1
|
Pantoja-Romero W, Lavín-Flores A, Morell G, Martínez M, Weiner BR, Coronas J. Solvent-Free Process for Preparing Metal-Organic Framework Composites Based on Carbon-Based Quantum Dots and Their Derivatives as Drug Delivery Systems for Andrographolide. Chemistry 2025:e202500655. [PMID: 40326208 DOI: 10.1002/chem.202500655] [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: 02/19/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/07/2025]
Abstract
Andrographolide (ADG) was conjugated with MIL-53(Al), ZIF-8, carbon-based quantum dots (CBQDs) and doped carbon-based quantum dots (D-CBQDs) using high-pressure (0.3 GPa) contact. This solvent-free approach is environmentally friendly, energy-efficient, and time-saving, resulting in ADG-MOFs-CBQDs/D-CBQDs with physical properties comparable to those produced by traditional liquid phase encapsulation. The resulting nanocomposites were characterized using SEM, XRD, TGA, FT-IR, and 1H NMR. The results indicate that ADG was partly encapsulated within the metal-organic framework (MOF) pores, while another portion was bound externally to CBQD/D-CBQDs, as evidenced by distinct drug signals in each analysis. This green synthesis approach simplifies the conjugation process. It significantly enhances the drug solubility, as indicated by changes in hydrophobicity observed via 1H NMR, surpassing the improvements achieved by ADG encapsulation within MOFs alone. Additionally, in preliminary tests, the materials exhibited significant cytotoxicity against PC3 cells compared to ADG (25.05 ± 0.06 µg/mL) after 48 hours of exposure.
Collapse
Affiliation(s)
- Wenndy Pantoja-Romero
- Department of Chemistry, University of Puerto Rico at Rio Piedras Campus, 601 Av. Universidad, San Juan, San Juan, Puerto Rico
- Molecular Sciences Research Center, University of Puerto Rico at Rio Piedras Campus, 1390 C. Juan Ponce de León, San Juan, 00926, Puerto Rico
| | - Alexis Lavín-Flores
- Department of Chemistry, University of Puerto Rico at Rio Piedras Campus, 601 Av. Universidad, San Juan, San Juan, Puerto Rico
- Molecular Sciences Research Center, University of Puerto Rico at Rio Piedras Campus, 1390 C. Juan Ponce de León, San Juan, 00926, Puerto Rico
| | - Gerardo Morell
- Molecular Sciences Research Center, University of Puerto Rico at Rio Piedras Campus, 1390 C. Juan Ponce de León, San Juan, 00926, Puerto Rico
- Department of Physics, University of Puerto Rico at Rio Piedras Campus, 601 Av. Universidad, San Juan, San Juan, Puerto Rico
| | - Magaly Martínez
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, 00936-3027, Puerto Rico
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico, San Juan, 00925-253, Puerto Rico
| | - Brad R Weiner
- Department of Chemistry, University of Puerto Rico at Rio Piedras Campus, 601 Av. Universidad, San Juan, San Juan, Puerto Rico
- Molecular Sciences Research Center, University of Puerto Rico at Rio Piedras Campus, 1390 C. Juan Ponce de León, San Juan, 00926, Puerto Rico
| | - Joaquín Coronas
- Instituto de Nanociencia y Materiales de Aragón (INMA), Universidad de Zaragoza-CSIC, Zaragoza, 50018, Spain
- Chemical and Environmental Engineering Department, Universidad de Zaragoza, Zaragoza, 50018, Spain
| |
Collapse
|
2
|
Kitahama K, Shigematsu Y, Sugawara E, Amori M, Amori G, Saito R, Ohmoto A, Yonese J, Takeuchi K, Inamura K. Clinicopathological characteristics of transcription factor-defined subtypes in bladder small cell carcinoma. BMC Cancer 2025; 25:766. [PMID: 40275187 PMCID: PMC12020276 DOI: 10.1186/s12885-025-14157-1] [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: 01/31/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Small cell carcinoma (SmCC) of the bladder is a rare and aggressive malignancy. Characterizing transcription factor (TF)-defined subtypes may provide insights into its biology and inform targeted therapies. This study investigates lineage-specific TF expression in bladder SmCC, its association with clinicopathological features, and comparisons with prostate SmCC. METHODS A retrospective analysis was conducted on 9 cases of bladder SmCC and 6 cases of prostate SmCC diagnosed at a single cancer hospital in Japan. Immunohistochemistry was performed for lineage-specific TFs (ASCL1, NEUROD1, POU2F3, and YAP1) and neuroendocrine and other markers. Statistical comparisons were made using Fisher's exact test and independent samples t-tests. RESULTS Combined SmCC morphology, including urothelial carcinoma (UC) (5 cases) and adenocarcinoma (2 cases), was more frequent in bladder SmCC than in prostate SmCC (78% [7 of 9 cases] vs. 17% [1 of 6 cases], p = 0.041). NEUROD1 was more frequently expressed in bladder SmCC than in prostate SmCC (67% [6 of 9 cases] vs. 0% [0 of 6 cases]; p = 0.028). NEUROD1 expression was more frequent in combined SmCC and UC bladder tumors than in other bladder SmCC tumors (100% [5 of 5 cases] vs. 25% [1 of 4 cases], p = 0.048). Conversely, HNF4A expression was absent in all combined SmCC and UC bladder tumors (0 of 5) but present in 75% (3 of 4) of other bladder SmCC tumors (p = 0.048). In 2 cases of bladder SmCC, NEUROD1 and POU2F3 were expressed in a mutually exclusive manner, with neuroendocrine markers expressed only in the NEUROD1-expressing component. CONCLUSIONS NEUROD1 is characteristically expressed in bladder SmCC, especially in SmCC combined with UC, suggesting a distinct phenotype from prostate SmCC. These findings highlight the potential for TF-based classification to improve diagnostic accuracy and inform therapeutic strategies.
Collapse
Affiliation(s)
- Keiichiro Kitahama
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Pathology, Kyorin University School of Medicine, Tokyo, Japan
| | - Yasuyuki Shigematsu
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Emiko Sugawara
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mahmut Amori
- Division of Tumor Pathology, Jichi Medical University, Shimotsuke, Japan
| | - Gulanbar Amori
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Division of Tumor Pathology, Jichi Medical University, Shimotsuke, Japan
| | - Rumiko Saito
- Department of Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Clinical Chemotherapy, The Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
- Graduate School of Engineering, Chiba Institute of Technology, Chiba, Japan
| | - Akihiro Ohmoto
- Department of Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Junji Yonese
- Department of Genitourinary Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kengo Takeuchi
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kentaro Inamura
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
- Division of Tumor Pathology, Jichi Medical University, Shimotsuke, Japan.
| |
Collapse
|
3
|
Haffner MC, Morris MJ, Ding CKC, Sayar E, Mehra R, Robinson B, True LD, Gleave M, Lotan TL, Aggarwal R, Huang J, Loda M, Nelson PS, Rubin MA, Beltran H. Framework for the Pathology Workup of Metastatic Castration-Resistant Prostate Cancer Biopsies. Clin Cancer Res 2025; 31:466-478. [PMID: 39589343 PMCID: PMC11790385 DOI: 10.1158/1078-0432.ccr-24-2061] [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: 07/01/2024] [Revised: 09/18/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Lineage plasticity and histologic transformation from prostate adenocarcinoma to neuroendocrine (NE) prostate cancer (NEPC) occur in up to 15% to 20% of patients with castration-resistant prostate cancer (CRPC) as a mechanism of treatment resistance and are associated with aggressive disease and poor prognosis. NEPC tumors typically display small cell carcinoma morphology with loss of androgen receptor (AR) expression and gain of NE lineage markers. However, there is a spectrum of phenotypes that are observed during the lineage plasticity process, and the clinical significance of mixed histologies or those that co-express AR and NE markers or lack all markers is not well defined. Translational research studies investigating NEPC have used variable definitions, making clinical trial design challenging. In this manuscript, we discuss the diagnostic workup of metastatic biopsies to help guide the reproducible classification of phenotypic CRPC subtypes. We recommend classifying CRPC tumors based on histomorphology (adenocarcinoma, small cell carcinoma, poorly differentiated carcinoma, other morphologic variant, or mixed morphology) and IHC markers with a priority for AR, NK3 homeobox 1, insulinoma-associated protein 1, synaptophysin, and cell proliferation based on Ki-67 positivity, with additional markers to be considered based on the clinical context. Ultimately, a unified workup of metastatic CRPC biopsies can improve clinical trial design and eventually practice.
Collapse
Affiliation(s)
- Michael C. Haffner
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Michael J. Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chien-Kuang C. Ding
- Department of Anatomic Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Erolcan Sayar
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
- Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
| | - Brian Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lawrence D. True
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Martin Gleave
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Tamara L. Lotan
- Departments of Pathology, Urology, Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rahul Aggarwal
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Jiaoti Huang
- Department of Pathology and Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Peter S. Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mark A. Rubin
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern and Inselspital, Bern, Switzerland
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
4
|
Furlano K, Keshavarzian T, Biernath N, Fendler A, de Santis M, Weischenfeldt J, Lupien M. Epigenomics-guided precision oncology: Chromatin variants in prostate tumor evolution. Int J Cancer 2025. [PMID: 39853587 DOI: 10.1002/ijc.35327] [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: 09/09/2024] [Revised: 12/17/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025]
Abstract
Prostate cancer is a common malignancy that in 5%-30% leads to treatment-resistant and highly aggressive disease. Metastasis-potential and treatment-resistance is thought to rely on increased plasticity of the cancer cells-a mechanism whereby cancer cells alter their identity to adapt to changing environments or therapeutic pressures to create cellular heterogeneity. To understand the molecular basis of this plasticity, genomic studies have uncovered genetic variants to capture clonal heterogeneity of primary tumors and metastases. As cellular plasticity is largely driven by non-genetic events, complementary studies in cancer epigenomics are now being conducted to identify chromatin variants. These variants, defined as genomic loci in cancer cells that show changes in chromatin state due to the loss or gain of epigenomic marks, inclusive of histone post-translational modifications, DNA methylation and histone variants, are considered the fundamental units of epigenomic heterogeneity. In prostate cancer chromatin variants hold the promise of guiding the new era of precision oncology. In this review, we explore the role of epigenomic heterogeneity in prostate cancer, focusing on how chromatin variants contribute to tumor evolution and therapy resistance. We therefore discuss their impact on cellular plasticity and stochastic events, highlighting the value of single-cell sequencing and liquid biopsy epigenomic assays to uncover new therapeutic targets and biomarkers. Ultimately, this review aims to support a new era of precision oncology, utilizing insights from epigenomics to improve prostate cancer patient outcomes.
Collapse
Affiliation(s)
- Kira Furlano
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Tina Keshavarzian
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Nadine Biernath
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Annika Fendler
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Maria de Santis
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Joachim Weischenfeldt
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
- Biotech Research & Innovation Centre (BRIC), The Finsen Laboratory, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Ueki H, Jimbo N, Terakawa T, Hara T, Tobe T, Hirata J, Wakita N, Okamura Y, Suzuki K, Bando Y, Chiba K, Teishima J, Nakano Y, Miyake H. Evaluating RB1 and p53 as diagnostic markers in treatment-related neuroendocrine prostate cancer through immunohistochemistry and genomic analysis of RB1 and TP53. Prostate 2024; 84:1506-1514. [PMID: 39279246 DOI: 10.1002/pros.24791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND The diagnosis of treatment-related neuroendocrine prostate cancer (t-NEPC) often involves a pathological assessment and immunohistochemistry (IHC) for neuroendocrine markers. Genomic alterations in RB1 and TP53 are frequently observed in NEPC and are believed to play a crucial role in the transformation of adenocarcinoma to NEPC. In this study, we examined the clinicopathologic, immunohistochemical, and genetic features of patients with t-NEPC to better understand their prognosis and diagnostic utility. METHODS This retrospective study reviewed the records of patients diagnosed with t-NEPC at Kobe University Hospital between October 2018 and December 2022. Clinical data, including age, serum neuroendocrine marker levels, and treatment history, were collected. IHC was performed for conventional neuroendocrine markers (synaptophysin, chromogranin A, and CD56) and RB1 and p53 expression. Next-generation sequencing (NGS) was conducted using FoundationOne® CDx to identify mutations in RB1 and TP53. RESULTS This study included 20 patients with t-NEPC. The median time from ADT initiation to development was 42.8 months. IHC revealed RB1 loss in 75% of cases and p53 abnormalities in 75% of cases. NGS identified RB1 mutations in 55% and TP53 mutations in 75% of cases. The concordance between NGS and IHC results was high, with 70% (14/20) agreement for RB1/RB1 and 80% (16/20) for p53/TP53. The immunostaining and genomic analysis of RB1/RB1 and p53/TP53 showed abnormal findings for the four negative cases for conventional neuroendocrine markers. CONCLUSIONS This study indicated high concordance between IHC and NGS findings for RB1/RB1 and p53/TP53 in t-NEPC. We provide a comprehensive benchmark of NGS performance compared with IHC, and these findings may help increase the diagnostic sensitivity of t-NEPC.
Collapse
Affiliation(s)
- Hideto Ueki
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoe Jimbo
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoaki Terakawa
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takuto Hara
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Taisuke Tobe
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Junichiro Hirata
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoto Wakita
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuyoshi Okamura
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kotaro Suzuki
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yukari Bando
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koji Chiba
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Jun Teishima
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuzo Nakano
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hideaki Miyake
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
6
|
Farinea G, Calabrese M, Carfì F, Saporita I, Poletto S, Delcuratolo MD, Turco F, Audisio M, Di Stefano FR, Tucci M, Buttigliero C. Impact of Neuroendocrine Differentiation (NED) on Enzalutamide and Abiraterone Efficacy in Metastatic Castration-Resistant Prostate Cancer (mCRPC): A Retrospective Analysis. Cells 2024; 13:1396. [PMID: 39195285 PMCID: PMC11352349 DOI: 10.3390/cells13161396] [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: 07/08/2024] [Revised: 08/01/2024] [Accepted: 08/10/2024] [Indexed: 08/29/2024] Open
Abstract
Neuroendocrine differentiation (NED) represents a possible androgen receptor pathway inhibitors (ARPI) resistance mechanism in metastatic castration resistance prostate cancer (mCRPC). As mCRPC with NED has been excluded from clinical trials evaluating ARPI efficacy, this study investigates the prognostic impact of NED in mCRPC patients treated with ARPIs. Methods: We retrospectively analyzed 327 mCRPC patient data treated with Enzalutamide or Abiraterone in the first and second or successive lines of treatment. NED was assessed using prostate biopsy samples through immunohistochemical staining. Results: NED was confirmed in 32/327 (9.8%) mCRPC patients. In the overall population, mCRPC with NED showed worse PFS (4.38 vs. 11.48 months HR 2.505 [1.71-3.68] p < 0.05), disease control rate (DCR), and PSA response. In the first line setting, mCRPC with NED demonstrated worse PFS (8.5 vs. 14.9 months HR 2.13 [1.18-3.88], p < 0.05). Similarly, in the second or successive lines, mCRPC with NED showed worse PFS (4.0 vs. 7.5 months HR 2.43 [1.45-4.05] p < 0.05), DCR, PSA response and OS (12.53 vs. 18.03 months HR 1.86 [1.12-3.10] p < 0.05). The adverse impact of NED on PFS was consistence across all subgroups; we also noted a trend of worse PFS in patients with high vs. low NED. Conclusions: In our study, mCRPC with NED treated with Enzalutamide or Abiraterone showed worse clinical outcomes. NED assessment should be considered to optimize treatment decisions in the mCRPC setting.
Collapse
Affiliation(s)
- Giovanni Farinea
- Department of Oncology, San Luigi Gonzaga University Hospital, University of Turin, 10043 Orbassano, Italy; (M.C.); (F.C.); (I.S.); (S.P.); (M.D.D.); (M.A.); (F.R.D.S.); (C.B.)
| | - Mariangela Calabrese
- Department of Oncology, San Luigi Gonzaga University Hospital, University of Turin, 10043 Orbassano, Italy; (M.C.); (F.C.); (I.S.); (S.P.); (M.D.D.); (M.A.); (F.R.D.S.); (C.B.)
| | - Federica Carfì
- Department of Oncology, San Luigi Gonzaga University Hospital, University of Turin, 10043 Orbassano, Italy; (M.C.); (F.C.); (I.S.); (S.P.); (M.D.D.); (M.A.); (F.R.D.S.); (C.B.)
| | - Isabella Saporita
- Department of Oncology, San Luigi Gonzaga University Hospital, University of Turin, 10043 Orbassano, Italy; (M.C.); (F.C.); (I.S.); (S.P.); (M.D.D.); (M.A.); (F.R.D.S.); (C.B.)
| | - Stefano Poletto
- Department of Oncology, San Luigi Gonzaga University Hospital, University of Turin, 10043 Orbassano, Italy; (M.C.); (F.C.); (I.S.); (S.P.); (M.D.D.); (M.A.); (F.R.D.S.); (C.B.)
| | - Marco Donatello Delcuratolo
- Department of Oncology, San Luigi Gonzaga University Hospital, University of Turin, 10043 Orbassano, Italy; (M.C.); (F.C.); (I.S.); (S.P.); (M.D.D.); (M.A.); (F.R.D.S.); (C.B.)
| | - Fabio Turco
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland;
| | - Marco Audisio
- Department of Oncology, San Luigi Gonzaga University Hospital, University of Turin, 10043 Orbassano, Italy; (M.C.); (F.C.); (I.S.); (S.P.); (M.D.D.); (M.A.); (F.R.D.S.); (C.B.)
| | - Francesco Rosario Di Stefano
- Department of Oncology, San Luigi Gonzaga University Hospital, University of Turin, 10043 Orbassano, Italy; (M.C.); (F.C.); (I.S.); (S.P.); (M.D.D.); (M.A.); (F.R.D.S.); (C.B.)
| | - Marcello Tucci
- Department of Medical Oncology, Cardinal Massaia Hospital, 14100 Asti, Italy;
| | - Consuelo Buttigliero
- Department of Oncology, San Luigi Gonzaga University Hospital, University of Turin, 10043 Orbassano, Italy; (M.C.); (F.C.); (I.S.); (S.P.); (M.D.D.); (M.A.); (F.R.D.S.); (C.B.)
| |
Collapse
|
7
|
Xu P, Yang JC, Chen B, Ning S, Zhang X, Wang L, Nip C, Shen Y, Johnson OT, Grigorean G, Phinney B, Liu L, Wei Q, Corey E, Tepper CG, Chen HW, Evans CP, Dall'Era MA, Gao AC, Gestwicki JE, Liu C. Proteostasis perturbation of N-Myc leveraging HSP70 mediated protein turnover improves treatment of neuroendocrine prostate cancer. Nat Commun 2024; 15:6626. [PMID: 39103353 PMCID: PMC11300456 DOI: 10.1038/s41467-024-50459-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: 11/30/2023] [Accepted: 07/11/2024] [Indexed: 08/07/2024] Open
Abstract
N-Myc is a key driver of neuroblastoma and neuroendocrine prostate cancer (NEPC). One potential way to circumvent the challenge of undruggable N-Myc is to target the protein homeostasis (proteostasis) system that maintains N-Myc levels. Here, we identify heat shock protein 70 (HSP70) as a top partner of N-Myc, which binds a conserved "SELILKR" motif and prevents the access of E3 ubiquitin ligase, STIP1 homology and U-box containing protein 1 (STUB1), possibly through steric hindrance. When HSP70's dwell time on N-Myc is increased by treatment with the HSP70 allosteric inhibitor, STUB1 is in close proximity with N-Myc and becomes functional to promote N-Myc ubiquitination on the K416 and K419 sites and forms polyubiquitination chains linked by the K11 and K63 sites. Notably, HSP70 inhibition significantly suppressed NEPC tumor growth, increased the efficacy of aurora kinase A (AURKA) inhibitors, and limited the expression of neuroendocrine-related pathways.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Joy C Yang
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Bo Chen
- Department of Urologic Surgery, University of California, Davis, CA, USA
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Shu Ning
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Xiong Zhang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA
| | - Leyi Wang
- Department of Urologic Surgery, University of California, Davis, CA, USA
- Graduate Group in Integrative Pathobiology, University of California, Davis, CA, USA
| | - Christopher Nip
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Yuqiu Shen
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Oleta T Johnson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | | | - Brett Phinney
- Proteomics Core Facility, University of California, Davis, CA, USA
| | - Liangren Liu
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Eva Corey
- Department of Urology, University of Washington, Washington, WA, USA
| | - Clifford G Tepper
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Christopher P Evans
- Department of Urologic Surgery, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Marc A Dall'Era
- Department of Urologic Surgery, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Allen C Gao
- Department of Urologic Surgery, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Chengfei Liu
- Department of Urologic Surgery, University of California, Davis, CA, USA.
- Graduate Group in Integrative Pathobiology, University of California, Davis, CA, USA.
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA.
| |
Collapse
|
8
|
Xiao M, Tong W, Xiao X, Pu X, Yi F. Systemic metastases in large cell neuroendocrine prostate cancer: a rare case report and literature review. Front Oncol 2024; 14:1398673. [PMID: 38812779 PMCID: PMC11133593 DOI: 10.3389/fonc.2024.1398673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/02/2024] [Indexed: 05/31/2024] Open
Abstract
Neuroendocrine prostate neoplasms, encompassing small cell carcinoma, carcinoid, and large cell carcinoma, are infrequently observed in malignant prostate tumors. The occurrence of large cell neuroendocrine prostate cancer (LCNEPC) is exceedingly rare. In this study, the patient initially presented with a persistent dysuria for a duration of one year, accompanied by a serum prostate-specific antigen (PSA) level of 17.83ng/mL. Prostate magnetic resonance imaging (MRI) and chest computed tomography (CT) scan showed that a neoplastic lesion was considered, and prostate biopsy confirmed prostate adenocarcinoma with a Gleason score of 7 (4 + 3). Then, thoracoscopic lung tumor resection was performed, and the pathological examination revealed the presence of primary moderately differentiated invasive adenocarcinoma of the lung and metastatic prostate adenocarcinoma, the Gleason score was 8 (4 + 4). After 1 year of endocrine therapy with goserelin acetate and bicalutamide, he underwent a laparoscopic radical prostatectomy (LRP), the pathological report indicated the presence of adenocarcinoma mixed with NE carcinoma. Two months after the LRP, the patient experienced gross hematuria and sacral tail pain. Further examination revealed multiple metastatic lesions throughout the body. He also underwent transurethral resection of bladder tumor (TURBT) for bladder tumor and received etoposide+ cisplatin chemotherapy three weeks post-surgery. The patient eventually died of multi-organ failure due to myelosuppression after chemotherapy. This case report presents an uncommon instance of LCNEPC with widespread systemic metastases, while also providing a comprehensive review of existing literature to facilitate improved management and treatment strategies for similar patients in subsequent cases.
Collapse
Affiliation(s)
- Maolin Xiao
- Department of Urology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | | | | | | | - Faxian Yi
- Department of Urology, Chongqing General Hospital, Chongqing University, Chongqing, China
| |
Collapse
|
9
|
Di Y, Song J, Song Z, Wang Y, Meng L. Prognostic nomogram to predict cancer-specific survival with small-cell carcinoma of the prostate: a multi-institutional study. Front Oncol 2024; 14:1349888. [PMID: 38800400 PMCID: PMC11116562 DOI: 10.3389/fonc.2024.1349888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
Objective The aim of this study is to examine the predictive factors for cancer-specific survival (CSS) in patients diagnosed with Small-Cell Carcinoma of the Prostate (SCCP) and to construct a prognostic model. Methods Cases were selected using the Surveillance, Epidemiology, and End Results (SEER) database. The Kaplan-Meier method was utilized to calculate survival rates, while Lasso and Cox regression were employed to analyze prognostic factors. An independent prognostic factor-based nomogram was created to forecast CSS at 12 and 24 months. The model's predictive efficacy was assessed using the consistency index (C-index), calibration curve, and decision curve analysis (DCA) in separate tests. Results Following the analysis of Cox and Lasso regression, age, race, Summary stage, and chemotherapy were determined to be significant risk factors (P < 0.05). In the group of participants who received training, the rate of 12-month CSS was 44.6%, the rate of 24-month CSS was 25.5%, and the median time for CSS was 10.5 months. The C-index for the training cohort was 0.7688 ± 0.024. As for the validation cohort, it was 0.661 ± 0.041. According to the nomogram, CSS was accurately predicted and demonstrated consistent and satisfactory predictive performance at both 12 months (87.3% compared to 71.2%) and 24 months (80.4% compared to 71.7%). As shown in the external validation calibration plot, the AUC for 12- and 24-month is 64.6% vs. 56.9% and 87.0% vs. 70.7%, respectively. Based on the calibration plot of the CSS nomogram at both the 12-month and 24-month marks, it can be observed that both the actual values and the nomogram predictions indicate a predominantly stable CSS. When compared to the AJCC staging system, DCA demonstrated a higher level of accuracy in predicting CSS through the use of a nomogram. Conclusion Clinical prognostic factors can be utilized with nomograms to forecast CSS in Small-Cell Carcinoma of the Prostate (SCCP).
Collapse
Affiliation(s)
- Yupeng Di
- Department of Radiotherapy, Air Force Medical Center, PLA, Beijing, China
| | - Jiazhao Song
- Department of Radiotherapy, Air Force Medical Center, PLA, Beijing, China
| | - Zhuo Song
- Department of Radiotherapy, Air Force Medical Center, PLA, Beijing, China
| | - Yingjie Wang
- Department of Radiotherapy, Air Force Medical Center, PLA, Beijing, China
| | - Lingling Meng
- Department of Radiation Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
10
|
Turnham DJ, Mullen MS, Bullock NP, Gilroy KL, Richards AE, Patel R, Quintela M, Meniel VS, Seaton G, Kynaston H, Clarkson RWE, Phesse TJ, Nelson PS, Haffner MC, Staffurth JN, Pearson HB. Development and Characterisation of a New Patient-Derived Xenograft Model of AR-Negative Metastatic Castration-Resistant Prostate Cancer. Cells 2024; 13:673. [PMID: 38667288 PMCID: PMC11049137 DOI: 10.3390/cells13080673] [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: 12/22/2023] [Revised: 03/26/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
As the treatment landscape for prostate cancer gradually evolves, the frequency of treatment-induced neuroendocrine prostate cancer (NEPC) and double-negative prostate cancer (DNPC) that is deficient for androgen receptor (AR) and neuroendocrine (NE) markers has increased. These prostate cancer subtypes are typically refractory to AR-directed therapies and exhibit poor clinical outcomes. Only a small range of NEPC/DNPC models exist, limiting our molecular understanding of this disease and hindering our ability to perform preclinical trials exploring novel therapies to treat NEPC/DNPC that are urgently needed in the clinic. Here, we report the development of the CU-PC01 PDX model that represents AR-negative mCRPC with PTEN/RB/PSMA loss and CTNN1B/TP53/BRCA2 genetic variants. The CU-PC01 model lacks classic NE markers, with only focal and/or weak expression of chromogranin A, INSM1 and CD56. Collectively, these findings are most consistent with a DNPC phenotype. Ex vivo and in vivo preclinical studies revealed that CU-PC01 PDX tumours are resistant to mCRPC standard-of-care treatments enzalutamide and docetaxel, mirroring the donor patient's treatment response. Furthermore, short-term CU-PC01 tumour explant cultures indicate this model is initially sensitive to PARP inhibition with olaparib. Thus, the CU-PC01 PDX model provides a valuable opportunity to study AR-negative mCRPC biology and to discover new treatment avenues for this hard-to-treat disease.
Collapse
Affiliation(s)
- Daniel J. Turnham
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
| | - Manisha S. Mullen
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
| | - Nicholas P. Bullock
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
| | | | - Anna E. Richards
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
| | - Radhika Patel
- Division of Human Biology and Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Marcos Quintela
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
| | - Valerie S. Meniel
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
| | - Gillian Seaton
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
| | - Howard Kynaston
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
- Department of Urology, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Richard W. E. Clarkson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
| | - Toby J. Phesse
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Peter S. Nelson
- Division of Human Biology and Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
- Department of Urology, University of Washington, Seattle, WA 98195, USA
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Michael C. Haffner
- Division of Human Biology and Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - John N. Staffurth
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
| |
Collapse
|
11
|
Briski LM, Aron M, Epstein JI, Russell DH, Assarzadegan N, Delma KS, O’Dell H, Rodriguez E, Montgomery EA, Kryvenko ON. Patterns of Immunoreactivity with TTF-1 Antibodies 8G7G3/1 and SPT24 Suggest Distinct Immunoprofiles Between Most Pulmonary and Nonpulmonary Small Cell Carcinomas. Int J Surg Pathol 2024; 32:230-238. [PMID: 37170625 PMCID: PMC11783247 DOI: 10.1177/10668969231171940] [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] [Indexed: 05/13/2023]
Abstract
Introduction. Small cell carcinoma can arise from various sites. Herein, we analyze the ability of 2 thyroid transcription factor-1 (TTF-1) antibodies (SPT24 and 8G7G3/1) to separate pulmonary from nonpulmonary small cell carcinoma. Materials and Methods. We analyzed 26 pulmonary and 83 nonpulmonary small cell carcinomas, and 14 Merkel cell carcinomas. Each tumor was stained with SPT24 and 8G7G3/1. Extent of nuclear staining was scored as diffuse (>50%), focal (11%-50%), rare (1%-10%), or negative (<1%). Results. All pulmonary small cell carcinomas were positive for SPT24 and 8G7G3/1. Four Merkel cell carcinomas (29%) were positive for SPT24 (ranging from rare-to-diffuse), while 2 (14%) showed rare expression with 8G7G3/1. For nonpulmonary small cell carcinomas, 69 (83%) were positive for SPT24 and 40 (48%) were positive for 8G7G3/1. For SPT24 positive tumors, the extent of 8G7G3/1 expression was equal in 17 (25%) and less in 52 tumors (75%), including 29 (42%) that were negative for 8G7G3/1. No nonpulmonary small cell carcinoma had more staining with 8G7G3/1 compared to SPT24. The differences in staining between 8G7G3/1 and SPT24 in the nonpulmonary cohort were statistically significant (P < 0.0001) with no significant difference between primary and metastatic lesions for 8G7G3/1 (P = 0.66) or SPT24 (P = 0.77). Conclusion. Most pulmonary small cell carcinomas are diffusely positive for both SPT24 and 8G7G3/1, whereas most nonpulmonary small cell carcinomas exhibit focal-to-no staining with 8G7G3/1 and significantly less staining with 8G7G3/1 compared to SPT24. However, these trends are not absolute and should be interpreted in conjunction with clinical and radiological findings.
Collapse
Affiliation(s)
- Laurence M. Briski
- Department of Pathology and Laboratory Medicine, University of Miami Hospital, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Manju Aron
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jonathan I. Epstein
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Daniel H. Russell
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Naziheh Assarzadegan
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Katiana S. Delma
- Department of Pathology and Laboratory Medicine, University of Miami Hospital, Miami, FL, USA
| | - Henry O’Dell
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Estelamari Rodriguez
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Oncology, University of Miami Hospital, Miami, FL, USA
| | - Elizabeth A. Montgomery
- Department of Pathology and Laboratory Medicine, University of Miami Hospital, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Oleksandr N. Kryvenko
- Department of Pathology and Laboratory Medicine, University of Miami Hospital, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Radiation Oncology, University of Miami Hospital, Miami, FL, USA
| |
Collapse
|
12
|
Kouroukli O, Bravou V, Giannitsas K, Tzelepi V. Tissue-Based Diagnostic Biomarkers of Aggressive Variant Prostate Cancer: A Narrative Review. Cancers (Basel) 2024; 16:805. [PMID: 38398199 PMCID: PMC10887410 DOI: 10.3390/cancers16040805] [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: 01/12/2024] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Prostate cancer (PC) is a common malignancy among elderly men, characterized by great heterogeneity in its clinical course, ranging from an indolent to a highly aggressive disease. The aggressive variant of prostate cancer (AVPC) clinically shows an atypical pattern of disease progression, similar to that of small cell PC (SCPC), and also shares the chemo-responsiveness of SCPC. The term AVPC does not describe a specific histologic subtype of PC but rather the group of tumors that, irrespective of morphology, show an aggressive clinical course, dictated by androgen receptor (AR) indifference. AR indifference represents an adaptive response to androgen deprivation therapy (ADT), driven by epithelial plasticity, an inherent ability of tumor cells to adapt to their environment by changing their phenotypic characteristics in a bi-directional way. The molecular profile of AVPC entails combined alterations in the tumor suppressor genes retinoblastoma protein 1 (RB1), tumor protein 53 (TP53), and phosphatase and tensin homolog (PTEN). The understanding of the biologic heterogeneity of castration-resistant PC (CRPC) and the need to identify the subset of patients that would potentially benefit from specific therapies necessitate the development of prognostic and predictive biomarkers. This review aims to discuss the possible pathophysiologic mechanisms of AVPC development and the potential use of emerging tissue-based biomarkers in clinical practice.
Collapse
Affiliation(s)
- Olga Kouroukli
- Department of Pathology, Evaggelismos General Hospital, 10676 Athens, Greece
| | - Vasiliki Bravou
- Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, 26504 Patras, Greece;
| | | | - Vasiliki Tzelepi
- Department of Pathology, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
13
|
Liao RS, Ruan HT, Jang A, Huynh M, Nadal Rios R, Hoffman-Censits JH, Wei S, Mian OY, Barata PC. Emerging Insights in Small-Cell Carcinoma of the Genitourinary Tract: From Diagnosis to Novel Therapeutic Horizons. Am Soc Clin Oncol Educ Book 2024; 44:e430336. [PMID: 38176691 DOI: 10.1200/edbk_430336] [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] [Indexed: 01/06/2024]
Abstract
Small-cell carcinomas (SCCs) of the genitourinary (GU) tract are rare malignancies with high metastatic potential. The most common primary sites are the bladder and prostate, but case reports of primary SCC of the kidney, ureter, and urethra also exist. The majority of patients present with gross hematuria, irritative or obstructive urinary symptoms, and symptoms of locoregionally advanced or metastatic disease at initial presentation. SCC of the bladder presents with nodal or metastatic involvement in the majority of cases and requires the use of platinum-based chemotherapy in combination with surgery and/or radiation. SCC of the prostate is most commonly seen in the metastatic castrate-resistant setting, and aggressive variant disease presents with a greater propensity for visceral metastases, osteolytic lesions, and relatively low serum prostate-specific antigen for volume of disease burden. Multiple retrospective and prospective randomized studies support the use of a multimodal approach combining platinum-based systemic therapy regimens with radiation and/or surgery for localized disease. This evidence-based strategy is reflected in multiple consensus guidelines. Emerging data suggest that small-cell bladder and prostate cancers transdifferentiate from a common progenitor of conventional urothelial bladder carcinoma and prostatic acinar adenocarcinoma, respectively. Areas of active basic research include efforts to identify the key genetic and epigenetic drivers involved in the emergence of small cell cancers to exploit them for novel therapies. Here, we review these efforts, discuss diagnosis and currently supported management strategies, and summarize ongoing clinical trials evaluating novel therapies to treat this rare, aggressive GU cancer.
Collapse
Affiliation(s)
- Ross S Liao
- Cleveland Clinic Glickman Urological and Kidney Institute, Cleveland, OH
| | - Hui Ting Ruan
- Cleveland Clinic Lerner Research Institute, Cleveland, OH
| | - Albert Jang
- University Hospitals Seidman Cancer Center, Cleveland, OH
| | - Melissa Huynh
- Western University Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | | | | | - Shuanzeng Wei
- Fox Chase Cancer Center, Department of Pathology, Philadelphia, PA
| | - Omar Y Mian
- Cleveland Clinic Foundation, Case Comprehensive Cancer Center, Cleveland, OH
| | - Pedro C Barata
- University Hospitals Seidman Cancer Center, Cleveland, OH
| |
Collapse
|
14
|
Thomas C. [Role of chemotherapy in metastatic castration-resistant prostate cancer (mCRPC) treatment: still standard or exception?]. UROLOGIE (HEIDELBERG, GERMANY) 2023; 62:1281-1288. [PMID: 37904040 DOI: 10.1007/s00120-023-02215-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 11/01/2023]
Abstract
BACKGROUND Taxanes have been used as monotherapy for metastatic prostate cancer for two decades. OBJECTIVES The current status of docetaxel and cabazitaxel in the treatment sequence for metastatic prostate cancer needs to be clarified. MATERIALS AND METHODS Overview of the existing literature regarding approval, dosage and new combination options for metastatic castration-resistant prostate cancer (mCRPC). RESULTS Taxanes represent one, but no longer the only treatment option for mCRPC. Previously, monotherapy was standard of care in the first and second line for mCRPC; nowadays taxanes are thrusted in the background due to new encouraging drug options. Based on the promising data of docetaxel in triple therapy setting for hormone-sensitive stage, its role as monotherapy in mCRPC needs to be clarified. Cabazitaxel is an alternative to PSMA radioligand therapy after failure of novel hormonal therapy (NHT) and docetaxel. Therapy adherence for taxanes can be significantly improved by dosage adjustments. Both treatment-related neuroendocrine prostate cancer (t-NEPC) and aggressive variant of prostate cancer (AVPC) represent a challenge for experienced uro-oncologists. Here, the combination of taxane plus platinum represents a promising option. CONCLUSIONS Taxanes are indicated in different stages of metastatic prostate cancer. Their use, particularly in combination with other drugs, appears to be promising. Traditional sequential taxane monotherapy regimens will be challenged by novel systemic therapy approaches.
Collapse
Affiliation(s)
- Christian Thomas
- Klinik und Poliklinik für Urologie, Universitätsklinikum Carl Gustav Carus Dresden, an der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.
| |
Collapse
|
15
|
Ji Y, Zhang W, Shen K, Su R, Liu X, Ma Z, Liu B, Hu C, Xue Y, Xin Z, Yang Y, Li A, Jiang Z, Jing N, Zhu HH, Dong L, Zhu Y, Dong B, Pan J, Wang Q, Xue W. The ELAVL3/MYCN positive feedback loop provides a therapeutic target for neuroendocrine prostate cancer. Nat Commun 2023; 14:7794. [PMID: 38016952 PMCID: PMC10684895 DOI: 10.1038/s41467-023-43676-3] [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: 04/06/2023] [Accepted: 11/16/2023] [Indexed: 11/30/2023] Open
Abstract
Neuroendocrine prostate cancer is a rapidly progressive and lethal disease characterized by early visceral metastasis, poor prognosis, and limited treatment options. Uncovering the oncogenic mechanisms could lead to the discovery of potential therapeutic avenues. Here, we demonstrate that the RNA-binding protein ELAVL3 is specifically upregulated in neuroendocrine prostate cancer and that overexpression of ELAVL3 alone is sufficient to induce the neuroendocrine phenotype in prostate adenocarcinoma. Mechanistically, ELAVL3 is transcriptionally regulated by MYCN and subsequently binds to and stabilizes MYCN and RICTOR mRNA. Moreover, ELAVL3 is shown to be released in extracellular vesicles and induce neuroendocrine differentiation of adenocarcinoma cells via an intercellular mechanism. Pharmacological inhibition of ELAVL3 with pyrvinium pamoate, an FDA-approved drug, effectively suppresses tumor growth, reduces metastatic risk, and improves survival in neuroendocrine prostate cancer mouse models. Our results identify ELAVL3 as a critical regulator of neuroendocrine differentiation in prostate cancer and propose a drug repurposing strategy for targeted therapies.
Collapse
Affiliation(s)
- Yiyi Ji
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Weiwei Zhang
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Kai Shen
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Ruopeng Su
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Xinyu Liu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Zehua Ma
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Bo Liu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Cong Hu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Yizheng Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Zhixiang Xin
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Yi Yang
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Ang Li
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Zhou Jiang
- Department of Pathology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Na Jing
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Helen He Zhu
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Dong
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Yinjie Zhu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Baijun Dong
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Jiahua Pan
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Qi Wang
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China.
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200120, China.
| | - Wei Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
16
|
Ji Y, Liu B, Chen L, Li A, Shen K, Su R, Zhang W, Zhu Y, Wang Q, Xue W. Repurposing ketotifen as a therapeutic strategy for neuroendocrine prostate cancer by targeting the IL-6/STAT3 pathway. Cell Oncol (Dordr) 2023; 46:1445-1456. [PMID: 37120492 DOI: 10.1007/s13402-023-00822-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2023] [Indexed: 05/01/2023] Open
Abstract
PURPOSE Neuroendocrine prostate cancer (NEPC), a highly aggressive subtype of prostate cancer displaying resistance to hormone therapy, presents a poor prognosis and limited therapeutic options. Here, we aimed to find novel medication therapies for NEPC and explore the underlying mechanism. METHODS A high-throughput drug screening utilizing an FDA-approved drug library was performed and ketotifen, an antihistamine agent, was identified as a potential therapeutic candidate for NEPC. The whole-transcriptome sequencing analysis was conducted to explore mechanism of ketotifen inhibitory in NEPC. Multiple cell biology and biochemistry experiments were performed to confirm the inhibitory effect of ketotifen in vitro. A spontaneous NEPC mice model (PBCre4:Ptenf/f;Trp53f/f;Rb1f/f) was used to reveal the inhibitory effect of ketotifen in vivo. RESULTS Our in vitro experiments demonstrated that ketotifen effectively suppressed neuroendocrine differentiation, reduced cell viability, and reversed the lineage switch via targeting the IL-6/STAT3 pathway. Our in vivo results showed that ketotifen significantly prolonged overall survival and reduced the risk of distant metastases in NEPC mice model. CONCLUSION Our findings repurpose ketotifen for antitumor applications and endorse its clinical development for NEPC therapy, offering a novel and promising therapeutic strategy for this formidable cancer subtype.
Collapse
Affiliation(s)
- Yiyi Ji
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Bo Liu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Lei Chen
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Ang Li
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Kai Shen
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Ruopeng Su
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Weiwei Zhang
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Yinjie Zhu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China.
| | - Qi Wang
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China.
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200120, China.
| | - Wei Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
17
|
Weng XT, Lin WL, Pan QM, Chen TF, Li SY, Gu CM. Aggressive variant prostate cancer: A case report and literature review. World J Clin Cases 2023; 11:6213-6222. [PMID: 37731555 PMCID: PMC10507546 DOI: 10.12998/wjcc.v11.i26.6213] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/29/2023] [Accepted: 08/15/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND Aggressive variant prostate cancer (AVPC) is a rare disease that progresses rapidly. The first-line treatment for AVPC is currently unknown. We examined a rare case of AVPC with rare brain and bladder metastases. A summary review of the mechanism of development, clinicopathological manifestations, associated treatments and prognosis of this disease is presented. CASE SUMMARY The patient was diagnosed with prostate cancer (PCA), and was actively treated with endocrine therapy, radiotherapy, chemotherapy, and traditional Chinese medicine. Unfortunately, he was insensitive to treatment, and the disease progressed rapidly. He died five years after being diagnosed with PCA. CONCLUSION We should reach consensus definitions of the AVPC and other androgen receptor-independent subtypes of PCA and develop new biomarkers to identify groups of high-risk variants. It is crucial to complete a puncture biopsy of the tumor or metastatic lesion as soon as possible in patients with advanced PCA who exhibit clinical features such as low Prostate-specific antigen levels, high carcinoembryonic antigen levels, and insensitivity to hormones to determine the pathological histological type and to create a more aggressive monitoring and treatment regimens.
Collapse
Affiliation(s)
- Xiang-Tao Weng
- Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong Province, China
| | - Wen-Li Lin
- Department of Urology, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong Province, China
| | - Qi-Man Pan
- Department of Urology, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong Province, China
| | - Tao-Fen Chen
- Department of Urology, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong Province, China
| | - Si-Yi Li
- Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong Province, China
| | - Chi-Ming Gu
- Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong Province, China
| |
Collapse
|
18
|
Mohanty SK, Lobo A, Williamson SR, Shah RB, Trpkov K, Varma M, Sirohi D, Aron M, Kandukari SR, Balzer BL, Luthringer DL, Ro J, Osunkoya AO, Desai S, Menon S, Nigam LK, Sardana R, Roy P, Kaushal S, Midha D, Swain M, Ambekar A, Mitra S, Rao V, Soni S, Jain K, Diwaker P, Pattnaik N, Sharma S, Chakrabarti I, Sable M, Jain E, Jain D, Samra S, Vankalakunti M, Mohanty S, Parwani AV, Sancheti S, Kumari N, Jha S, Dixit M, Malik V, Arora S, Munjal G, Gopalan A, Magi-Galluzzi C, Dhillon J. Reporting Trends, Practices, and Resource Utilization in Neuroendocrine Tumors of the Prostate Gland: A Survey among Thirty-Nine Genitourinary Pathologists. Int J Surg Pathol 2023; 31:993-1005. [PMID: 35946087 DOI: 10.1177/10668969221116629] [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] [Indexed: 11/15/2022]
Abstract
Background. Neuroendocrine differentiation in the prostate gland ranges from clinically insignificant neuroendocrine differentiation detected with markers in an otherwise conventional prostatic adenocarcinoma to a lethal high-grade small/large cell neuroendocrine carcinoma. The concept of neuroendocrine differentiation in prostatic adenocarcinoma has gained considerable importance due to its prognostic and therapeutic ramifications and pathologists play a pivotal role in its recognition. However, its awareness, reporting, and resource utilization practice patterns among pathologists are largely unknown. Methods. Representative examples of different spectrums of neuroendocrine differentiation along with a detailed questionnaire were shared among 39 urologic pathologists using the survey monkey software. Participants were specifically questioned about the use and awareness of the 2016 WHO classification of neuroendocrine tumors of the prostate, understanding of the clinical significance of each entity, and use of different immunohistochemical (IHC) markers. De-identified respondent data were analyzed. Results. A vast majority (90%) of the participants utilize IHC markers to confirm the diagnosis of small cell neuroendocrine carcinoma. A majority (87%) of the respondents were in agreement regarding the utilization of type of IHC markers for small cell neuroendocrine carcinoma for which 85% of the pathologists agreed that determination of the site of origin of a high-grade neuroendocrine carcinoma is not critical, as these are treated similarly. In the setting of mixed carcinomas, 62% of respondents indicated that they provide quantification and grading of the acinar component. There were varied responses regarding the prognostic implication of focal neuroendocrine cells in an otherwise conventional acinar adenocarcinoma and for Paneth cell-like differentiation. The classification of large cell neuroendocrine carcinoma was highly varied, with only 38% agreement in the illustrated case. Finally, despite the recommendation not to perform neuroendocrine markers in the absence of morphologic evidence of neuroendocrine differentiation, 62% would routinely utilize IHC in the work-up of a Gleason score 5 + 5 = 10 acinar adenocarcinoma and its differentiation from high-grade neuroendocrine carcinoma. Conclusion. There is a disparity in the practice utilization patterns among the urologic pathologists with regard to diagnosing high-grade neuroendocrine carcinoma and in understanding the clinical significance of focal neuroendocrine cells in an otherwise conventional acinar adenocarcinoma and Paneth cell-like neuroendocrine differentiation. There seems to have a trend towards overutilization of IHC to determine neuroendocrine differentiation in the absence of neuroendocrine features on morphology. The survey results suggest a need for further refinement and development of standardized guidelines for the classification and reporting of neuroendocrine differentiation in the prostate gland.
Collapse
Affiliation(s)
- Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, India
| | - Anandi Lobo
- Department of Pathology and Laboratory Medicine, Kapoor Urology Center and Pathology Laboratory, Raipur, India
| | | | - Rajal B Shah
- Department of Pathology, UT Southwestern University, Dallas, TX, USA
| | - Kiril Trpkov
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Murali Varma
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| | - Deepika Sirohi
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Manju Aron
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Shivani R Kandukari
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Bonnie L Balzer
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daniel L Luthringer
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jae Ro
- Department of Pathology and Genomic Medicine, Methodist Hospital, Houston, TX, USA
| | - Adeboye O Osunkoya
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Sangeeta Desai
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Santosh Menon
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Lovelesh K Nigam
- Department of Pathology and Division of Renal and Urologic Pathology, Lal Pathology Laboratory, New Delhi, India
| | - Rohan Sardana
- Department of Pathology, Ampath Pathological Laboratory, Hyderabad, India
| | - Paromita Roy
- Department of Oncopathology, Tata Medical Center, Kolkata, India
| | - Seema Kaushal
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Divya Midha
- Department of Oncopathology, Tata Medical Center, Kolkata, India
| | - Minakshi Swain
- Department of Pathology and Laboratory Medicine, Apollo Hospital, Hyderabad, India
| | - Asawari Ambekar
- Department of Pathology and Laboratory Medicine, Apollo Hospital, Mumbai, India
| | - Suvradeep Mitra
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Vishal Rao
- Department of Pathology and Laboratory Medicine, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, India
| | - Shailesh Soni
- Department of Pathology and Laboratory Medicine, Muljibhai Patel Urological Hospital, Gujarat, India
| | - Kavita Jain
- Department of Pathology and Laboratory Medicine, Max Superspeciality Hospital, New Delhi, India
| | - Preeti Diwaker
- Department of Pathology, University College of Medical Sciences, New Delhi, India
| | - Niharika Pattnaik
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, India
| | - Shivani Sharma
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | | | - Mukund Sable
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Ekta Jain
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | - Deepika Jain
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | - Spinder Samra
- Department of Pathology, Dubbo Base Hospital, Dubbo, NSW, Australia
| | - Mahesha Vankalakunti
- Department of Pathology and Laboratory Medicine, Manipal Hospital, Bangalore, India
| | - Subhashis Mohanty
- Department of Histopathology, SUM Ultimate Medicare, Bhubaneswar, India
| | - Anil V Parwani
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Sankalp Sancheti
- Department of Pathology and Laboratory Medicine, Homi Bhabha Cancer Hospital & Research Centre, Punjab (A Unit of Tata Memorial Centre, Mumbai), India
| | - Niraj Kumari
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Raebareli, India
| | - Shilpy Jha
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, India
| | - Mallika Dixit
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | - Vipra Malik
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | - Samriti Arora
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | - Gauri Munjal
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | - Anuradha Gopalan
- Department of Pathology, Memorial Sloan Kettering Cancer, New York, NY, USA
| | | | | |
Collapse
|
19
|
Cohen D, Hazut Krauthammer S, Fahoum I, Kesler M, Even-Sapir E. PET radiotracers for whole-body in vivo molecular imaging of prostatic neuroendocrine malignancies. Eur Radiol 2023; 33:6502-6512. [PMID: 37052659 DOI: 10.1007/s00330-023-09619-8] [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: 09/13/2022] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 04/14/2023]
Abstract
Prostatic neuroendocrine malignancies represent a spectrum of diseases. Treatment-induced neuroendocrine differentiation (tiNED) in hormonally treated adenocarcinoma has been the subject of a large amount of recent research. However, the identification of neuroendocrine features in treatment-naïve prostatic tumor raises a differential diagnosis between prostatic adenocarcinoma with de novo neuroendocrine differentiation (dNED) versus one of the primary prostatic neuroendocrine tumors (P-NETs) and carcinomas (P-NECs). While [18F]FDG is being used as the main PET radiotracer in oncologic imaging and reflects cellular glucose metabolism, other molecules labeled with positron-emitting isotopes, mainly somatostatin-analogues labeled with 68Ga and prostate-specific membrane antigen (PSMA)-ligands labeled with either 18F or 68Ga, are now routinely used in departments of nuclear medicine and molecular imaging, and may be advantageous in imaging prostatic neuroendocrine malignancies. Still, the selection of the preferred PET radiotracer in such cases might be challenging. In the current review, we summarize and discuss published data on these different entities from clinical, biological, and molecular imaging standpoints. Specifically, we review the roles that [18F]FDG, radiolabeled somatostatin-analogues, and radiolabeled PSMA-ligands play in these entities in order to provide the reader with practical recommendations regarding the preferred PET radiotracers for imaging each entity. In cases of tiNED, we conclude that PSMA expression may be low and that [18F]FDG or radiolabeled somatostatin-analogues should be preferred for imaging. In cases of prostatic adenocarcinoma with dNED, we present data that support the superiority of radiolabeled PSMA-ligands. In cases of primary neuroendocrine malignancies, the use of [18F]FDG for imaging high-grade P-NECs and radiolabeled somatostatin-analogues for imaging well-differentiated P-NETs is recommended. KEY POINTS: • The preferred PET radiotracer for imaging prostatic neuroendocrine malignancies depends on the specific clinical scenario and pathologic data. • When neuroendocrine features result from hormonal therapy for prostate cancer, PET-CT should be performed with [18F]FDG or radiolabeled somatostatin-analogue rather than with radiolabeled PSMA-ligand. • When neuroendocrine features are evident in newly diagnosed prostate cancer, differentiating adenocarcinoma from primary neuroendocrine malignancy is challenging but crucial for selection of PET radiotracer and for clinical management.
Collapse
Affiliation(s)
- Dan Cohen
- Department of Nuclear Medicine, Tel-Aviv Sourasky Medical Center, 6 Weizmann St, 6423906, Tel Aviv, Israel.
| | - Shir Hazut Krauthammer
- Department of Nuclear Medicine, Tel-Aviv Sourasky Medical Center, 6 Weizmann St, 6423906, Tel Aviv, Israel
| | - Ibrahim Fahoum
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Mikhail Kesler
- Department of Nuclear Medicine, Tel-Aviv Sourasky Medical Center, 6 Weizmann St, 6423906, Tel Aviv, Israel
| | - Einat Even-Sapir
- Department of Nuclear Medicine, Tel-Aviv Sourasky Medical Center, 6 Weizmann St, 6423906, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
20
|
Huang Z, Tang Y, Wei Y, Qian J, Kang Y, Wang D, Xu M, Nie L, Chen X, Chen N, Zhou Q. Prognostic Significance of Chromogranin A Expression in the Initial and Second Biopsies in Metastatic Prostate Cancer. J Clin Med 2023; 12:jcm12103362. [PMID: 37240468 DOI: 10.3390/jcm12103362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/27/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Neuroendocrine differentiation (NED) characterized by the expression of neuroendocrine markers, such as chromogranin A (CgA), is frequently observed in advanced prostate cancer (PCa), the prognostic significance of which is still controversial. Here we specifically addressed the issue of the potential prognostic value of CgA expression in advanced-stage PCa patients with distant metastases and its change over time from metastatic hormone-sensitive (mHSPC) to metastatic castration-resistant prostate cancer (mCRPC). CgA expression was assessed immunohistochemically in initial biopsies of mHSPC, as well as in second biopsies of mCRPC in sixty-eight patients, and its correlation with prognosis (together with conventional clinicopathologic parameters) was analyzed using the Kaplan-Meier method and Cox proportional hazard model. We found that CgA expression was an independent adverse prognostic factor for both mHSPC (CgA positivity ≥ 1%, HR = 2.16, 95% CI: 1.04-4.26, p = 0.031) and mCRPC (CgA ≥ 10%, HR = 20.19, 95% CI: 3.04-329.9, p = 0.008). CgA positivity generally increased from mHSPC to mCRPC and was a negative prognosticator. The assessment of CgA expression may help with the clinical evaluation of advanced-stage patients with distant metastases.
Collapse
Affiliation(s)
- Zhuo Huang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Tang
- Department of Pathology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Yuyan Wei
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingyu Qian
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yifan Kang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Duohao Wang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Miao Xu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ling Nie
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xueqin Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ni Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiao Zhou
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
Chen L, Ji Y, Li A, Liu B, Shen K, Su R, Ma Z, Zhang W, Wang Q, Zhu Y, Xue W. High-throughput drug screening identifies fluoxetine as a potential therapeutic agent for neuroendocrine prostate cancer. Front Oncol 2023; 13:1085569. [PMID: 36994207 PMCID: PMC10042075 DOI: 10.3389/fonc.2023.1085569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
IntroductionNeuroendocrine prostate cancer (NEPC) is an aggressive subtype of prostate cancer with poor prognosis and resistance to hormone therapy, which has limited therapeutic approaches. Therefore, this study aimed to identify a novel treatment for NEPC and provide evidence of its inhibitory effects.MethodsWe performed a high-throughput drug screening and identified fluoxetine, originally an FDA-approved antidepressant, as candidate therapeutic agent for NEPC. We carried out both in vitro and in vivo experiments to demonstrate the inhibitory effects of fluoxetine on NEPC models and its mechanism in detail.ResultsOur results demonstrated that fluoxetine effectively curbed the neuroendocrine differentiation and inhibited cell viability by targeting the AKT pathway. Preclinical test in NEPC mice model (PBCre4: Ptenf/f; Trp53f/f; Rb1f/f) showed that fluoxetine effectively prolonged the overall survival and reduced the risk of tumor distant metastases.DiscussionThis work repurposed fluoxetine for antitumor application, and supported its clinical development for NEPC therapy, which may provide a promising therapeutic strategy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qi Wang
- *Correspondence: Qi Wang, ; Yinjie Zhu,
| | | | | |
Collapse
|
22
|
Kusakabe M, Taguchi A, Tanikawa M, Wagatsuma R, Yamazaki M, Tsuchimochi S, Toyohara Y, Kawata A, Baba S, Ueno T, Sone K, Mori-Uchino M, Ikemura M, Matsunaga H, Nagamatsu T, Wada-Hiraike O, Kawazu M, Ushiku T, Takeyama H, Oda K, Kawana K, Mano H, Osuga Y. Cells with stem-like properties are associated with the development of HPV18-positive cervical cancer. Cancer Sci 2023; 114:885-895. [PMID: 36404139 PMCID: PMC9986059 DOI: 10.1111/cas.15664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/22/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
The cellular origins of cervical cancer and the histological differentiation of human papillomavirus (HPV)-infected cells remain unexplained. To gain new insights into the carcinogenesis and histological differentiation of HPV-associated cervical cancer, we focused on cervical cancer with mixed histological types. We conducted genomic and transcriptomic analyses of cervical cancers with mixed histological types. The commonality of the cellular origins of these cancers was inferred using phylogenetic analysis and by assessing the HPV integration sites. Carcinogenesis was estimated by analyzing human gene expression profiles in different histological types. Among 42 cervical cancers with known HPV types, mixed histological types were detected in four cases, and three of them were HPV18-positive. Phylogenetic analysis of these three cases revealed that the different histological types had a common cell of origin. Moreover, the HPV-derived transcriptome and HPV integration sites were common among different histological types, suggesting that HPV integration could occur before differentiation into each histological type. Human gene expression profiles indicated that HPV18-positive cancer retained immunologically cold components with stem cell properties. Mixed cervical cancer has a common cellular origin among different histological types, and progenitor cells with stem-like properties may be associated with the development of HPV18-positive cervical cancer.
Collapse
Affiliation(s)
- Misako Kusakabe
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ayumi Taguchi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Michihiro Tanikawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryota Wagatsuma
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan.,CBBD-OIL, AIST-Waseda University, Tokyo, Japan
| | - Miki Yamazaki
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan.,CBBD-OIL, AIST-Waseda University, Tokyo, Japan
| | - Saki Tsuchimochi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yusuke Toyohara
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akira Kawata
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Satoshi Baba
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshihide Ueno
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Kenbun Sone
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mayuyo Mori-Uchino
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masako Ikemura
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroko Matsunaga
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masahito Kawazu
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan.,CBBD-OIL, AIST-Waseda University, Tokyo, Japan.,Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan.,Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
| | - Katsutoshi Oda
- Division of Integrative Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kei Kawana
- Department of Obstetrics and Gynecology, Nihon University School of medicine, Tokyo, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
23
|
Abdulfatah E, Fine SW, Lotan TL, Mehra R. Reprint of: de novo neuroendocrine features in prostate cancer. Hum Pathol 2023; 133:115-125. [PMID: 36894369 DOI: 10.1016/j.humpath.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/01/2022] [Indexed: 03/09/2023]
Abstract
Neuroendocrine tumors of the prostate are rare and encompass a group of entities that are classified based on a combination of morphological and immunohistochemical features. Despite the 2016 World Health Organization classification of prostatic neuroendocrine tumors, variants have been reported that do not fit well in the categorization scheme. While the majority of these tumors arise in the setting of castration-resistant prostate cancer (postandrogen deprivation therapy), de novo cases may occur. In this review, we highlight the most significant pathological and immunohistochemical features, emerging biomarkers, and molecular features of such tumors.
Collapse
Affiliation(s)
- Eman Abdulfatah
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Samson W Fine
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21211, USA
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA; Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, 48109, USA; Michigan Center for Translational Pathology, Ann Arbor, MI, 48104, USA.
| |
Collapse
|
24
|
Kufe D. Dependence on MUC1-C in Progression of Neuroendocrine Prostate Cancer. Int J Mol Sci 2023; 24:3719. [PMID: 36835130 PMCID: PMC9967814 DOI: 10.3390/ijms24043719] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/11/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Castration resistant prostate cancer (CRPC) is responsive to androgen receptor (AR) axis targeted agents; however, patients invariably relapse with resistant disease that often progresses to neuroendocrine prostate cancer (NEPC). Treatment-related NEPC (t-NEPC) is highly aggressive with limited therapeutic options and poor survival outcomes. The molecular basis for NEPC progression remains incompletely understood. The MUC1 gene evolved in mammals to protect barrier tissues from loss of homeostasis. MUC1 encodes the transmembrane MUC1-C subunit, which is activated by inflammation and contributes to wound repair. However, chronic activation of MUC1-C contributes to lineage plasticity and carcinogenesis. Studies in human NEPC cell models have demonstrated that MUC1-C suppresses the AR axis and induces the Yamanaka OSKM pluripotency factors. MUC1-C interacts directly with MYC and activates the expression of the BRN2 neural transcription factor (TF) and other effectors, such as ASCL1, of the NE phenotype. MUC1-C also induces the NOTCH1 stemness TF in promoting the NEPC cancer stem cell (CSC) state. These MUC1-C-driven pathways are coupled with activation of the SWI/SNF embryonic stem BAF (esBAF) and polybromo-BAF (PBAF) chromatin remodeling complexes and global changes in chromatin architecture. The effects of MUC1-C on chromatin accessibility integrate the CSC state with the control of redox balance and induction of self-renewal capacity. Importantly, targeting MUC1-C inhibits NEPC self-renewal, tumorigenicity and therapeutic resistance. This dependence on MUC1-C extends to other NE carcinomas, such as SCLC and MCC, and identify MUC1-C as a target for the treatment of these aggressive malignancies with the anti-MUC1 agents now under clinical and preclinical development.
Collapse
Affiliation(s)
- Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
25
|
Molecular Classification of Extrapulmonary Neuroendocrine Carcinomas With Emphasis on POU2F3-positive Tuft Cell Carcinoma. Am J Surg Pathol 2023; 47:183-193. [PMID: 36253891 PMCID: PMC9833113 DOI: 10.1097/pas.0000000000001977] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Extrapulmonary neuroendocrine carcinomas (EP-NECs) are associated with a poor clinical outcome, and limited information is available on the biology and treatment of EP-NECs. We studied EP-NECs by applying the recent novel findings from studies of pulmonary neuroendocrine carcinomas, including POU2F3, the master regulator of tuft cell variant of small cell lung carcinomas. A cohort of 190 patients with surgically resected EP-NECs or poorly differentiated carcinomas (PDCs) were established. Immunohistochemistry (IHC) for POU2F3 along with ASCL1, NEUROD1, YAP1, and conventional neuroendocrine markers was performed on tissue microarrays. Selected cases with or without POU2F3 expression were subjected to targeted gene expression profiling using nCounter PanCancer Pathway panel. POU2F3-positive tuft cell carcinomas were present in 12.6% of EP-NEC/PDCs, with variable proportions according to organ systems. POU2F3 expression was negatively correlated with the expression levels of ASCL1, NEUROD1, and conventional neuroendocrine markers ( P <0.001), enabling IHC-based molecular classification into ASCL1-dominant, NEUROD1-dominant, POU2F3-dominant, YAP1-dominant, and not otherwise specified subtypes. Compared wih POU2F3-negative cases, POU2F3-positive tuft cell carcinomas showed markedly higher expression levels of PLCG2 and BCL2 , which was also validated in the entire cohort by IHC. In addition to POU2F3, YAP1-positive tumors were a distinct subtype among EP-NEC/PDCs, characterized by unique T-cell inflamed microenvironment. We found rare extrapulmonary POU2F3-positive tumors arising from previously unappreciated cells of origin. Our data show novel molecular pathologic features of EP-NEC/PDCs including potential therapeutic vulnerabilities, thereby emphasizing the need for focusing on unique features of EP-NEC/PDCs.
Collapse
|
26
|
Yabusaki R, Yoshimura K, Taku K, Suzuki M. Prostate cancer recurring as small‐cell carcinoma with a
BRCA2
somatic mutation. IJU Case Rep 2022; 5:489-492. [PMID: 36341199 PMCID: PMC9626344 DOI: 10.1002/iju5.12523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Small‐cell carcinoma of the prostate has a poor prognosis, and treatment options for the refractory disease are unclear. Case presentation A 68‐year‐old man with prostate cancer was referred to our hospital. He was treated with combined androgen blockade (bicalutamide and degarelix acetate). The disease progressed to castration‐resistant prostate cancer, but with additional treatment, prostate‐specific antigen levels remained below 0.02 ng/mL. However, computed tomography revealed enlarged right inguinal lymph nodes; moreover, his neuron‐specific enolase levels were elevated. Histopathologic analysis of a biopsied lymph node confirmed small‐cell carcinoma. After administering cytotoxic chemotherapy (etoposide plus cisplatin and amrubicin), the patient temporarily improved before relapsing. After genetic testing of the biopsy specimen revealed a BRCA2 deletion, we administered the oral PARP‐2 inhibitor olaparib, which has achieved partial remission for 8 months. Conclusion PARP‐2 inhibition may improve the survival of patients with BRCA2‐positive small‐cell carcinoma of the prostate.
Collapse
Affiliation(s)
- Ryo Yabusaki
- Department of Urology Shizuoka General Hospital Shizuoka Japan
| | - Koji Yoshimura
- Department of Urology Shizuoka General Hospital Shizuoka Japan
| | - Keisei Taku
- Department of Oncology Shizuoka General Hospital Shizuoka Japan
| | - Makoto Suzuki
- Department of Pathology Shizuoka General Hospital Shizuoka Japan
| |
Collapse
|
27
|
Kost Y, Mattis DM, Muskat A, Nazarian R, Amin B, Cheng H, McLellan BN. Cutaneous metastasis as the first sign of small-cell cancer: An unexpected presentation in a patient with concurrent prostate cancer. JAAD Case Rep 2022; 26:104-106. [PMID: 35958435 PMCID: PMC9357704 DOI: 10.1016/j.jdcr.2022.05.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
| | | | | | | | | | | | - Beth N. McLellan
- Correspondence to: Beth N. McLellan, MD, Department of Medicine, Division of Dermatology, Montefiore Medical Center, Albert Einstein College of Medicine, 3411 Wayne Avenue, Bronx, NY 10467.
| |
Collapse
|
28
|
Abdulfatah E, Fine SW, Lotan T, Mehra R. De Novo Neuroendocrine Features in Prostate Cancer. Hum Pathol 2022; 127:112-122. [PMID: 35810832 DOI: 10.1016/j.humpath.2022.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/01/2022] [Indexed: 12/22/2022]
Abstract
Neuroendocrine tumors of the prostate are rare and encompass a group of entities that are classified based on a combination of morphological and immunohistochemical features. Despite the 2016 World Health Organization classification of prostatic neuroendocrine tumors, variants have been reported that do not fit well in the categorization scheme. While the majority of these tumors arise in the setting of castration-resistant prostate cancer (postandrogen deprivation therapy), de novo cases may occur. In this review, we highlight the most significant pathological and immunohistochemical features, emerging biomarkers, and molecular features of such tumors.
Collapse
Affiliation(s)
- Eman Abdulfatah
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Samson W Fine
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tamara Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, Ann Arbor, MI, USA.
| |
Collapse
|
29
|
Gopalan A, Al-Ahmadie H, Chen YB, Sarungbam J, Sirintrapun SJ, Tickoo SK, Reuter VE, Fine SW. Neuroendocrine Differentiation in the Setting of Prostatic Carcinoma: Contemporary Assessment of a Consecutive Series. Histopathology 2022; 81:246-254. [PMID: 35758203 PMCID: PMC9327588 DOI: 10.1111/his.14707] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/20/2022] [Accepted: 06/05/2022] [Indexed: 11/28/2022]
Abstract
AIM Clinicopathologic characterization of contemporary series of neuroendocrine (NE) differentiation in the setting of prostatic carcinoma (PCa). METHODS & RESULTS We reviewed institutional databases for in-house cases with history of PCa and histopathologic evidence of NE differentiation during the disease course. 79 cases identified: 32 primary and 47 metastases. Metastatic lesions were in liver [n=15], lymph node [n=9], bone [n=6], lung [n=3], brain [n=1], other sites [n=13]. 63 of 76 (82%) cases with NE differentiation and available history were post-therapy: 6 post-radiation therapy (RT), 24 post- androgen-deprivation therapy (ADT) and 33 post-RT+ADT. Morphologic assessment [n=79]: a. 23 pure small cell/high-grade NE carcinoma (HGNEC): 20/23 metastatic; b. 10 combined high-grade PCa and small cell/HGNEC: 9/10 primary; c. 15 PCa with diffuse NE immunohistochemistry (IHC) marker positivity/differentiation, associated with nested to sheet-like growth of cells with abundant cytoplasm and prominent nucleoli, yet diffuse positivity for at least one prostatic and one NE IHC marker: all metastatic; d. 11 PCa with patchy NE differentiation, displaying more than single cell positivity for NE IHC: 5 primary / 6 metastatic; e. 9 PCa with focal NE marker positive cells: 4 primary / 5 metastatic; f. 11 PCa with 'Paneth cell-like' change: all primary. CONCLUSIONS In this contemporary series, the majority of NE differentiation in the setting of PCa was seen post-therapy. We highlight tendencies of small cell/HGNEC and PCa with diffuse NE differentiation by IHC to occur in metastatic settings, while morphologically combined high grade PCa+small cell/HGNEC and 'Paneth cell-like' change occur in primary disease.
Collapse
Affiliation(s)
- Anuradha Gopalan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hikmat Al-Ahmadie
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ying-Bei Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Judy Sarungbam
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - S Joseph Sirintrapun
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Satish K Tickoo
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Victor E Reuter
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samson W Fine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
30
|
Single-cell proteomics defines the cellular heterogeneity of localized prostate cancer. Cell Rep Med 2022; 3:100604. [PMID: 35492239 PMCID: PMC9044103 DOI: 10.1016/j.xcrm.2022.100604] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/30/2021] [Accepted: 03/21/2022] [Indexed: 11/16/2022]
Abstract
Localized prostate cancer exhibits multiple genomic alterations and heterogeneity at the proteomic level. Single-cell technologies capture important cell-to-cell variability responsible for heterogeneity in biomarker expression that may be overlooked when molecular alterations are based on bulk tissue samples. This study aims to identify prognostic biomarkers and describe the heterogeneity of prostate cancer and the associated microenvironment by simultaneously quantifying 36 proteins using single-cell mass cytometry analysis of over 1.6 million cells from 58 men with localized prostate cancer. We perform this task, using a high-dimensional clustering pipeline named Franken to describe subpopulations of immune, stromal, and prostate cells, including changes occurring in tumor tissues and high-grade disease that provide insights into the coordinated progression of prostate cancer. Our results further indicate that men with localized disease already harbor rare subpopulations that typically occur in castration-resistant and metastatic disease.
Single-cell proteomics of localized prostate cancer defines disease heterogeneity Malignant and benign prostate tissues differ in rare cell-type proportional shifts T cells and proliferating macrophages are associated with high-grade PCa Rare CD15+ epithelial cells are amplified in high-grade PCa
Collapse
|
31
|
Galea LA, Mow C, Fine SW, Manohar P. Primary Prostatic Carcinoma With De Novo Diffuse Neuroendocrine Differentiation. Int J Surg Pathol 2022; 30:232-236. [PMID: 34338584 PMCID: PMC9887995 DOI: 10.1177/10668969211035844] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The 2016 World Health Organization classification of prostate cancer with neuroendocrine (NE) differentiation includes NE cells in usual prostate cancer, adenocarcinoma with Paneth cell-like NE differentiation, well-differentiated NE tumor (carcinoid), small cell NE carcinoma, and large cell NE carcinoma. In this article, we report a rare case of primary prostatic carcinoma with de novo diffuse NE differentiation presenting with bilateral hydronephrosis in a 79-year-old man. This case did not fit into any of the existing classifications. The clinical, radiological, morphological, and immunohistochemical findings and response to androgen deprivation therapy (ADT) are presented. The proposed pathogenesis of NE differentiation via transdifferentiation from conventional prostatic adenocarcinoma whereby genomic alterations, coupled with ADT can induce lineage plasticity resulting in NE differentiation is described.
Collapse
Affiliation(s)
- Laurence A. Galea
- Melbourne Pathology (Sonic Healthcare), Melbourne, Victoria, Australia
| | - Christopher Mow
- Melbourne Pathology (Sonic Healthcare), Melbourne, Victoria, Australia
| | | | - Paul Manohar
- Monash Health, Melbourne, Victoria, Australia,Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Golestani ZB, Soltani S, Kalantari MR, Ghorbani HR, Aghaee A. Rare Case of Small Cell Metastatic Prostatic Adenocarcinoma, Properly Staged by 68Ga-PSMA PET/CT Scan. Clin Nucl Med 2022; 47:e259-e261. [PMID: 35025791 DOI: 10.1097/rlu.0000000000004033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Small cell carcinoma (SCC) of the prostate is a very rare entity and one of most aggressive neuroendocrine malignancies of prostate. We present a 60-year-old man with pathology-proven prostatic SCC by transrectal biopsy with ultrasonography guidance and Gleason score of 5 + 5. He was referred to perform 68Ga-PSMA PET/CT scan for staging. The scan showed PSMA uptake in the prostate bed and multiple large pelvic lymph nodes. As we know, 68Ga-PSMA uptake in the de novo form of SCC of prostate is not commonly found in the literature.
Collapse
Affiliation(s)
| | | | - Mahmoud Reza Kalantari
- Department of Pathology, Faculty of Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | |
Collapse
|
33
|
Shi HJ, Fan ZN, Zhang JS, Xiong BB, Wang HF, Wang JS. Small-cell carcinoma of the prostate with negative CD56, NSE, Syn, and CgA indicators: A case report. World J Clin Cases 2022; 10:1630-1638. [PMID: 35211603 PMCID: PMC8855255 DOI: 10.12998/wjcc.v10.i5.1630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/14/2021] [Accepted: 12/31/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Small-cell carcinoma of the prostate (SCCP) is a clinically rare malignant tumor, accounting for < 1% of all prostate tumors. However, negativity for all SCCP neuroendocrine markers is rare. Herein, we report a case of SCCP with completely negative neuroendocrine markers and explore its clinicopathologic features, thus improving the understanding of its clinical diagnosis and management.
CASE SUMMARY We report the case of a 48-year-old patient with SCCP negative for common sensitive neuroendocrine-staining indicators. Dysuria was the first symptom, and rectal examination revealed a hard prostate, palpable nodules, diffuse prostate enlargement, no pressure pain, no blood staining in the finger sleeve, 1.33 ng/mL total prostate-specific antigen level, and a free-to-total prostate-specific antigen ratio of 0.21 ng/mL. Ultrasound suggested a prostate size of 5.3 cm × 5.8 cm × 5.6 cm, and magnetic resonance imaging suggested prostate cancer. The lower posterior bladder wall, rectal mesentery, and bilateral seminal vesicles were invaded, with multiple lymph node metastases in the pelvis. A whole-body bone scan suggested an abnormally active multiple bone metabolism and possible bone metastases. Head and lungs computed tomography revealed no significant nodal shadow. Following a pathological diagnosis of SCCP after a prostate puncture, with negative indicators of common sensitive neuroendocrine staining, chemotherapy was administered; the patient died 4-5 mo after SCCP diagnosis.
CONCLUSION SCCP is a rare disease characterized by atypical clinical symptoms, limited treatment options, a short survival period, and a poor prognosis.
Collapse
Affiliation(s)
- Hong-Jin Shi
- Department of Urology, The Second Affiliated Hospital, Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Zhi-Nan Fan
- Department of Urology, The Second Affiliated Hospital, Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Jin-Song Zhang
- Department of Urology, The Second Affiliated Hospital, Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Bo-Bo Xiong
- Department of Urology, The Second Affiliated Hospital, Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Hai-Feng Wang
- Department of Urology, The Second Affiliated Hospital, Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Jian-Song Wang
- Department of Urology, The Second Affiliated Hospital, Kunming Medical University, Kunming 650000, Yunnan Province, China
| |
Collapse
|
34
|
Singh N, Ramnarine VR, Song JH, Pandey R, Padi SKR, Nouri M, Olive V, Kobelev M, Okumura K, McCarthy D, Hanna MM, Mukherjee P, Sun B, Lee BR, Parker JB, Chakravarti D, Warfel NA, Zhou M, Bearss JJ, Gibb EA, Alshalalfa M, Karnes RJ, Small EJ, Aggarwal R, Feng F, Wang Y, Buttyan R, Zoubeidi A, Rubin M, Gleave M, Slack FJ, Davicioni E, Beltran H, Collins C, Kraft AS. The long noncoding RNA H19 regulates tumor plasticity in neuroendocrine prostate cancer. Nat Commun 2021; 12:7349. [PMID: 34934057 PMCID: PMC8692330 DOI: 10.1038/s41467-021-26901-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
Neuroendocrine (NE) prostate cancer (NEPC) is a lethal subtype of castration-resistant prostate cancer (PCa) arising either de novo or from transdifferentiated prostate adenocarcinoma following androgen deprivation therapy (ADT). Extensive computational analysis has identified a high degree of association between the long noncoding RNA (lncRNA) H19 and NEPC, with the longest isoform highly expressed in NEPC. H19 regulates PCa lineage plasticity by driving a bidirectional cell identity of NE phenotype (H19 overexpression) or luminal phenotype (H19 knockdown). It contributes to treatment resistance, with the knockdown of H19 re-sensitizing PCa to ADT. It is also essential for the proliferation and invasion of NEPC. H19 levels are negatively regulated by androgen signaling via androgen receptor (AR). When androgen is absent SOX2 levels increase, driving H19 transcription and facilitating transdifferentiation. H19 facilitates the PRC2 complex in regulating methylation changes at H3K27me3/H3K4me3 histone sites of AR-driven and NEPC-related genes. Additionally, this lncRNA induces alterations in genome-wide DNA methylation on CpG sites, further regulating genes associated with the NEPC phenotype. Our clinical data identify H19 as a candidate diagnostic marker and predictive marker of NEPC with elevated H19 levels associated with an increased probability of biochemical recurrence and metastatic disease in patients receiving ADT. Here we report H19 as an early upstream regulator of cell fate, plasticity, and treatment resistance in NEPC that can reverse/transform cells to a treatable form of PCa once therapeutically deactivated.
Collapse
MESH Headings
- Androgen Antagonists/therapeutic use
- Animals
- Benzamides/pharmacology
- Benzamides/therapeutic use
- Biomarkers, Tumor/metabolism
- Carcinoma, Neuroendocrine/diagnosis
- Carcinoma, Neuroendocrine/drug therapy
- Carcinoma, Neuroendocrine/genetics
- Carcinoma, Neuroendocrine/pathology
- Cell Line, Tumor
- Cell Lineage/genetics
- Cell Nucleus/metabolism
- Cell Plasticity/genetics
- Cell Proliferation/genetics
- Cohort Studies
- DNA Methylation/genetics
- Disease Models, Animal
- Drug Resistance, Neoplasm/genetics
- Epigenesis, Genetic/drug effects
- Gene Expression Regulation, Neoplastic
- Genome, Human
- Histones/metabolism
- Humans
- Male
- Neoplasm Grading
- Neoplasm Invasiveness
- Neoplastic Stem Cells/metabolism
- Nitriles/pharmacology
- Nitriles/therapeutic use
- Organoids/metabolism
- Organoids/pathology
- Phenylthiohydantoin/pharmacology
- Phenylthiohydantoin/therapeutic use
- Phylogeny
- Polycomb Repressive Complex 2/metabolism
- Promoter Regions, Genetic/genetics
- Prostatic Neoplasms/diagnosis
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/pathology
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Receptors, Androgen/metabolism
- SOXB1 Transcription Factors/metabolism
- Transcription, Genetic/drug effects
Collapse
Affiliation(s)
- Neha Singh
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
| | - Varune R Ramnarine
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Jin H Song
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Ritu Pandey
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Sathish K R Padi
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Molecular Biology and Biophysics, UConn Health Center, Farmington, CT, 06030, USA
| | - Mannan Nouri
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Virginie Olive
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Maxim Kobelev
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Koichi Okumura
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Physiology, University of Arizona, Tucson, AZ, 85724, USA
| | - David McCarthy
- Ribomed Biotechnologies, Inc., 8821N. 7th St. STE 300, Phoenix, AZ, 85020, USA
| | - Michelle M Hanna
- Ribomed Biotechnologies, Inc., 8821N. 7th St. STE 300, Phoenix, AZ, 85020, USA
| | - Piali Mukherjee
- Epigenomics Core Facility, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Belinda Sun
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Benjamin R Lee
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
| | - J Brandon Parker
- Department of Obstetrics and Gynecology, Division of Reproductive Science in Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Debabrata Chakravarti
- Department of Obstetrics and Gynecology, Division of Reproductive Science in Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Noel A Warfel
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Muhan Zhou
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
| | - Jeremiah J Bearss
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
| | - Ewan A Gibb
- Decipher Biosciences, Inc, Vancouver, BC, Canada
| | - Mohammed Alshalalfa
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - R Jefferey Karnes
- Department of Urology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Eric J Small
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Rahul Aggarwal
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Felix Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Yuzhuo Wang
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ralph Buttyan
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Amina Zoubeidi
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Mark Rubin
- Department for BioMedical Research, University of Bern, 3008, Bern, Switzerland
| | - Martin Gleave
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Frank J Slack
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | | | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, 44 Binney St, Boston, MA, 02115, USA
| | - Colin Collins
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
| | - Andrew S Kraft
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA.
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA.
| |
Collapse
|
35
|
Slabáková E, Kahounová Z, Procházková J, Souček K. Regulation of Neuroendocrine-like Differentiation in Prostate Cancer by Non-Coding RNAs. Noncoding RNA 2021; 7:ncrna7040075. [PMID: 34940756 PMCID: PMC8704250 DOI: 10.3390/ncrna7040075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022] Open
Abstract
Neuroendocrine prostate cancer (NEPC) represents a variant of prostate cancer that occurs in response to treatment resistance or, to a much lesser extent, de novo. Unravelling the molecular mechanisms behind transdifferentiation of cancer cells to neuroendocrine-like cancer cells is essential for development of new treatment opportunities. This review focuses on summarizing the role of small molecules, predominantly microRNAs, in this phenomenon. A published literature search was performed to identify microRNAs, which are reported and experimentally validated to modulate neuroendocrine markers and/or regulators and to affect the complex neuroendocrine phenotype. Next, available patients’ expression datasets were surveyed to identify deregulated microRNAs, and their effect on NEPC and prostate cancer progression is summarized. Finally, possibilities of miRNA detection and quantification in body fluids of prostate cancer patients and their possible use as liquid biopsy in prostate cancer monitoring are discussed. All the addressed clinical and experimental contexts point to an association of NEPC with upregulation of miR-375 and downregulation of miR-34a and miR-19b-3p. Together, this review provides an overview of different roles of non-coding RNAs in the emergence of neuroendocrine prostate cancer.
Collapse
|
36
|
Kanayama M, Luo J. Delineating the Molecular Events Underlying Development of Prostate Cancer Variants with Neuroendocrine/Small Cell Carcinoma Characteristics. Int J Mol Sci 2021; 22:12742. [PMID: 34884545 PMCID: PMC8657721 DOI: 10.3390/ijms222312742] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/18/2022] Open
Abstract
The treatment landscape of prostate cancer has changed dramatically following the advent of novel systemic therapies, most of which target the androgen receptor (AR). Agents such as abiraterone, enzalutamide, apalutamide, darolutamide were designed to further suppress androgen receptor signaling following gonadal suppression achieved by first-line androgen deprivation therapies. These potent AR targeting agents are increasingly used in the earlier stages of the disease spectrum with the goal of delaying disease progression and extending survival. Although these therapies are effective in controlling prostate tumors dependent on or addicted to AR signaling, prostate tumors surviving the onslaught of potent treatments may evolve and develop drug resistance. A substantial proportion of treatment failures can be explained by the development of treatment-induced aggressive prostate cancer variants such as neuroendocrine/small cell carcinoma. These emerging disease entities demand detailed characterization and precise definitions. We postulate that these treatment-induced prostate cancer entities should be defined molecularly to overcome the drawbacks associated with the current clinical and pathological definitions. A precise molecular definition conforms with current knowledge on the molecular evolution of this disease entity and will enable early detection and early intervention.
Collapse
Affiliation(s)
- Mayuko Kanayama
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | | |
Collapse
|
37
|
PCK1 regulates neuroendocrine differentiation in a positive feedback loop of LIF/ZBTB46 signalling in castration-resistant prostate cancer. Br J Cancer 2021; 126:778-790. [PMID: 34815524 DOI: 10.1038/s41416-021-01631-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/24/2021] [Accepted: 11/03/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Castration-resistant prostate cancer (CRPC) patients frequently develop neuroendocrine differentiation, with high mortality and no effective treatment. However, the regulatory mechanism that connects neuroendocrine differentiation and metabolic adaptation in response to therapeutic resistance of prostate cancer remain to be unravelled. METHODS By unbiased cross-correlation between RNA-sequencing, database signatures, and ChIP analysis, combining in vitro cell lines and in vivo animal models, we identified that PCK1 is a pivotal regulator in therapy-induced neuroendocrine differentiation of prostate cancer through a LIF/ZBTB46-driven glucose metabolism pathway. RESULTS Upregulation of PCK1 supports cell proliferation and reciprocally increases ZBTB46 levels to promote the expression of neuroendocrine markers that are conducive to the development of neuroendocrine characteristic CRPC. PCK1 and neuroendocrine marker expressions are regulated by the ZBTB46 transcription factor upon activation of LIF signalling. Targeting PCK1 can reduce the neuroendocrine phenotype and decrease the growth of prostate cancer cells in vitro and in vivo. CONCLUSION Our study uncovers LIF/ZBTB46 signalling activation as a key mechanism for upregulating PCK1-driven glucose metabolism and neuroendocrine differentiation of CRPC, which may yield significant improvements in prostate cancer treatment after ADT using PCK1 inhibitors.
Collapse
|
38
|
Taher A, Jensen CT, Yedururi S, Surasi DS, Faria SC, Bathala TK, Mujtaba B, Bhosale P, Wagner-Bartak N, Morani AC. Imaging of Neuroendocrine Prostatic Carcinoma. Cancers (Basel) 2021; 13:5765. [PMID: 34830919 PMCID: PMC8616225 DOI: 10.3390/cancers13225765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 12/27/2022] Open
Abstract
Neuroendocrine prostate cancer (NEPC) is an aggressive subtype of prostate cancer that typically has a high metastatic potential and poor prognosis in comparison to the adenocarcinoma subtype. Although it can arise de novo, NEPC much more commonly occurs as a mechanism of treatment resistance during therapy for conventional prostatic adenocarcinoma, the latter is also termed as castration-resistant prostate cancer (CRPC). The incidence of NEPC increases after hormonal therapy and they represent a challenge, both in the radiological and pathological diagnosis, as well as in the clinical management. This article provides a comprehensive imaging review of prostatic neuroendocrine tumors.
Collapse
Affiliation(s)
- Ahmed Taher
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holocombe Blvd., Houston, TX 77030, USA; (A.T.); (C.T.J.); (S.Y.); (S.C.F.); (T.K.B.); (B.M.); (P.B.); (N.W.-B.)
| | - Corey T. Jensen
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holocombe Blvd., Houston, TX 77030, USA; (A.T.); (C.T.J.); (S.Y.); (S.C.F.); (T.K.B.); (B.M.); (P.B.); (N.W.-B.)
| | - Sireesha Yedururi
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holocombe Blvd., Houston, TX 77030, USA; (A.T.); (C.T.J.); (S.Y.); (S.C.F.); (T.K.B.); (B.M.); (P.B.); (N.W.-B.)
| | - Devaki Shilpa Surasi
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA;
| | - Silvana C. Faria
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holocombe Blvd., Houston, TX 77030, USA; (A.T.); (C.T.J.); (S.Y.); (S.C.F.); (T.K.B.); (B.M.); (P.B.); (N.W.-B.)
| | - Tharakeshwar K. Bathala
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holocombe Blvd., Houston, TX 77030, USA; (A.T.); (C.T.J.); (S.Y.); (S.C.F.); (T.K.B.); (B.M.); (P.B.); (N.W.-B.)
| | - Bilal Mujtaba
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holocombe Blvd., Houston, TX 77030, USA; (A.T.); (C.T.J.); (S.Y.); (S.C.F.); (T.K.B.); (B.M.); (P.B.); (N.W.-B.)
| | - Priya Bhosale
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holocombe Blvd., Houston, TX 77030, USA; (A.T.); (C.T.J.); (S.Y.); (S.C.F.); (T.K.B.); (B.M.); (P.B.); (N.W.-B.)
| | - Nicolaus Wagner-Bartak
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holocombe Blvd., Houston, TX 77030, USA; (A.T.); (C.T.J.); (S.Y.); (S.C.F.); (T.K.B.); (B.M.); (P.B.); (N.W.-B.)
| | - Ajaykumar C. Morani
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holocombe Blvd., Houston, TX 77030, USA; (A.T.); (C.T.J.); (S.Y.); (S.C.F.); (T.K.B.); (B.M.); (P.B.); (N.W.-B.)
| |
Collapse
|
39
|
Wang Q, Li Z, Yang J, Peng S, Zhou Q, Yao K, Cai W, Xie Z, Qin F, Li H, Chen X, Li K, Huang H. Loss of NEIL3 activates radiotherapy resistance in the progression of prostate cancer. Cancer Biol Med 2021; 19:j.issn.2095-3941.2020.0550. [PMID: 34591415 PMCID: PMC9425180 DOI: 10.20892/j.issn.2095-3941.2020.0550] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE To explore the genetic changes in the progression of castration-resistant prostate cancer (CRPC) and neuroendocrine prostate cancer (NEPC) and the reason why these cancers resist existing therapies. METHODS We employed our CRPC cell line microarray and other CRPC or NEPC datasets to screen the target gene NEIL3. Lentiviral transfection and RNA interference were used to construct overexpression and knockdown cell lines. Cell and animal models of radiotherapy were established by using a medical electron linear accelerator. Flow cytometry was used to detect apoptosis or cell cycle progression. Western blot and qPCR were used to detect changes in the protein and RNA levels. RESULTS TCGA and clinical patient datasets indicated that NEIL3 was downregulated in CRPC and NEPC cell lines, and NEIL3 was correlated with a high Gleason score but a good prognosis. Further functional studies demonstrated that NEIL3 had no effect on the proliferation and migration of PCa cells. However, cell and animal radiotherapy models revealed that NEIL3 could facilitate the radiotherapy sensitivity of PCa cells, while loss of NEIL3 activated radiotherapy resistance. Mechanistically, we found that NEIL3 negatively regulated the expression of ATR, and higher NEIL3 expression repressed the ATR/CHK1 pathway, thus regulating the cell cycle. CONCLUSIONS We demonstrated that NEIL3 may serve as a diagnostic or therapeutic target for therapy-resistant patients.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Department of Pathology, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zean Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jin Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Department of Radiation Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shirong Peng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Qianghua Zhou
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Kai Yao
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Wenli Cai
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Zhongqiu Xie
- Department of Pathology, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Fujun Qin
- Department of Pathology, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Hui Li
- Department of Pathology, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Xu Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Kaiwen Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China
| |
Collapse
|
40
|
Wang Y, Wang Y, Ci X, Choi SYC, Crea F, Lin D, Wang Y. Molecular events in neuroendocrine prostate cancer development. Nat Rev Urol 2021; 18:581-596. [PMID: 34290447 PMCID: PMC10802813 DOI: 10.1038/s41585-021-00490-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2021] [Indexed: 02/07/2023]
Abstract
Neuroendocrine prostate cancer (NEPC) is a lethal subtype of prostate cancer. NEPC arises de novo only rarely; the disease predominantly develops from adenocarcinoma in response to drug-induced androgen receptor signalling inhibition, although the mechanisms behind this transdifferentiation are a subject of debate. The survival of patients with NEPC is poor, and few effective treatment options are available. To improve clinical outcomes, understanding of the biology and molecular mechanisms regulating NEPC development is crucial. Various NEPC molecular drivers make temporal contributions during NEPC development, and despite the limited treatment options available, several novel targeted therapeutics are currently under research.
Collapse
Affiliation(s)
- Yong Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yu Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Xinpei Ci
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Stephen Y C Choi
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Francesco Crea
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Dong Lin
- Vancouver Prostate Centre, Vancouver, BC, Canada.
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada.
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada.
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada.
| |
Collapse
|
41
|
Park SH, Fong KW, Mong E, Martin MC, Schiltz GE, Yu J. Going beyond Polycomb: EZH2 functions in prostate cancer. Oncogene 2021; 40:5788-5798. [PMID: 34349243 PMCID: PMC8487936 DOI: 10.1038/s41388-021-01982-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023]
Abstract
The Polycomb group (PcG) protein Enhancer of Zeste Homolog 2 (EZH2) is one of the three core subunits of the Polycomb Repressive Complex 2 (PRC2). It harbors histone methyltransferase activity (MTase) that specifically catalyze histone 3 lysine 27 (H3K27) methylation on target gene promoters. As such, PRC2 are epigenetic silencers that play important roles in cellular identity and embryonic stem cell maintenance. In the past two decades, mounting evidence supports EZH2 mutations and/or over-expression in a wide array of hematological cancers and solid tumors, including prostate cancer. Further, EZH2 is among the most upregulated genes in neuroendocrine prostate cancers, which become abundant due to the clinical use of high-affinity androgen receptor pathway inhibitors. While numerous studies have reported epigenetic functions of EZH2 that inhibit tumor suppressor genes and promote tumorigenesis, discordance between EZH2 and H3K27 methylation has been reported. Further, enzymatic EZH2 inhibitors have shown limited efficacy in prostate cancer, warranting a more comprehensive understanding of EZH2 functions. Here we first review how canonical functions of EZH2 as a histone MTase are regulated and describe the various mechanisms of PRC2 recruitment to the chromatin. We further outline non-histone substrates of EZH2 and discuss post-translational modifications to EZH2 itself that may affect substrate preference. Lastly, we summarize non-canonical functions of EZH2, beyond its MTase activity and/or PRC2, as a transcriptional cofactor and discuss prospects of its therapeutic targeting in prostate cancer.
Collapse
Affiliation(s)
- Su H Park
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ka-Wing Fong
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Ezinne Mong
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - M Cynthia Martin
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Gary E Schiltz
- Department of Chemistry, Northwestern University, Evanston, IL, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Jindan Yu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
42
|
Berchuck JE, Viscuse PV, Beltran H, Aparicio A. Clinical considerations for the management of androgen indifferent prostate cancer. Prostate Cancer Prostatic Dis 2021; 24:623-637. [PMID: 33568748 PMCID: PMC8353003 DOI: 10.1038/s41391-021-00332-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/16/2020] [Accepted: 01/20/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND Many systemic therapies for advanced prostate cancer work by disrupting androgen receptor signaling. Androgen indifferent prostate cancer (AIPC) variants, including aggressive variant prostate cancer (AVPC), neuroendocrine prostate cancer (NEPC), and double-negative prostate cancer (DNPC), are increasingly common and often overlapping resistance phenotypes following treatment with androgen receptor signaling inhibitors in men with metastatic castration-resistant prostate cancer and are associated with poor outcomes. Understanding the underlying biology and identifying effective therapies for AIPC is paramount for improving survival for men with prostate cancer. METHODS In this review, we summarize the current knowledge on AIPC variants, including our current understanding of the clinical, morphologic, and molecular features as well as current therapeutic approaches. We also explore emerging therapies and biomarkers aimed at improving outcomes for men with AIPC. RESULTS AND CONCLUSIONS Establishing consensus definitions, developing novel biomarkers for early and accurate detection, further characterization of molecular drivers of each phenotype, and developing effective therapies will be critical to improving outcomes for men with AIPC. Significant progress has been made toward defining the clinical and molecular characteristics of AVPC, NEPC, and DNPC. Novel diagnostic approaches, including cell-free DNA, circulating tumor cells, and molecular imaging are promising tools for detecting AIPC in clinical practice. Building on previous treatment advances, several clinical trials are underway evaluating novel therapeutic approaches in patients with AIPC informed by an understanding of variant-specific biology. In this review, we discuss how these recent and ongoing studies will help to improve diagnosis, prognosis, and therapy for men with AIPC.
Collapse
Affiliation(s)
- Jacob E Berchuck
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Paul V Viscuse
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Ana Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
43
|
AbdullGaffar B, Hotait H. The value of intercluster single cell streaks, triplet signet ring cells, and a two-cell population in urine small cell neuroendocrine carcinoma. Cytopathology 2021; 32:700-704. [PMID: 34029408 DOI: 10.1111/cyt.12997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/10/2021] [Accepted: 04/26/2021] [Indexed: 01/02/2023]
Affiliation(s)
| | - Hassan Hotait
- Cytology Unit, Dubai Hospital, Dubai, United Arab Emirates
| |
Collapse
|
44
|
Bell PD, Huber AR, Agostini-Vulaj D. Clinicopathologic features of metastatic small cell carcinoma of the prostate to the liver: a series of four cases. Diagn Pathol 2021; 16:35. [PMID: 33892760 PMCID: PMC8067396 DOI: 10.1186/s13000-021-01096-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 04/13/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Small cell neuroendocrine carcinoma of the prostate (SCNECP) is a rare, aggressive subtype of prostate carcinoma. Most SCNECP arise from conventional prostate adenocarcinoma (CPAC) treated with androgen deprivation therapy (ADT). CASE PRESENTATIONS We identified four cases of CPAC treated with ADT, which evolved to SCNECP with liver metastasis. The average interval between the diagnosis of CPAC and SCNECP was 102 months (range: 12 to 168). Histologically, the tumors showed nests of cells with high nuclear:cytoplasmic ratios, granular chromatin, and frequent mitoses. All cases were synaptophysin, chromogranin, and AE1/AE3 positive, with a Ki-67 labeling index ≥70%. NKX3.1 was negative in all but one case and TTF-1 was positive in half. Weak ERG positivity by IHC was seen in one case which also demonstrated the TMPRSS2-ERG gene rearrangement; all other cases were negative for ERG by IHC. Serum prostate specific antigen (PSA) levels were normal to near-normal in all. The median interval between the diagnosis of SCNECP and death was 3.25 months (range: 0.75 to 26). CONCLUSIONS Our case series highlights the importance of considering a prostate primary, even in the setting of normal PSA levels and loss of prostate markers, when diagnosing neuroendocrine carcinoma in the liver. Further, we emphasize the significance of diagnosing SCNECP that metastasizes to the liver, as it portends a particularly dismal prognosis.
Collapse
Affiliation(s)
- Phoenix D. Bell
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642 USA
| | - Aaron R. Huber
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642 USA
| | - Diana Agostini-Vulaj
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642 USA
| |
Collapse
|
45
|
Undesirable Status of Prostate Cancer Cells after Intensive Inhibition of AR Signaling: Post-AR Era of CRPC Treatment. Biomedicines 2021; 9:biomedicines9040414. [PMID: 33921329 PMCID: PMC8069212 DOI: 10.3390/biomedicines9040414] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 12/19/2022] Open
Abstract
Recent advances in prostate cancer (PC) research unveiled real androgen receptor (AR) functions in castration-resistant PC (CRPC). Moreover, AR still accelerates PC cell proliferation via the activation of several mechanisms (e.g., mutation, variants, and amplifications in CRPC). New-generation AR signaling-targeted agents, inhibiting extremely the activity of AR, were developed based on these incontrovertible mechanisms of AR-induced CRPC progression. However, long-term administration of AR signaling-targeted agents subsequently induces the major problem that AR (complete)-independent CRPC cells present neither AR nor prostate-specific antigen, including neuroendocrine differentiation as a subtype of AR-independent CRPC. Moreover, there are few treatments effective for AR-independent CRPC with solid evidence. This study focuses on the transformation mechanisms of AR-independent from AR-dependent CRPC cells and potential treatment strategy for AR-independent CRPC and discusses them based on a review of basic and clinical literature.
Collapse
|
46
|
Galosi AB, Palagonia E, Scarcella S, Cimadamore A, Lacetera V, Delle Fave RF, Antezza A, Dell'Atti L. Detection limits of significant prostate cancer using multiparametric MR and digital rectal examination in men with low serum PSA: Up-date of the Italian Society of Integrated Diagnostic in Urology. ACTA ACUST UNITED AC 2021; 93:92-100. [PMID: 33754619 DOI: 10.4081/aiua.2021.1.92] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 11/22/2022]
Abstract
Reasons why significant prostate cancer is still missed in early stage were investigated at the 22nd National SIEUN (Italian Society of integrated diagnostic in Urology, Andrology, Nephrology) congress took place from 30th November to 1st December 2020, in virtual modality. Even if multiparametric magnetic resonance (MR) has been introduced in the clinical practice several, limitations are emerging in patient with regular digital rectal examination (DRE) and serum prostate specific antigen (PSA) levels approaching the normal limits. The present paper summarizes highlights observed in those cases where significant prostate cancer may be missed by PSA or imaging and DRE. The issue of multidisciplinary interest had been subdivided and deepened under four main topics: biochemical, clinical, pathological and radiological point of view with a focus on PI-RADS 3 lesions.
Collapse
Affiliation(s)
- Andrea B Galosi
- Division of Urology, School of Medicine, Università Politecnica delle Marche, Ancona.
| | - Erika Palagonia
- Division of Urology, School of Medicine, Università Politecnica delle Marche, Ancona.
| | - Simone Scarcella
- Division of Urology, School of Medicine, Università Politecnica delle Marche, Ancona.
| | - Alessia Cimadamore
- Division of Pathology, School of Medicine, Università Politecnica delle Marche, Ancona.
| | - Vito Lacetera
- Division of Urology, Azienda Ospedaliera Marche Nord, Pesaro.
| | - Rocco F Delle Fave
- Division of Urology, School of Medicine, Università Politecnica delle Marche, Ancona.
| | - Angelo Antezza
- Division of Urology, School of Medicine, Università Politecnica delle Marche, Ancona.
| | - Lucio Dell'Atti
- Division of Urology, School of Medicine, Università Politecnica delle Marche, Ancona.
| |
Collapse
|
47
|
Kryvenko ON. Small Cell-like Change in Central Zone Histology-A New Observation Mimicking Cribriform Intraductal Prostatic Adenocarcinoma. Int J Surg Pathol 2021; 29:635-637. [PMID: 33729852 DOI: 10.1177/10668969211003966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A small cell-like change in prostate has been described in high-grade prostatic intraepithelial neoplasia (PIN), intraductal prostatic adenocarcinoma, and invasive prostate cancer. It occurs when these processes have a cribriform architecture. To date, small cell-like change has not been described in benign glands. Herein, I describe such a change in cribriform central zone histology from a radical prostatectomy with a spatially remote treatment naïve Grade Group 3 prostate cancer. The cancer did not have cribriform morphology or intraductal prostatic adenocarcinoma. The small cell-like change was positive for racemase in PIN-4 cocktail and no nuclei were highlighted by Ki-67. This is the first report of a small cell-like change in benign prostate tissue. Although rare, such finding in cribriform architecture of central zone histology can potentially be misinterpreted as a neoplastic process.
Collapse
|
48
|
Aggarwal R, Romero GR, Friedl V, Weinstein A, Foye A, Huang J, Feng F, Stuart JM, Small EJ. Clinical and genomic characterization of Low PSA Secretors: a unique subset of metastatic castration resistant prostate cancer. Prostate Cancer Prostatic Dis 2021; 24:81-87. [PMID: 32286548 DOI: 10.1038/s41391-020-0228-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/29/2020] [Accepted: 03/20/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Metastatic disease burden out of proportion to serum PSA has been used as a marker of aggressive phenotype prostate cancer but is not well defined as a distinct subgroup. We sought to prospectively characterize the molecular features and clinical outcomes of Low PSA Secretors. METHODS Eligible metastatic castration resistant prostate cancer (mCRPC) patients without prior small cell histology underwent metastatic tumor biopsy with molecular characterization. Low PSA secretion was defined as serum PSA < 2, 5, or 10 ng/mL plus >5 metastases with radiographic progression at study entry. Clinical and molecular features were compared between low PSA vs. normal secretors in a post-hoc fashion. RESULTS 183 patients were enrolled, including 15 (8%) identified as Low PSA Secretors using optimal PSA cut point of 5 ng/mL. Biopsies from Low PSA Secretors demonstrated higher t-SCNC and RB1 loss and lower AR transcriptional signature scores compared with normal secretors. Genomic loss of RB1 and/or TP53 was more common in Low PSA Secretors (80% vs. 41%). Overall survival (OS) was shorter in Low PSA Secretors (median OS = 26.7 vs. 46.0 months, hazard ratio = 2.465 (95% CI: 0.982-6.183). Progression-free survival (PFS) on post-biopsy treatment with AR-targeted therapy was shorter than with chemotherapy (median PFS 6.2 vs. 4.1 months). CONCLUSIONS Low PSA secretion in relation to metastatic tumor burden may be a readily available clinical selection tool for de-differentiated mCRPC with molecular features consistent with t-SCNC. Prospective validation is warranted.
Collapse
Affiliation(s)
- Rahul Aggarwal
- University of California San Francisco, San Francisco, CA, USA.
| | | | - Verena Friedl
- University of California Santa Cruz, Santa Cruz, CA, USA
| | | | - Adam Foye
- University of California San Francisco, San Francisco, CA, USA
| | | | - Felix Feng
- University of California San Francisco, San Francisco, CA, USA
| | | | - Eric J Small
- University of California Santa Cruz, Santa Cruz, CA, USA
| |
Collapse
|
49
|
Cheng WC, Wang HJ. Current advances of targeting epigenetic modifications in neuroendocrine prostate cancer. Tzu Chi Med J 2021; 33:224-232. [PMID: 34386358 PMCID: PMC8323647 DOI: 10.4103/tcmj.tcmj_220_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/05/2020] [Accepted: 10/06/2020] [Indexed: 11/15/2022] Open
Abstract
Neuroendocrine prostate cancer (NEPC) is the most lethal malignancy of prostate cancer (PCa). Treatment with next-generation androgen receptor (AR) pathway inhibitors (ARPIs) has successfully extended patients' lifespan. However, with the emergence of drug resistance, PCa tumors increasingly adapt to potent ARPI therapies by transitioning to alternative cellular lineage. Such therapy-induced drug resistance is largely driven from the cellular plasticity of PCa cells to alter their phenotypes of AR independence for cell growth and survival. Some of the resistant PCa cells undergo cellular reprogramming to form neuroendocrine phenotypes. Recent evidences suggest that this cellular reprogramming or the lineage plasticity is driven by dysregulation of the epigenome and transcriptional networks. Aberrant DNA methylation and altered expression of epigenetic modifiers, such as enhancer of zeste-homolog 2, transcription factors, histone demethylases, are hallmarks of NEPC. In this review, we discuss the nature of the epigenetic and transcriptional landscapes of PCa cells which lose their AR independence and transition to the neuroendocrine lineage. We also discuss how oncogenic signaling and metabolic reprogramming fuel epigenetic and transcriptional alterations. In addition, the current state of epigenetic therapies for NEPC is addressed.
Collapse
Affiliation(s)
- Wen-Chi Cheng
- SDGs Teaching and Research Headquarters, Tzu Chi University, Hualien, Taiwan
| | - Hung-Jung Wang
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,Doctoral Degree Program in Translational Medicine, Tzu Chi University and Academia Sinica, Hualien, Taiwan
| |
Collapse
|
50
|
Butler W, Huang J. Neuroendocrine cells of the prostate: Histology, biological functions, and molecular mechanisms. PRECISION CLINICAL MEDICINE 2021; 4:25-34. [PMID: 33842835 PMCID: PMC8023015 DOI: 10.1093/pcmedi/pbab003] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/23/2021] [Accepted: 01/24/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is a common cause of cancer-related mortality in men worldwide. Although most men are diagnosed with low grade, indolent tumors that are potentially curable, a significant subset develops advanced disease where hormone therapy is required to target the androgen receptor (AR). Despite its initial effect, hormone therapy eventually fails and the tumor progresses to lethal stages even through continued inhibition of AR. This review article focuses on the role of PCa cellular heterogeneity in therapy resistance and disease progression. Although AR-positive luminal-type cells represent the vast majority of PCa cells, there exists a minor component of AR-negative neuroendocrine (NE) cells that are resistant to hormonal therapy and are enriched by the treatment. In addition, it is now well accepted that a significant subset of hormonally treated tumors recur as small cell neuroendocrine carcinoma (SCNC), further highlighting the importance of targeting NE cells in addition to the more abundant luminal-type cancer cells. Although it has been long recognized that NE cells are present in PCa, their underlying function in benign prostate and molecular mechanisms contributing to PCa progression remains poorly understood. In this article, we review the morphology and function of NE cells in benign prostate and PCa as well as underlying molecular mechanisms. In addition, we review the major reported mechanisms for transformation from common adenocarcinoma histology to the highly lethal SCNC, a significant clinical challenge in the management of advanced PCa.
Collapse
Affiliation(s)
- William Butler
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|