A Compound that Inhibits Glycolysis in Prostate Cancer Controls Growth of Advanced Prostate Cancer.

Purpose Metastatic castration-resistant prostate cancer remains incurable regardless of recent therapeutic advances. Prostate cancer tumors display highly glycolytic phenotypes as the cancer progresses. Nonspecific inhibitors of glycolysis have not been utilized successfully for chemotherapy, because of their penchant to cause systemic toxicity. This study reports the preclinical activity, safety, and pharmacokinetics of a novel small molecule preclinical candidate, BKIDC 1553, with antiglycolytic activity. Experimental design We tested a large battery of prostate cancer cell lines for inhibition of cell proliferation, in vitro. Cell cycle, metabolic and enzymatic assays were used to demonstrate their mechanism of action. A human PDX model implanted in mice was studied for sensitivity to our BKIDC preclinical candidate. A battery of pharmacokinetic experiments, absorption, distribution, metabolism, and excretion experiments, and in vitro and in vivo toxicology experiments were carried out to assess readiness for clinical trials. Results We demonstrate a new class of small molecule inhibitors where anti-glycolytic activity in prostate cancer cell lines is mediated through inhibition of hexokinase 2. These compounds display selective growth inhibition across multiple prostate cancer models. We describe a lead BKIDC-1553 that demonstrates promising activity in a preclinical xenograft model of advanced prostate cancer, equivalent to that of enzalutamide. BKIDC-1553 demonstrates safety and pharmacologic properties consistent with a compound that can be taken into human studies with expectations of a good safety margin and predicted dosing for efficacy. Conclusion This work supports testing BKIDC-1553 and its derivatives in clinical trials for patients with advanced prostate cancer. Translational Relevance The lead compound, BKIDC-1553 demonstrates in vitro activity against most advanced prostate cancers and in vitro activity in a SCID human xenograft prostate cancer model. It also demonstrates pharmacokinetic and safety properties consistent with a late preclinical candidate that can be taken into clinical trials after good-laboratory-practice (GLP) confirmatory toxicology, formulation, and good manufacturing is performed and a FDA investigative new drug application is accepted. Since this compound has a unique mechanism of action, it will be a welcome addition to the chemotherapy of advanced prostate cancer, which generally becomes resistant to the available androgen-directed therapy, and should supplant rescue cytotoxic chemotherapy regimens that are very toxic and fail to deliver lasting response.

[1]  M. Lupien,et al.  Chronic hypoxia favours adoption to a castration-resistant cell state in prostate cancer , 2023, Oncogene.

[2]  B. Layden,et al.  Aiding Cancer’s “Sweet Tooth”: Role of Hexokinases in Metabolic Reprogramming , 2023, Life.

[3]  Xue Zhang,et al.  Time to hit pause on mitochondria-targeting cancer therapies , 2023, Nature Medicine.

[4]  M. Loda,et al.  Druggable Metabolic Vulnerabilities Are Exposed and Masked during Progression to Castration Resistant Prostate Cancer , 2022, Biomolecules.

[5]  M. Puhr,et al.  Metabolic changes during prostate cancer development and progression , 2022, Journal of Cancer Research and Clinical Oncology.

[6]  Yan Zhou,et al.  The development of small-molecule inhibitors targeting hexokinase 2. , 2022, Drug discovery today.

[7]  Youqing Wang,et al.  Prostate Cancer Incidence and Mortality: Global Status and Temporal Trends in 89 Countries From 2000 to 2019 , 2022, Frontiers in Public Health.

[8]  C. Dang,et al.  Targeting cancer metabolism in the era of precision oncology , 2021, Nature reviews. Drug discovery.

[9]  E. Boccardo,et al.  Metabolic Reprogramming and Cancer , 2021, Essential Aspects of Immunometabolism in Health and Disease.

[10]  P. Choyke,et al.  Metabolic reprogramming in prostate cancer , 2021, British Journal of Cancer.

[11]  K. Rahbar,et al.  Lutetium-177-PSMA-617 for Metastatic Castration-Resistant Prostate Cancer. , 2021, The New England journal of medicine.

[12]  William J. Israelsen,et al.  Pyruvate Kinase M1 Suppresses Development and Progression of Prostate Adenocarcinoma , 2021, bioRxiv.

[13]  A. Rasola,et al.  Hexokinase 2 in Cancer: A Prima Donna Playing Multiple Characters , 2021, International journal of molecular sciences.

[14]  G. Zadra,et al.  Metabolic reprogramming as an emerging mechanism of resistance to endocrine therapies in prostate cancer , 2021, Cancer drug resistance.

[15]  S. Larson,et al.  Quantification of Metastatic Prostate Cancer Whole-Body Tumor Burden with 18F-FDG PET Parameters and Associations with Overall Survival After First-Line Abiraterone or Enzalutamide: A Single-Center Retrospective Cohort Study , 2021, The Journal of Nuclear Medicine.

[16]  A. Chinnaiyan,et al.  Accelerating precision medicine in metastatic prostate cancer , 2020, Nature Cancer.

[17]  S. Plymate,et al.  Androgen Receptor Signaling and Metabolic and Cellular Plasticity During Progression to Castration Resistant Prostate Cancer , 2020, Frontiers in Oncology.

[18]  B. Faubert,et al.  Metabolic reprogramming and cancer progression , 2020, Science.

[19]  T. Guthrie,et al.  Prostate cancer. , 2020, American family physician.

[20]  D. Hardie,et al.  AMP-Activated Protein Kinase: Friend or Foe in Cancer? , 2020, Annual Review of Cancer Biology.

[21]  Qi Wang,et al.  Hexokinase 2 Depletion Confers Sensitization to Metformin and Inhibits Glycolysis in Lung Squamous Cell Carcinoma , 2020, Frontiers in Oncology.

[22]  P. Myler,et al.  Structures of glyceraldehyde 3‐phosphate dehydrogenase in Neisseria gonorrhoeae and Chlamydia trachomatis , 2020, Protein science : a publication of the Protein Society.

[23]  Xin Chen,et al.  Identifying strategies to target the metabolic flexibility of tumours , 2020, Nature Metabolism.

[24]  John F. Trant,et al.  Differential Expression of Glucose Transporters and Hexokinases in Prostate Cancer with a Neuroendocrine Gene Signature: A Mechanistic Perspective for 18F-FDG Imaging of PSMA-Suppressed Tumors , 2019, The Journal of Nuclear Medicine.

[25]  H. Herschman,et al.  A tumor agnostic therapeutic strategy for Hexokinase 1 Null/Hexokinase 2 positive cancers. , 2019, Cancer research.

[26]  A. Gori,et al.  Hexokinase 2 displacement from mitochondria‐associated membranes prompts Ca2+‐dependent death of cancer cells , 2019, bioRxiv.

[27]  M. Loda,et al.  The Role of Lineage Plasticity in Prostate Cancer Therapy Resistance , 2019, Clinical Cancer Research.

[28]  A. Tutt,et al.  A decade of clinical development of PARP inhibitors in perspective , 2019, Annals of oncology : official journal of the European Society for Medical Oncology.

[29]  Dustin J Maly,et al.  Bumped kinase inhibitor 1369 is effective against Cystoisospora suis in vivo and in vitro , 2019, International Journal for Parasitology: Drugs and Drug Resistance.

[30]  M. Loda,et al.  Inhibition of de novo lipogenesis targets androgen receptor signaling in castration-resistant prostate cancer , 2018, Proceedings of the National Academy of Sciences.

[31]  T. Graeber,et al.  A precision therapeutic strategy for hexokinase 1-null, hexokinase 2-positive cancers , 2018, Cancer & Metabolism.

[32]  J. D. de Bono,et al.  Treatment of Advanced Prostate Cancer-A Review of Current Therapies and Future Promise. , 2018, Cold Spring Harbor perspectives in medicine.

[33]  N. Hay,et al.  Selective eradication of cancer displaying hyperactive Akt by exploiting the metabolic consequences of Akt activation , 2018, eLife.

[34]  M. Antoniewicz A guide to 13C metabolic flux analysis for the cancer biologist , 2018, Experimental & Molecular Medicine.

[35]  A. Zoubeidi,et al.  Cellular plasticity and the neuroendocrine phenotype in prostate cancer , 2018, Nature Reviews Urology.

[36]  Christopher P. Long,et al.  Hexokinase-2 depletion inhibits glycolysis and induces oxidative phosphorylation in hepatocellular carcinoma and sensitizes to metformin , 2018, Nature Communications.

[37]  S. Nehmeh,et al.  Positron Emission Tomography/Computed Tomography–Based Assessments of Androgen Receptor Expression and Glycolytic Activity as a Prognostic Biomarker for Metastatic Castration-Resistant Prostate Cancer , 2017, JAMA oncology.

[38]  Michael D. Nyquist,et al.  Androgen Receptor Pathway-Independent Prostate Cancer Is Sustained through FGF Signaling. , 2017, Cancer cell.

[39]  R. Shaw,et al.  AMPK: Mechanisms of Cellular Energy Sensing and Restoration of Metabolic Balance. , 2017, Molecular cell.

[40]  Dustin J Maly,et al.  Kinobead and Single-Shot LC-MS Profiling Identifies Selective PKD Inhibitors. , 2017, Journal of proteome research.

[41]  Matthew G. Vander Heiden,et al.  Understanding the Intersections between Metabolism and Cancer Biology , 2017, Cell.

[42]  H. Jadvar Is There Use for FDG-PET in Prostate Cancer? , 2016, Seminars in nuclear medicine.

[43]  Dustin J Maly,et al.  Development of an Orally Available and Central Nervous System (CNS) Penetrant Toxoplasma gondii Calcium-Dependent Protein Kinase 1 (TgCDPK1) Inhibitor with Minimal Human Ether-a-go-go-Related Gene (hERG) Activity for the Treatment of Toxoplasmosis. , 2016, Journal of medicinal chemistry.

[44]  M. Brand,et al.  Determining Maximum Glycolytic Capacity Using Extracellular Flux Measurements , 2016, PloS one.

[45]  Birgit Kasch,et al.  Next Generation , 2005, Im OP.

[46]  Meagan E. Sullender,et al.  Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9 , 2015, Nature Biotechnology.

[47]  V. Arora,et al.  Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer , 2015, Nature Reviews Cancer.

[48]  Hua Tang,et al.  SD-208, a Novel Protein Kinase D Inhibitor, Blocks Prostate Cancer Cell Proliferation and Tumor Growth In Vivo by Inducing G2/M Cell Cycle Arrest , 2015, PloS one.

[49]  Hans Clevers,et al.  Organoid Cultures Derived from Patients with Advanced Prostate Cancer , 2014, Cell.

[50]  Li-Ju Chang,et al.  Hexokinase 2-mediated Warburg effect is required for PTEN- and p53-deficiency-driven prostate cancer growth. , 2014, Cell reports.

[51]  Neville E. Sanjana,et al.  Improved vectors and genome-wide libraries for CRISPR screening , 2014, Nature Methods.

[52]  P. Wipf,et al.  New Pyrazolopyrimidine Inhibitors of Protein Kinase D as Potent Anticancer Agents for Prostate Cancer Cells , 2013, PloS one.

[53]  Abhishek K. Jha,et al.  Hexokinase 2 is required for tumor initiation and maintenance and its systemic deletion is therapeutic in mouse models of cancer. , 2013, Cancer cell.

[54]  P. Sorger,et al.  Profiling phospho-signaling networks in breast cancer using reverse-phase protein arrays , 2013, Oncogene.

[55]  I Syndikus,et al.  Alpha emitter radium-223 and survival in metastatic prostate cancer. , 2013, The New England journal of medicine.

[56]  R. Snyder,et al.  Bacterial mutagenicity screening in the pharmaceutical industry. , 2013, Mutation research.

[57]  Gareth J Waldron,et al.  Reducing safety-related drug attrition: the use of in vitro pharmacological profiling , 2012, Nature Reviews Drug Discovery.

[58]  Adam A. Margolin,et al.  The Cancer Cell Line Encyclopedia enables predictive modeling of anticancer drug sensitivity , 2012, Nature.

[59]  W. V. Van Voorhis,et al.  Immobilized metal-affinity chromatography protein-recovery screening is predictive of crystallographic structure success , 2011, Acta crystallographica. Section F, Structural biology and crystallization communications.

[60]  D. Hanahan,et al.  Hallmarks of Cancer: The Next Generation , 2011, Cell.

[61]  Paul J. Harradine,et al.  Control and Measurement of Plasma pH in Equilibrium Dialysis: Influence on Drug Plasma Protein Binding , 2011, Drug Metabolism and Disposition.

[62]  David G. Nicholls,et al.  Quantitative Microplate-Based Respirometry with Correction for Oxygen Diffusion , 2009, Analytical chemistry.

[63]  Timothy N C Wells,et al.  Glycogen Synthase Kinase 3 Is a Potential Drug Target for African Trypanosomiasis Therapy , 2008, Antimicrobial Agents and Chemotherapy.

[64]  L. Kifle,et al.  Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. , 2006, Cell metabolism.

[65]  Huadong Tang,et al.  A MATHEMATICAL DESCRIPTION OF THE FUNCTIONALITY OF CORRECTION FACTORS USED IN ALLOMETRY FOR PREDICTING HUMAN DRUG CLEARANCE , 2005, Drug Metabolism and Disposition.

[66]  F. Studier,et al.  Protein production by auto-induction in high density shaking cultures. , 2005, Protein expression and purification.

[67]  R. Vessella,et al.  LuCaP 35: A new model of prostate cancer progression to androgen independence , 2003, The Prostate.

[68]  J. Mordenti,et al.  Man versus beast: pharmacokinetic scaling in mammals. , 1986, Journal of pharmaceutical sciences.

[69]  W. Förster,et al.  Dependence of pharmacokinetic parameters on the body weight. , 1977, International journal of clinical pharmacology and biopharmacy.

[70]  B. Landau,et al.  Certain metabolic and pharmacologic effects in cancer patients given infusions of 2-deoxy-D-glucose. , 1958, Journal of the National Cancer Institute.

[71]  F. Collins,et al.  Principles of Biochemistry , 1937, The Indian Medical Gazette.

[72]  O. Warburg über den Stoffwechsel der Carcinomzelle , 1925, Klinische Wochenschrift.

[73]  Jacob D. Jaffe,et al.  Next-generation characterization of the Cancer Cell Line Encyclopedia , 2019, Nature.

[74]  O. Meyuhas Ribosomal Protein S6 Phosphorylation: Four Decades of Research. , 2015, International review of cell and molecular biology.

[75]  Donald Wlodkowic,et al.  Apoptosis and beyond: cytometry in studies of programmed cell death. , 2011, Methods in cell biology.

[76]  Daniel Lindmark,et al.  Four decades of research , 2009 .

[77]  Å. Lernmark,et al.  Systematic screening of potential beta-cell imaging agents. , 2004, Biochemical and biophysical research communications.

[78]  Y. Le Marchand-Brustel,et al.  Assays of glucose entry, glucose transporter amount, and translocation. , 2001, Methods in molecular biology.

[79]  J. Balian,et al.  Interspecies scaling: predicting clearance of drugs in humans. Three different approaches. , 1996, Xenobiotica; the fate of foreign compounds in biological systems.

[80]  P. Rabinovitch DNA content histogram and cell-cycle analysis. , 1994, Methods in cell biology.