Skip to main content
Top
Published in: Cancer and Metastasis Reviews 1-2/2019

01-06-2019 | Acidosis

Causes, consequences, and therapy of tumors acidosis

Authors: Smitha R. Pillai, Mehdi Damaghi, Yoshinori Marunaka, Enrico Pierluigi Spugnini, Stefano Fais, Robert J. Gillies

Published in: Cancer and Metastasis Reviews | Issue 1-2/2019

Login to get access

Abstract

While cancer is commonly described as “a disease of the genes,” it is also associated with massive metabolic reprogramming that is now accepted as a disease “Hallmark.” This programming is complex and often involves metabolic cooperativity between cancer cells and their surrounding stroma. Indeed, there is emerging clinical evidence that interrupting a cancer’s metabolic program can improve patients’ outcomes. The most commonly observed and well-studied metabolic adaptation in cancers is the fermentation of glucose to lactic acid, even in the presence of oxygen, also known as “aerobic glycolysis” or the “Warburg Effect.” Much has been written about the mechanisms of the Warburg effect, and this remains a topic of great debate. However, herein, we will focus on an important sequela of this metabolic program: the acidification of the tumor microenvironment. Rather than being an epiphenomenon, it is now appreciated that this acidosis is a key player in cancer somatic evolution and progression to malignancy. Adaptation to acidosis induces and selects for malignant behaviors, such as increased invasion and metastasis, chemoresistance, and inhibition of immune surveillance. However, the metabolic reprogramming that occurs during adaptation to acidosis also introduces therapeutic vulnerabilities. Thus, tumor acidosis is a relevant therapeutic target, and we describe herein four approaches to accomplish this: (1) neutralizing acid directly with buffers, (2) targeting metabolic vulnerabilities revealed by acidosis, (3) developing acid-activatable drugs and nanomedicines, and (4) inhibiting metabolic processes responsible for generating acids in the first place.
Literature
2.
go back to reference Toyomura, T., Oka, T., Yamaguchi, C., Wada, Y., & Futai, M. (2000). Three subunit a isoforms of mouse vacuolar H(+)-ATPase. Preferential expression of the a3 isoform during osteoclast differentiation. The Journal of Biological Chemistry, 275, 8760–8765.CrossRefPubMed Toyomura, T., Oka, T., Yamaguchi, C., Wada, Y., & Futai, M. (2000). Three subunit a isoforms of mouse vacuolar H(+)-ATPase. Preferential expression of the a3 isoform during osteoclast differentiation. The Journal of Biological Chemistry, 275, 8760–8765.CrossRefPubMed
3.
go back to reference Boedtkjer, E., Moreira, J. M. A., Mele, M., Vahl, P., Wielenga, V. T., Christiansen, P. M., Jensen, V. E. D., Pedersen, S. F., & Aalkjaer, C. (2013). Contribution of Na+,HCO3 −-cotransport to cellular pH control in human breast cancer: a role for the breast cancer susceptibility locus NBCn1 (SLC4A7). International Journal of Cancer, 132, 1288–1299. https://doi.org/10.1002/ijc.27782.PubMedCrossRef Boedtkjer, E., Moreira, J. M. A., Mele, M., Vahl, P., Wielenga, V. T., Christiansen, P. M., Jensen, V. E. D., Pedersen, S. F., & Aalkjaer, C. (2013). Contribution of Na+,HCO3 -cotransport to cellular pH control in human breast cancer: a role for the breast cancer susceptibility locus NBCn1 (SLC4A7). International Journal of Cancer, 132, 1288–1299. https://​doi.​org/​10.​1002/​ijc.​27782.PubMedCrossRef
4.
10.
go back to reference Shiozaki, A., Hikami, S., Ichikawa, D., Kosuga, T., Shimizu, H., Kudou, M., Yamazato, Y., Kobayashi, T., Shoda, K., Arita, T., Konishi, H., Komatsu, S., Kubota, T., Fujiwara, H., Okamoto, K., Kishimoto, M., Konishi, E., Marunaka, Y., & Otsuji, E. (2018). Anion exchanger 2 suppresses cellular movement and has prognostic significance in esophageal squamous cell carcinoma. Oncotarget, 9, 25993–26006. https://doi.org/10.18632/oncotarget.25417.PubMedPubMedCentralCrossRef Shiozaki, A., Hikami, S., Ichikawa, D., Kosuga, T., Shimizu, H., Kudou, M., Yamazato, Y., Kobayashi, T., Shoda, K., Arita, T., Konishi, H., Komatsu, S., Kubota, T., Fujiwara, H., Okamoto, K., Kishimoto, M., Konishi, E., Marunaka, Y., & Otsuji, E. (2018). Anion exchanger 2 suppresses cellular movement and has prognostic significance in esophageal squamous cell carcinoma. Oncotarget, 9, 25993–26006. https://​doi.​org/​10.​18632/​oncotarget.​25417.PubMedPubMedCentralCrossRef
16.
go back to reference Barathova, M., Takacova, M., Holotnakova, T., Gibadulinova, A., Ohradanova, A., Zatovicova, M., Hulikova, A., Kopacek, J., Parkkila, S., Supuran, C. T., Pastorekova, S., & Pastorek, J. (2008). Alternative splicing variant of the hypoxia marker carbonic anhydrase IX expressed independently of hypoxia and tumour phenotype. British Journal of Cancer, 98, 129–136. https://doi.org/10.1038/sj.bjc.6604111.PubMedCrossRef Barathova, M., Takacova, M., Holotnakova, T., Gibadulinova, A., Ohradanova, A., Zatovicova, M., Hulikova, A., Kopacek, J., Parkkila, S., Supuran, C. T., Pastorekova, S., & Pastorek, J. (2008). Alternative splicing variant of the hypoxia marker carbonic anhydrase IX expressed independently of hypoxia and tumour phenotype. British Journal of Cancer, 98, 129–136. https://​doi.​org/​10.​1038/​sj.​bjc.​6604111.PubMedCrossRef
19.
go back to reference Helmlinger, G., Yuan, F., Dellian, M., & Jain, R. K. (1997). Interstitial pH and pO2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation. Nature Medicine, 3, 177–182.CrossRefPubMed Helmlinger, G., Yuan, F., Dellian, M., & Jain, R. K. (1997). Interstitial pH and pO2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation. Nature Medicine, 3, 177–182.CrossRefPubMed
22.
go back to reference Gatenby, R. A., & Gawlinski, E. T. (1996). A reaction-diffusion model of cancer invasion. Cancer Research, 56, 5745–5753.PubMed Gatenby, R. A., & Gawlinski, E. T. (1996). A reaction-diffusion model of cancer invasion. Cancer Research, 56, 5745–5753.PubMed
23.
go back to reference Stubbs, M., McSheehy, P. M., Griffiths, J. R., & Bashford, C. L. (2000). Causes and consequences of tumour acidity and implications for treatment. Molecular Medicine Today, 6, 15–19.CrossRefPubMed Stubbs, M., McSheehy, P. M., Griffiths, J. R., & Bashford, C. L. (2000). Causes and consequences of tumour acidity and implications for treatment. Molecular Medicine Today, 6, 15–19.CrossRefPubMed
25.
go back to reference Faubert, B., Li, K. Y., Cai, L., Hensley, C. T., Kim, J., Zacharias, L. G., Yang, C., Do, Q. N., Doucette, S., Burguete, D., Li, H., Huet, G., Yuan, Q., Wigal, T., Butt, Y., Ni, M., Torrealba, J., Oliver, D., Lenkinski, R. E., Malloy, C. R., Wachsmann, J. W., Young, J. D., Kernstine, K., & DeBerardinis, R. J. (2017). Lactate metabolism in human lung tumors. Cell, 171, 358–371 e359. https://doi.org/10.1016/j.cell.2017.09.019.PubMedPubMedCentralCrossRef Faubert, B., Li, K. Y., Cai, L., Hensley, C. T., Kim, J., Zacharias, L. G., Yang, C., Do, Q. N., Doucette, S., Burguete, D., Li, H., Huet, G., Yuan, Q., Wigal, T., Butt, Y., Ni, M., Torrealba, J., Oliver, D., Lenkinski, R. E., Malloy, C. R., Wachsmann, J. W., Young, J. D., Kernstine, K., & DeBerardinis, R. J. (2017). Lactate metabolism in human lung tumors. Cell, 171, 358–371 e359. https://​doi.​org/​10.​1016/​j.​cell.​2017.​09.​019.PubMedPubMedCentralCrossRef
26.
go back to reference Nelson, S. J., Kurhanewicz, J., Vigneron, D. B., Larson, P. E. Z., Harzstark, A. L., Ferrone, M., van Criekinge, M., Chang, J. W., Bok, R., Park, I., Reed, G., Carvajal, L., Small, E. J., Munster, P., Weinberg, V. K., Ardenkjaer-Larsen, J. H., Chen, A. P., Hurd, R. E., Odegardstuen, L. I., Robb, F. J., Tropp, J., & Murray, J. A. (2013). Metabolic imaging of patients with prostate cancer using hyperpolarized [1-(1)(3)C]pyruvate. Science Translational Medicine, 5, 198ra108. https://doi.org/10.1126/scitranslmed.3006070.PubMedPubMedCentralCrossRef Nelson, S. J., Kurhanewicz, J., Vigneron, D. B., Larson, P. E. Z., Harzstark, A. L., Ferrone, M., van Criekinge, M., Chang, J. W., Bok, R., Park, I., Reed, G., Carvajal, L., Small, E. J., Munster, P., Weinberg, V. K., Ardenkjaer-Larsen, J. H., Chen, A. P., Hurd, R. E., Odegardstuen, L. I., Robb, F. J., Tropp, J., & Murray, J. A. (2013). Metabolic imaging of patients with prostate cancer using hyperpolarized [1-(1)(3)C]pyruvate. Science Translational Medicine, 5, 198ra108. https://​doi.​org/​10.​1126/​scitranslmed.​3006070.PubMedPubMedCentralCrossRef
28.
go back to reference Morita, T., Nagaki, T., Fukuda, I., & Okumura, K. (1992). Clastogenicity of low pH to various cultured mammalian cells. Mutation Research, 268, 297–305.CrossRefPubMed Morita, T., Nagaki, T., Fukuda, I., & Okumura, K. (1992). Clastogenicity of low pH to various cultured mammalian cells. Mutation Research, 268, 297–305.CrossRefPubMed
33.
go back to reference Glunde, K., Guggino, S. E., Solaiyappan, M., Pathak, A. P., Ichikawa, Y., & Bhujwalla, Z. M. (2003). Extracellular acidification alters lysosomal trafficking in human breast cancer cells. Neoplasia, 5, 533–545.CrossRefPubMedPubMedCentral Glunde, K., Guggino, S. E., Solaiyappan, M., Pathak, A. P., Ichikawa, Y., & Bhujwalla, Z. M. (2003). Extracellular acidification alters lysosomal trafficking in human breast cancer cells. Neoplasia, 5, 533–545.CrossRefPubMedPubMedCentral
34.
go back to reference Damaghi, M., Tafreshi, N. K., Lloyd, M. C., Sprung, R., Estrella, V., Wojtkowiak, J. W., Morse, D. L., Koomen, J. M., Bui, M. M., Gatenby, R. A., & Gillies, R. J. (2015). Chronic acidosis in the tumour microenvironment selects for overexpression of LAMP2 in the plasma membrane. Nature Communications, 6, 8752. https://doi.org/10.1038/ncomms9752.PubMedCrossRef Damaghi, M., Tafreshi, N. K., Lloyd, M. C., Sprung, R., Estrella, V., Wojtkowiak, J. W., Morse, D. L., Koomen, J. M., Bui, M. M., Gatenby, R. A., & Gillies, R. J. (2015). Chronic acidosis in the tumour microenvironment selects for overexpression of LAMP2 in the plasma membrane. Nature Communications, 6, 8752. https://​doi.​org/​10.​1038/​ncomms9752.PubMedCrossRef
35.
go back to reference Rothberg, J. M., Bailey, K. M., Wojtkowiak, J. W., Ben-Nun, Y., Bogyo, M., Weber, E., Moin, K., Blum, G., Mattingly, R. R., Gillies, R. J., & Sloane, B. F. (2013). Acid-mediated tumor proteolysis: contribution of cysteine cathepsins. Neoplasia, 15, 1125–1137.CrossRefPubMedPubMedCentral Rothberg, J. M., Bailey, K. M., Wojtkowiak, J. W., Ben-Nun, Y., Bogyo, M., Weber, E., Moin, K., Blum, G., Mattingly, R. R., Gillies, R. J., & Sloane, B. F. (2013). Acid-mediated tumor proteolysis: contribution of cysteine cathepsins. Neoplasia, 15, 1125–1137.CrossRefPubMedPubMedCentral
36.
go back to reference Rozhin, J., Sameni, M., Ziegler, G., & Sloane, B. F. (1994). Pericellular pH affects distribution and secretion of cathepsin B in malignant cells. Cancer Research, 54, 6517–6525.PubMed Rozhin, J., Sameni, M., Ziegler, G., & Sloane, B. F. (1994). Pericellular pH affects distribution and secretion of cathepsin B in malignant cells. Cancer Research, 54, 6517–6525.PubMed
37.
go back to reference Wojtkowiak, J. W., Rothberg, J. M., Kumar, V., Schramm, K. J., Haller, E., Proemsey, J. B., Lloyd, M. C., Sloane, B. F., & Gillies, R. J. (2012). Chronic autophagy is a cellular adaptation to tumor acidic pH microenvironments. Cancer Research, 72, 3938–3947.CrossRefPubMedPubMedCentral Wojtkowiak, J. W., Rothberg, J. M., Kumar, V., Schramm, K. J., Haller, E., Proemsey, J. B., Lloyd, M. C., Sloane, B. F., & Gillies, R. J. (2012). Chronic autophagy is a cellular adaptation to tumor acidic pH microenvironments. Cancer Research, 72, 3938–3947.CrossRefPubMedPubMedCentral
39.
go back to reference Logozzi, M., Angelini, D. F., Iessi, E., Mizzoni, D., di Raimo, R., Federici, C., Lugini, L., Borsellino, G., Gentilucci, A., Pierella, F., Marzio, V., Sciarra, A., Battistini, L., & Fais, S. (2017). Increased PSA expression on prostate cancer exosomes in in vitro condition and in cancer patients. Cancer Letters, 403, 318–329. https://doi.org/10.1016/j.canlet.2017.06.036.PubMedCrossRef Logozzi, M., Angelini, D. F., Iessi, E., Mizzoni, D., di Raimo, R., Federici, C., Lugini, L., Borsellino, G., Gentilucci, A., Pierella, F., Marzio, V., Sciarra, A., Battistini, L., & Fais, S. (2017). Increased PSA expression on prostate cancer exosomes in in vitro condition and in cancer patients. Cancer Letters, 403, 318–329. https://​doi.​org/​10.​1016/​j.​canlet.​2017.​06.​036.PubMedCrossRef
43.
44.
go back to reference Busco, G., Cardone, R. A., Greco, M. R., Bellizzi, A., Colella, M., Antelmi, E., Mancini, M. T., Dell'Aquila, M. E., Casavola, V., Paradiso, A., & Reshkin, S. J. (2010). NHE1 promotes invadopodial ECM proteolysis through acidification of the peri-invadopodial space. The FASEB Journal, 24, 3903–3915. https://doi.org/10.1096/fj.09-149518.CrossRefPubMed Busco, G., Cardone, R. A., Greco, M. R., Bellizzi, A., Colella, M., Antelmi, E., Mancini, M. T., Dell'Aquila, M. E., Casavola, V., Paradiso, A., & Reshkin, S. J. (2010). NHE1 promotes invadopodial ECM proteolysis through acidification of the peri-invadopodial space. The FASEB Journal, 24, 3903–3915. https://​doi.​org/​10.​1096/​fj.​09-149518.CrossRefPubMed
51.
go back to reference Lardner, A. (2001). The effects of extracellular pH on immune function. Journal of Leukocyte Biology, 69, 522–530.PubMed Lardner, A. (2001). The effects of extracellular pH on immune function. Journal of Leukocyte Biology, 69, 522–530.PubMed
52.
go back to reference Calcinotto, A., Filipazzi, P., Grioni, M., Iero, M., de Milito, A., Ricupito, A., Cova, A., Canese, R., Jachetti, E., Rossetti, M., Huber, V., Parmiani, G., Generoso, L., Santinami, M., Borghi, M., Fais, S., Bellone, M., & Rivoltini, L. (2012). Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes. Cancer Research, 72, 2746–2756. https://doi.org/10.1158/0008-5472.CAN-11-1272.PubMedCrossRef Calcinotto, A., Filipazzi, P., Grioni, M., Iero, M., de Milito, A., Ricupito, A., Cova, A., Canese, R., Jachetti, E., Rossetti, M., Huber, V., Parmiani, G., Generoso, L., Santinami, M., Borghi, M., Fais, S., Bellone, M., & Rivoltini, L. (2012). Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes. Cancer Research, 72, 2746–2756. https://​doi.​org/​10.​1158/​0008-5472.​CAN-11-1272.PubMedCrossRef
53.
go back to reference Grundstrom, S., Dohlsten, M., & Sundstedt, A. (2000). IL-2 unresponsiveness in anergic CD4+ T cells is due to defective signaling through the common gamma-chain of the IL-2 receptor. Journal of Immunology, 164, 1175–1184.CrossRef Grundstrom, S., Dohlsten, M., & Sundstedt, A. (2000). IL-2 unresponsiveness in anergic CD4+ T cells is due to defective signaling through the common gamma-chain of the IL-2 receptor. Journal of Immunology, 164, 1175–1184.CrossRef
54.
go back to reference Wells, A. D., Walsh, M. C., Sankaran, D., & Turka, L. A. (2000). T cell effector function and anergy avoidance are quantitatively linked to cell division. Journal of Immunology, 165, 2432–2443.CrossRef Wells, A. D., Walsh, M. C., Sankaran, D., & Turka, L. A. (2000). T cell effector function and anergy avoidance are quantitatively linked to cell division. Journal of Immunology, 165, 2432–2443.CrossRef
55.
go back to reference Demotte, N., Stroobant, V., Courtoy, P. J., van der Smissen, P., Colau, D., Luescher, I. F., Hivroz, C., Nicaise, J., Squifflet, J. L., Mourad, M., Godelaine, D., Boon, T., & van der Bruggen, P. (2008). Restoring the association of the T cell receptor with CD8 reverses anergy in human tumor-infiltrating lymphocytes. Immunity, 28, 414–424. https://doi.org/10.1016/j.immuni.2008.01.011.PubMedCrossRef Demotte, N., Stroobant, V., Courtoy, P. J., van der Smissen, P., Colau, D., Luescher, I. F., Hivroz, C., Nicaise, J., Squifflet, J. L., Mourad, M., Godelaine, D., Boon, T., & van der Bruggen, P. (2008). Restoring the association of the T cell receptor with CD8 reverses anergy in human tumor-infiltrating lymphocytes. Immunity, 28, 414–424. https://​doi.​org/​10.​1016/​j.​immuni.​2008.​01.​011.PubMedCrossRef
60.
63.
go back to reference Ibrahim-Hashim, A., Robertson-Tessi, M., Enriquez-Navas, P. M., Damaghi, M., Balagurunathan, Y., Wojtkowiak, J. W., Russell, S., Yoonseok, K., Lloyd, M. C., Bui, M. M., Brown, J. S., Anderson, A. R. A., Gillies, R. J., & Gatenby, R. A. (2017). Defining cancer subpopulations by adaptive strategies rather than molecular properties provides novel insights into intratumoral evolution. Cancer Research, 77, 2242–2254. https://doi.org/10.1158/0008-5472.CAN-16-2844.PubMedPubMedCentralCrossRef Ibrahim-Hashim, A., Robertson-Tessi, M., Enriquez-Navas, P. M., Damaghi, M., Balagurunathan, Y., Wojtkowiak, J. W., Russell, S., Yoonseok, K., Lloyd, M. C., Bui, M. M., Brown, J. S., Anderson, A. R. A., Gillies, R. J., & Gatenby, R. A. (2017). Defining cancer subpopulations by adaptive strategies rather than molecular properties provides novel insights into intratumoral evolution. Cancer Research, 77, 2242–2254. https://​doi.​org/​10.​1158/​0008-5472.​CAN-16-2844.PubMedPubMedCentralCrossRef
64.
go back to reference Pilot, C., Mahipal, A., & Gillies, R. J. (2018). Buffer therapy → buffer diet. Journal of Nutrition & Food Sciences, 8, 684–688.CrossRef Pilot, C., Mahipal, A., & Gillies, R. J. (2018). Buffer therapy → buffer diet. Journal of Nutrition & Food Sciences, 8, 684–688.CrossRef
66.
go back to reference Ramlau, R., et al. (2017). Phase I/II dose escalation study of L-DOS47 as a monotherapy in non-squamous non-small cell lung cancer patients. Journal of Thoracic Oncology, 12, S1017–S1072.CrossRef Ramlau, R., et al. (2017). Phase I/II dose escalation study of L-DOS47 as a monotherapy in non-squamous non-small cell lung cancer patients. Journal of Thoracic Oncology, 12, S1017–S1072.CrossRef
67.
go back to reference Bushinsky, D. A., Hostetter, T., Klaerner, G., Stasiv, Y., Lockey, C., McNulty, S., Lee, A., Parsell, D., Mathur, V., Li, E., Buysse, J., & Alpern, R. (2018). Randomized, controlled trial of TRC101 to increase serum bicarbonate in patients with CKD. Clinical Journal of the American Society of Nephrology, 13, 26–35. https://doi.org/10.2215/CJN.07300717.PubMedCrossRef Bushinsky, D. A., Hostetter, T., Klaerner, G., Stasiv, Y., Lockey, C., McNulty, S., Lee, A., Parsell, D., Mathur, V., Li, E., Buysse, J., & Alpern, R. (2018). Randomized, controlled trial of TRC101 to increase serum bicarbonate in patients with CKD. Clinical Journal of the American Society of Nephrology, 13, 26–35. https://​doi.​org/​10.​2215/​CJN.​07300717.PubMedCrossRef
69.
go back to reference Boyd, N. H., Walker K., Fried J., Hackney J. R., McDonald P. C., Benavides G. A., Spina R., Audia A., Scott S. E., Libby C. J., Tran A. N., Bevensee M. O., Griguer C., Nozell S., Gillespie G. Y., Nabors B., Bhat K. P., Bar E. E., Darley-Usmar V., Xu B., Gordon E., Cooper S. J., Dedhar S., Hjelmeland A. B.. (2017) Addition of carbonic anhydrase 9 inhibitor SLC-0111 to temozolomide treatment delays glioblastoma growth in vivo. JCI Insight 2, doi:https://doi.org/10.1172/jci.insight.92928. Boyd, N. H., Walker K., Fried J., Hackney J. R., McDonald P. C., Benavides G. A., Spina R., Audia A., Scott S. E., Libby C. J., Tran A. N., Bevensee M. O., Griguer C., Nozell S., Gillespie G. Y., Nabors B., Bhat K. P., Bar E. E., Darley-Usmar V., Xu B., Gordon E., Cooper S. J., Dedhar S., Hjelmeland A. B.. (2017) Addition of carbonic anhydrase 9 inhibitor SLC-0111 to temozolomide treatment delays glioblastoma growth in vivo. JCI Insight 2, doi:https://​doi.​org/​10.​1172/​jci.​insight.​92928.
71.
go back to reference Polanski, R., Hodgkinson, C. L., Fusi, A., Nonaka, D., Priest, L., Kelly, P., Trapani, F., Bishop, P. W., White, A., Critchlow, S. E., Smith, P. D., Blackhall, F., Dive, C., & Morrow, C. J. (2014). Activity of the monocarboxylate transporter 1 inhibitor AZD3965 in small cell lung cancer. Clinical Cancer Research, 20, 926–937. https://doi.org/10.1158/1078-0432.CCR-13-2270.PubMedCrossRef Polanski, R., Hodgkinson, C. L., Fusi, A., Nonaka, D., Priest, L., Kelly, P., Trapani, F., Bishop, P. W., White, A., Critchlow, S. E., Smith, P. D., Blackhall, F., Dive, C., & Morrow, C. J. (2014). Activity of the monocarboxylate transporter 1 inhibitor AZD3965 in small cell lung cancer. Clinical Cancer Research, 20, 926–937. https://​doi.​org/​10.​1158/​1078-0432.​CCR-13-2270.PubMedCrossRef
73.
go back to reference Noble, R. A., Bell, N., Blair, H., Sikka, A., Thomas, H., Phillips, N., Nakjang, S., Miwa, S., Crossland, R., Rand, V., Televantou, D., Long, A., Keun, H. C., Bacon, C. M., Bomken, S., Critchlow, S. E., & Wedge, S. R. (2017). Inhibition of monocarboxyate transporter 1 by AZD3965 as a novel therapeutic approach for diffuse large B-cell lymphoma and Burkitt lymphoma. Haematologica, 102, 1247–1257. https://doi.org/10.3324/haematol.2016.163030.PubMedPubMedCentralCrossRef Noble, R. A., Bell, N., Blair, H., Sikka, A., Thomas, H., Phillips, N., Nakjang, S., Miwa, S., Crossland, R., Rand, V., Televantou, D., Long, A., Keun, H. C., Bacon, C. M., Bomken, S., Critchlow, S. E., & Wedge, S. R. (2017). Inhibition of monocarboxyate transporter 1 by AZD3965 as a novel therapeutic approach for diffuse large B-cell lymphoma and Burkitt lymphoma. Haematologica, 102, 1247–1257. https://​doi.​org/​10.​3324/​haematol.​2016.​163030.PubMedPubMedCentralCrossRef
75.
go back to reference Doherty, J. R., Yang, C., Scott, K. E. N., Cameron, M. D., Fallahi, M., Li, W., Hall, M. A., Amelio, A. L., Mishra, J. K., Li, F., Tortosa, M., Genau, H. M., Rounbehler, R. J., Lu, Y., Dang, C. V., Kumar, K. G., Butler, A. A., Bannister, T. D., Hooper, A. T., Unsal-Kacmaz, K., Roush, W. R., & Cleveland, J. L. (2014). Blocking lactate export by inhibiting the Myc target MCT1 disables glycolysis and glutathione synthesis. Cancer Research, 74, 908–920. https://doi.org/10.1158/0008-5472.CAN-13-2034.PubMedCrossRef Doherty, J. R., Yang, C., Scott, K. E. N., Cameron, M. D., Fallahi, M., Li, W., Hall, M. A., Amelio, A. L., Mishra, J. K., Li, F., Tortosa, M., Genau, H. M., Rounbehler, R. J., Lu, Y., Dang, C. V., Kumar, K. G., Butler, A. A., Bannister, T. D., Hooper, A. T., Unsal-Kacmaz, K., Roush, W. R., & Cleveland, J. L. (2014). Blocking lactate export by inhibiting the Myc target MCT1 disables glycolysis and glutathione synthesis. Cancer Research, 74, 908–920. https://​doi.​org/​10.​1158/​0008-5472.​CAN-13-2034.PubMedCrossRef
77.
78.
go back to reference Mele, L., Paino, F., Papaccio, F., Regad, T., Boocock, D., Stiuso, P., Lombardi, A., Liccardo, D., Aquino, G., Barbieri, A., Arra, C., Coveney, C., la Noce, M., Papaccio, G., Caraglia, M., Tirino, V., & Desiderio, V. (2018). A new inhibitor of glucose-6-phosphate dehydrogenase blocks pentose phosphate pathway and suppresses malignant proliferation and metastasis in vivo. Cell Death & Disease, 9, 572. https://doi.org/10.1038/s41419-018-0635-5.CrossRef Mele, L., Paino, F., Papaccio, F., Regad, T., Boocock, D., Stiuso, P., Lombardi, A., Liccardo, D., Aquino, G., Barbieri, A., Arra, C., Coveney, C., la Noce, M., Papaccio, G., Caraglia, M., Tirino, V., & Desiderio, V. (2018). A new inhibitor of glucose-6-phosphate dehydrogenase blocks pentose phosphate pathway and suppresses malignant proliferation and metastasis in vivo. Cell Death & Disease, 9, 572. https://​doi.​org/​10.​1038/​s41419-018-0635-5.CrossRef
79.
go back to reference Cremon, C., Stanghellini, V., Barbaro, M. R., Cogliandro, R. F., Bellacosa, L., Santos, J., Vicario, M., Pigrau, M., Alonso Cotoner, C., Lobo, B., Azpiroz, F., Bruley des Varannes, S., Neunlist, M., DeFilippis, D., Iuvone, T., Petrosino, S., di Marzo, V., & Barbara, G. (2017). Randomised clinical trial: the analgesic properties of dietary supplementation with palmitoylethanolamide and polydatin in irritable bowel syndrome. Alimentary Pharmacology & Therapeutics, 45, 909–922. https://doi.org/10.1111/apt.13958.CrossRef Cremon, C., Stanghellini, V., Barbaro, M. R., Cogliandro, R. F., Bellacosa, L., Santos, J., Vicario, M., Pigrau, M., Alonso Cotoner, C., Lobo, B., Azpiroz, F., Bruley des Varannes, S., Neunlist, M., DeFilippis, D., Iuvone, T., Petrosino, S., di Marzo, V., & Barbara, G. (2017). Randomised clinical trial: the analgesic properties of dietary supplementation with palmitoylethanolamide and polydatin in irritable bowel syndrome. Alimentary Pharmacology & Therapeutics, 45, 909–922. https://​doi.​org/​10.​1111/​apt.​13958.CrossRef
81.
go back to reference Hitosugi, T., Zhou, L., Elf, S., Fan, J., Kang, H. B., Seo, J. H., Shan, C., Dai, Q., Zhang, L., Xie, J., Gu, T. L., Jin, P., Alečković, M., LeRoy, G., Kang, Y., Sudderth, J. A., DeBerardinis, R. J., Luan, C. H., Chen, G. Z., Muller, S., Shin, D. M., Owonikoko, T. K., Lonial, S., Arellano, M. L., Khoury, H. J., Khuri, F. R., Lee, B. H., Ye, K., Boggon, T. J., Kang, S., He, C., & Chen, J. (2012). Phosphoglycerate mutase 1 coordinates glycolysis and biosynthesis to promote tumor growth. Cancer Cell, 22, 585–600. https://doi.org/10.1016/j.ccr.2012.09.020.PubMedPubMedCentralCrossRef Hitosugi, T., Zhou, L., Elf, S., Fan, J., Kang, H. B., Seo, J. H., Shan, C., Dai, Q., Zhang, L., Xie, J., Gu, T. L., Jin, P., Alečković, M., LeRoy, G., Kang, Y., Sudderth, J. A., DeBerardinis, R. J., Luan, C. H., Chen, G. Z., Muller, S., Shin, D. M., Owonikoko, T. K., Lonial, S., Arellano, M. L., Khoury, H. J., Khuri, F. R., Lee, B. H., Ye, K., Boggon, T. J., Kang, S., He, C., & Chen, J. (2012). Phosphoglycerate mutase 1 coordinates glycolysis and biosynthesis to promote tumor growth. Cancer Cell, 22, 585–600. https://​doi.​org/​10.​1016/​j.​ccr.​2012.​09.​020.PubMedPubMedCentralCrossRef
82.
go back to reference Liberti, M. V., Dai, Z., Wardell, S. E., Baccile, J. A., Liu, X., Gao, X., Baldi, R., Mehrmohamadi, M., Johnson, M. O., Madhukar, N. S., Shestov, A. A., Chio, I. I. C., Elemento, O., Rathmell, J. C., Schroeder, F. C., McDonnell, D. P., & Locasale, J. W. (2017). A predictive model for selective targeting of the Warburg effect through GAPDH inhibition with a natural product. Cell Metabolism, 26, 648–659 e648. https://doi.org/10.1016/j.cmet.2017.08.017.PubMedPubMedCentralCrossRef Liberti, M. V., Dai, Z., Wardell, S. E., Baccile, J. A., Liu, X., Gao, X., Baldi, R., Mehrmohamadi, M., Johnson, M. O., Madhukar, N. S., Shestov, A. A., Chio, I. I. C., Elemento, O., Rathmell, J. C., Schroeder, F. C., McDonnell, D. P., & Locasale, J. W. (2017). A predictive model for selective targeting of the Warburg effect through GAPDH inhibition with a natural product. Cell Metabolism, 26, 648–659 e648. https://​doi.​org/​10.​1016/​j.​cmet.​2017.​08.​017.PubMedPubMedCentralCrossRef
83.
go back to reference Harriman, G., Greenwood, J., Bhat, S., Huang, X., Wang, R., Paul, D., Tong, L., Saha, A. K., Westlin, W. F., Kapeller, R., & Harwood, H. J., Jr. (2016). Acetyl-CoA carboxylase inhibition by ND-630 reduces hepatic steatosis, improves insulin sensitivity, and modulates dyslipidemia in rats. Proceedings of the National Academy of Sciences of the United States of America, 113, E1796–E1805. https://doi.org/10.1073/pnas.1520686113.PubMedPubMedCentralCrossRef Harriman, G., Greenwood, J., Bhat, S., Huang, X., Wang, R., Paul, D., Tong, L., Saha, A. K., Westlin, W. F., Kapeller, R., & Harwood, H. J., Jr. (2016). Acetyl-CoA carboxylase inhibition by ND-630 reduces hepatic steatosis, improves insulin sensitivity, and modulates dyslipidemia in rats. Proceedings of the National Academy of Sciences of the United States of America, 113, E1796–E1805. https://​doi.​org/​10.​1073/​pnas.​1520686113.PubMedPubMedCentralCrossRef
84.
go back to reference Kim, C. W., Addy, C., Kusunoki, J., Anderson, N. N., Deja, S., Fu, X., Burgess, S. C., Li, C., Ruddy, M., Chakravarthy, M., Previs, S., Milstein, S., Fitzgerald, K., Kelley, D. E., & Horton, J. D. (2017). Acetyl CoA carboxylase inhibition reduces hepatic steatosis but elevates plasma triglycerides in mice and humans: a bedside to bench investigation. Cell Metabolism, 26, 576. https://doi.org/10.1016/j.cmet.2017.08.011.PubMedCrossRef Kim, C. W., Addy, C., Kusunoki, J., Anderson, N. N., Deja, S., Fu, X., Burgess, S. C., Li, C., Ruddy, M., Chakravarthy, M., Previs, S., Milstein, S., Fitzgerald, K., Kelley, D. E., & Horton, J. D. (2017). Acetyl CoA carboxylase inhibition reduces hepatic steatosis but elevates plasma triglycerides in mice and humans: a bedside to bench investigation. Cell Metabolism, 26, 576. https://​doi.​org/​10.​1016/​j.​cmet.​2017.​08.​011.PubMedCrossRef
88.
go back to reference Mazarico, J. M., Sanchez-Arevalo Lobo, V. J., Favicchio, R., Greenhalf, W., Costello, E., Carrillo-de Santa Pau, E., Marques, M., Lacal, J. C., Aboagye, E., & Real, F. X. (2016). Choline kinase alpha (CHKalpha) as a therapeutic target in pancreatic ductal adenocarcinoma: expression, predictive value, and sensitivity to inhibitors. Molecular Cancer Therapeutics, 15, 323–333. https://doi.org/10.1158/1535-7163.MCT-15-0214.PubMedCrossRef Mazarico, J. M., Sanchez-Arevalo Lobo, V. J., Favicchio, R., Greenhalf, W., Costello, E., Carrillo-de Santa Pau, E., Marques, M., Lacal, J. C., Aboagye, E., & Real, F. X. (2016). Choline kinase alpha (CHKalpha) as a therapeutic target in pancreatic ductal adenocarcinoma: expression, predictive value, and sensitivity to inhibitors. Molecular Cancer Therapeutics, 15, 323–333. https://​doi.​org/​10.​1158/​1535-7163.​MCT-15-0214.PubMedCrossRef
89.
go back to reference Zaytseva, Y. Y., Rychahou, P. G., le, A. T., Scott, T. L., Flight, R. M., Kim, J. T., Harris, J., Liu, J., Wang, C., Morris, A. J., Sivakumaran, T. A., Fan, T., Moseley, H., Gao, T., Lee, E. Y., Weiss, H. L., Heuer, T. S., Kemble, G., & Evers, M. (2018). Preclinical evaluation of novel fatty acid synthase inhibitors in primary colorectal cancer cells and a patient-derived xenograft model of colorectal cancer. Oncotarget, 9, 24787–24800. https://doi.org/10.18632/oncotarget.25361.PubMedPubMedCentralCrossRef Zaytseva, Y. Y., Rychahou, P. G., le, A. T., Scott, T. L., Flight, R. M., Kim, J. T., Harris, J., Liu, J., Wang, C., Morris, A. J., Sivakumaran, T. A., Fan, T., Moseley, H., Gao, T., Lee, E. Y., Weiss, H. L., Heuer, T. S., Kemble, G., & Evers, M. (2018). Preclinical evaluation of novel fatty acid synthase inhibitors in primary colorectal cancer cells and a patient-derived xenograft model of colorectal cancer. Oncotarget, 9, 24787–24800. https://​doi.​org/​10.​18632/​oncotarget.​25361.PubMedPubMedCentralCrossRef
90.
go back to reference Wang, B. Y., Zhang, J., Wang, J. L., Sun, S., Wang, Z. H., Wang, L. P., Zhang, Q. L., Lv, F. F., Cao, E. Y., Shao, Z. M., Fais, S., & Hu, X. C. (2015). Intermittent high dose proton pump inhibitor enhances the antitumor effects of chemotherapy in metastatic breast cancer. Journal of Experimental & Clinical Cancer Research, 34, 85. https://doi.org/10.1186/s13046-015-0194-x.CrossRef Wang, B. Y., Zhang, J., Wang, J. L., Sun, S., Wang, Z. H., Wang, L. P., Zhang, Q. L., Lv, F. F., Cao, E. Y., Shao, Z. M., Fais, S., & Hu, X. C. (2015). Intermittent high dose proton pump inhibitor enhances the antitumor effects of chemotherapy in metastatic breast cancer. Journal of Experimental & Clinical Cancer Research, 34, 85. https://​doi.​org/​10.​1186/​s13046-015-0194-x.CrossRef
92.
go back to reference Walsh, M., Fais, S., Spugnini, E. P., Harguindey, S., Abu Izneid, T., Scacco, L., Williams, P., Allegrucci, C., Rauch, C., & Omran, Z. (2015). Proton pump inhibitors for the treatment of cancer in companion animals. Journal of Experimental & Clinical Cancer Research, 34, 93. https://doi.org/10.1186/s13046-015-0204-z.CrossRef Walsh, M., Fais, S., Spugnini, E. P., Harguindey, S., Abu Izneid, T., Scacco, L., Williams, P., Allegrucci, C., Rauch, C., & Omran, Z. (2015). Proton pump inhibitors for the treatment of cancer in companion animals. Journal of Experimental & Clinical Cancer Research, 34, 93. https://​doi.​org/​10.​1186/​s13046-015-0204-z.CrossRef
94.
go back to reference Barkey, N. M., Preihs, C., Cornnell, H. H., Martinez, G., Carie, A., Vagner, J., Xu, L., Lloyd, M. C., Lynch, V. M., Hruby, V. J., Sessler, J. L., Sill, K. N., Gillies, R. J., & Morse, D. L. (2013). Development and in vivo quantitative magnetic resonance imaging of polymer micelles targeted to the melanocortin 1 receptor. Journal of Medicinal Chemistry, 56, 6330–6338. https://doi.org/10.1021/jm4005576.PubMedCrossRef Barkey, N. M., Preihs, C., Cornnell, H. H., Martinez, G., Carie, A., Vagner, J., Xu, L., Lloyd, M. C., Lynch, V. M., Hruby, V. J., Sessler, J. L., Sill, K. N., Gillies, R. J., & Morse, D. L. (2013). Development and in vivo quantitative magnetic resonance imaging of polymer micelles targeted to the melanocortin 1 receptor. Journal of Medicinal Chemistry, 56, 6330–6338. https://​doi.​org/​10.​1021/​jm4005576.PubMedCrossRef
104.
go back to reference Antosh, M. P., Wijesinghe, D. D., Shrestha, S., Lanou, R., Huang, Y. H., Hasselbacher, T., Fox, D., Neretti, N., Sun, S., Katenka, N., Cooper, L. N., Andreev, O. A., & Reshetnyak, Y. K. (2015). Enhancement of radiation effect on cancer cells by gold-pHLIP. Proceedings of the National Academy of Sciences of the United States of America, 112, 5372–5376. https://doi.org/10.1073/pnas.1501628112.PubMedPubMedCentralCrossRef Antosh, M. P., Wijesinghe, D. D., Shrestha, S., Lanou, R., Huang, Y. H., Hasselbacher, T., Fox, D., Neretti, N., Sun, S., Katenka, N., Cooper, L. N., Andreev, O. A., & Reshetnyak, Y. K. (2015). Enhancement of radiation effect on cancer cells by gold-pHLIP. Proceedings of the National Academy of Sciences of the United States of America, 112, 5372–5376. https://​doi.​org/​10.​1073/​pnas.​1501628112.PubMedPubMedCentralCrossRef
108.
go back to reference Rahier, N. J., Molinier, N., Long, C., Deshmukh, S. K., Kate, A. S., Ranadive, P., Verekar, S. A., Jiotode, M., Lavhale, R. R., Tokdar, P., Balakrishnan, A., Meignan, S., Robichon, C., Gomes, B., Aussagues, Y., Samson, A., Sautel, F., & Bailly, C. (2015). Anticancer activity of koningic acid and semisynthetic derivatives. Bioorganic & Medicinal Chemistry, 23, 3712–3721. https://doi.org/10.1016/j.bmc.2015.04.004.CrossRef Rahier, N. J., Molinier, N., Long, C., Deshmukh, S. K., Kate, A. S., Ranadive, P., Verekar, S. A., Jiotode, M., Lavhale, R. R., Tokdar, P., Balakrishnan, A., Meignan, S., Robichon, C., Gomes, B., Aussagues, Y., Samson, A., Sautel, F., & Bailly, C. (2015). Anticancer activity of koningic acid and semisynthetic derivatives. Bioorganic & Medicinal Chemistry, 23, 3712–3721. https://​doi.​org/​10.​1016/​j.​bmc.​2015.​04.​004.CrossRef
110.
go back to reference Peng, X., Gong, F., Chen, Y., Jiang, Y., Liu, J., Yu, M., Zhang, S., Wang, M., Xiao, G., & Liao, H. (2014). Autophagy promotes paclitaxel resistance of cervical cancer cells: involvement of Warburg effect activated hypoxia-induced factor 1-alpha-mediated signaling. Cell Death & Disease, 5, e1367. https://doi.org/10.1038/cddis.2014.297.CrossRef Peng, X., Gong, F., Chen, Y., Jiang, Y., Liu, J., Yu, M., Zhang, S., Wang, M., Xiao, G., & Liao, H. (2014). Autophagy promotes paclitaxel resistance of cervical cancer cells: involvement of Warburg effect activated hypoxia-induced factor 1-alpha-mediated signaling. Cell Death & Disease, 5, e1367. https://​doi.​org/​10.​1038/​cddis.​2014.​297.CrossRef
115.
go back to reference Sun, X., du, R., Zhang, L., Zhang, G., Zheng, X., Qian, J., Tian, X., Zhou, J., He, J., Wang, Y., Wu, Y., Zhong, K., Cai, D., Zou, D., & Wu, Z. (2017). A pH-responsive yolk-like nanoplatform for tumor targeted dual-mode magnetic resonance imaging and chemotherapy. ACS Nano, 11, 7049–7059. https://doi.org/10.1021/acsnano.7b02675.PubMedCrossRef Sun, X., du, R., Zhang, L., Zhang, G., Zheng, X., Qian, J., Tian, X., Zhou, J., He, J., Wang, Y., Wu, Y., Zhong, K., Cai, D., Zou, D., & Wu, Z. (2017). A pH-responsive yolk-like nanoplatform for tumor targeted dual-mode magnetic resonance imaging and chemotherapy. ACS Nano, 11, 7049–7059. https://​doi.​org/​10.​1021/​acsnano.​7b02675.PubMedCrossRef
121.
go back to reference Izumi, H., Torigoe, T., Ishiguchi, H., Uramoto, H., Yoshida, Y., Tanabe, M., Ise, T., Murakami, T., Yoshida, T., Nomoto, M., & Kohno, K. (2003). Cellular pH regulators: potentially promising molecular targets for cancer chemotherapy. Cancer Treatment Reviews, 29, 541–549.CrossRefPubMed Izumi, H., Torigoe, T., Ishiguchi, H., Uramoto, H., Yoshida, Y., Tanabe, M., Ise, T., Murakami, T., Yoshida, T., Nomoto, M., & Kohno, K. (2003). Cellular pH regulators: potentially promising molecular targets for cancer chemotherapy. Cancer Treatment Reviews, 29, 541–549.CrossRefPubMed
124.
go back to reference De Milito, A., Marino, M. L., & Fais, S. (2012). A rationale for the use of proton pump inhibitors as antineoplastic agents. Current Pharmaceutical Design, 18, 1395–1406.CrossRefPubMed De Milito, A., Marino, M. L., & Fais, S. (2012). A rationale for the use of proton pump inhibitors as antineoplastic agents. Current Pharmaceutical Design, 18, 1395–1406.CrossRefPubMed
126.
129.
go back to reference Johnson, P. J. (1998). Dermatologic tumors (excluding sarcoids). The Veterinary Clinics of North America. Equine Practice, 14, 625–658, viii.CrossRefPubMed Johnson, P. J. (1998). Dermatologic tumors (excluding sarcoids). The Veterinary Clinics of North America. Equine Practice, 14, 625–658, viii.CrossRefPubMed
133.
go back to reference Papagerakis, S., Bellile, E., Peterson, L. A., Pliakas, M., Balaskas, K., Selman, S., Hanauer, D., Taylor, J. M. G., Duffy, S., & Wolf, G. (2014). Proton pump inhibitors and histamine 2 blockers are associated with improved overall survival in patients with head and neck squamous carcinoma. Cancer Prevention Research (Philadelphia, Pa.), 7, 1258–1269. https://doi.org/10.1158/1940-6207.CAPR-14-0002.CrossRef Papagerakis, S., Bellile, E., Peterson, L. A., Pliakas, M., Balaskas, K., Selman, S., Hanauer, D., Taylor, J. M. G., Duffy, S., & Wolf, G. (2014). Proton pump inhibitors and histamine 2 blockers are associated with improved overall survival in patients with head and neck squamous carcinoma. Cancer Prevention Research (Philadelphia, Pa.), 7, 1258–1269. https://​doi.​org/​10.​1158/​1940-6207.​CAPR-14-0002.CrossRef
137.
go back to reference Sennoune, S. R., Bakunts, K., Martínez, G. M., Chua-Tuan, J. L., Kebir, Y., Attaya, M. N., & Martínez-Zaguilán, R. (2004). Vacuolar H+-ATPase in human breast cancer cells with distinct metastatic potential: distribution and functional activity. American Journal of Physiology. Cell Physiology, 286, C1443–C1452. https://doi.org/10.1152/ajpcell.00407.2003.CrossRefPubMed Sennoune, S. R., Bakunts, K., Martínez, G. M., Chua-Tuan, J. L., Kebir, Y., Attaya, M. N., & Martínez-Zaguilán, R. (2004). Vacuolar H+-ATPase in human breast cancer cells with distinct metastatic potential: distribution and functional activity. American Journal of Physiology. Cell Physiology, 286, C1443–C1452. https://​doi.​org/​10.​1152/​ajpcell.​00407.​2003.CrossRefPubMed
142.
go back to reference Pouyssegur, J., Franchi, A., & Pages, G. (2001). pHi, aerobic glycolysis and vascular endothelial growth factor in tumour growth. Novartis Found Symp, 240, 186–196; discussion 196–188.PubMed Pouyssegur, J., Franchi, A., & Pages, G. (2001). pHi, aerobic glycolysis and vascular endothelial growth factor in tumour growth. Novartis Found Symp, 240, 186–196; discussion 196–188.PubMed
143.
go back to reference Chiche, J., Fur, Y. L., Vilmen, C., Frassineti, F., Daniel, L., Halestrap, A. P., Cozzone, P. J., Pouysségur, J., & Lutz, N. W. (2012). In vivo pH in metabolic-defective Ras-transformed fibroblast tumors: key role of the monocarboxylate transporter, MCT4, for inducing an alkaline intracellular pH. International Journal of Cancer, 130, 1511–1520. https://doi.org/10.1002/ijc.26125.PubMedCrossRef Chiche, J., Fur, Y. L., Vilmen, C., Frassineti, F., Daniel, L., Halestrap, A. P., Cozzone, P. J., Pouysségur, J., & Lutz, N. W. (2012). In vivo pH in metabolic-defective Ras-transformed fibroblast tumors: key role of the monocarboxylate transporter, MCT4, for inducing an alkaline intracellular pH. International Journal of Cancer, 130, 1511–1520. https://​doi.​org/​10.​1002/​ijc.​26125.PubMedCrossRef
144.
146.
go back to reference Commisso, C., Davidson, S. M., Soydaner-Azeloglu, R. G., Parker, S. J., Kamphorst, J. J., Hackett, S., Grabocka, E., Nofal, M., Drebin, J. A., Thompson, C. B., Rabinowitz, J. D., Metallo, C. M., Vander Heiden, M. G., & Bar-Sagi, D. (2013). Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature, 497, 633–637. https://doi.org/10.1038/nature12138.PubMedPubMedCentralCrossRef Commisso, C., Davidson, S. M., Soydaner-Azeloglu, R. G., Parker, S. J., Kamphorst, J. J., Hackett, S., Grabocka, E., Nofal, M., Drebin, J. A., Thompson, C. B., Rabinowitz, J. D., Metallo, C. M., Vander Heiden, M. G., & Bar-Sagi, D. (2013). Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature, 497, 633–637. https://​doi.​org/​10.​1038/​nature12138.PubMedPubMedCentralCrossRef
147.
go back to reference Hosogi, S., Miyazaki, H., Nakajima, K. I., Ashihara, E., Niisato, N., Kusuzaki, K., & Marunaka, Y. (2012). An inhibitor of Na(+)/H(+) exchanger (NHE), ethyl-isopropyl amiloride (EIPA), diminishes proliferation of MKN28 human gastric cancer cells by decreasing the cytosolic Cl(−) concentration via DIDS-sensitive pathways. Cellular Physiology and Biochemistry, 30, 1241–1253. https://doi.org/10.1159/000343315.PubMedCrossRef Hosogi, S., Miyazaki, H., Nakajima, K. I., Ashihara, E., Niisato, N., Kusuzaki, K., & Marunaka, Y. (2012). An inhibitor of Na(+)/H(+) exchanger (NHE), ethyl-isopropyl amiloride (EIPA), diminishes proliferation of MKN28 human gastric cancer cells by decreasing the cytosolic Cl(−) concentration via DIDS-sensitive pathways. Cellular Physiology and Biochemistry, 30, 1241–1253. https://​doi.​org/​10.​1159/​000343315.PubMedCrossRef
148.
go back to reference Kellen, J. A., Mirakian, A., & Kolin, A. (1988). Antimetastatic effect of amiloride in an animal tumour model. Anticancer Research, 8, 1373–1376.PubMed Kellen, J. A., Mirakian, A., & Kolin, A. (1988). Antimetastatic effect of amiloride in an animal tumour model. Anticancer Research, 8, 1373–1376.PubMed
150.
go back to reference Dimmer, K. S., Friedrich, B., Lang, F., Deitmer, J. W., & Broer, S. (2000). The low-affinity monocarboxylate transporter MCT4 is adapted to the export of lactate in highly glycolytic cells. The Biochemical Journal, 350(Pt 1), 219–227.CrossRefPubMedPubMedCentral Dimmer, K. S., Friedrich, B., Lang, F., Deitmer, J. W., & Broer, S. (2000). The low-affinity monocarboxylate transporter MCT4 is adapted to the export of lactate in highly glycolytic cells. The Biochemical Journal, 350(Pt 1), 219–227.CrossRefPubMedPubMedCentral
157.
go back to reference Manning Fox, J. E., Meredith, D., & Halestrap, A. P. (2000). Characterisation of human monocarboxylate transporter 4 substantiates its role in lactic acid efflux from skeletal muscle. The Journal of Physiology, 529(Pt 2), 285–293.PubMedPubMedCentral Manning Fox, J. E., Meredith, D., & Halestrap, A. P. (2000). Characterisation of human monocarboxylate transporter 4 substantiates its role in lactic acid efflux from skeletal muscle. The Journal of Physiology, 529(Pt 2), 285–293.PubMedPubMedCentral
159.
go back to reference Mathupala, S. P., Parajuli, P., & Sloan, A. E. (2004). Silencing of monocarboxylate transporters via small interfering ribonucleic acid inhibits glycolysis and induces cell death in malignant glioma: an in vitro study. Neurosurgery, 55, 1410–1419; discussion 1419.CrossRefPubMed Mathupala, S. P., Parajuli, P., & Sloan, A. E. (2004). Silencing of monocarboxylate transporters via small interfering ribonucleic acid inhibits glycolysis and induces cell death in malignant glioma: an in vitro study. Neurosurgery, 55, 1410–1419; discussion 1419.CrossRefPubMed
160.
163.
go back to reference Baenke, F., Dubuis, S., Brault, C., Weigelt, B., Dankworth, B., Griffiths, B., Jiang, M., Mackay, A., Saunders, B., Spencer-Dene, B., Ros, S., Stamp, G., Reis-Filho, J. S., Howell, M., Zamboni, N., & Schulze, A. (2015). Functional screening identifies MCT4 as a key regulator of breast cancer cell metabolism and survival. The Journal of Pathology, 237, 152–165. https://doi.org/10.1002/path.4562.PubMedCrossRef Baenke, F., Dubuis, S., Brault, C., Weigelt, B., Dankworth, B., Griffiths, B., Jiang, M., Mackay, A., Saunders, B., Spencer-Dene, B., Ros, S., Stamp, G., Reis-Filho, J. S., Howell, M., Zamboni, N., & Schulze, A. (2015). Functional screening identifies MCT4 as a key regulator of breast cancer cell metabolism and survival. The Journal of Pathology, 237, 152–165. https://​doi.​org/​10.​1002/​path.​4562.PubMedCrossRef
168.
go back to reference Pastorek, J., et al. (1994). Cloning and characterization of MN, a human tumor-associated protein with a domain homologous to carbonic anhydrase and a putative helix-loop-helix DNA binding segment. Oncogene, 9, 2877–2888.PubMed Pastorek, J., et al. (1994). Cloning and characterization of MN, a human tumor-associated protein with a domain homologous to carbonic anhydrase and a putative helix-loop-helix DNA binding segment. Oncogene, 9, 2877–2888.PubMed
170.
go back to reference Ilies, M. A., Vullo, D., Pastorek, J., Scozzafava, A., Ilies, M., Caproiu, M. T., Pastorekova, S., & Supuran, C. T. (2003). Carbonic anhydrase inhibitors. Inhibition of tumor-associated isozyme IX by halogenosulfanilamide and halogenophenylaminobenzolamide derivatives. Journal of Medicinal Chemistry, 46, 2187–2196. https://doi.org/10.1021/jm021123s.PubMedCrossRef Ilies, M. A., Vullo, D., Pastorek, J., Scozzafava, A., Ilies, M., Caproiu, M. T., Pastorekova, S., & Supuran, C. T. (2003). Carbonic anhydrase inhibitors. Inhibition of tumor-associated isozyme IX by halogenosulfanilamide and halogenophenylaminobenzolamide derivatives. Journal of Medicinal Chemistry, 46, 2187–2196. https://​doi.​org/​10.​1021/​jm021123s.PubMedCrossRef
173.
go back to reference Andreucci, E., Peppicelli, S., Carta, F., Brisotto, G., Biscontin, E., Ruzzolini, J., Bianchini, F., Biagioni, A., Supuran, C. T., & Calorini, L. (2017). Carbonic anhydrase IX inhibition affects viability of cancer cells adapted to extracellular acidosis. J Mol Med (Berl), 95, 1341–1353. https://doi.org/10.1007/s00109-017-1590-9.CrossRef Andreucci, E., Peppicelli, S., Carta, F., Brisotto, G., Biscontin, E., Ruzzolini, J., Bianchini, F., Biagioni, A., Supuran, C. T., & Calorini, L. (2017). Carbonic anhydrase IX inhibition affects viability of cancer cells adapted to extracellular acidosis. J Mol Med (Berl), 95, 1341–1353. https://​doi.​org/​10.​1007/​s00109-017-1590-9.CrossRef
174.
go back to reference Lou, Y., McDonald, P. C., Oloumi, A., Chia, S., Ostlund, C., Ahmadi, A., Kyle, A., auf dem Keller, U., Leung, S., Huntsman, D., Clarke, B., Sutherland, B. W., Waterhouse, D., Bally, M., Roskelley, C., Overall, C. M., Minchinton, A., Pacchiano, F., Carta, F., Scozzafava, A., Touisni, N., Winum, J. Y., Supuran, C. T., & Dedhar, S. (2011). Targeting tumor hypoxia: suppression of breast tumor growth and metastasis by novel carbonic anhydrase IX inhibitors. Cancer Research, 71, 3364–3376. https://doi.org/10.1158/0008-5472.CAN-10-4261.PubMedCrossRef Lou, Y., McDonald, P. C., Oloumi, A., Chia, S., Ostlund, C., Ahmadi, A., Kyle, A., auf dem Keller, U., Leung, S., Huntsman, D., Clarke, B., Sutherland, B. W., Waterhouse, D., Bally, M., Roskelley, C., Overall, C. M., Minchinton, A., Pacchiano, F., Carta, F., Scozzafava, A., Touisni, N., Winum, J. Y., Supuran, C. T., & Dedhar, S. (2011). Targeting tumor hypoxia: suppression of breast tumor growth and metastasis by novel carbonic anhydrase IX inhibitors. Cancer Research, 71, 3364–3376. https://​doi.​org/​10.​1158/​0008-5472.​CAN-10-4261.PubMedCrossRef
Metadata
Title
Causes, consequences, and therapy of tumors acidosis
Authors
Smitha R. Pillai
Mehdi Damaghi
Yoshinori Marunaka
Enrico Pierluigi Spugnini
Stefano Fais
Robert J. Gillies
Publication date
01-06-2019
Publisher
Springer US
Keyword
Acidosis
Published in
Cancer and Metastasis Reviews / Issue 1-2/2019
Print ISSN: 0167-7659
Electronic ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-019-09792-7

Other articles of this Issue 1-2/2019

Cancer and Metastasis Reviews 1-2/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine