Cancer Letters

Cancer Letters

Volume 477, 1 May 2020, Pages 76-87
Cancer Letters

Mini-review
The metabolic landscape of urological cancers: New therapeutic perspectives

https://doi.org/10.1016/j.canlet.2020.02.034Get rights and content

Highlights

  • Metabolic alterations are critical for cancer development and progression.

  • In urological tumors, metabolic genes upregulation is associated with poor prognosis.

  • Metabolic inhibitors have arisen as novel target therapies.

Abstract

Deregulation of cell metabolism is an established cancer hallmark that contributes to tumor initiation and progression, as well as tumor heterogeneity. In solid tumors, alterations in different metabolic pathways, including glycolysis, pentose phosphate pathway, glutaminolysis and fatty acid metabolism, support the high proliferative rates and macromolecule biosynthesis of cancer cells. Despite advances in therapy, urothelial tumors still exhibit high recurrence and mortality rates, especially in advanced stages of disease. These tumors harbor gene mutations and expression patterns which play an important role in metabolic reprogramming. Taking into account the unique metabolic features underlying carcinogenesis in these cancers, new and promising therapeutic targets based on metabolic alterations must be considered. Furthermore, the combination of metabolic inhibitors with conventional targeted therapies may improve effectiveness of treatments. This review will summarize the metabolic alterations present in urological tumors and the results with metabolic inhibitors currently available.

Introduction

Malignant solid tumors are often heterogeneous, with highly disorganized structures composed by several types of cells in the tumor microenvironment (TME) [1]. Accordingly, solid tumors exhibit diverse metabolic features due several intrinsic factors and also signals imposed by the microenvironment [2]. Importantly, tumor metabolic heterogeneity and flexibility among tumors and within distinct regions of the same tumor, support the presence of a metabolic symbiosis and complementarity between tumor cells and the TME [3]. This metabolic symbiosis has high clinical relevance, since it can help to explain the mechanisms behind the tumor therapeutic vulnerabilities, including treatment failures, and may predict clinical outcomes.

Section snippets

Cancer metabolism

Deregulation of energetic metabolism constitutes a well-established cancer hallmark [4]. The high proliferative rates of cancer cells impose a high demand of energy and macromolecule precursor synthesis to sustain tumor growth and progression [5]. This requirement is ensured by the presence of tumor metabolic reprogramming, a feature present in several tumors.

Prostate cancer

Prostate cancer (PCa) is the most commonly diagnosed cancer among men and the second leading cause of cancer-related death in developed countries [26]. PCa screening relies on prostate-specific antigen (PSA), a widely used prostate cancer marker, in combination to digital rectal examination (DRE) and transrectal ultrasound biopsy [27]. Despite all studies, the molecular pathogenesis of these tumors is not yet fully known. However, fusion chromosomal rearrangements with specific prostate

Conclusions

Considering the unique metabolic features and metabolism-related enzyme deregulation present in urological cancers, new and promising metabolic therapeutic targets should be explored, especially in the treatment of advanced and metastatic disease, where they are most needed. Also, the combination of metabolic inhibitors with conventional standard therapies may improve effectiveness of patients’ treatment.

Until now, in PCa and RCC, metabolic inhibitors demonstrate promising results in

Funding

This work was supported by the Research Center, Portuguese Oncology Institute of Porto (CI-IPOP): PI 112-CI-IPOP 92-2018-MCTKidCan. VM-G received a fellowship from POCI-01-0145-FEDER-29043: Development of novel prognostic and predictive epigenetic biomarkers for malignant testicular germ cell tumors and AL from UID/DTP/00776/POCI-01-0145-FEDER-006868.

Author contributions

AL and VM-G conceptualized the review and drafted the manuscript. AL and VM-G collected and analyzed the information and elaborated the figures. RH, FB and CJ revised the manuscript. All authors read and approved the final manuscript.

CRediT authorship contribution statement

Vera Miranda-Gonçalves: Conceptualization, Writing - original draft. Ana Lameirinhas: Conceptualization, Writing - original draft. Rui Henrique: Writing - original draft. Fátima Baltazar: Writing - review & editing. Carmen Jerónimo: Writing - review & editing.

Declaration of competing interest

None.

References (155)

  • M.C. Franz et al.

    Zinc transporters in prostate cancer

    Mol. Aspect. Med.

    (2013)
  • T. Pan et al.

    Elevated expression of glutaminase confers glucose utilization via glutaminolysis in prostate cancer

    Biochem. Biophys. Res. Commun.

    (2015)
  • D. Ackerman et al.

    Hypoxia, lipids, and cancer: surviving the harsh tumor microenvironment

    Trends Cell Biol.

    (2014)
  • G. Zadra et al.

    The fat side of prostate cancer

    Biochim. Biophys. Acta

    (2013)
  • Z.T. Schug et al.

    Acetyl-CoA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress

    Canc. Cell

    (2015)
  • D.K. Nomura et al.

    Monoacylglycerol lipase exerts dual control over endocannabinoid and fatty acid pathways to support prostate cancer

    Chem. Biol.

    (2011)
  • D. Robinson et al.

    Integrative clinical genomics of advanced prostate cancer

    Cell

    (2015)
  • E. Scott

    Chemohormonal therapy in metastatic hormone-sensitive prostate cancer. Sweeney CJ, chen YH, carducci M, liu G, jarrard DF, eisenberger M, wong YN, hahn N, kohli M, cooney MM, dreicer R, vogelzang NJ, picus J, shevrin D, hussain M, garcia JA, DiPaola RS. Department of medicine; department of biostatistics and computational biology; dana-farber cancer institute, boston; harvard medical school, boston; johns hopkins university, baltimore; university of Wisconsin carbone cancer center; school of medicine and public health; madison; fox chase cancer center, temple university health system, philadelphia; Indiana university melvin and bren simon cancer center, indianapolis; mayo clinic, rochester, MN; university hospitals case medical center, seidman cancer center; cleveland clinic taussig cancer institute; both in cleveland; university of Virginia cancer center, charlottesville; comprehensive cancer centers of Nevada, las vegas; siteman cancer center, Washington university school of medicine, st. Louis; NorthShore university health system, evanston, IL; university of Michigan comprehensive cancer center, ann arbor; rutgers cancer institute of New Jersey, new brunswick.N engl J med. 2015 aug 20;373(8):737-746. [Epub 2015 aug 5]. Doi: 10.1056/NEJMoa1503747

    Urol. Oncol.

    (2017)
  • N.D. James et al.

    Survival with newly diagnosed metastatic prostate cancer in the "docetaxel era": data from 917 patients in the control arm of the STAMPEDE trial (MRC PR08, CRUK/06/019)

    Eur. Urol.

    (2015)
  • N.B. Haas et al.

    Hereditary renal cancer syndromes

    Adv. Chron. Kidney Dis.

    (2014)
  • D.J. Sanchez et al.

    Genetic and metabolic hallmarks of clear cell renal cell carcinoma

    Biochim. Biophys. Acta Rev. Canc

    (2018)
  • J. Schodel et al.

    Hypoxia, hypoxia-inducible transcription factors, and renal cancer

    Eur. Urol.

    (2016)
  • M.A. Selak et al.

    Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-alpha prolyl hydroxylase

    Canc. Cell

    (2005)
  • J.S. Isaacs et al.

    HIF overexpression correlates with biallelic loss of fumarate hydratase in renal cancer: novel role of fumarate in regulation of HIF stability

    Canc. Cell

    (2005)
  • H.Y. Lim et al.

    Metabolic signatures of renal cell carcinoma

    Biochem. Biophys. Res. Commun.

    (2015)
  • A. Horiguchi et al.

    Fatty acid synthase over expression is an indicator of tumor aggressiveness and poor prognosis in renal cell carcinoma

    J. Urol.

    (2008)
  • B. Escudier et al.

    Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up

    Ann. Oncol.

    (2016)
  • K. Nath et al.

    Mechanism of antineoplastic activity of lonidamine

    Biochim. Biophys. Acta

    (2016)
  • A. Kinnaird et al.

    Metabolic modulation of clear-cell renal cell carcinoma with dichloroacetate, an inhibitor of pyruvate dehydrogenase kinase

    Eur. Urol.

    (2016)
  • P.A. Gameiro et al.

    In vivo HIF-mediated reductive carboxylation is regulated by citrate levels and sensitizes VHL-deficient cells to glutamine deprivation

    Cell Metabol.

    (2013)
  • O. Warburg

    On the origin of cancer cells

    Science

    (1956)
  • O. Warburg

    On respiratory impairment in cancer cells

    Science

    (1956)
  • R.A. Gatenby et al.

    Why do cancers have high aerobic glycolysis?

    Nat. Rev. Canc.

    (2004)
  • C. Zhang et al.

    Glucose-6-phosphate dehydrogenase: a biomarker and potential therapeutic target for cancer

    Anticanc. Agents Med. Chem.

    (2014)
  • F. Hirschhaeuser et al.

    Lactate: a metabolic key player in cancer

    Canc. Res..

    (2011)
  • K.L. Eales et al.

    Hypoxia and metabolic adaptation of cancer cells

    Oncogenesis

    (2016)
  • N.C. Denko

    Hypoxia, HIF1 and glucose metabolism in the solid tumour

    Nat. Rev. Canc.

    (2008)
  • J. Chiche et al.

    Tumour hypoxia induces a metabolic shift causing acidosis: a common feature in cancer

    J. Cell Mol. Med.

    (2010)
  • F. Bost et al.

    Energy disruptors: rising stars in anticancer therapy?

    Oncogenesis

    (2016)
  • L. Yang et al.

    Glutaminolysis: a hallmark of cancer metabolism

    Annu. Rev. Biomed. Eng.

    (2017)
  • Y.K. Choi et al.

    Targeting glutamine metabolism for cancer treatment

    Biomol. Ther. (Seoul)

    (2018)
  • D.R. Wise et al.

    Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction

    Proc. Natl. Acad. Sci. U. S. A.

    (2008)
  • S. Beloribi-Djefaflia et al.

    Lipid metabolic reprogramming in cancer cells

    Oncogenesis

    (2016)
  • C. Cheng et al.

    Lipid metabolism reprogramming and its potential targets in cancer

    Canc. Commun.

    (2018)
  • J.J. Kamphorst et al.

    Quantitative analysis of acetyl-CoA production in hypoxic cancer cells reveals substantial contribution from acetate

    Canc. Metabol.

    (2014)
  • F. Rohrig et al.

    The multifaceted roles of fatty acid synthesis in cancer

    Nat. Rev. Canc.

    (2016)
  • K. Zaugg et al.

    Carnitine palmitoyltransferase 1C promotes cell survival and tumor growth under conditions of metabolic stress

    Genes Dev.

    (2011)
  • R.L. Siegel et al.

    Cancer statistics, 2018

    CA: A Canc. J. Clin.

    (2018)
  • I. Graça et al.

    Epigenetic modulators as therapeutic targets in prostate cancer

    Clin. Epigenet.

    (2016)
  • K.E. Knudsen et al.

    Starving the addiction: new opportunities for durable suppression of AR signaling in prostate cancer

    Clin. Canc. Res.

    (2009)
  • Cited by (0)

    1

    Equal contribution.

    2

    Joint Senior authors.

    View full text