1
|
Ren S, Li J, Dorado J, Sierra A, González-Díaz H, Duardo A, Shen B. From molecular mechanisms of prostate cancer to translational applications: based on multi-omics fusion analysis and intelligent medicine. Health Inf Sci Syst 2024; 12:6. [PMID: 38125666 PMCID: PMC10728428 DOI: 10.1007/s13755-023-00264-5] [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: 08/24/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Prostate cancer is the most common cancer in men worldwide and has a high mortality rate. The complex and heterogeneous development of prostate cancer has become a core obstacle in the treatment of prostate cancer. Simultaneously, the issues of overtreatment in early-stage diagnosis, oligometastasis and dormant tumor recognition, as well as personalized drug utilization, are also specific concerns that require attention in the clinical management of prostate cancer. Some typical genetic mutations have been proved to be associated with prostate cancer's initiation and progression. However, single-omic studies usually are not able to explain the causal relationship between molecular alterations and clinical phenotypes. Exploration from a systems genetics perspective is also lacking in this field, that is, the impact of gene network, the environmental factors, and even lifestyle behaviors on disease progression. At the meantime, current trend emphasizes the utilization of artificial intelligence (AI) and machine learning techniques to process extensive multidimensional data, including multi-omics. These technologies unveil the potential patterns, correlations, and insights related to diseases, thereby aiding the interpretable clinical decision making and applications, namely intelligent medicine. Therefore, there is a pressing need to integrate multidimensional data for identification of molecular subtypes, prediction of cancer progression and aggressiveness, along with perosonalized treatment performing. In this review, we systematically elaborated the landscape from molecular mechanism discovery of prostate cancer to clinical translational applications. We discussed the molecular profiles and clinical manifestations of prostate cancer heterogeneity, the identification of different states of prostate cancer, as well as corresponding precision medicine practices. Taking multi-omics fusion, systems genetics, and intelligence medicine as the main perspectives, the current research results and knowledge-driven research path of prostate cancer were summarized.
Collapse
Affiliation(s)
- Shumin Ren
- Department of Urology and Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041 China
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
| | - Jiakun Li
- Department of Urology and Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Julián Dorado
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
| | - Alejandro Sierra
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
- IKERDATA S.L., ZITEK, University of Basque Country UPVEHU, Rectorate Building, 48940 Leioa, Spain
| | - Humbert González-Díaz
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
- IKERDATA S.L., ZITEK, University of Basque Country UPVEHU, Rectorate Building, 48940 Leioa, Spain
| | - Aliuska Duardo
- Department of Computer Science and Information Technology, University of A Coruña, 15071 A Coruña, Spain
- IKERDATA S.L., ZITEK, University of Basque Country UPVEHU, Rectorate Building, 48940 Leioa, Spain
| | - Bairong Shen
- Department of Urology and Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041 China
| |
Collapse
|
2
|
Parajuli KR, Jung Y, Taichman RS. Abscisic acid signaling through LANCL2 and PPARγ induces activation of p38MAPK resulting in dormancy of prostate cancer metastatic cells. Oncol Rep 2024; 51:39. [PMID: 38624012 PMCID: PMC10804438 DOI: 10.3892/or.2024.8698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/12/2023] [Indexed: 04/17/2024] Open
Abstract
Prostate cancer (PCa) is one the most common malignancies in men. The high incidence of bone metastasis years after primary therapy suggests that disseminated tumor cells must become dormant, but maintain their ability to proliferate in the bone marrow. Abscisic acid (ABA) is a stress response molecule best known for its regulation of seed germination, stomal opening, root shoot growth and other stress responses in plants. ABA is also synthesized by mammalian cells and has been linked to human disease. The aim of the present study was to examine the role of ABA in regulating tumor dormancy via signaling through lanthionine synthetase C‑like protein 2 (LANCL2) and peroxisome proliferator activated receptor γ (PPARγ) receptors. ABA signaling in human PCa cell lines was studied using targeted gene knockdown (KD), western blotting, quantitative PCR, cell proliferation, migration, invasion and soft agar assays, as well as co‑culture assays with bone marrow stromal cells. The data demonstrated that ABA signaling increased the expression of p21, p27 and p16, while inhibiting viability, migration, invasion and colony size in a reversable manner without toxicity. ABA also induced p38MAPK activation and NR2F1 signaling. Targeted gene KD of LANCL2 and PPARγ abrogated the cellular responses to ABA. Taken together, these data demonstrate that ABA may induce dormancy in PCa cell lines through LANCL2 and PPARγ signaling, and suggest novel targets to manage metastatic PCa growth.
Collapse
Affiliation(s)
- Keshab Raj Parajuli
- Department of Periodontology, University of Alabama at Birmingham School of Dentistry, Birmingham, AL 35294, USA
| | - Younghun Jung
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Russell S. Taichman
- Department of Periodontology, University of Alabama at Birmingham School of Dentistry, Birmingham, AL 35294, USA
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| |
Collapse
|
3
|
de la Calle CM, Shee K, Yang H, Lonergan PE, Nguyen HG. The endoplasmic reticulum stress response in prostate cancer. Nat Rev Urol 2022; 19:708-726. [PMID: 36168057 DOI: 10.1038/s41585-022-00649-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2022] [Indexed: 11/09/2022]
Abstract
In order to proliferate in unfavourable conditions, cancer cells can take advantage of the naturally occurring endoplasmic reticulum-associated unfolded protein response (UPR) via three highly conserved signalling arms: IRE1α, PERK and ATF6. All three arms of the UPR have key roles in every step of tumour progression: from cancer initiation to tumour growth, invasion, metastasis and resistance to therapy. At present, no cure for metastatic prostate cancer exists, as targeting the androgen receptor eventually results in treatment resistance. New research has uncovered an important role for the UPR in prostate cancer tumorigenesis and crosstalk between the UPR and androgen receptor signalling pathways. With an improved understanding of the mechanisms by which cancer cells exploit the endoplasmic reticulum stress response, targetable points of vulnerability can be uncovered.
Collapse
Affiliation(s)
- Claire M de la Calle
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Kevin Shee
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Heiko Yang
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Peter E Lonergan
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, St. James's Hospital, Dublin, Ireland
- Department of Surgery, Trinity College, Dublin, Ireland
| | - Hao G Nguyen
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
4
|
Lee S, Mendoza TR, Burner DN, Muldong MT, Wu CCN, Arreola-Villanueva C, Zuniga A, Greenburg O, Zhu WY, Murtadha J, Koutouan E, Pineda N, Pham H, Kang SG, Kim HT, Pineda G, Lennon KM, Cacalano NA, Jamieson CHM, Kane CJ, Kulidjian AA, Gaasterland T, Jamieson CAM. Novel Dormancy Mechanism of Castration Resistance in Bone Metastatic Prostate Cancer Organoids. Int J Mol Sci 2022; 23:ijms23063203. [PMID: 35328625 PMCID: PMC8952299 DOI: 10.3390/ijms23063203] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/07/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Advanced prostate cancer (PCa) patients with bone metastases are treated with androgen pathway directed therapy (APDT). However, this treatment invariably fails and the cancer becomes castration resistant. To elucidate resistance mechanisms and to provide a more predictive pre-clinical research platform reflecting tumor heterogeneity, we established organoids from a patient-derived xenograft (PDX) model of bone metastatic prostate cancer, PCSD1. APDT-resistant PDX-derived organoids (PDOs) emerged when cultured without androgen or with the anti-androgen, enzalutamide. Transcriptomics revealed up-regulation of neurogenic and steroidogenic genes and down-regulation of DNA repair, cell cycle, circadian pathways and the severe acute respiratory syndrome (SARS)-CoV-2 host viral entry factors, ACE2 and TMPRSS2. Time course analysis of the cell cycle in live cells revealed that enzalutamide induced a gradual transition into a reversible dormant state as shown here for the first time at the single cell level in the context of multi-cellular, 3D living organoids using the Fucci2BL fluorescent live cell cycle tracker system. We show here a new mechanism of castration resistance in which enzalutamide induced dormancy and novel basal-luminal-like cells in bone metastatic prostate cancer organoids. These PDX organoids can be used to develop therapies targeting dormant APDT-resistant cells and host factors required for SARS-CoV-2 viral entry.
Collapse
MESH Headings
- Androgens/pharmacology
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensin-Converting Enzyme 2/metabolism
- Animals
- Benzamides/pharmacology
- Bone Neoplasms/genetics
- Bone Neoplasms/metabolism
- Bone Neoplasms/secondary
- COVID-19/genetics
- COVID-19/metabolism
- COVID-19/virology
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Male
- Mice
- Nitriles/pharmacology
- Organoids/metabolism
- Phenylthiohydantoin/pharmacology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- SARS-CoV-2/metabolism
- SARS-CoV-2/physiology
- Serine Endopeptidases/genetics
- Serine Endopeptidases/metabolism
- Transplantation, Heterologous
- Virus Internalization
Collapse
Affiliation(s)
- Sanghee Lee
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
- Rady Children’s Hospital, San Diego, CA 92123, USA
| | - Theresa R. Mendoza
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Danielle N. Burner
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Michelle T. Muldong
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Christina C. N. Wu
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (G.P.); (K.M.L.)
| | - Catalina Arreola-Villanueva
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Abril Zuniga
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Olga Greenburg
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - William Y. Zhu
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Jamillah Murtadha
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Evodie Koutouan
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Naomi Pineda
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Hao Pham
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Sung-Gu Kang
- Department of Urology, Korea University College of Medicine, Seongbuk-Gu, Seoul 02841, Korea;
| | - Hyun Tae Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| | - Gabriel Pineda
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (G.P.); (K.M.L.)
| | - Kathleen M. Lennon
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (G.P.); (K.M.L.)
| | - Nicholas A. Cacalano
- Department of Radiation Oncology, University of California, Los Angeles, CA 90095, USA;
| | - Catriona H. M. Jamieson
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
- Department of Urology, Korea University College of Medicine, Seongbuk-Gu, Seoul 02841, Korea;
| | - Christopher J. Kane
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | | | - Terry Gaasterland
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA 92093, USA;
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Christina A. M. Jamieson
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
- Correspondence: ; Tel.: +1-858-534-2921
| |
Collapse
|
5
|
Cackowski FC, Heath EI. Prostate cancer dormancy and recurrence. Cancer Lett 2022; 524:103-108. [PMID: 34624433 PMCID: PMC8694498 DOI: 10.1016/j.canlet.2021.09.037] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 09/13/2021] [Accepted: 09/24/2021] [Indexed: 01/03/2023]
Abstract
Prostate cancer can progress rapidly after diagnosis, but can also become undetectable after curative intent radiation or surgery, only to recur years or decades later. This capacity to lie dormant and recur long after a patient was thought to be cured, is relatively unique to prostate cancer, with estrogen receptor positive breast cancer being the other common and well-studied example. Most investigators agree that the bone marrow is an important site for dormant tumor cells, given the frequency of bone metastases and that multiple studies have reported disseminated tumor cells in patients with localized disease. However, while more difficult to study, lymph nodes and the prostate bed are likely to be important reservoirs as well. Dormant tumor cells may be truly quiescent and in the G0 phase of the cell cycle, which is commonly called cellular dormancy. However, tumor growth may also be held in check through a balance of proliferation and cell death (tumor mass dormancy). For induction of cellular dormancy, prostate cancer cells respond to signals from their microenvironment, including TGF-β2, BMP-7, GAS6, and Wnt-5a, which result in signals transduced in part through p38 MAPK and pluripotency associated transcription factors including SOX2 and NANOG, which likely affect the epi-genome through histone modification. Clinical use of adjuvant radiation or androgen deprivation has been modestly successful to prevent recurrence. With the rapid pace of discovery in this field, systemic adjuvant therapy is likely to continue to improve in the future.
Collapse
Affiliation(s)
- Frank C Cackowski
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA.
| | - Elisabeth I Heath
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
6
|
Hugenschmidt H, Labori KJ, Brunborg C, Verbeke CS, Seeberg LT, Bendigtsen Schirmer C, Renolen A, Borgen E, Naume B, Wiedswang G. Cytokeratin-positive cells in the bone marrow from patients with pancreatic, periampullary malignancy and benign pancreatic disease show no prognostic information. BMC Cancer 2020; 20:1107. [PMID: 33198661 PMCID: PMC7667773 DOI: 10.1186/s12885-020-07510-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 10/07/2020] [Indexed: 12/11/2022] Open
Abstract
Background Pancreatic and periampullary carcinoma are aggressive tumours where preoperative assessment is challenging. Disseminated tumour cells (DTC) in the bone marrow (BM) are associated with impaired prognosis in a variety of epithelial cancers. In a cohort of patients with presumed resectable pancreatic and periampullary carcinoma, we evaluated the frequency and the potential prognostic impact of the preoperative presence of DTC, defined as cytokeratin-positive cells detected by immunocytochemistry (ICC). Methods Preoperative BM samples from 242 patients selected for surgical resection of presumed resectable pancreatic and periampullary carcinoma from 09/2009 to 12/2014, were analysed for presence of CK-positive cells by ICC. The median observation time was 21.5 months. Overall survival (OS) and disease-free survival (DFS) were calculated by Kaplan-Meier and Cox regression analysis. Results Successful resections of malignant tumours were performed in 179 of the cases, 30 patients resected had benign pancreatic disease based on postoperative histology, and 33 were deemed inoperable intraoperatively due to advanced disease. Overall survival for patients with resected carcinoma was 21.1 months (95% CI: 18.0–24.1), for those with benign disease OS was 101 months (95% CI: 69.4–132) and for those with advanced disease OS was 8.8 months (95% CI: 4.3–13.3). The proportion of patients with detected CK-positive cells was 6/168 (3.6%) in resected malignant cases, 2/31 (6.5%) in advanced disease and 4/29 (13.8%) in benign disease. The presence of CK-positive cells was not correlated to OS or DFS, neither in the entire cohort nor in the subgroup negative for circulating tumour cells (CTC). Conclusions The results indicate that CK-positive cells may be present in both patients with malignant and benign diseases of the pancreas. Detection of CK-positive cells was not associated with differences in prognosis for the entire cohort or any of the subgroups analysed. Trial registration clinicaltrials.gov (NCT01919151).
Collapse
Affiliation(s)
- Harald Hugenschmidt
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway. .,Department of Transplantation Surgery, Oslo University Hospital, PO.Box 4950, NO-0424, Oslo, Nydalen, Norway. .,Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway.
| | - Knut Jørgen Labori
- Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Cathrine Brunborg
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Caroline Sophie Verbeke
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Lars Thomas Seeberg
- Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway.,Department of Gastrointestinal Surgery, Vestfold Hospital Trust, Tønsberg, Norway
| | | | - Anne Renolen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Elin Borgen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Bjørn Naume
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Deparment of Oncology, Oslo University Hospital, Oslo, Norway
| | - Gro Wiedswang
- Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
7
|
Anwar A, Siddiqui R, Khan NA. Whole Organism Model to Study Molecular Mechanisms of Differentiation and Dedifferentiation. BIOLOGY 2020; 9:E79. [PMID: 32316619 PMCID: PMC7235994 DOI: 10.3390/biology9040079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/25/2022]
Abstract
Cancer recurrence has remained a significant challenge, despite advances in therapeutic approaches. In part, this is due to our incomplete understanding of the biology of cancer stem cells and the underlying molecular mechanisms. The phenomenon of differentiation and dedifferentiation (phenotypic switching) is not only unique to stem cells but it is also observed in several other organisms, as well as evolutionary-related microbes. Here, we propose the use of a primitive eukaryotic unicellular organism, Acanthamoeba castellanii, as a model to study the molecular mechanisms of cellular differentiation and dedifferentiation.
Collapse
Affiliation(s)
- Areeba Anwar
- Department of Biological Sciences, School of Science and Technology, Sunway University, Bandar Sunway 47500, Malaysia;
| | - Ruqaiyyah Siddiqui
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, University City 26666, UAE;
| | - Naveed Ahmed Khan
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, University City 26666, UAE;
| |
Collapse
|
8
|
Kim K, Marquez-Palencia M, Malladi S. Metastatic Latency, a Veiled Threat. Front Immunol 2019; 10:1836. [PMID: 31447846 PMCID: PMC6691038 DOI: 10.3389/fimmu.2019.01836] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Metastatic relapse is observed in cancer patients with no clinical evidence of disease for months to decades after initial diagnosis and treatment. Disseminated cancer cells that are capable of entering reversible cell cycle arrest are believed to be responsible for these late metastatic relapses. Dynamic interactions between the latent disseminated tumor cells and their surrounding microenvironment aid cancer cell survival and facilitate escape from immune surveillance. Here, we highlight findings from preclinical models that provide a conceptual framework to define and target the latent metastatic phase of tumor progression. The hope is by identifying patients harboring latent metastatic cells and providing therapeutic options to eliminate metastatic seeds prior to their emergence will result in long lasting cures.
Collapse
Affiliation(s)
- Kangsan Kim
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, United States.,Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, United States
| | - Mauricio Marquez-Palencia
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, United States.,Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, United States
| | - Srinivas Malladi
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, United States.,Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
9
|
Worrede A, Meucci O, Fatatis A. Limiting tumor seeding as a therapeutic approach for metastatic disease. Pharmacol Ther 2019; 199:117-128. [PMID: 30877019 PMCID: PMC6571062 DOI: 10.1016/j.pharmthera.2019.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/21/2019] [Indexed: 12/16/2022]
Abstract
Here we propose that therapeutic targeting of circulating tumor cells (CTCs), which are widely understood to be the seeds of metastasis, would represent an effective strategy towards limiting numerical expansion of secondary lesions and containing overall tumor burden in cancer patients. However, the molecular mediators of tumor seeding have not been well characterized. This is in part due to the limited number of pre-clinical in vivo approaches that appropriately interrogate the mechanisms by which cancer cells home to arresting organs. It is critical that we continue to investigate the mediators of tumor seeding as it is evident that the ability of CTCs to colonize in distant sites is what drives disease progression even after the primary tumor has been ablated by local modalities. In addition to slowing disease progression, containing metastatic spread by impeding tumor cell seeding may also provide a clinical benefit by increasing the duration of the residence of CTCs in systemic circulation thereby increasing their exposure to pharmacological agents commonly used in the treatment of patients such as chemotherapy and immunotherapies. In this review we will examine the current state of knowledge about the mechanisms of tumor cells seeding as well as explore how targeting this stage of metastatic spreading may provide therapeutic benefit to patients with advanced disease.
Collapse
Affiliation(s)
- Asurayya Worrede
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15(th) Street, Philadelphia, PA, USA
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15(th) Street, Philadelphia, PA, USA
| | - Alessandro Fatatis
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15(th) Street, Philadelphia, PA, USA; Program in Prostate Cancer, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Murray NP, Aedo S, Fuentealba C, Reyes E, Salazar A, Lopez MA, Minzer S, Orrego S, Guzman E. Subtypes of minimal residual disease, association with Gleason score, risk and time to biochemical failure in pT2 prostate cancer treated with radical prostatectomy. Ecancermedicalscience 2019; 13:934. [PMID: 31281431 PMCID: PMC6605630 DOI: 10.3332/ecancer.2019.934] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Indexed: 01/27/2023] Open
Abstract
INTRODUCTION The Gleason score is a strong prognostic factor for treatment failure in pathologically organ-confined prostate cancer (pT2) treated by radical prostatectomy (RP). However, within each Gleason score, there is clinical heterogeneity with respect to treatment outcome, even in patients with the same pathological stage and prostate-specific antigen (PSA) at diagnosis. This may be due to minimal residual disease (MRD) remaining after surgery. We hypothesise that the sub-type of MRD determines the risk of and timing of treatment failure, is a biological classification, and may explain in part clinical heterogeneity. We present a study of pT2 patients treated with RP, the subtypes of MRD for each Gleason score and clinical outcomes. PATIENTS AND METHODS Patients with Gleason ≤6 (G6) or Gleason 7 (G7) pT2 cancer participated in the study. One month after surgery, blood was taken for circulating prostate cell (CPCs); mononuclear cells were obtained by differential gel centrifugation and identified using immunocytochemistry with anti-PSA. The detection of one CPC/sample was defined as a positive test. Touch-preparations from bone-marrow biopsies were used to detect micro-metastasis using immunocytochemistry with anti-PSA. Biochemical failure was defined as a PSA >0.2 ng/mL. Patients were classified as: Group A MRD negative (CPC and micro-metastasis negative), Group B (only micro-metastasis positive) and Group C (CPC positive). Biochemical failure-free survival (BFFS) using Kaplan-Meier and time to failure using Restricted Mean Survival Time (RMST) after 10 years of follow-up were calculated for each group based on the Gleason score. RESULTS Of a cohort of 253 men, four were excluded for having Gleason 8 or 9 prostate cancer, leaving a study group of 249 men of whom 52 had G7 prostate cancer. G7 patients had a higher frequency of MRD (69% versus 36%) and worse prognosis. G6 and G7 patients negative for MRD had similar BBFS rates, 98% at 10 years, time to failure 9.9 years. Group C, G6 patients had a higher BFFS and longer time to failure compared to G7 patients (19% versus 5% and 7 versus 3 years). Group B showed similar results up to 5 years, thereafter G6 had a lower BFFS 63% versus 90%. CONCLUSIONS G7 and G6 pT2 patients have different patterns of MRD and relapse. Risk stratification using MRD sub-types may help to define the need for adjuvant therapy. This needs confirmation with large randomised long-term trials.
Collapse
Affiliation(s)
- Nigel P Murray
- Faculty of Medicine, University Finis Terrae, Pedro de Valdivia 1509, Providencia, Santiago 7501015, Chile
- Urology Service, Hospital de Carabineros, Simón Bolívar 2200, Ñuñoa, Santiago 7770199, Chile
| | - Socrates Aedo
- Faculty of Medicine, University Finis Terrae, Pedro de Valdivia 1509, Providencia, Santiago 7501015, Chile
| | - Cynthia Fuentealba
- Urology Service, Hospital de Carabineros, Simón Bolívar 2200, Ñuñoa, Santiago 7770199, Chile
| | - Eduardo Reyes
- Faculty of Medicine, University Diego Portales, Manuel Rodríguez Sur 415, Santiago 8370179, Chile
- Urology Service, Hospital DIPRECA, Vital Apoquindo 1200, Las Condes, Santiago 7601003, Chile
| | - Anibal Salazar
- Urology Service, Hospital de Carabineros, Simón Bolívar 2200, Ñuñoa, Santiago 7770199, Chile
| | - Marco Antonio Lopez
- Faculty of Medicine, University Mayor, San Pio X 2422, Providencia, Santiago 7510041, Chile
| | - Simona Minzer
- Faculty of Medicine, University Mayor, San Pio X 2422, Providencia, Santiago 7510041, Chile
| | - Shenda Orrego
- Faculty of Medicine, University Mayor, San Pio X 2422, Providencia, Santiago 7510041, Chile
| | - Eghon Guzman
- Faculty of Medicine, University Mayor, San Pio X 2422, Providencia, Santiago 7510041, Chile
| |
Collapse
|
11
|
Yang LY, Shan YM, Zhang Y, Zhou EH, Chen XP, Zhang H. Aurora kinase A induces chemotherapy resistance through revival of dormant cells in laryngeal squamous cell carcinoma. Head Neck 2019; 41:2239-2248. [PMID: 30706572 DOI: 10.1002/hed.25689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 12/12/2018] [Accepted: 01/15/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Chemotherapy resistance was an important tumor metastasis mechanism. METHODS Cell Counting Kit-8 assay and plate colony formation assay were applied to examine the proliferation of laryngeal squamous cell carcinoma (LSCC). Immunofluorescent staining and Western blotting were carried out to show the expression of related proteins. Wound healing, migration, and invasion assays were used to examine the mobility, migration, and invasion of LSCC. RESULTS Downregulated Aurora kinase A (AURKA) increased chemotherapy sensitivity and reduced the ability of mobility, migration, and invasion of Hep2 cells, while upregulated AURKA possessed opposite results. Hep2/5-Fu cells possessed dormancy-like properties and upregulated AURKA in Hep2/5-Fu cells (Hep2/5-Fu/AURKA cells) revived dormant state. Furthermore, Erk1/2 was restrained in Hep2/5-Fu cells and activated in Hep2/5-Fu/AURKA cells. Moreover, Erk1/2 accelerated the ability of mobility, migration, and invasion in Hep2/5-Fu/AURKA cells. CONCLUSION AURKA activated dormant state to induce chemotherapy resistance and promoted metastasis of LSCC through Erk1/2 pathway.
Collapse
Affiliation(s)
- Li-Yun Yang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, China
| | - Ya-Min Shan
- Department of Otolaryngology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yi Zhang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, China
| | - En-Hui Zhou
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, China
| | - Xiao-Ping Chen
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, China
| | - Hao Zhang
- Department of Otolaryngology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
12
|
Levesque C, Nelson PS. Cellular Constituents of the Prostate Stroma: Key Contributors to Prostate Cancer Progression and Therapy Resistance. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a030510. [PMID: 28490538 DOI: 10.1101/cshperspect.a030510] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Reciprocal signaling between prostate stroma and its epithelium are fundamental to organ development and homeostasis. Similarly, interactions between tumor cells and stromal constituents are central to key aspects of carcinogenesis and malignancy growth involving tumor cell invasion, dissemination, and growth in distant sites. The prostate stroma is complex with several distinct resident cell types, infiltrating nonresident cell types and an amalgam of structural matrix factors, matricellular proteins, metabolites, growth factors, and cytokines. Of importance, the stroma is dynamic with changes in composition as a cause or consequence of intrinsic and extrinsic factors. In the context of epithelial neoplasia, the prostate stroma undergoes phenotypic changes with a loss of well-differentiated smooth muscle cell population and the expansion of cancer-associated fibroblast populations. This reactive stroma further coevolves with tumor progression. Recent studies show the role of tumor microenvironment components in therapy resistance and highlight the importance of a thorough knowledge of cross talk between tumor cells and microenvironment niches to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Christine Levesque
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Peter S Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| |
Collapse
|
13
|
Gourdin T, Sonpavde G. Utility of cell-free nucleic acid and circulating tumor cell analyses in prostate cancer. Asian J Androl 2018; 20:230-237. [PMID: 29578115 PMCID: PMC5952476 DOI: 10.4103/aja.aja_1_18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer is characterized by bone metastases and difficulty of objectively measuring disease burden. In this context, cell-free circulating tumor DNA (ctDNA) and circulating tumor cell (CTC) quantitation and genomic profiling afford the ability to noninvasively and serially monitor the tumor. Recent data suggest that ctDNA and CTC quantitation are prognostic for survival. Indeed, CTC enumeration using the CellSearch® platform is validated as a prognostic factor and warrants consideration as a stratification factor in randomized trials. Changes in quantities of CTCs using CellSearch also are prognostic and may be employed to detect a signal of activity of new agents. Molecular profiling of both CTCs and ctDNA for androgen receptor (AR) variants has been associated with outcomes in the setting of novel androgen inhibitors. Serial profiling to detect the evolution of new alterations may inform drug development and help develop precision medicine. The costs of these assays and the small quantities in which they are detectable in blood are a limitation, and novel platforms are required to address this challenge. The presence of multiple platforms to assay CTCs and ctDNA also warrants the consideration of a mechanism to allow comparison of data across platforms. Further validation and the continued development and standardization of these promising modalities will facilitate their adoption in the clinic.
Collapse
Affiliation(s)
| | - Guru Sonpavde
- Dana Farber Cancer Institute, Genitourinary Oncology Section, Boston, MA 02215, USA
| |
Collapse
|
14
|
Yang LY, He CY, Chen XH, Su LP, Liu BY, Zhang H. Aurora kinase A revives dormant laryngeal squamous cell carcinoma cells via FAK/PI3K/Akt pathway activation. Oncotarget 2018; 7:48346-48359. [PMID: 27356739 PMCID: PMC5217022 DOI: 10.18632/oncotarget.10233] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 06/09/2016] [Indexed: 12/20/2022] Open
Abstract
Revival of dormant tumor cells may be an important tumor metastasis mechanism. We hypothesized that aurora kinase A (AURKA), a cell cycle control kinase, promotes the transition of laryngeal squamous cell carcinoma (LSCC) cells from G0 phase to active division. We therefore investigated whether AURKA could revive dormant tumor cells to promote metastasis. Western blotting revealed that AURKA expression was persistently low in dormant laryngeal cancer Hep2 (D-Hep2) cells and high in non-dormant (T-Hep2) cells. Decreasing AURKA expression in T-Hep2 cells induced dormancy and reduced FAK/PI3K/Akt pathway activity. Increasing AURKA expression in D-Hep2 cells increased FAK/PI3K/Akt pathway activity and enhanced cellular proliferation, migration, invasion and metastasis. In addition, FAK/PI3K/Akt pathway inhibition caused dormancy-like behavior and reduced cellular mobility, migration and invasion. We conclude that AURKA may revive dormant tumor cells via FAK/PI3K/Akt pathway activation, thereby promoting migration and invasion in laryngeal cancer. AURKA/FAK/PI3K/Akt inhibitors may thus represent potential targets for clinical LSCC treatment.
Collapse
Affiliation(s)
- Li-Yun Yang
- Department of Otolaryngology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Chang-Yu He
- Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xue-Hua Chen
- Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Li-Ping Su
- Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Bing-Ya Liu
- Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hao Zhang
- Department of Otolaryngology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
15
|
Mooney SM, Talebian V, Jolly MK, Jia D, Gromala M, Levine H, McConkey BJ. The GRHL2/ZEB Feedback Loop-A Key Axis in the Regulation of EMT in Breast Cancer. J Cell Biochem 2017; 118:2559-2570. [PMID: 28266048 DOI: 10.1002/jcb.25974] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 03/03/2017] [Indexed: 12/17/2022]
Abstract
More than 90% of cancer-related deaths are caused by metastasis. Epithelial-to-Mesenchymal Transition (EMT) causes tumor cell dissemination while the reverse process, Mesenchymal-to-Epithelial Transition (MET) allows cancer cells to grow and establish a potentially deadly metastatic lesion. Recent evidence indicates that in addition to E and M, cells can adopt a stable hybrid Epithelial/Mesenchymal (E/M) state where they can move collectively leading to clusters of Circulating Tumor Cells-the "bad actors" of metastasis. EMT is postulated to occur in all four major histological breast cancer subtypes. Here, we identify a set of genes strongly correlated with CDH1 in 877 cancer cell lines, and differentially expressed genes in cell lines overexpressing ZEB1, SNAIL, and TWIST. GRHL2 and ESRP1 appear in both these sets and also correlate with CDH1 at the protein level in 40 breast cancer specimens. Next, we find that GRHL2 and CD24 expression coincide with an epithelial character in human mammary epithelial cells. Further, we show that high GRHL2 expression is highly correlated with worse relapse-free survival in all four subtypes of breast cancer. Finally, we integrate CD24, GRHL2, and ESRP1 into a mathematical model of EMT regulation to validate the role of these players in EMT. Our data analysis and modeling results highlight the relationships among multiple crucial EMT/MET drivers including ZEB1, GRHL2, CD24, and ESRP1, particularly in basal-like breast cancers, which are most similar to triple-negative breast cancer (TNBC) and are considered the most dangerous subtype. J. Cell. Biochem. 118: 2559-2570, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Steven M Mooney
- Department of Biology, University of Waterloo, Waterloo N2L3G1, ON, Canada
| | - Vida Talebian
- Department of Biology, University of Waterloo, Waterloo N2L3G1, ON, Canada
| | - Mohit Kumar Jolly
- Center for Theoretical Biological Physics, Rice University, Houston 77005, Texas.,Department of Bioengineering, Rice University, Houston 77005, Texas
| | - Dongya Jia
- Center for Theoretical Biological Physics, Rice University, Houston 77005, Texas.,Program in Systems/Synthetic/Physical Biology, Rice University, Houston 77005, Texas
| | - Monica Gromala
- Department of Biology, University of Waterloo, Waterloo N2L3G1, ON, Canada
| | - Herbert Levine
- Center for Theoretical Biological Physics, Rice University, Houston 77005, Texas.,Program in Systems/Synthetic/Physical Biology, Rice University, Houston 77005, Texas.,Department of Physics and Astronomy, Rice University, Houston 77005, Texas
| | - Brendan J McConkey
- Department of Biology, University of Waterloo, Waterloo N2L3G1, ON, Canada
| |
Collapse
|
16
|
Mooney SM, Jolly MK, Levine H, Kulkarni P. Phenotypic plasticity in prostate cancer: role of intrinsically disordered proteins. Asian J Androl 2017; 18:704-10. [PMID: 27427552 PMCID: PMC5000791 DOI: 10.4103/1008-682x.183570] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
A striking characteristic of cancer cells is their remarkable phenotypic plasticity, which is the ability to switch states or phenotypes in response to environmental fluctuations. Phenotypic changes such as a partial or complete epithelial to mesenchymal transition (EMT) that play important roles in their survival and proliferation, and development of resistance to therapeutic treatments, are widely believed to arise due to somatic mutations in the genome. However, there is a growing concern that such a deterministic view is not entirely consistent with multiple lines of evidence, which indicate that stochasticity may also play an important role in driving phenotypic plasticity. Here, we discuss how stochasticity in protein interaction networks (PINs) may play a key role in determining phenotypic plasticity in prostate cancer (PCa). Specifically, we point out that the key players driving transitions among different phenotypes (epithelial, mesenchymal, and hybrid epithelial/mesenchymal), including ZEB1, SNAI1, OVOL1, and OVOL2, are intrinsically disordered proteins (IDPs) and discuss how plasticity at the molecular level may contribute to stochasticity in phenotypic switching by rewiring PINs. We conclude by suggesting that targeting IDPs implicated in EMT in PCa may be a new strategy to gain additional insights and develop novel treatments for this disease, which is the most common form of cancer in adult men.
Collapse
Affiliation(s)
- Steven M Mooney
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Mohit Kumar Jolly
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005; Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Herbert Levine
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005; Department of Bioengineering, Rice University, Houston, TX 77005; Department of Physics and Astronomy, Rice University, Houston, TX 77005, USA
| | - Prakash Kulkarni
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland 20850, USA
| |
Collapse
|
17
|
Chattopadhyay I, Wang J, Qin M, Gao L, Holtz R, Vessella RL, Leach RW, Gelman IH. Src promotes castration-recurrent prostate cancer through androgen receptor-dependent canonical and non-canonical transcriptional signatures. Oncotarget 2017; 8:10324-10347. [PMID: 28055971 PMCID: PMC5354662 DOI: 10.18632/oncotarget.14401] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 12/05/2016] [Indexed: 11/25/2022] Open
Abstract
Progression of prostate cancer (PC) to castration-recurrent growth (CRPC) remains dependent on sustained expression and transcriptional activity of the androgen receptor (AR). A major mechanism contributing to CRPC progression is through the direct phosphorylation and activation of AR by Src-family (SFK) and ACK1 tyrosine kinases. However, the AR-dependent transcriptional networks activated by Src during CRPC progression have not been elucidated. Here, we show that activated Src (Src527F) induces androgen-independent growth in human LNCaP cells, concomitant with its ability to induce proliferation/survival genes normally induced by dihydrotestosterone (DHT) in androgen-dependent LNCaP and VCaP cells. Src induces additional gene signatures unique to CRPC cell lines, LNCaP-C4-2 and CWR22Rv1, and to CRPC LuCaP35.1 xenografts. By comparing the Src-induced AR-cistrome and/or transcriptome in LNCaP to those in CRPC and LuCaP35.1 tumors, we identified an 11-gene Src-regulated CRPC signature consisting of AR-dependent, AR binding site (ARBS)-associated genes whose expression is altered by DHT in LNCaP[Src527F] but not in LNCaP cells. The differential expression of a subset (DPP4, BCAT1, CNTNAP4, CDH3) correlates with earlier PC metastasis onset and poorer survival, with the expression of BCAT1 required for Src-induced androgen-independent proliferation. Lastly, Src enhances AR binding to non-canonical ARBS enriched for FOXO1, TOP2B and ZNF217 binding motifs; cooperative AR/TOP2B binding to a non-canonical ARBS was both Src- and DHT-sensitive and correlated with increased levels of Src-induced phosphotyrosyl-TOP2B. These data suggest that CRPC progression is facilitated via Src-induced sensitization of AR to intracrine androgen levels, resulting in the engagement of canonical and non-canonical ARBS-dependent gene signatures.
Collapse
MESH Headings
- Androgen Antagonists/pharmacology
- Binding Sites
- Cell Line, Tumor
- Cell Proliferation
- Dihydrotestosterone/pharmacology
- Disease Progression
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Phosphorylation
- Promoter Regions, Genetic
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/enzymology
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/pathology
- Receptors, Androgen/drug effects
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Signal Transduction
- Time Factors
- Transcription, Genetic/drug effects
- Transcriptome
- Transfection
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Indranil Chattopadhyay
- Department of Life Sciences, School of Basic and Applied Science, Central University of Tamil Nadu, Thiruvarur, Tamil Nadu, India
| | - Jianmin Wang
- Department of Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Maochun Qin
- Department of Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Lingqiu Gao
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Renae Holtz
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Robert W. Leach
- Lewis-Sigler Institute for Integrative Genomics, Princeton, NJ, USA
| | - Irwin H. Gelman
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
18
|
Linde N, Fluegen G, Aguirre-Ghiso JA. The Relationship Between Dormant Cancer Cells and Their Microenvironment. Adv Cancer Res 2016; 132:45-71. [PMID: 27613129 PMCID: PMC5342905 DOI: 10.1016/bs.acr.2016.07.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The majority of cancer deaths are due to metastases that can occur years or decades after primary tumor diagnosis and treatment. Disseminated tumor cells (DTCs) surviving in a dormant state in target organs appear to explain the timing of this phenomenon. Knowledge on this process is important as it might provide a window of opportunity to prevent recurrences by eradicating dormant DTCs and/or by maintaining DTCs in a dormant state. Importantly, this research might offer markers of dormancy for early monitoring of metastatic relapse. However, our understanding of the mechanisms underlying the regulation of entry into and exit from dormancy is still limited and crippling any therapeutic opportunity. While cancer cell-intrinsic signaling pathways have been linked to dormancy regulation, it is likely that these pathways and the switch controlling reactivation from dormancy are regulated by microenvironmental cues. Here we review and discuss recent findings on how the microenvironment regulates cancer dormancy and raise new questions that may help advance the field.
Collapse
Affiliation(s)
- N Linde
- Tisch Cancer Institute, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY, United States.
| | - G Fluegen
- Tisch Cancer Institute, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY, United States.
| | - J A Aguirre-Ghiso
- Tisch Cancer Institute, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY, United States.
| |
Collapse
|
19
|
van der Toom EE, Verdone JE, Pienta KJ. Disseminated tumor cells and dormancy in prostate cancer metastasis. Curr Opin Biotechnol 2016; 40:9-15. [PMID: 26900985 DOI: 10.1016/j.copbio.2016.02.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/30/2016] [Accepted: 02/02/2016] [Indexed: 12/20/2022]
Abstract
It has been reported that disseminated tumor cells (DTCs) can be found in the majority of prostate cancer (PCa) patients, even at the time of primary treatment with no clinical evidence of metastatic disease. This suggests that these cells escaped the primary tumor early in the disease and exist in a dormant state in distant organs until they develop in some patients as overt metastases. Understanding the mechanisms by which cancer cells exit the primary tumor, survive the circulation, settle in a distant organ, and exist in a quiescent state is critical to understanding tumorigenesis, developing new prognostic assays, and designing new therapeutic modalities to prevent and treat clinical metastases.
Collapse
Affiliation(s)
- Emma E van der Toom
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA; VUmc School of Medical Sciences, VU University, Amsterdam, The Netherlands
| | - James E Verdone
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kenneth J Pienta
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|