PI3K/AKT/mTOR Dysregulation and Reprogramming Metabolic Pathways in Renal Cancer: Crosstalk with the VHL/HIF Axis

Renal cell carcinoma (RCC) represents 85–95% of kidney cancers and is the most frequent type of renal cancer in adult patients. It accounts for 3% of all cancer cases and is in 7th place among the most frequent histological types of cancer. Clear cell renal cell carcinoma (ccRCC), accounts for 75% of RCCs and has the most kidney cancer-related deaths. One-third of the patients with ccRCC develop metastases. Renal cancer presents cellular alterations in sugars, lipids, amino acids, and nucleic acid metabolism. RCC is characterized by several metabolic dysregulations including oxygen sensing (VHL/HIF pathway), glucose transporters (GLUT 1 and GLUT 4) energy sensing, and energy nutrient sensing cascade. Metabolic reprogramming represents an important characteristic of the cancer cells to survive in nutrient and oxygen-deprived environments, to proliferate and metastasize in different body sites. The phosphoinositide 3-kinase-AKT-mammalian target of the rapamycin (PI3K/AKT/mTOR) signaling pathway is usually dysregulated in various cancer types including renal cancer. This molecular pathway is frequently correlated with tumor growth and survival. The main aim of this review is to present renal cancer types, dysregulation of PI3K/AKT/mTOR signaling pathway members, crosstalk with VHL/HIF axis, and carbohydrates, lipids, and amino acid alterations.

[1]  M. Maggi,et al.  Radiogenomics in Renal Cancer Management—Current Evidence and Future Prospects , 2023, International journal of molecular sciences.

[2]  Hao Huang,et al.  A novel lipid metabolism gene signature for clear cell renal cell carcinoma using integrated bioinformatics analysis , 2023, Frontiers in Cell and Developmental Biology.

[3]  O. Andronic,et al.  Oral Arginine Supplementation in Healthy Individuals Performing Regular Resistance Training , 2023, Healthcare.

[4]  L. Spain,et al.  Navigating the Current Landscape of Non-Clear Cell Renal Cell Carcinoma: A Review of the Literature , 2023, Current oncology.

[5]  Mohammad H Al-thnaibat,et al.  Etiologies, Gross Appearance, Histopathological Patterns, Prognosis, and Best Treatments for Subtypes of Renal Carcinoma: An Educational Review , 2022, Cureus.

[6]  Guo Ci Teo,et al.  Histopathologic and proteogenomic heterogeneity reveals features of clear cell renal cell carcinoma aggressiveness. , 2022, Cancer cell.

[7]  Mark W. Ball,et al.  Tumorigenesis Mechanisms Found in Hereditary Renal Cell Carcinoma: A Review , 2022, Genes.

[8]  R. Bardan,et al.  Contemporary Clinical Definitions, Differential Diagnosis, and Novel Predictive Tools for Renal Cell Carcinoma , 2022, Biomedicines.

[9]  J. Carles,et al.  Update in collecting duct carcinoma: Current aspects of the clinical and molecular characterization of an orphan disease , 2022, Frontiers in Oncology.

[10]  N. Khurana,et al.  Primary leiomyosarcoma kidney – A rare entity with a diagnostic challenge , 2022, Journal of cancer research and therapeutics.

[11]  D. Sahu,et al.  Differential expression profiling of onco and tumor-suppressor genes from major-signaling pathways in Wilms’ tumor , 2022, Pediatric Surgery International.

[12]  W. Kaelin Von Hippel–Lindau disease: insights into oxygen sensing, protein degradation, and cancer , 2022, The Journal of clinical investigation.

[13]  Tao Lin,et al.  Immune-related gene signature associates with immune landscape and predicts prognosis accurately in patients with Wilms tumour , 2022, Frontiers in Immunology.

[14]  M. Tretiakova What’s new in kidney tumor pathology 2022: WHO 5th edition updates , 2022, Journal of pathology and translational medicine.

[15]  Yang Liu,et al.  A Randomized Study on the Effect of Metformin Combined with Intensive-Exercise Diet Therapy on Glucose and Lipid Metabolism and Islet Function in Patients with Renal Cell Carcinoma and Diabetes , 2022, Disease markers.

[16]  R. Wilson,et al.  Molecular Heterogeneity in Pediatric Malignant Rhabdoid Tumors in Patients With Multi-Organ Involvement , 2022, Frontiers in Oncology.

[17]  M. Rubin,et al.  The 2022 World Health Organization Classification of Tumours of the Urinary System and Male Genital Organs-Part A: Renal, Penile, and Testicular Tumours. , 2022, European urology.

[18]  A. Hong,et al.  Recent Advances in Renal Medullary Carcinoma , 2022, International journal of molecular sciences.

[19]  N. Ahmed,et al.  Precision Medicine: An Optimal Approach to Patient Care in Renal Cell Carcinoma , 2022, Frontiers in Medicine.

[20]  Jinu Kim,et al.  Mitochondrial metabolic reprogramming by SIRT3 regulation ameliorates drug resistance in renal cell carcinoma , 2022, PloS one.

[21]  Ming Wu,et al.  C1q/tumour necrosis factor-related protein-3 alleviates high-glucose-induced lipid accumulation and necroinflammation in renal tubular cells by activating the adenosine monophosphate-activated protein kinase pathway. , 2022, The international journal of biochemistry & cell biology.

[22]  U. Hofmann,et al.  Nicotinamide‐N‐methyltransferase is a promising metabolic drug target for primary and metastatic clear cell renal cell carcinoma , 2022, Clinical and translational medicine.

[23]  R. Mehrazin,et al.  Radiogenomics in Clear Cell Renal Cell Carcinoma: A Review of the Current Status and Future Directions , 2022, Cancers.

[24]  H. Chang,et al.  Identification of HGD and GSTZ1 as Biomarkers Involved Metabolic Reprogramming in Kidney Renal Clear Cell Carcinoma , 2022, International journal of molecular sciences.

[25]  M. Chintagumpala,et al.  Outcomes based on histopathologic response to preoperative chemotherapy in children with bilateral Wilms tumor: A prospective study (COG AREN0534) , 2022, Cancer.

[26]  S. Daneshmand,et al.  Pediatric genitourinary tumors: Distribution, demographics, and outcomes , 2022, Pediatric investigation.

[27]  T. Darré,et al.  Renal Leiomyosarcoma, a Rare Presentation , 2022, Journal of kidney cancer and VHL.

[28]  M. Baranowski,et al.  Grade‐dependent changes in sphingolipid metabolism in clear cell renal cell carcinoma , 2022, Journal of cellular biochemistry.

[29]  H. Al-Ali,et al.  Adaptive and maladaptive roles of lipid droplets in health and disease. , 2022, American journal of physiology. Cell physiology.

[30]  Xinjian Li,et al.  Fumarate inhibits PTEN to promote tumorigenesis and therapeutic resistance of type2 papillary renal cell carcinoma. , 2022, Molecular cell.

[31]  Teruhiko Yoshida,et al.  A novel pathogenic variant of the FH gene in a family with hereditary leiomyomatosis and renal cell carcinoma , 2022, Human Genome Variation.

[32]  Shivani Akula,et al.  The role of mediator subunit 12 in tumorigenesis and cancer therapeutics , 2022, Oncology letters.

[33]  Erguang Li,et al.  Single-cell chromatin accessibility landscape in kidney identifies additional cell-of-origin in heterogenous papillary renal cell carcinoma , 2022, Nature Communications.

[34]  Yunwei Han,et al.  Biological Role and Mechanism of Lipid Metabolism Reprogramming Related Gene ECHS1 in Cancer , 2022, Technology in cancer research & treatment.

[35]  Wei Yan,et al.  FOXF2 Regulates PRUNE2 Transcription in the Pathogenesis of Colorectal Cancer , 2022, Technology in cancer research & treatment.

[36]  Xudong Yang,et al.  Role of Metabolic Reprogramming of Long non-coding RNA in Clear Cell Renal Cell Carcinoma , 2022, Journal of Cancer.

[37]  R. Mehra,et al.  Chromophobe renal cell carcinoma: Novel molecular insights and clinicopathologic updates , 2021, Asian journal of urology.

[38]  N. Nonomura,et al.  Comparison of molecular profiles of upper tract urothelial carcinoma vs. urinary bladder cancer in the era of targeted therapy: a narrative review , 2021, Translational andrology and urology.

[39]  H. Daw,et al.  Overview of histologic variants of urothelial carcinoma: current trends and narrative review on treatment outcomes , 2021, Translational andrology and urology.

[40]  T. Deguchi,et al.  Derivative Chromosome 3 Loss from t(3;6)(q12;q14) Followed by Differential VHL Mutations Underlie Multifocal ccRCC , 2022, Cancer Genomics & Proteomics.

[41]  Beata Franczyk,et al.  Characteristics of Clear Cell Papillary Renal Cell Carcinoma (ccpRCC) , 2021, International journal of molecular sciences.

[42]  P. Russo,et al.  Papillary Renal Cell Carcinoma: A Single Institutional Study of 199 Cases Addressing Classification, Clinicopathologic and Molecular Features, and Treatment Outcome , 2021, Modern Pathology.

[43]  A. Italiano,et al.  Improving Immunotherapy Efficacy in Soft-Tissue Sarcomas: A Biomarker Driven and Histotype Tailored Review , 2021, Frontiers in Immunology.

[44]  Yidong Fan,et al.  SETD8 stabilized by USP17 epigenetically activates SREBP1 pathway to drive lipogenesis and oncogenesis of ccRCC. , 2021, Cancer letters.

[45]  Kui-Sheng Chen,et al.  Targeting PI3K/Akt signal transduction for cancer therapy , 2021, Signal Transduction and Targeted Therapy.

[46]  Degang Ding,et al.  Revealing potential lipid biomarkers in clear cell renal cell carcinoma using targeted quantitative lipidomics , 2021, Lipids in health and disease.

[47]  L. Matherly,et al.  Lipid metabolism reprogramming in renal cell carcinoma , 2021, Cancer and Metastasis Reviews.

[48]  E. Henske,et al.  Renal Cell Carcinoma in Tuberous Sclerosis Complex , 2021, Genes.

[49]  Guangzhen Wu,et al.  The Uniqueness of Clear Cell Renal Cell Carcinoma: Summary of the Process and Abnormality of Glucose Metabolism and Lipid Metabolism in ccRCC , 2021, Frontiers in Oncology.

[50]  Laurentiu M. Pop,et al.  Renal Lipid Metabolism Abnormalities in Obesity and Clear Cell Renal Cell Carcinoma , 2021, Metabolites.

[51]  Fengzhi Li,et al.  Kidney cancer biomarkers and targets for therapeutics: survivin (BIRC5), XIAP, MCL-1, HIF1α, HIF2α, NRF2, MDM2, MDM4, p53, KRAS and AKT in renal cell carcinoma , 2021, Journal of experimental & clinical cancer research : CR.

[52]  Yan Lin,et al.  Deficiency of the X-inactivation escaping gene KDM5C in clear cell renal cell carcinoma promotes tumorigenicity by reprogramming glycogen metabolism and inhibiting ferroptosis , 2021, Theranostics.

[53]  M. Weber,et al.  Primary renal sarcomas: imaging features and discrimination from non-sarcoma renal tumors , 2021, European Radiology.

[54]  C. Rübe,et al.  Prognostic Factors for Wilms Tumor Recurrence: A Review of the Literature , 2021, Cancers.

[55]  Hui Zheng,et al.  VHL regulates the sensitivity of clear cell renal cell carcinoma to SIRT4-mediated metabolic stress via HIF-1α/HO-1 pathway , 2021, Cell Death & Disease.

[56]  Sounak Gupta An update on the pathology of collecting duct & papillary renal cell carcinoma with a discussion of SNP-Arrays as an emerging laboratory technique. , 2021, Urologic oncology.

[57]  S. Pal,et al.  "Collecting duct carcinoma of the kidney: diagnosis and implications for management". , 2021, Urologic oncology.

[58]  S. Yuan,et al.  Targeting GLS1 to cancer therapy through glutamine metabolism , 2021, Clinical and Translational Oncology.

[59]  T. Choueiri,et al.  Metabolic reprogramming in renal cancer: Events of a metabolic disease. , 2021, Biochimica et biophysica acta. Reviews on cancer.

[60]  M. Björkholm,et al.  Regulatory region mutations of TERT, PLEKHS1 and GPR126 genes as urinary biomarkers in upper tract urothelial carcinomas , 2021, Journal of Cancer.

[61]  F. Baltazar,et al.  Cancer Cells’ Metabolism Dynamics in Renal Cell Carcinoma Patients’ Outcome: Influence of GLUT-1-Related hsa-miR-144 and hsa-miR-186 , 2021, Cancers.

[62]  R. Garje,et al.  Comprehensive Review of Chromophobe Renal Cell Carcinoma. , 2021, Critical reviews in oncology/hematology.

[63]  L. Pellerin,et al.  Inhibition of eIF5A hypusination reprogrammes metabolism and glucose handling in mouse kidney , 2021, Cell Death & Disease.

[64]  M. Simon,et al.  Glycogen metabolism is dispensable for tumor progression in clear cell renal cell carcinoma , 2021, Nature Metabolism.

[65]  A. Jemal,et al.  Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries , 2021, CA: a cancer journal for clinicians.

[66]  G. Sauter,et al.  Y-chromosome loss is frequent in male renal tumors , 2021, Annals of translational medicine.

[67]  Nora C. Toussaint,et al.  The Tumor Profiler Study: integrated, multi-omic, functional tumor profiling for clinical decision support. , 2021, Cancer cell.

[68]  M. Gessler,et al.  Characteristics and outcome of pediatric renal cell carcinoma patients registered in the International Society of Pediatric Oncology (SIOP) 93‐01, 2001 and UK‐IMPORT database: A report of the SIOP‐Renal Tumor Study Group , 2021, International journal of cancer.

[69]  M. V. Heiden,et al.  Cell Programmed Nutrient Partitioning in the Tumor Microenvironment , 2020, bioRxiv.

[70]  J. Mora,et al.  Comprehensive Biology and Genetics Compendium of Wilms Tumor Cell Lines with Different WT1 Mutations , 2020, Cancers.

[71]  C. Porta,et al.  Integration of Lipidomics and Transcriptomics Reveals Reprogramming of the Lipid Metabolism and Composition in Clear Cell Renal Cell Carcinoma , 2020, Metabolites.

[72]  M. Yaspo,et al.  Endocytosis-Mediated Replenishment of Amino Acids Favors Cancer Cell Proliferation and Survival in Chromophobe Renal Cell Carcinoma , 2020, Cancer Research.

[73]  Jong-il Kim,et al.  Treatment strategy for papillary renal cell carcinoma type 2: a case series of seven patients treated based on next generation sequencing data , 2020, Annals of translational medicine.

[74]  W. Kaelin,et al.  Targeting the HIF2–VEGF axis in renal cell carcinoma , 2020, Nature Medicine.

[75]  Yifan Chen,et al.  Research progress of mTOR inhibitors. , 2020, European journal of medicinal chemistry.

[76]  G. Lopes,et al.  Unusual Presentation of Renal Medullary Carcinoma With Undiagnosed Sickle Cell Trait , 2020, Cureus.

[77]  J. Hsieh,et al.  Targeting Metabolic Pathways in Kidney Cancer , 2020, Cancer journal.

[78]  F. Tang,et al.  The tumour microenvironment and metabolism in renal cell carcinoma targeted or immune therapy , 2020, Journal of cellular physiology.

[79]  U. Testa,et al.  Genetic Alterations in Renal Cancers: Identification of The Mechanisms Underlying Cancer Initiation and Progression and of Therapeutic Targets , 2020, Medicines.

[80]  G. De Velasco,et al.  Molecular characterization of chromophobe renal cell carcinoma reveals mTOR pathway alterations in patients with poor outcome , 2020, Modern Pathology.

[81]  E. Philip,et al.  Genomic profiling in renal cell carcinoma , 2020, Nature Reviews Nephrology.

[82]  A. Papavassiliou,et al.  Polycystin-1 induces activation of the PI3K/AKT/mTOR pathway and promotes angiogenesis in renal cell carcinoma. , 2020, Cancer letters.

[83]  W. Rathmell,et al.  Beyond glycolysis: Hypoxia signaling as a master regulator of alternative metabolic pathways and the implications in clear cell renal cell carcinoma. , 2020, Cancer letters.

[84]  A. Kallam,et al.  Clear Cell Renal Carcinoma , 2020 .

[85]  Tao Tao,et al.  mTOR signaling pathway and mTOR inhibitors in cancer: progress and challenges , 2020, Cell & Bioscience.

[86]  Christian M. Metallo,et al.  Cystine transporter regulation of pentose phosphate pathway dependency and disulfide stress exposes a targetable metabolic vulnerability in cancer , 2020, Nature Cell Biology.

[87]  James R. Anderson,et al.  Activity of Vincristine and Irinotecan in Diffuse Anaplastic Wilms Tumor and Therapy Outcomes of Stage II to IV Disease: Results of the Children's Oncology Group AREN0321 Study. , 2020, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[88]  F. Baltazar,et al.  The metabolic landscape of urological cancers: new therapeutic perspectives. , 2020, Cancer letters.

[89]  L. Cantley,et al.  Insulin–PI3K signalling: an evolutionarily insulated metabolic driver of cancer , 2020, Nature Reviews Endocrinology.

[90]  Hiromi I. Wettersten,et al.  Reprogramming of Metabolism in Kidney Cancer. , 2020, Seminars in nephrology.

[91]  C. Creighton Proteomic signatures of clear cell renal cell carcinoma , 2019, Nature Reviews Nephrology.

[92]  M. Broggini,et al.  Downregulation of class II phosphoinositide 3-kinase PI3K-C2β delays cell division and potentiates the effect of docetaxel on cancer cell growth , 2019, Journal of Experimental & Clinical Cancer Research.

[93]  Marcello Polesel,et al.  Targeting Glycolysis in Proliferative Kidney Diseases. , 2019, American journal of physiology. Renal physiology.

[94]  Bing Su,et al.  Sin1-mediated mTOR signaling in cell growth, metabolism and immune response , 2019, National science review.

[95]  Sungpil Yoon,et al.  Knockdown of Pyruvate Kinase M2 Inhibits Cell Proliferation, Metabolism, and Migration in Renal Cell Carcinoma , 2019, International journal of molecular sciences.

[96]  Guo Ci Teo,et al.  Integrated Proteogenomic Characterization of Clear Cell Renal Cell Carcinoma , 2019, Cell.

[97]  Fatima Shirly Anitha,et al.  Atypical Presentation of Renal Leiomyosarcoma: A Case Report , 2019, Cureus.

[98]  Yi Xiao,et al.  Glutathione Metabolism in Renal Cell Carcinoma Progression and Implications for Therapies , 2019, International Journal of Molecular Sciences.

[99]  Xianrang Song,et al.  Physiological functions of Wilms’ tumor 1‐associating protein and its role in tumourigenesis , 2019, Journal of cellular biochemistry.

[100]  A. Carnero,et al.  c‐MYB‐ and PGC1a‐dependent metabolic switch induced by MYBBP1A loss in renal cancer , 2019, Molecular oncology.

[101]  J. Katzenellenbogen,et al.  Free Fatty Acids Rewire Cancer Metabolism in Obesity-Associated Breast Cancer via Estrogen Receptor and mTOR Signaling. , 2019, Cancer research.

[102]  R. Henrique,et al.  The Complex Interplay between Metabolic Reprogramming and Epigenetic Alterations in Renal Cell Carcinoma , 2019, Genes.

[103]  Scott R. Kennedy,et al.  Abnormal oxidative metabolism in a quiet genomic background underlies clear cell papillary renal cell carcinoma , 2019, eLife.

[104]  Y. Akao,et al.  Synthetic miR-143 Exhibited an Anti-Cancer Effect via the Downregulation of K-RAS Networks of Renal Cell Cancer Cells In Vitro and In Vivo , 2019, Molecular therapy : the journal of the American Society of Gene Therapy.

[105]  E. Bruner,et al.  Renal Medullary Carcinoma. , 2019, Archives of pathology & laboratory medicine.

[106]  T. A. Al Hussain,et al.  Papillary Renal Cell Carcinoma (PRCC): An Update. , 2019, Advances in anatomic pathology.

[107]  P. Dzięgiel,et al.  The Usefulness of Immunohistochemistry in the Differential Diagnosis of Lesions Originating from the Myometrium , 2019, International journal of molecular sciences.

[108]  Gemma Sangüesa,et al.  mTOR is a Key Protein Involved in the Metabolic Effects of Simple Sugars , 2019, International journal of molecular sciences.

[109]  W. Linehan,et al.  Updated Recommendations on the Diagnosis, Management, and Clinical Trial Eligibility Criteria for Patients With Renal Medullary Carcinoma. , 2019, Clinical genitourinary cancer.

[110]  A. Fan,et al.  The ‘Achilles Heel’ of Metabolism in Renal Cell Carcinoma: Glutaminase Inhibition as a Rational Treatment Strategy , 2019, Kidney cancer.

[111]  Yuquan Wei,et al.  Glutaminase 1 expression in colorectal cancer cells is induced by hypoxia and required for tumor growth, invasion, and metastatic colonization , 2019, Cell Death & Disease.

[112]  W. Rathmell,et al.  Hypoxia, angiogenesis, and metabolism in the hereditary kidney cancers , 2019, The Journal of clinical investigation.

[113]  A. Nukui,et al.  Immunohistochemical expressionof sodium-dependent glucose transporter - 2 (SGLT-2) in clear cell renal carcinoma: possible prognostic implications , 2019, International braz j urol : official journal of the Brazilian Society of Urology.

[114]  D. Barupal,et al.  Arginine reprogramming in ADPKD results in arginine-dependent cystogenesis. , 2018, American journal of physiology. Renal physiology.

[115]  C. Greenwood,et al.  Translational and HIF-1α-Dependent Metabolic Reprogramming Underpin Metabolic Plasticity and Responses to Kinase Inhibitors and Biguanides. , 2018, Cell metabolism.

[116]  N. Agarwal,et al.  Unclassified renal cell carcinoma: diagnostic difficulties and treatment modalities , 2018, Research and reports in urology.

[117]  Y. Takuwa,et al.  The class II phosphoinositide 3-kinases PI3K-C2α and PI3K-C2β differentially regulate clathrin-dependent pinocytosis in human vascular endothelial cells , 2018, The Journal of Physiological Sciences.

[118]  D. Smiraglia,et al.  Dietary Protein Restriction Reprograms Tumor-Associated Macrophages and Enhances Immunotherapy , 2018, Clinical Cancer Research.

[119]  E. Paraskeva,et al.  Expression of AGPAT2, an enzyme involved in the glycerophospholipid/triacylglycerol biosynthesis pathway, is directly regulated by HIF-1 and promotes survival and etoposide resistance of cancer cells under hypoxia. , 2018, Biochimica et biophysica acta. Molecular and cell biology of lipids.

[120]  M. Simon,et al.  Genetic and metabolic hallmarks of clear cell renal cell carcinoma. , 2018, Biochimica et biophysica acta. Reviews on cancer.

[121]  V. Haucke,et al.  Autoregulation of Class II Alpha PI3K Activity by Its Lipid-Binding PX-C2 Domain Module. , 2018, Molecular cell.

[122]  G. Medina-Gómez,et al.  Maintenance of Kidney Metabolic Homeostasis by PPAR Gamma , 2018, International journal of molecular sciences.

[123]  D. Felsher,et al.  The glutathione redox system is essential to prevent ferroptosis caused by impaired lipid metabolism in clear cell renal cell carcinoma , 2018, Oncogene.

[124]  Yu Cheng,et al.  PTEN in kidney cancer: A review and meta-analysis. , 2018, Clinica chimica acta; international journal of clinical chemistry.

[125]  Y. T. Yeung,et al.  Signaling Pathways in Inflammation and Anti-inflammatory Therapies. , 2018, Current pharmaceutical design.

[126]  E. Lam,et al.  Renal cell carcinoma: a review of biology and pathophysiology , 2018, F1000Research.

[127]  R. Weiss Metabolomics and Metabolic Reprogramming in Kidney Cancer. , 2018, Seminars in nephrology.

[128]  A. Hartmann,et al.  Metabolic and Lipidomic Reprogramming in Renal Cell Carcinoma Subtypes Reflects Regions of Tumor Origin. , 2018, European urology focus.

[129]  Hao Yang,et al.  SREBP1-driven lipid desaturation supports clear cell renal cell carcinoma growth through regulation of NF-κB signaling. , 2018, Biochemical and biophysical research communications.

[130]  Ping Zhang,et al.  The pro-apoptosis effect of sinomenine in renal carcinoma via inducing autophagy through inactivating PI3K/AKT/mTOR pathway. , 2018, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie.

[131]  E. Burdmann,et al.  Assessment of Kidney Function in Patients With Cancer. , 2018, Advances in chronic kidney disease.

[132]  B. Delahunt,et al.  Clear cell renal cell carcinoma: validation of World Health Organization/International Society of Urological Pathology grading , 2017, Histopathology.

[133]  S. Welford,et al.  HIF drives lipid deposition and cancer in ccRCC via repression of fatty acid metabolism , 2017, Nature Communications.

[134]  H. Nishiyama,et al.  Epidemiology of urothelial carcinoma , 2017, International journal of urology : official journal of the Japanese Urological Association.

[135]  Ligong Chen,et al.  Zoledronate dysregulates fatty acid metabolism in renal tubular epithelial cells to induce nephrotoxicity , 2017, Archives of Toxicology.

[136]  Lewis C. Cantley,et al.  The PI3K Pathway in Human Disease , 2017, Cell.

[137]  B. Caldwell,et al.  Current Management for Pediatric Urologic Oncology. , 2017, Advances in pediatrics.

[138]  L. Elo,et al.  HIF prolyl hydroxylase PHD3 regulates translational machinery and glucose metabolism in clear cell renal cell carcinoma , 2017, Cancer & metabolism.

[139]  Emanuel J. V. Gonçalves,et al.  Post-translational regulation of metabolism in fumarate hydratase deficient cancer cells , 2017, bioRxiv.

[140]  R. Weiss,et al.  Metabolic reprogramming in clear cell renal cell carcinoma , 2017, Nature Reviews Nephrology.

[141]  Xiaoqiang Guo,et al.  The Emerging Role of Histone Demethylases in Renal Cell Carcinoma , 2017, Journal of kidney cancer and VHL.

[142]  G. Stephanopoulos,et al.  Glutaminase and poly(ADP-ribose) polymerase inhibitors suppress pyrimidine synthesis and VHL-deficient renal cancers , 2017, The Journal of clinical investigation.

[143]  Y. Oda,et al.  Oncogenic roles of SMARCB1/INI1 and its deficient tumors , 2017, Cancer science.

[144]  Charles Swanton,et al.  Renal cell carcinoma , 2017, Nature Reviews Disease Primers.

[145]  E. Grande,et al.  Targeting HIF-2 α in clear cell renal cell carcinoma: A promising therapeutic strategy. , 2017, Critical reviews in oncology/hematology.

[146]  M. Aoki,et al.  Oncogenic Roles of the PI3K/AKT/mTOR Axis. , 2017, Current topics in microbiology and immunology.

[147]  U. Tateishi,et al.  The acceleration of glucose accumulation in renal cell carcinoma assessed by FDG PET/CT demonstrated acquisition of resistance to tyrosine kinase inhibitor therapy , 2017, BMC Cancer.

[148]  N. Schultz,et al.  Molecular analysis of aggressive renal cell carcinoma with unclassified histology reveals distinct subsets , 2016, Nature Communications.

[149]  K. Gardner,et al.  Targeting renal cell carcinoma with a HIF-2 antagonist , 2016, Nature.

[150]  J. Backer The intricate regulation and complex functions of the Class III phosphoinositide 3-kinase Vps34. , 2016, The Biochemical journal.

[151]  H. Verheul,et al.  Novel drugs that target the metabolic reprogramming in renal cell cancer , 2016, Cancer & Metabolism.

[152]  M. Schipma,et al.  PBRM1 Regulates the Expression of Genes Involved in Metabolism and Cell Adhesion in Renal Clear Cell Carcinoma , 2016, PloS one.

[153]  H. Moch,et al.  Hypoxia, Hypoxia-inducible Transcription Factors, and Renal Cancer. , 2016, European urology.

[154]  Chris Sander,et al.  Multilevel Genomics-Based Taxonomy of Renal Cell Carcinoma. , 2016, Cell reports.

[155]  R. Piñeiro,et al.  Class II phosphoinositide 3-kinase C2β regulates a novel signaling pathway involved in breast cancer progression , 2016, Oncotarget.

[156]  D. Hsu,et al.  Cystine Deprivation Triggers Programmed Necrosis in VHL-Deficient Renal Cell Carcinomas. , 2016, Cancer research.

[157]  Alessandro Conti,et al.  Metabolic Alterations in Renal and Prostate Cancer. , 2016, Current drug metabolism.

[158]  Steven J. M. Jones,et al.  Comprehensive Molecular Characterization of Papillary Renal-Cell Carcinoma. , 2016, The New England journal of medicine.

[159]  C. Arteaga,et al.  The PI3K/AKT Pathway as a Target for Cancer Treatment. , 2016, Annual review of medicine.

[160]  J. Fish,et al.  Lanzkowsky's Manual of Pediatric Hematology and Oncology , 2016 .

[161]  Liping Han,et al.  Prognostic significance of two lipid metabolism enzymes, HADHA and ACAT2, in clear cell renal cell carcinoma , 2016, Tumor Biology.

[162]  Xuejun Jiang,et al.  Akt inhibition attenuates rasfonin-induced autophagy and apoptosis through the glycolytic pathway in renal cancer cells , 2015, Cell Death and Disease.

[163]  R. Montironi,et al.  Metabolic alterations in renal cell carcinoma. , 2015, Cancer treatment reviews.

[164]  K. Pritchard-Jones,et al.  Advances in Wilms Tumor Treatment and Biology: Progress Through International Collaboration. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[165]  C. Kieda,et al.  The Role of Hypoxia and Cancer Stem Cells in Renal Cell Carcinoma Pathogenesis , 2015, Stem Cell Reviews and Reports.

[166]  W. Sperl,et al.  Energy metabolism in neuroblastoma and Wilms tumor. , 2015, Translational pediatrics.

[167]  R. Cetin-Atalay,et al.  The PI3K/AKT/mTOR interactive pathway. , 2015, Molecular bioSystems.

[168]  P. Hawkins,et al.  PI3K signalling in inflammation. , 2015, Biochimica et biophysica acta.

[169]  V. Muglia,et al.  Renal cell carcinoma: histological classification and correlation with imaging findings* , 2015, Radiologia brasileira.

[170]  D. Ribatti,et al.  Metabolomic profile of glycolysis and the pentose phosphate pathway identifies the central role of glucose-6-phosphate dehydrogenase in clear cell-renal cell carcinoma , 2015, Oncotarget.

[171]  P. Oefner,et al.  Distinct von Hippel-Lindau gene and hypoxia-regulated alterations in gene and protein expression patterns of renal cell carcinoma and their effects on metabolism , 2015, Oncotarget.

[172]  H. Öztürk Prognostic features of renal sarcomas (Review) , 2014, Oncology letters.

[173]  W. Rathmell,et al.  Renal cell carcinoma , 2014, BMJ : British Medical Journal.

[174]  F. Kawakami,et al.  Clear cell papillary renal cell carcinoma: a review. , 2014, International journal of clinical and experimental pathology.

[175]  R. Nussbaum,et al.  Renal Cell Carcinoma in Tuberous Sclerosis Complex , 2014, The American journal of surgical pathology.

[176]  Yingying Wang,et al.  GRP78 enhances the glutamine metabolism to support cell survival from glucose deficiency by modulating the β-catenin signaling , 2014, Oncotarget.

[177]  U. Völker,et al.  Kidney protein profiling of Wilms' tumor patients by analysis of formalin-fixed paraffin-embedded tissue samples. , 2014, Clinica chimica acta; international journal of clinical chemistry.

[178]  Jiri Polivka,et al.  Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway. , 2014, Pharmacology & therapeutics.

[179]  B. Ku,et al.  Increased expression of transglutaminase 2 drives glycolytic metabolism in renal carcinoma cells , 2014, Amino Acids.

[180]  R. Sun,et al.  Hypoxic regulation of glutamine metabolism through HIF1 and SIAH2 supports lipid synthesis that is necessary for tumor growth. , 2014, Cell metabolism.

[181]  C. Rommel,et al.  PI3K and cancer: lessons, challenges and opportunities , 2014, Nature Reviews Drug Discovery.

[182]  S. Gabriel,et al.  Discovery and saturation analysis of cancer genes across 21 tumor types , 2014, Nature.

[183]  D. Mukhopadhyay,et al.  Inhibition of GSK-3 Induces Differentiation and Impaired Glucose Metabolism in Renal Cancer , 2013, Molecular Cancer Therapeutics.

[184]  C. Betz,et al.  Where is mTOR and what is it doing there? , 2013, The Journal of cell biology.

[185]  F. Marchi,et al.  Genomic Signatures Predict Poor Outcome in Undifferentiated Pleomorphic Sarcomas and Leiomyosarcomas , 2013, PloS one.

[186]  W. Linehan,et al.  Aerobic glycolysis: a novel target in kidney cancer , 2013, Expert review of anticancer therapy.

[187]  W Marston Linehan,et al.  Molecular Pathways: Fumarate Hydratase-Deficient Kidney Cancer—Targeting the Warburg Effect in Cancer , 2013, Clinical Cancer Research.

[188]  E. Hirsch,et al.  Targeting PI3K in Cancer: Any Good News? , 2013, Front. Oncol..

[189]  W. Linehan,et al.  Renal Medullary Carcinoma: Molecular, Immunohistochemistry, and Morphologic Correlation , 2013, The American journal of surgical pathology.

[190]  G. Stephanopoulos,et al.  In vivo HIF-mediated reductive carboxylation is regulated by citrate levels and sensitizes VHL-deficient cells to glutamine deprivation. , 2013, Cell metabolism.

[191]  H. Axelson,et al.  Kidney cancer. , 2013, Seminars in cancer biology.

[192]  F. Khuri,et al.  Targeting the PI3K/AKT/mTOR pathway: biomarkers of success and tribulation. , 2013, American Society of Clinical Oncology educational book. American Society of Clinical Oncology. Annual Meeting.

[193]  Jing Pan,et al.  Identification of somatic mutations in non-small cell lung carcinomas using whole-exome sequencing. , 2012, Carcinogenesis.

[194]  V. Haase,et al.  Renal cancer: oxygen meets metabolism. , 2012, Experimental cell research.

[195]  R. Stanton Glucose‐6‐phosphate dehydrogenase, NADPH, and cell survival , 2012, IUBMB life.

[196]  B. Hemmings,et al.  PI3K/AKT, MAPK and AMPK signalling: protein kinases in glucose homeostasis , 2012, Expert Reviews in Molecular Medicine.

[197]  W. Marston Linehan,et al.  Reductive carboxylation supports growth in tumor cells with defective mitochondria , 2011, Nature.

[198]  J. Montani,et al.  PI3Kγ within a nonhematopoietic cell type negatively regulates diet-induced thermogenesis and promotes obesity and insulin resistance , 2011, Proceedings of the National Academy of Sciences.

[199]  M. Rosenblum,et al.  Clear-cell papillary renal cell carcinoma: molecular and immunohistochemical analysis with emphasis on the von Hippel–Lindau gene and hypoxia-inducible factor pathway-related proteins , 2011, Modern Pathology.

[200]  E. Jacinto,et al.  mTOR complex 2 signaling and functions , 2011, Cell cycle.

[201]  V. Huff,et al.  Wilms' tumours: about tumour suppressor genes, an oncogene and a chameleon gene , 2011, Nature Reviews Cancer.

[202]  F. Kim,et al.  Molecular aspects of renal cell carcinoma: a review. , 2011, American journal of cancer research.

[203]  J. Tabernero,et al.  Targeting the PI3K/Akt/mTOR Pathway – Beyond Rapalogs , 2010, Oncotarget.

[204]  M. van Engeland,et al.  VHL and HIF signalling in renal cell carcinogenesis , 2010, The Journal of pathology.

[205]  W. Linehan,et al.  The genetic basis of kidney cancer: a metabolic disease , 2010, Nature Reviews Urology.

[206]  M. Adamo,et al.  Current Perspectives on Akt Akt-ivation and Akt-ions , 2009, Experimental biology and medicine.

[207]  D. Sabatini,et al.  mTOR signaling at a glance , 2009, Journal of Cell Science.

[208]  W. Krek,et al.  Activation of a HIF1alpha-PPARgamma axis underlies the integration of glycolytic and lipid anabolic pathways in pathologic cardiac hypertrophy. , 2009, Cell metabolism.

[209]  W. Linehan,et al.  Targeting the Met signaling pathway in renal cancer , 2009, Expert review of anticancer therapy.

[210]  N. Sebire,et al.  Paediatric renal tumours: recent developments, new entities and pathological features , 2009, Histopathology.

[211]  M. Hall,et al.  mTOR-what does it do? , 2008, Transplantation proceedings.

[212]  J. McKiernan,et al.  Epidemiology, clinical staging, and presentation of renal cell carcinoma. , 2008, The Urologic clinics of North America.

[213]  J. Frank,et al.  Absence of fumarate hydratase mutation in a family with cutaneous leiomyosarcoma and renal cancer , 2008, International journal of dermatology.

[214]  V. Huff,et al.  Wilms tumor genetics: Mutations in WT1, WTX, and CTNNB1 account for only about one‐third of tumors , 2008, Genes, chromosomes & cancer.

[215]  P. Vogt,et al.  Oncogenic signaling of class I PI3K isoforms , 2008, Oncogene.

[216]  P. Hawkins,et al.  PI3K Class IB Pathway , 2007, Science's STKE.

[217]  C. Kolbitsch,et al.  PTEN expression in renal cell carcinoma and oncocytoma and prognosis , 2007, Pathology.

[218]  C. Godinot,et al.  Mitochondria and reactive oxygen species in renal cancer. , 2007, Biochimie.

[219]  W. Linehan,et al.  Pseudohypoxic Pathways in Renal Cell Carcinoma , 2007, Clinical Cancer Research.

[220]  C. Godinot,et al.  Actuality of Warburg’s views in our understanding of renal cancer metabolism , 2007, Journal of bioenergetics and biomembranes.

[221]  David M Sabatini,et al.  Defining the role of mTOR in cancer. , 2007, Cancer cell.

[222]  W. Linehan,et al.  HIF and fumarate hydratase in renal cancer , 2007, British Journal of Cancer.

[223]  P. Hawkins,et al.  Signalling through Class I PI3Ks in mammalian cells. , 2006, Biochemical Society transactions.

[224]  N. Breslow,et al.  Treatment of anaplastic histology Wilms' tumor: results from the fifth National Wilms' Tumor Study. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[225]  D. Guertin,et al.  Rictor, a Novel Binding Partner of mTOR, Defines a Rapamycin-Insensitive and Raptor-Independent Pathway that Regulates the Cytoskeleton , 2004, Current Biology.

[226]  J. Blenis,et al.  mTOR Controls Cell Cycle Progression through Its Cell Growth Effectors S6K1 and 4E-BP1/Eukaryotic Translation Initiation Factor 4E , 2004, Molecular and Cellular Biology.

[227]  L. Cantley,et al.  Rheb fills a GAP between TSC and TOR. , 2003, Trends in biochemical sciences.

[228]  H. Tabuchi,et al.  Loss of von Hippel-Lindau protein causes cell density dependent deregulation of CyclinD1 expression through Hypoxia-inducible factor , 2003, Oncogene.

[229]  S. Emr,et al.  Retromer function in endosome-to-Golgi retrograde transport is regulated by the yeast Vps34 PtdIns 3-kinase , 2002, Journal of Cell Science.

[230]  Lewis C Cantley,et al.  The phosphoinositide 3-kinase pathway. , 2002, Science.

[231]  H. Lehr,et al.  Loss of tumor suppressor protein PTEN during renal carcinogenesis , 2002, International journal of cancer.

[232]  M. Wymann,et al.  Membrane transport in Caenorhabditis elegans: an essential role for VPS34 at the nuclear membrane , 2002, The EMBO journal.

[233]  Alfonso Bellacosa,et al.  AKT plays a central role in tumorigenesis , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[234]  T. Glasz,et al.  [Papillary renal cell carcinoma]. , 2001, Orvosi hetilap.

[235]  M. J. Fry Phosphoinositide 3-kinase signalling in breast cancer: how big a role might it play? , 2001, Breast Cancer Research.

[236]  K. Kaestner,et al.  Insulin Resistance and a Diabetes Mellitus-Like Syndrome in Mice Lacking the Protein Kinase Akt2 (PKBβ) , 2001 .

[237]  Takeshi Noda,et al.  Two Distinct Vps34 Phosphatidylinositol 3–Kinase Complexes Function in Autophagy and Carboxypeptidase Y Sorting inSaccharomyces cerevisiae , 2001, The Journal of cell biology.

[238]  John F. Timms,et al.  Cellular function of phosphoinositide 3-kinases: Implications for development, immunity, homeostasis, and cancer , 2001 .

[239]  Phillip T. Hawkins,et al.  Crystal Structure and Functional Analysis of Ras Binding to Its Effector Phosphoinositide 3-Kinase γ , 2000, Cell.

[240]  H. Chapuis,et al.  A specific function for phosphatidylinositol 3-kinase α (p85α-p110α) in cell survival and for phosphatidylinositol 3-kinase β (p85α-p110β) in de novo DNA synthesis of human colon carcinoma cells , 2000, Oncogene.

[241]  S. Emr,et al.  Phosphoinositide 3-Kinases and Their FYVE Domain-containing Effectors as Regulators of Vacuolar/Lysosomal Membrane Trafficking Pathways* , 1999, The Journal of Biological Chemistry.

[242]  Holger Moch,et al.  The Heidelberg classification of renal cell tumours , 1997, The Journal of pathology.

[243]  S. Scherer,et al.  Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas , 1997, Nature Genetics.

[244]  W. Linehan,et al.  Chromosome imbalances in papillary renal cell carcinoma and first cytogenetic data of familial cases analyzed by comparative genomic hybridization. , 1996, Cytogenetics and cell genetics.

[245]  H. Brauch,et al.  Loss of alleles of loci on the short arm of chromosome 3 in renal cell carcinoma , 1988, Nature.

[246]  J. Higginson,et al.  International Agency for Research on Cancer. , 1968, WHO chronicle.

[247]  H. Grabstald,et al.  Renal‐cell cancer , 1966, CA: a cancer journal for clinicians.

[248]  M. Karno,et al.  Renal cell carcinoma. , 1956, Bulletin. Tufts-New England Medical Center.