1
|
Habault J, Franco JL, Ha S, Schneider JA, Voisin M, Wise DR, Wong KK, Garabedian MJ, Kirshenbaum K, Logan SK. In Vivo Efficacy of a Macrocyclic Peptoid-Peptide Hybrid That Selectively Modulates the Beta-Catenin/TCF Interaction to Inhibit Prostate Cancer. Prostate 2025; 85:646-658. [PMID: 39956770 DOI: 10.1002/pros.24868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/07/2025] [Accepted: 01/29/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Prostate cancer is the most common form of male cancer and can initially be treated as a localized disease. Although the 5-year survival rate at diagnosis approaches 100 percent, a subset of patients will subsequently develop resistance to treatment. This may ultimately lead to metastatic castration resistant prostate cancer (mCRPC), for which the prognosis is much less favorable. The importance of the Wnt/β-catenin pathway in treatment-resistant prostate cancer has inspired efforts to exploit the interaction of β-catenin with its transcription binding partners as a therapeutic strategy for prostate cancer. METHODS Peptoid-peptide macrocycles are attractive design scaffolds for disrupting protein-protein interactions. In this study, we evaluate a library of these macrocycles and demonstrate their selectivity for the β-catenin/TCF (T Cell Factor) interaction. RESULTS Importantly, we show that the macrocycles do not significantly alter the binding of β-catenin to cell surface protein, E-cadherin. Our lead sequence, Macrocycle 13, (MC13) was also tolerant of modifications aimed to improve aqueous solubility while retaining activity. Herein, we demonstrate in vivo proof of principle for using peptidomimetic macrocycles to target the β-catenin/TCF interaction. Treated prostate cancer mouse xenografts show markedly diminished tumor growth and decreased levels of myc protein. MC13 also inhibits growth in an organoid model with genetic alterations frequently found in prostate cancer. Transcriptome analysis of prostate cancer cells treated with MC13 reveals downregulation of key pathways, including Wnt/β-catenin and c-myc. Furthermore, chromatin immunoprecipitation (ChIP) analysis shows reduced β-catenin at its target genes, axin2 and c-myc. CONCLUSION Our findings underscore the therapeutic potential of peptoid-peptide macrocycle inhibition of β-catenin in prostate cancer.
Collapse
Affiliation(s)
- Justine Habault
- Department of Microbiology, NYU Grossman School of Medicine, New York, New York, USA
| | | | - Susan Ha
- Department of Urology, NYU Grossman School of Medicine, New York, New York, USA
| | - Jeffry A Schneider
- Department of Urology, NYU Grossman School of Medicine, New York, New York, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, New York, USA
| | - Maud Voisin
- Department of Microbiology, NYU Grossman School of Medicine, New York, New York, USA
| | - David R Wise
- Department of Medicine, Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Kwok-Kin Wong
- Department of Medicine, Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Michael J Garabedian
- Department of Microbiology, NYU Grossman School of Medicine, New York, New York, USA
- Department of Urology, NYU Grossman School of Medicine, New York, New York, USA
| | - Kent Kirshenbaum
- Chemistry Department, New York University, New York, New York, USA
| | - Susan K Logan
- Department of Urology, NYU Grossman School of Medicine, New York, New York, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
2
|
Mishra A, Huang SB, Dubash T, Burr R, Edd JF, Wittner BS, Cunneely QE, Putaturo VR, Deshpande A, Antmen E, Gopinathan KA, Otani K, Miyazawa Y, Kwak JE, Guay SY, Kelly J, Walsh J, Nieman LT, Galler I, Chan P, Lawrence MS, Sullivan RJ, Bardia A, Micalizzi DS, Sequist LV, Lee RJ, Franses JW, Ting DT, Brunker PAR, Maheswaran S, Miyamoto DT, Haber DA, Toner M. Tumor cell-based liquid biopsy using high-throughput microfluidic enrichment of entire leukapheresis product. Nat Commun 2025; 16:32. [PMID: 39746954 PMCID: PMC11696112 DOI: 10.1038/s41467-024-55140-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 11/28/2024] [Indexed: 01/04/2025] Open
Abstract
Circulating Tumor Cells (CTCs) in blood encompass DNA, RNA, and protein biomarkers, but clinical utility is limited by their rarity. To enable tumor epitope-agnostic interrogation of large blood volumes, we developed a high-throughput microfluidic device, depleting hematopoietic cells through high-flow channels and force-amplifying magnetic lenses. Here, we apply this technology to analyze patient-derived leukapheresis products, interrogating a mean blood volume of 5.83 liters from seven patients with metastatic cancer. High CTC yields (mean 10,057 CTCs per patient; range 100 to 58,125) reveal considerable intra-patient heterogeneity. CTC size varies within patients, with 67% overlapping in diameter with WBCs. Paired single-cell DNA and RNA sequencing identifies subclonal patterns of aneuploidy and distinct signaling pathways within CTCs. In prostate cancers, a subpopulation of small aneuploid cells lacking epithelial markers is enriched for neuroendocrine signatures. Pooling of CNV-confirmed CTCs enables whole exome sequencing with high mutant allele fractions. High-throughput CTC enrichment thus enables cell-based liquid biopsy for comprehensive monitoring of cancer.
Collapse
Affiliation(s)
- Avanish Mishra
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Shih-Bo Huang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Howard Hughes Medical Institute, Bethesda, MD, 20815, USA
| | - Taronish Dubash
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Risa Burr
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Jon F Edd
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ben S Wittner
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Quinn E Cunneely
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Victor R Putaturo
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Akansha Deshpande
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ezgi Antmen
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Kaustav A Gopinathan
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Keisuke Otani
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yoshiyuki Miyazawa
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Ji Eun Kwak
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Sara Y Guay
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Justin Kelly
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - John Walsh
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Linda T Nieman
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Isabella Galler
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - PuiYee Chan
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Michael S Lawrence
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Ryan J Sullivan
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Aditya Bardia
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
- Hematology/Oncology, University of California, Los Angeles, USA
| | - Douglas S Micalizzi
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Lecia V Sequist
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Richard J Lee
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Joseph W Franses
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - David T Ting
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Patricia A R Brunker
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Shyamala Maheswaran
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - David T Miyamoto
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA.
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| | - Daniel A Haber
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA.
- Howard Hughes Medical Institute, Bethesda, MD, 20815, USA.
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA.
| | - Mehmet Toner
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
- Shriners Children's Boston, Boston, MA, 02114, USA.
| |
Collapse
|
3
|
Purcell C, Srinivasan PR, Pinho-Schwermann M, MacDonald WJ, Ding E, El-Deiry WS. Neuroendocrine prostate cancer drivers SOX2 and BRN2 confer differential responses to imipridones ONC201, ONC206, and ONC212 in prostate cancer cell lines. Am J Transl Res 2024; 16:7972-7982. [PMID: 39822524 PMCID: PMC11733388 DOI: 10.62347/nbnq6383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/10/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVES Prostate cancer (PCa) is a leading cause of cancer death in men worldwide. Approximately 30% of castrate-resistant PCa becomes refractory to therapy due to neuroendocrine differentiation (NED) that is present in <1% of de-novo tumors. First-in-class imipridone ONC201/TIC10 therapy has shown clinical activity against midline gliomas, neuroendocrine tumors, and PCa. We explored whether NED promotes sensitivity to imipridones ONC201 and ONC206 by inducibly overexpressing SOX2 and BRN2, well-known neuroendocrine drivers. METHODS Inducible SOX2 or BRN2 systems were cloned into human PCa cell lines LNCaP and DU145. Inducible cell lines were characterized based on protein expression, morphology, and migration. The sensitivity of the inducible cell lines to imipridone therapy was determined by viability, cell growth, or clonogenic assays. RESULTS Slight protection from ONC201 or ONC206 with SOX2 and BRN2 overexpression was observed in the inducible LNCaP cells but not in the DU145 cells. At 2 months, there was an apparent increase in CLpP expression in LNCaP SOX2-overexpressing cells, though this did not confer enhanced sensitivity to ONC201. DU145 SOX2-overexpressing cells had a significantly reduced ONC201 sensitivity than DU145 control cells. CONCLUSIONS The results suggest that treatment of castrate-resistant prostate cancer by imipridones may not be substantially affected by neuroendocrine differentiation as a therapy-resistance mechanism. The results support further testing of imipridones across subtypes of androgen-sensitive and castrate-resistant prostate cancer.
Collapse
Affiliation(s)
- Connor Purcell
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Praveen R Srinivasan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Maximilian Pinho-Schwermann
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - William J MacDonald
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Elizabeth Ding
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- The Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Hematology-Oncology Division, Department of Medicine, Rhode Island Hospital and Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| |
Collapse
|
4
|
Azam H, Veale C, Zitzmann K, Marcone S, Gallagher WM, Prencipe M. Identification of druggable targets from the interactome of the Androgen Receptor and Serum Response Factor pathways in prostate cancer. PLoS One 2024; 19:e0309491. [PMID: 39671399 PMCID: PMC11642960 DOI: 10.1371/journal.pone.0309491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/23/2024] [Indexed: 12/15/2024] Open
Abstract
BACKGROUND The Androgen Receptor (AR) pathway is crucial in driving the progression of prostate cancer (PCa) to an advanced state. Despite the introduction of second-generation AR antagonists, such as enzalutamide, majority of patients develop resistance. Several mechanisms of resistance have been identified, including the constitutive activation of the AR pathway, the emergence of AR spliced variants, and the influence of other signalling pathways. The Serum Response Factor (SRF) was previously identified as a possible player of resistance involved in a crosstalk with the AR signalling pathway. Elevated SRF levels in PCa patients were associated with disease progression and resistance to enzalutamide. However, the molecular mediators of the crosstalk between SRF and AR still need to be elucidated. The objective of this study was to identify common interactors of the AR/SRF crosstalk as therapeutic targets. METHODS Here we used affinity purification mass spectrometry (MS) following immunoprecipitation of SRF and AR, to identify proteins that interact with both SRF and AR. The list of common interactors was expanded using STRING. Four common interactors were functionally validated using MTT assays. RESULTS Seven common interactors were identified, including HSP70, HSP0AA1, HSP90AB1, HSAP5, PRDX1 and GAPDH. Pathway analysis revealed that the PI3k/AKT pathway was the most enriched in the AR/SRF network. Moreover, pharmacological inhibition of several proteins in this network, including HSP70, HSP90, PI3k and AKT, significantly decreased cellular viability of PCa cells. CONCLUSIONS This study identified a list of AR/SRF common interactors that represent a pipeline of druggable targets for the treatment of PCa.
Collapse
Affiliation(s)
- Haleema Azam
- Cancer Biology and Therapeutics Laboratory, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Colin Veale
- Cancer Biology and Therapeutics Laboratory, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Kim Zitzmann
- Cancer Biology and Therapeutics Laboratory, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Simone Marcone
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St. James’s Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - William M. Gallagher
- Cancer Biology and Therapeutics Laboratory, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Maria Prencipe
- Cancer Biology and Therapeutics Laboratory, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
5
|
Raith F, O’Donovan DH, Lemos C, Politz O, Haendler B. Addressing the Reciprocal Crosstalk between the AR and the PI3K/AKT/mTOR Signaling Pathways for Prostate Cancer Treatment. Int J Mol Sci 2023; 24:ijms24032289. [PMID: 36768610 PMCID: PMC9917236 DOI: 10.3390/ijms24032289] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
The reduction in androgen synthesis and the blockade of the androgen receptor (AR) function by chemical castration and AR signaling inhibitors represent the main treatment lines for the initial stages of prostate cancer. Unfortunately, resistance mechanisms ultimately develop due to alterations in the AR pathway, such as gene amplification or mutations, and also the emergence of alternative pathways that render the tumor less or, more rarely, completely independent of androgen activation. An essential oncogenic axis activated in prostate cancer is the phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, as evidenced by the frequent alterations of the negative regulator phosphatase and tensin homolog (PTEN) and by the activating mutations in PI3K subunits. Additionally, crosstalk and reciprocal feedback loops between androgen signaling and the PI3K/AKT/mTOR signaling cascade that activate pro-survival signals and play an essential role in disease recurrence and progression have been evidenced. Inhibitors addressing different players of the PI3K/AKT/mTOR pathway have been evaluated in the clinic. Only a limited benefit has been reported in prostate cancer up to now due to the associated side effects, so novel combination approaches and biomarkers predictive of patient response are urgently needed. Here, we reviewed recent data on the crosstalk between AR signaling and the PI3K/AKT/mTOR pathway, the selective inhibitors identified, and the most advanced clinical studies, with a focus on combination treatments. A deeper understanding of the complex molecular mechanisms involved in disease progression and treatment resistance is essential to further guide therapeutic approaches with improved outcomes.
Collapse
Affiliation(s)
- Fabio Raith
- Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany
| | - Daniel H. O’Donovan
- Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany
| | - Clara Lemos
- Bayer Research and Innovation Center, Bayer US LLC, 238 Main Street, Cambridge, MA 02142, USA
| | - Oliver Politz
- Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany
| | - Bernard Haendler
- Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany
- Correspondence: ; Tel.: +49-30-2215-41198
| |
Collapse
|
6
|
Mossa F, Robesti D, Sumankalai R, Corey E, Titus M, Kang Y, Zhang J, Briganti A, Montorsi F, Vellano CP, Marszaleck JR, Frigo DE, Logothetis CJ, Gujral TS, Dondossola E. Subtype and Site Specific-Induced Metabolic Vulnerabilities in Prostate Cancer. Mol Cancer Res 2023; 21:51-61. [PMID: 36112348 PMCID: PMC9812897 DOI: 10.1158/1541-7786.mcr-22-0250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/26/2022] [Accepted: 09/14/2022] [Indexed: 02/03/2023]
Abstract
Aberrant metabolic functions play a crucial role in prostate cancer progression and lethality. Currently, limited knowledge is available on subtype-specific metabolic features and their implications for treatment. We therefore investigated the metabolic determinants of the two major subtypes of castration-resistant prostate cancer [androgen receptor-expressing prostate cancer (ARPC) and aggressive variant prostate cancer (AVPC)]. Transcriptomic analyses revealed enrichment of gene sets involved in oxidative phosphorylation (OXPHOS) in ARPC tumor samples compared with AVPC. Unbiased screening of metabolic signaling pathways in patient-derived xenograft models by proteomic analyses further supported an enrichment of OXPHOS in ARPC compared with AVPC, and a skewing toward glycolysis by AVPC. In vitro, ARPC C4-2B cells depended on aerobic respiration, while AVPC PC3 cells relied more heavily on glycolysis, as further confirmed by pharmacologic interference using IACS-10759, a clinical-grade inhibitor of OXPHOS. In vivo studies confirmed IACS-10759's inhibitory effects in subcutaneous and bone-localized C4-2B tumors, and no effect in subcutaneous PC3 tumors. Unexpectedly, IACS-10759 inhibited PC3 tumor growth in bone, indicating microenvironment-induced metabolic reprogramming. These results suggest that castration-resistant ARPC and AVPC exhibit different metabolic dependencies, which can further undergo metabolic reprogramming in bone. IMPLICATIONS These vulnerabilities may be exploited with mechanistically novel treatments, such as those targeting OXPHOS alone or possibly in combination with existing therapies. In addition, our findings underscore the impact of the tumor microenvironment in reprogramming prostate cancer metabolism.
Collapse
Affiliation(s)
- Federica Mossa
- David H. Koch Center for Applied Research of Genitourinary Cancers and Genitourinary Medical Oncology Department, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030
| | - Daniele Robesti
- David H. Koch Center for Applied Research of Genitourinary Cancers and Genitourinary Medical Oncology Department, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030,Department of Urology, Urological Research Institute, Vita Salute San Raffaele University, San Raffaele Scientific Institute, Milan, Italy, 20152
| | - Ramachandran Sumankalai
- David H. Koch Center for Applied Research of Genitourinary Cancers and Genitourinary Medical Oncology Department, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, WA 98195
| | - Mark Titus
- David H. Koch Center for Applied Research of Genitourinary Cancers and Genitourinary Medical Oncology Department, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030
| | - Yuqi Kang
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109; Department of Molecular and Cellular Biology, University of Washington, Seattle, WA 98195
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030
| | - Alberto Briganti
- Department of Urology, Urological Research Institute, Vita Salute San Raffaele University, San Raffaele Scientific Institute, Milan, Italy, 20152
| | - Francesco Montorsi
- Department of Urology, Urological Research Institute, Vita Salute San Raffaele University, San Raffaele Scientific Institute, Milan, Italy, 20152
| | - Christopher P. Vellano
- TRACTION Platform, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030
| | - Joseph R. Marszaleck
- TRACTION Platform, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030
| | - Daniel E. Frigo
- David H. Koch Center for Applied Research of Genitourinary Cancers and Genitourinary Medical Oncology Department, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030,Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030
| | - Christopher J Logothetis
- David H. Koch Center for Applied Research of Genitourinary Cancers and Genitourinary Medical Oncology Department, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030
| | - Taranjit S. Gujral
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109; Department of Molecular and Cellular Biology, University of Washington, Seattle, WA 98195
| | - Eleonora Dondossola
- David H. Koch Center for Applied Research of Genitourinary Cancers and Genitourinary Medical Oncology Department, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030,Corresponding author: Eleonora Dondossola, 1515 Holcombe Blvd, Houston, Texas, 77030, +17137459200
| |
Collapse
|
7
|
Comparative Proteomic and Transcriptomic Analysis of the Impact of Androgen Stimulation and Darolutamide Inhibition. Cancers (Basel) 2022; 15:cancers15010002. [PMID: 36611998 PMCID: PMC9817687 DOI: 10.3390/cancers15010002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/22/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Several inhibitors of androgen receptor (AR) function are approved for prostate cancer treatment, and their impact on gene transcription has been described. However, the ensuing effects at the protein level are far less well understood. We focused on the AR signaling inhibitor darolutamide and confirmed its strong AR binding and antagonistic activity using the high throughput cellular thermal shift assay (CETSA HT). Then, we generated comprehensive, quantitative proteomic data from the androgen-sensitive prostate cancer cell line VCaP and compared them to transcriptomic data. Following treatment with the synthetic androgen R1881 and darolutamide, global mass spectrometry-based proteomics and label-free quantification were performed. We found a generally good agreement between proteomic and transcriptomic data upon androgen stimulation and darolutamide inhibition. Similar effects were found both for the detected expressed genes and their protein products as well as for the corresponding biological programs. However, in a few instances there was a discrepancy in the magnitude of changes induced on gene expression levels compared to the corresponding protein levels, indicating post-transcriptional regulation of protein abundance. Chromatin immunoprecipitation DNA sequencing (ChIP-seq) and Hi-C chromatin immunoprecipitation (HiChIP) revealed the presence of androgen-activated AR-binding regions and long-distance AR-mediated loops at these genes.
Collapse
|