thyroid carcinoma (MTC) is a rare calcitonin-producing neuroendocrine tumor arising from neural crest-derived parafollicular C cells (calcitonin-producing cells) of the thyroid gland (1), a component of multiple endocrine neoplasia type 2 or MEN2 syndromes (2), accounting for approximately 4% of all thyroid cancers. Although 25-30% of MTC cases are inherited disorders, the remaining cases consist of sporadic forms of the disease (3). The 3-year survival rate of patients with differentiated thyroid cancer and radioactive iodine resistance is less than 50%, with little response obtained from standard cytotoxic chemotherapies (4). In endocrine malignancies, such as thyroid carcinoma, the treatment of choice is surgery; however, this approach is only successful in early stage disease. MTC is an incurable disease once metastases become unresectable (5). Treatment options for advanced endocrine malignancies remain unsatisfactory and are associated with poor patient prognoses. Cytotoxic chemotherapy and radiation therapy, which are associated with significant toxicity, often show only limited and transient efficacy (6). Tyrosine kinase inhibitors particularly effecting the RET (rearranged during transfection) proto-oncogene gain of function activity, such as with vandetanib, sorafenib and sunitinib, appear to be promising. However, the low rate of partial responses and the absence of complete responses in all of the various trials of monotherapy emphasize the need for novel and more effective single or combination of agents with acceptable toxicity (3). Case report This study presents a male MTC patient with lung metastases, who developed new tumors seven years after surgical intervention and successful chemotherapy with dimethyltriazenoimidazole carboximide (DTIC) and 5-fluorouracil. Initially diagnosed in 2001, the 51-year-old patient with MTC and lymph node involvement had a sporadic form of the disease, while his twin brother, who was also negative for any genetic factor, was disease-free.…

A Novel Form of Dichloroacetate Therapy for Patients With Advanced Cancer: A Report of 3 Cases

Akbar Khan, MD; Denis Marier, ND; Eric Marsden, ND; Douglas Andrews, ND; Isaac Eliaz, MD Akbar Khan, MD, is the medical director of Medicor Cancer Centres, Inc, in Toronto, Ontario, Canada. Denis Marier, ND, is the director of Canadian Clinic for Integrative Medicine in Windsor, Ontario. Eric Marsden, ND, is the director of Marsden Center of Naturopathic Excellence in Maple, Ontario. Douglas Andrews, ND, is a staff naturopathic doctor at Medicor Cancer Centres, Inc. Isaac Eliaz, MD, is the director of Amitabha Medical Clinic, in Santa Rosa, California. Corresponding author: Akbar Khan, MD E-mail address: [email protected] Abstract Oral dichloroacetate sodium (DCA) is currently under investigation as a single agent and as an adjuvant for treatment of various cancers. One of the factors limiting its clinical use in a continuous oral regimen is a doserelated, reversible neurotoxicity, including peripheral neuropathy and encephalopathy. The intravenous (IV) route has a number of potential advantages, including (1) pulsed dosing to achieve higher concentrations than feasible with oral use, (2) a longer washout period to reduce the potential for neurotoxicity, and (3) a bypassing of the digestive system, which is particularly significant for advanced-stage cancer patients. Data were available on high-dose IV DCA (up to 100 mg/kg/dose) that have confirmed its safety, both in healthy volunteers and in critically ill patients, allowing the authors to begin offlabel treatment of cancer patients. In several of their patients treated with IV DCA, the authors observed clinical, hematological, or radiological responses. This article presents 3 cases with patients who had recurrent cancers and for whom all conventional therapies had failed: (1) a 79-y-old male patient with colon cancer who had liver metastases, (2) a 43-y-old male patient with angiosarcoma who had pancreatic and bone metastases, and (3) a 10-y-old male patient with pancreatic neuroendocrine carcinoma who had liver metastases. (Altern Ther Health Med. 2014;20(suppl 2):21-28.) Oral sodium dichloroacetate (DCA) is a drug that is currently under investigation as a single agent and an adjunctive cancer treatment.1 As of this writing, an ongoing phase I trial of oral DCA for recurrent or metastatic solid tumors is occurring at the University of Alberta and 2 trials of oral DCA for head and neck cancers are occurring at Stanford University. DCA has been extensively studied by Stacpoole2-5 for the treatment of congenital lactic acidosis, which includes a group of inherited mitochondrial diseases. The safety profile for use of oral DCA in humans has been established through this body of work. The drug has been found to be relatively safe, with no hematologic, cardiac, pulmonary, or renal toxicity.6 The main toxicity is neurological, primarily peripheral neuropathy, and this condition is reversible.7 DCA-induced delirium has been observed and is rapidly reversible upon discontinuation of the drug.8 An asymptomatic but reversible elevation of liver enzymes can occur in a small percentage of patients.9 In January 2007, Bonnet et al10 published a groundbreaking paper that demonstrated that DCA was effective in treating human breast, lung, and brain cancers in vitro and in vivo (in…

Prolonged Survival After Dichloroacetate Treatment of Non-Small-Cell Lung Carcinoma-Related Leptomeningeal Carcinomatosis

Walter Lemmoa, c, Gerard Tanb Manuscript accepted for publication March 04, 2016 a LEMMO Integrated Cancer Care Inc., 327 Renfrew Street, Vancouver, BC V5K 5G5, Canadab Gerard Tan & Associates Integrative Cancer Centre, Suite 105-2295 West Broadway, Vancouver, BC V6K 2E4, Canadac Corresponding Author: Walter Lemmo, ND, FABNO, LEMMO Integrated Cancer Care Inc., 327 Renfrew Street, Vancouver, BC V5K 5G5, Canada. Email: [email protected] Abstract Here we present an observational case report of a 49-year-old female, non-smoker, having a poor performance status with non-small-cell lung cancer and leptomeningeal carcinomatosis (LMC), who upon introduction of oral dichloroacetate (DCA) survived approximately 64 weeks (454 days) following palliative whole brain radiation without the need for chemotherapy or further targeted therapy to specifically address the LMC. To our knowledge, this is the first case report incorporating the use of DCA in LMC. Our findings are discussed in the context of previously reported applications of DCA in malignancies of the central nervous system. Keywords: Dichloroacetate; Dichloroacetic acid; Non-small-cell lung cancer; Leptomeningeal carcinomatosisdoi: http://dx.doi.org/10.14740/jmc2456w Articles © The authors | Journal compilation © J Med Cases and Elmer Press Inc™ | www.journalmc.orgThis is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited INTRODUCTION Leptomeningeal carcinomatosis (LMC) can be a challenging comorbidity of various malignancies, in particular breast and lung cancer [1, 2]. Common treatment protocols favor chemotherapeutic approaches, including intrathecal (IT) applications [3, 4], targeted agents [5], palliative radiation [6, 7], and the use of a ventriculoperitoneal (VP) shunt to alleviate hydrocephalus complications [8-10]. There is a great need for research into new treatment modalities that are convenient, low risk, and efficacious, as the median survival continues to be only a few months for patients with advanced non-small-cell lung cancer (NSCLC). Case Report A 49-year-old non-smoker woman presented in May 2006 with inoperable NSCLC IIIb diffusely involving the right lung along with a right pleural effusion. Three cycles of gemcitabine and cisplatin beginning on July 26, 2006 were deemed ineffective. On September 18, 2006, she responded to talc pleurodesis. Subsequently, on October 5, second line paclitaxel was initiated but due to significant toxicity, it was replaced by nab-paclitaxel on October 26; the fourth and final doses were on January 2, 2007 due to progressive disease. She was then switched to erlotinib 100 mg/day on January 23, 2007, which continued until February 2009. Epidermal growth factor receptor (EGFR) mutational status was unknown as, at the time, this test was not subsidized by the Medical Service Plan of British Columbia, Canada. Despite stable appearing chest X-ray imaging between February 27, 2007 and December 29, 2007, and computerized tomography (CT) chest and abdomen imaging revealing no further abnormal findings as of April 28, 2008, carcinoembryonic antigen (CEA) continued to gradually rise beginning June 28, 2007 with a value of 28, to 170 on April 28, 2008. The CEA rise, in this case, appeared to correlate with…

Case Report of Long Term Complete Remission of Metastatic Renal Squamous Cell Carcinoma after Palliative Radiotherapy and Adjuvant Dichloroacetate

Akbar Khan Medical Director, Medicor Cancer Centres Inc., Toronto, Canada Abstract Renal squamous cell carcinoma is a rare form of renal cancer which is considered incurable once metastases develop. Prognosis is poor and average survival of advanced stage disease is typically in the range of several months, despite all available conventional therapies. We describe the case of a 72 year old female with metastatic renal squamous cell carcinoma who had a radical nephrectomy with positive surgical margins, renal vein invasion and metastases to multiple abdominal lymph nodes. She received a course of palliative radiotherapy to the abdomen with 4500cGy in 25 fractions over 5 weeks. Following radiotherapy, she was treated with a cyclic regimen of oral sodium dichloroacetate (“DCA”). Treatment was discontinued after 3 months due to development of peripheral neuropathy. Follow-up imaging upon completion of DCA treatment revealed no sign of metastatic disease. The neuropathy gradually improved and computed tomography imaging four years later demonstrated no cancer recurrence. The patient continues to feel well with no clinical evidence of recurrence five years after completion of therapy, and is living a normal and active life. Key words: Dichloroacetate, radiotherapy, renal squamous cell carcinoma, remission Copyright © 2012 Akbar Khan. This is an open access article distributed under the Creative Commons Attribution License unported 3.0, which permits unrestricted use, distribution, and reproduction in any medium, provided that original work is properly cited. Contact author: Akbar Khan E-mail: [email protected] INTRODUCTION Renal squamous cell carcinoma (“RSCC”) is a rare form of renal cancer which originates from the renal pelvis. RSCC comprises about 0.5 - 0.8% of all malignant renal tumours (Bhaijee 2012). Although surgery is sometimes curative for localized disease, metastatic renal squamous cell carcinoma (“mRSCC”) is considered incurable (Holmang et al. 2007). Multiple publications from physicians with experience treating RSCC have established that this cancer type is radio-resistant, and that systemic chemotherapy provides little benefit. (Bhandari et al. 2010), (Di Battista et al. 2012), (Kimura et al. 2000), (Li and Cheung 1987). Average survival of advanced stage disease is extremely poor (in the range of several months) and five year survival is reported to be less than 10% (Holmang et al. 2007). In a review of 15 cases, Lee et al. reported a median survival of 3.5 months (Lee et al. 1998). A Medline search was conducted (Medline RSCC 2012) to determine if any cases of long-term complete remission of mRSCC had been recorded. This revealed over 200 citations of RSCC but only one published case of 5 year complete remission/cure (Carlson 1960). In this case, the patient had complete nephro-ureterectomy, including resection of peri-renal fat and a cuff of bladder. Pathology did not demonstrate any involvement of surgical margins and there were no metastases reported. Sodium dichloroacetate (“DCA”) is a drug that has been extensively studied for the treatment of congenital lactic acidosis which is comprised of a group of inherited mitochondrial diseases (Stacpoole et al. 2006), (Stacpoole et al. 1992), (Stacpoole et al. 1988). The safety profile of DCA…

Targeting metabolism with arsenic trioxide and dichloroacetate in breast cancer cells

Ramon C Sun1,2, Philip G Board1 and Anneke C Blackburn1* 1 Molecular Genetics Group, Department of Translational Biosciences, John Curtin School of Medical Research, Building 131, Australian National University, P.O. Box 334, Canberra ACT 0200, AUSTRALIA 2 Department of Radiation Oncology, Stanford School of Medicine, Stanford CA 94305 USA.1*Correspondence: [email protected]© 2011 Sun et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.Received: 3 May 2011 Accepted: 18 November 2011Published: 18 November 2011 Abstract BackgroundCancer cells have a different metabolic profile compared to normal cells. The Warburg effect (increased aerobic glycolysis) and glutaminolysis (increased mitochondrial activity from glutamine catabolism) are well known hallmarks of cancer and are accompanied by increased lactate production, hyperpolarized mitochondrial membrane and increased production of reactive oxygen species. MethodsIn this study we target the Warburg effect with dichloroacetate (DCA) and the increased mitochondrial activity of glutaminolysis with arsenic trioxide (ATO) in breast cancer cells, measuring cell proliferation, cell death and mitochondrial characteristics. ResultsThe combination of DCA and ATO was more effective at inhibiting cell proliferation and inducing cell death than either drug alone. We examined the effect of these treatments on mitochondrial membrane potential, reactive oxygen species production and ATP levels and have identified new molecular mechanisms within the mitochondria for both ATO and DCA: ATO reduces mitochondrial function through the inhibition of cytochrome C oxidase (complex IV of the electron transport chain) while DCA up-regulates ATP synthase b subunit expression. The potentiation of ATO cytotoxicity by DCA is correlated with strong suppression of the expression of c-Myc and HIF-1a, and decreased expression of the survival protein Bcl-2. ConclusionThis study is the first to demonstrate that targeting two key metabolic hallmarks of cancer is an effective anti-cancer strategy with therapeutic potential. KeywordsDichloroacetate, breast cancer, electron transport chain, mitochondria, arsenic trioxide Introduction Arsenic trioxide (ATO) has been used as a therapeutic agent for over 2000 years. Originating from China [1], it is currently being used against acute promyeloid leukemia (APL) in patients who have relapsed following alltrans-retinoic acid/anthracycline therapy and is being promoted for first line therapy of de novo APL [2-4]. ATO is known as a hyper-reactive molecule and could potentially bind to thiol groups in many proteins [2,5]. Its ability to bind to the thiol-rich, mutant protein PML-RAR-α produced from a chromosome translocation in APL has made it an effective drug in APL [2,5,6]. ATO has been shown to induce apoptosis in a variety of cancer cell lines in vitro and in vivo [7,8], but it has been difficult to consider ATO for clinical use in tumor types other than APL due to the lack of knowledge of the molecular targets that result in its cytotoxicity. In the past 10 years, physiological changes within cancer cells in response to ATO treatment have been well characterized, and many clinical trials for new applications of ATO are…

Non-Hodgkin’s Lymphoma Reversal with Dichloroacetate

Dana F. Flavin1, 2 1Klinik im Alpenpark, Defreggerweg 2-6, Ringsee, 83707 Tegernsee, Germany2 Foundation for Collaborative Medicine and Research, 24 Midwood Drive, Greenwich, CT 06830, USA Correspondence should be addressed to Dana F. Flavin, [email protected]: 4 June 2010Accepted: 23 July 2010Academic Editor: Michael A. CarducciCopyright © 2010 Dana F. Flavin. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. In June 2007, a 48-year-old male patient, diagnosed with Stage 4 Non-Hodgkin’s Follicular Lymphoma (NHL), was treated for 3 months with conventional chemotherapy resulting in a complete remission. Almost one year later tumors returned in the nasopharynx and neck lymph glands. Refusing all suggested chemotherapies, the patient began self-administering dichloroacetate (DCA) 900 mg daily with a PET scan showing complete remission four months later. Since his last PET scan, May, 2009, he remains tumor-free from continuous DCA usage. INTRODUCTION Non-Hodgkin’s lymphoma (NHL), a cancer of the lymph system that can start anywhere in body, affects 400,000+ people in the United States with 66,000 new cases in 2009 [1]. NHL often presents as a low grade fever with sweating, swollen lymph nodes, general malaise, and fatigue. Although it responds well to established therapies, including chemotherapy and radiation [2], more aggressive newer treatments are being developed, including chemotherapy with whole body radiation followed by stem cell transplants [3]. While these treatments have resulted in complete remission in some patients [4], other patients, aware of the quality of life compromises sustained with aggressive therapies [3], seek alternate avenues of treatment with professionals or on their own, many of which are nonconventional or in experimental stages. One such therapy is dichloroacetate (DCA) [5]. DCA is a by-product of water chlorination [6, 7] that inhibits aerobic glycolysis. It has been used in medicine for over 30 years [8] as an investigational drug to treat severe metabolic disorders such as diabetes and hypercholesterolemia [5, 9] as well as the treatment of congenital lactic acidosis in North American children [10]. The bioavailability [11] and pharmacokinetics [12] of DCA have been well researched over several decades in adults [6], children [13, 14], and animals [15]. As a medicinal, DCA is generally well tolerated from dosages between 10 mg/Kg and 50 mg/Kg, although prolonged exposure is associated with peripheral neuropathy [16]. Its activation of the pyruvate dehydrogenase enzyme (PDH) of the mitochondria decreases glycolysis and reactivates glucose oxidation, a favorable approach to ameliorate lactic acidosis [9]. Cancer cells predominantly utilize a system of glycolysis for energy instead of the glucose oxidation used by healthy cells. Cancer appears to be a form of intracellular lactic acidosis caused by a block in the oxidation of glucose at the level of PDH (pyruvate dehydrogenase). The glycolysis metabolism of glucose increases cancer cells’ lactic acid and reduces the intracellular pH [7] resulting major shifts in the intracellular biochemistry. Aerobic glycolysis, known as the “Warburg Effect” [17], inactivates mitochondrial respiration which allows cancer…

Dichloroacetate inhibits neuroblastoma growth by specifically acting against malignant undifferentiated cells

Serena Vella1*, Matteo Conti2*, Roberta Tasso1, Ranieri Cancedda1,3 and Aldo Pagano1,3 1 Oncology, Biology, and Genetics Department (DOBiG), University of Genoa, Genoa-Italy2 Laboratory of Clinical Pharmacology and Toxicology, Ospedale S. Maria delle Croci, 48100 Ravenna-Italy3 National Institute for Cancer Research (IST) Genoa, Largo R. Benzi, 10, 16132 Genoa-Italy The small, water soluble molecule Dichloroacetate (DCA) is recently arousing lively interests in the field of cancer therapy for it has been shown to be able to inhibit the growth of human tumors acting specifically on the mitochondria of cancer cells without perturbing the physiology of nonmalignant cells. Neuroblastoma was one of the tumor types on which DCA was considered ineffective as it is composed of cells with few recognized mitochondrial anomalies. Neuroblastoma, however, is composed of different cell types in terms of metabolism, phenotype and malignant potential. Despite the above prediction, in this work, we show that (i) DCA exhibits an unexpected anticancer effect on NB tumor cells and (ii) this effect is selectively directed to very malignant NB cells, whereas the more differentiated/less malignant NB cells are refractory to DCA treatment. This result supports the need of a detailed investigation of DCA anticancer properties against this tumor type with the final aim of its possible use as therapeutic agent. The small molecule/orphan drug DCA recently stepped into the limelight for its capacity to restrict glioblastoma multiforme (GBM) tumor growth at dosages compatible with no side effects.1–4 Thus, considering its well-tolerated toxicity together with its low cost, DCA is arousing lively interest for its potential use in cancer therapy and in the cure of certain tumor types.5 Indeed, although DCA has been shown to be effective in small cell lung carcinoma,6 breast,7 prostate8 and endometrial9 cancers and glioblastoma cell lines,2 the efficacy of this small molecule as anticancer treatment has been so far clinically demonstrated only in human GBM so that the proved efficacy of DCA on other malignancies still remains to be evaluated.10 In detail, due to its mechanism of action, DCA is expected to be ineffective on those tumors characterized by a low mitochondrial polarization such as oat cell lung cancer, lymphomas, neuroblastoma (NB) and sarcomas.5 DCA as an inhibitor of the mitochondrial enzyme pyruvate dehydrogenase kinase (PDK) activates pyruvate dehydrogenase (PDH), a gatekeeper enzyme that regulates the flux of pyruvate into the mitochondria, increasing the ratio of glucose oxidation to glycolysis.4–6 Bonnet et al. showed that this oxidative phosphorylation boosting is selectively pro-apoptotic in cancer cells, leading to a decrease in their typical mitochondrial hyperpolarization associated with apoptosis resistance.6 Despite NB being initially considered one tumor type on which DCA is most likely ineffective, due to its specific small cell feature and presumed absence of mitochondrial membrane hyperpolarization,5 proliferating NB cells are sustained by a glycolytic phenotype.11 We studied the possible efficacy of DCA treatment in inhibiting the growth of human NB nodules generated in NOD-SCID mice. Surprisingly, we observed that DCA significantly restricts in vivo tumor growth. In human NB tumors, there are three distinct cell…

Long-term stabilization of metastatic melanoma with sodium dichloroacetate

Akbar Khan, Doug Andrews, Jill Shainhouse, Anneke C Blackburn Akbar Khan, Douglas Andrews, Medicor Cancer Centres Inc., Toronto, ON M2N 6N4, Canada Jill Shainhouse, Insight Naturopathic Clinic, Toronto, ON M4P 1N9, Canada Anneke C Blackburn, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia  Author contributions: Khan A treated the patient and wrote most of the case report; Andrews D assisted in development of the natural medication protocol for reduction of DCA side effects, and wrote a portion of the case report; Shainhouse J treated the patient with natural therapy; Blackburn AC interpreted the case report in the context of the literature on in vitro and in vivo DCA research, wrote parts of the introduction and discussion, and reviewed the manuscript overall. Informed consent statement: The patient described in this manuscript has given consent to publish her case anonymously.  Conflict-of-interest statement: One of the authors (Khan) administers dichloroacetate therapy for cancer patients through Medicor Cancer Centres at a cost, and without profit. The clinic is owned by a family member of this author. The other authors have nothing to disclose. Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/ Manuscript source: Invited manuscript Correspondence to: Akbar Khan, MD, Medical Director, Medicor Cancer Centres Inc., 4576 Yonge St., Suite 301, Toronto, ON M2N 6N4, Canada. [email protected]: +1-416-2270037Fax: +1-416-2271915   Received: January 30, 2017  Peer-review started: February 12, 2017  First decision: March 28, 2017  Revised: May 5, 2017  Accepted: May 30, 2017  Article in press: May 31, 2017  Published online: August 10, 2017 Abstract Sodium dichloroacetate (DCA) has been studied as a metabolic cancer therapy since 2007, based on a publication from Bonnet et al demonstrating that DCA can induce apoptosis (programmed cell death) in human breast, lung and brain cancer cells. Classically, the response of cancer to a medical therapy in human research is measured by Response Evaluation Criterial for Solid Tumours definitions, which define “response” by the degree of tumour reduction, or tumour disappearance on imaging, however disease stabilization is also a beneficial clinical outcome. It has been shown that DCA can function as a cytostatic agent in vitro and in vivo, without causing apoptosis. A case of a 32-year-old male is presented in which DCA therapy, with no concurrent conventional therapy, resulted in regression and stabilization of recurrent metastatic melanoma for over 4 years’ duration, with trivial side effects. This case demonstrates that DCA can be used to reduce disease volume and maintain longterm stability in patients with advanced melanoma. Key words: Dichloroacetate; Cancer; BRAF; Melanoma; Cytostatic © The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved. Core tip: Sodium dichloroacetate (DCA) has been studied as a metabolic cancer therapy since 2007. It has been shown that…

Long-term stabilization of stage 4 colon cancer using sodium dichloroacetate therapy (Case report)

Akbar Khan, Doug Andrews, Anneke C Blackburn Akbar Khan, Douglas Andrews, Medicor Cancer Centres Inc., Toronto, ON M2N 6N4, Canada Anneke C Blackburn, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia  Author contributions: Khan A treated the patient and wrote most of the case report; Andrews D treated the patient, designed the natural therapy protocols, and co-wrote the case report; Blackburn AC performed in vitro and in vivo work demonstrating DCA’s effects as a cytostatic agent, and wrote the parts of the case report dealing with the in vitro and in vivo DCA research. Institutional review board statement: Not applicable.  Informed consent statement: The patient described in this manuscript has given consent to publish her case anonymously.  Conflict-of-interest statement: One of the authors (Khan) administers dichloroacetate therapy for cancer patients through Medicor Cancer Centres at a cost, and without profit. The clinic is owned by a family member of this author. The other authors have nothing to disclose. Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/ Manuscript source: Invited manuscript Correspondence to: Akbar Khan, MD, Medical Director, Medicor Cancer Centres Inc., 4576 Yonge St., Suite 301, Toronto, ON M2N 6N4, Canada. [email protected]: +1-416-2270037Fax: +1-416-2271915  Received: April 30, 2016  Peer-review started: May 3, 2016  First decision: June 17, 2016  Revised: July 23, 2016  Accepted: August 6, 2016  Article in press: August 8, 2016  Published online: October 16, 2016 Abstract Oral dichloroacetate sodium (DCA) has been investi-gated as a novel metabolic therapy for various cancers since 2007, based on data from Bonnet et al that DCA can trigger apoptosis of human lung, breast and brain cancer cells. Response to therapy in human studies is measured by standard response evaluation criteria for solid tumours definitions, which define “response” by the degree of tumour reduction, or tumour disappearance on imaging.However, Blackburn et al have demonstrated that DCA can also act as a cytostatic agent in vitro and in vivo , without causing apoptosis (programmed cell death). A case is presented in which oral DCA therapy resulted in tum our stabilization of stage 4 colon cancer in a 57 years old female for a period of nearly 4 years, with no serious toxicity. Since the natural history of stage 4 colon cancer consists of steady progression leading to disability and death, this case highlights a novel use of DCA as a cytostatic agent with a potential to maintain long-term stability of advanced-stage cancer. Key words: Dichloroacetate; Cancer; Colon; Colorectal; Cytostatic; Stabilization; Growth inhibition; Intravenous © The Author(s) 2016. Published by Baishideng Publishing  Group Inc. All rights reserved. Core tip: Oral dichloroacetate sodium (DCA) has been investigated as a novel metabolic therapy for various cancers. Response to therapy in human studies is measured by standard…

No more pages to load