1
|
Saeed MA, Peng B, Kim K, Rawat K, Kuehm LM, Siegel ZR, Borkowski A, Habib N, Van Tine B, Sheikh N, Tuyen V, Thorek DLJ, Fehniger TA, Pachynski RK. High-Dimensional Analyses Reveal IL15 Enhances Activation of Sipuleucel-T Lymphocyte Subsets and Reverses Immunoresistance. Cancer Immunol Res 2024; 12:559-574. [PMID: 38407894 DOI: 10.1158/2326-6066.cir-23-0652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/21/2023] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
Sipuleucel-T (sip-T) is the only FDA-approved autologous cellular immunotherapy for metastatic castration-resistant prostate cancer (mCRPC). To elucidate parameters of the response profile to this therapy, we report high-dimensional analyses of sip-T using cytometry by time of flight (CyTOF) and show a lymphoid predominance, with CD3+ T cells constituting the highest proportion (median ∼60%) of sip-T, followed by B cells, and natural killer (NK) and NKT cells. We hypothesized that treatment of sip-T with homeostatic cytokines known to activate/expand effector lymphocytes could augment efficacy against prostate tumors. Of the cytokines tested, IL15 was the most effective at enhancing activation and proliferation of effector lymphocytes, as well as augmenting tumor cytotoxicity in vitro. Co-culture of sip-T with IL15 and control or prostate-relevant antigens showed substantial activation and expansion of CD8+ T cells and NKT cells in an antigen-specific manner. Adoptive transfer of IL15-treated sip-T into NSG mice resulted in more potent prostate tumor growth inhibition compared with control sip-T. Evaluation of tumor-infiltrating lymphocytes revealed a 2- to 14-fold higher influx of sip-T and a significant increase in IFNγ producing CD8+ T cells and NKT cells within the tumor microenvironment in the IL15 group. In conclusion, we put forward evidence that IL15 treatment can enhance the functional antitumor immunity of sip-T, providing rationale for combining IL15 or IL15 agonists with sip-T to treat patients with mCRPC.
Collapse
Affiliation(s)
- Muhammad A Saeed
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Bo Peng
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Kevin Kim
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Kavita Rawat
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Lindsey M Kuehm
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Zoe R Siegel
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Ariel Borkowski
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Nabih Habib
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Brian Van Tine
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
| | | | - Vu Tuyen
- Dendreon Pharmaceuticals LLC, Seattle, Washington
| | - Daniel L J Thorek
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
- Bursky Center for Human Immunology and Immunotherapy, Washington University School of Medicine, St Louis, Missouri
| | - Russell K Pachynski
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
- Bursky Center for Human Immunology and Immunotherapy, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
2
|
Zhanghuang C, Zhu J, Li Y, Wang J, Ma J, Li L, Yao Z, Ji F, Wu C, Tang H, Xie Y, Yan B, Yang Z. Prognostic significance of surgery and radiotherapy in elderly patients with localized prostate cancer: establishing and time-based external validation a nomogram from SEER-based study. BMC Urol 2024; 24:12. [PMID: 38184526 PMCID: PMC10771675 DOI: 10.1186/s12894-023-01384-6] [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: 02/02/2023] [Accepted: 11/28/2023] [Indexed: 01/08/2024] Open
Abstract
OBJECTIVE Prostate cancer (PC) is a significant disease affecting men's health worldwide. More than 60% of patients over 65 years old and more than 80% are diagnosed with localized PC. The current choice of treatment modalities for localized PC and whether overtreatment is controversial. Therefore, we wanted to construct a nomogram to predict the risk factors associated with cancer-specific survival (CSS) and overall survival (OS) in elderly patients with localized PC while assessing the survival differences in surgery and radiotherapy for elderly patients with localized PC. METHODS Data of patients with localized PC over 65 years were obtained from the Surveillance, Epidemiology, and End Results (SEER) database. Univariate and multivariate Cox regression models were used to determine independent risk factors for CSS and OS. Nomograms predicting CSS and OS were built using multivariate Cox regression models. The consistency index (C-index), the area under the subject operating characteristic curve (AUC), and the calibration curve were used to test the accuracy and discrimination of the prediction model. Decision curve analysis (DCA) was used to test the potential clinical value of this model. RESULTS A total of 90,434 patients over 65 years and diagnosed with localized PC from 2010 to 2018 were included in the study. All patients were randomly assigned to the training set (n = 63,328) and the validation set (n = 27,106). Univariate and multivariate Cox regression model analysis showed that age, race, marriage, T stage, surgical, radiotherapy, prostate-specific antigen (PSA), and Gleason score (GS) were independent risk factors for predicting CSS in elderly patients with localized PC. Age, race, marriage, surgery, radiotherapy, PSA, and GS were independent risk factors for predicting OS in elderly patients with localized PC. The c-index of the training and validation sets for the predicted CSS is 0.802(95%CI:0.788-0.816) and 0.798(95%CI:0.776-0.820, respectively). The c-index of the training and validation sets for predicting OS is 0.712(95%:0.704-0.720) and 0.724(95%:0.714-0.734). It shows that the nomograms have excellent discriminatory ability. The AUC and the calibration curves also show good accuracy and discriminability. CONCLUSION We have developed new nomograms to predict CSS and OS in elderly patients with localized PC. After internal validation and external temporal validation with reasonable accuracy, reliability and potential clinical value, the model can be used for clinically assisted decision-making.
Collapse
Affiliation(s)
- Chenghao Zhanghuang
- Department of Urology, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University), Kunming, People's Republic of China
- Department of Oncology; Yunnan Children solid Tumor Treatment Center, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University), Kunming Children's Solid Tumor Diagnosis and Treatment Center, Kunming, People's Republic of China
- Yunnan Key Laboratory of Children's Major Disease Research, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University); Yunnan Province Clinical Research Center for Children's Health and Disease, Kunming Children's Solid Tumor Diagnosis and Treatment Center, Yunnan Clinical Medical Center for Pediatric Diseases, Kunming, People's Republic of China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation base of Child development and Critical Disorders; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400015, China
| | - Jianjun Zhu
- Department of Oncology; Yunnan Children solid Tumor Treatment Center, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University), Kunming Children's Solid Tumor Diagnosis and Treatment Center, Kunming, People's Republic of China
| | - Ye Li
- Department of Oncology; Yunnan Children solid Tumor Treatment Center, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University), Kunming Children's Solid Tumor Diagnosis and Treatment Center, Kunming, People's Republic of China
| | - Jinkui Wang
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation base of Child development and Critical Disorders; Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400015, China
| | - Jing Ma
- Yunnan Key Laboratory of Children's Major Disease Research, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University); Yunnan Province Clinical Research Center for Children's Health and Disease, Kunming Children's Solid Tumor Diagnosis and Treatment Center, Yunnan Clinical Medical Center for Pediatric Diseases, Kunming, People's Republic of China
- Department of Otolaryngology, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University), Kunming, People's Republic of China
| | - Li Li
- Yunnan Key Laboratory of Children's Major Disease Research, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University); Yunnan Province Clinical Research Center for Children's Health and Disease, Kunming Children's Solid Tumor Diagnosis and Treatment Center, Yunnan Clinical Medical Center for Pediatric Diseases, Kunming, People's Republic of China
| | - Zhigang Yao
- Department of Urology, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University), Kunming, People's Republic of China
| | - Fengming Ji
- Department of Urology, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University), Kunming, People's Republic of China
| | - Chengchuang Wu
- Department of Urology, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University), Kunming, People's Republic of China
| | - Haoyu Tang
- Department of Urology, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University), Kunming, People's Republic of China
| | - Yucheng Xie
- Department of Pathology, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University), Kunming, People's Republic of China
| | - Bing Yan
- Department of Urology, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University), Kunming, People's Republic of China.
- Yunnan Key Laboratory of Children's Major Disease Research, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University); Yunnan Province Clinical Research Center for Children's Health and Disease, Kunming Children's Solid Tumor Diagnosis and Treatment Center, Yunnan Clinical Medical Center for Pediatric Diseases, Kunming, People's Republic of China.
| | - Zhen Yang
- Department of Oncology; Yunnan Children solid Tumor Treatment Center, Kunming Children's Hospital (Children's Hospital affiliated to Kunming Medical University), Kunming Children's Solid Tumor Diagnosis and Treatment Center, Kunming, People's Republic of China.
| |
Collapse
|
3
|
Choi SW, Sun AK, Cheung JPY, Ho JCY. Circulating Tumour Cells in the Prediction of Bone Metastasis. Cancers (Basel) 2024; 16:252. [PMID: 38254743 PMCID: PMC10813668 DOI: 10.3390/cancers16020252] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Bone is the most common organ for the development of metastases in many primary tumours, including those of the breast, prostate and lung. In most cases, bone metastasis is incurable, and treatment is predominantly palliative. Much research has focused on the role of Circulating Tumour Cells (CTCs) in the mechanism of metastasis to the bone, and methods have been developed to isolate and count CTCs from peripheral blood. Several methods are currently being used in the study of CTCs, but only one, the CellSearchTM system has been approved by the United States Food and Drug Administration for clinical use. This review summarises the advantages and disadvantages, and outlines which clinical studies have used these methods. Studies have found that CTC numbers are predictive of bone metastasis in breast, prostate and lung cancer. Further work is required to incorporate information on CTCs into current staging systems to guide treatment in the prevention of tumour progression into bone.
Collapse
Affiliation(s)
- Siu-Wai Choi
- Department of Orthopaedics and Tramatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Aria Kaiyuan Sun
- Department of Anaesthesiology, School of Clinical Medicine, Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (A.K.S.); (J.C.-Y.H.)
| | - Jason Pui-Yin Cheung
- Department of Orthopaedics and Tramatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jemmi Ching-Ying Ho
- Department of Anaesthesiology, School of Clinical Medicine, Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (A.K.S.); (J.C.-Y.H.)
| |
Collapse
|
4
|
Ślusarczyk A, Baboudjian M, Zapała P, Yanagisawa T, Miszczyk M, Chlosta M, Krumpoeck P, Moschini M, Gandaglia G, Ploussard G, Rivas JG, Życzkowski M, Karakiewicz PI, Radziszewski P, Leapman MS, Shariat SF, Rajwa P. Survival outcomes of patients treated with local therapy for nonmetastatic prostate cancer with high prostate-specific antigen concentrations. Prostate 2023; 83:1504-1515. [PMID: 37545342 DOI: 10.1002/pros.24609] [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: 04/21/2023] [Revised: 06/25/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Patients with nonmetastatic prostate cancer (nmPCa) and high prostate-specific antigen (PSA) levels due to the high likelihood of metastasis pose a clinical dilemma regarding their optimal treatment and long-term outcomes after initial local therapy. We aimed to evaluate the oncologic outcomes of patients treated with radical prostatectomy (RP) or radiotherapy (RT) for nmPCa with high PSA levels. METHODS We queried the Surveillance, Epidemiology, and End Results (SEER) database to identify patients diagnosed with nmPCa who received RP or RT from 2004 through 2015. We included nmPCa patients with high PSA levels categorized as ≥50 and ≥98 ng/mL, the highest level recorded in SEER. We used the Kaplan-Meier method and Cox proportional hazards to analyze cancer-specific (CSS) and overall survival (OS). RESULTS We included 6177 patients with nmPCa and PSA ≥ 50 ng/mL at diagnosis; 1698 (27%) had PSA ≥ 98 ng/mL. Of these, 1658 (26.8%) underwent RP and 4519 (73.16%) patients received primary RT. Within a median of 113 months (interquartile range 74-150 months), the 5- and 10-year CSS estimates were 92.3% and 81.5% respectively; 10-year OS was 61%. In the PSA ≥ 98 ng/mL subgroup 5- and 10-year CSS estimates were 89.2% and 76%, respectively. In multivariable analyses for CSS, ISUP grade group (p < 0.001), N stage (p < 0.001), treatment with RP (hazard ratio [HR] = 0.60, 95% confidence interval [CI] 0.43-0.83, p < 0.001), and patient's age (p < 0.05) were associated with improved CSS. In the whole cohort of patients with PSA ≥ 50 ng/mL and RP subgroup, PSA failed to retain its independent prognostic value for CSS. CONCLUSIONS Patients treated with local therapy for nmPCa with very high PSA at diagnosis have relatively good long-term oncological outcomes. Therefore, among well-selected patients with nmPCa, high PSA levels alone should not preclude the use of radical local therapy. Potential selection bias limits inferences about the relative effectiveness of specific local therapies in this setting.
Collapse
Affiliation(s)
- Aleksander Ślusarczyk
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, Warsaw, Poland
| | | | - Piotr Zapała
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, Warsaw, Poland
| | - Takafumi Yanagisawa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Marcin Miszczyk
- IIIrd Radiotherapy and Chemotherapy Department, Maria Skłodowska-Curie National Research Institute of Oncology, Warszawa, Poland
| | - Marcin Chlosta
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Urology, Jagiellonian University in Cracow, Kraków, Poland
| | - Paul Krumpoeck
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Marco Moschini
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giorgio Gandaglia
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Juan G Rivas
- Department of Urology, Clinico San Carlos Hospital, Madrid, Spain
| | - Marcin Życzkowski
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Pierre I Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, University of Montreal Health Centre, Montreal, Canada
| | - Piotr Radziszewski
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, Warsaw, Poland
| | - Michael S Leapman
- Department of Urology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Shahrokh F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Weill Cornell Medical College, New York, New York, USA
- Department of Urology, University of Texas Southwestern, Dallas, Texas, USA
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
- Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria
- Department of Urology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Urology, Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan
| | - Paweł Rajwa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| |
Collapse
|
5
|
Wang J, Ben-David R, Mehrazin R, Yang W, Tewari AK, Kyprianou N. Novel signatures of prostate cancer progression and therapeutic resistance. Expert Opin Ther Targets 2023; 27:1195-1206. [PMID: 38108262 DOI: 10.1080/14728222.2023.2293757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION The extensive heterogeneity of prostate cancer (PCa) and multilayered complexity of progression to castration-resistant prostate cancer (CRPC) have contributed to the challenges of accurately monitoring advanced disease. Profiling of the tumor microenvironment with large-scale transcriptomic studies have identified gene signatures that predict biochemical recurrence, lymph node invasion, metastases, and development of therapeutic resistance through critical determinants driving CRPC. AREAS COVERED This review encompasses understanding of the role of different molecular determinants of PCa progression to lethal disease including the phenotypic dynamic of cell plasticity, EMT-MET interconversion, and signaling-pathways driving PCa cells to advance and metastasize. The value of liquid biopsies encompassing circulating tumor cells and extracellular vesicles to detect disease progression and emergence of therapeutic resistance in patients progressing to lethal disease is discussed. Relevant literature was added from PubMed portal. EXPERT OPINION Despite progress in the tumor-targeted therapeutics and biomarker discovery, distant metastasis and therapeutic resistance remain the major cause of mortality in patients with advanced CRPC. No single signature can encompass the tremendous phenotypic and genomic heterogeneity of PCa, but rather multi-threaded omics-derived and phenotypic markers tailored and validated into a multimodal signature.
Collapse
Affiliation(s)
- Jason Wang
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Reuben Ben-David
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Reza Mehrazin
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wei Yang
- Department of Pathology, Stony Brook University, New York, NY, USA
| | - Ashutosh K Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natasha Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology & Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
6
|
Farré X, Mousavi SM, Marcos-Gragera R, Malaker K. A Global Perspective on the Controversy of Gleason Score 6 Prostate Cancer Reporting: The Potential Role of Population-Based Cancer Registries. JCO Glob Oncol 2023; 9:e2200427. [PMID: 37099738 DOI: 10.1200/go.22.00427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023] Open
Affiliation(s)
- Xavier Farré
- Xavier Farré, MD, PhD, Department of Health, Public Health Agency of Catalonia, Lleida, Spain; Seyed Mohsen Mousavi, MD, East Switzerland Cancer Registry, St Gallen, Switzerland; Rafael Marcos-Gragera, MD, PhD, Epidemiology Unit and Girona Cancer Registry, Catalan Institute of Oncology, Girona Biomedical Research Institute Dr Josep Trueta, Girona, Spain; and Kamal Malaker, MD, PhD, North Arc Health Consultancy Services, Singapore, Singapore
| | - Seyed Mohsen Mousavi
- Xavier Farré, MD, PhD, Department of Health, Public Health Agency of Catalonia, Lleida, Spain; Seyed Mohsen Mousavi, MD, East Switzerland Cancer Registry, St Gallen, Switzerland; Rafael Marcos-Gragera, MD, PhD, Epidemiology Unit and Girona Cancer Registry, Catalan Institute of Oncology, Girona Biomedical Research Institute Dr Josep Trueta, Girona, Spain; and Kamal Malaker, MD, PhD, North Arc Health Consultancy Services, Singapore, Singapore
| | - Rafael Marcos-Gragera
- Xavier Farré, MD, PhD, Department of Health, Public Health Agency of Catalonia, Lleida, Spain; Seyed Mohsen Mousavi, MD, East Switzerland Cancer Registry, St Gallen, Switzerland; Rafael Marcos-Gragera, MD, PhD, Epidemiology Unit and Girona Cancer Registry, Catalan Institute of Oncology, Girona Biomedical Research Institute Dr Josep Trueta, Girona, Spain; and Kamal Malaker, MD, PhD, North Arc Health Consultancy Services, Singapore, Singapore
| | - Kamal Malaker
- Xavier Farré, MD, PhD, Department of Health, Public Health Agency of Catalonia, Lleida, Spain; Seyed Mohsen Mousavi, MD, East Switzerland Cancer Registry, St Gallen, Switzerland; Rafael Marcos-Gragera, MD, PhD, Epidemiology Unit and Girona Cancer Registry, Catalan Institute of Oncology, Girona Biomedical Research Institute Dr Josep Trueta, Girona, Spain; and Kamal Malaker, MD, PhD, North Arc Health Consultancy Services, Singapore, Singapore
| |
Collapse
|
7
|
Wang JJ, Sun N, Lee YT, Kim M, Vagner T, Rohena-Rivera K, Wang Z, Chen Z, Zhang RY, Lee J, Zhang C, Tang H, Widjaja J, Zhang TX, Qi D, Teng PC, Jan YJ, Hou KC, Hamann C, Sandler HM, Daskivich TJ, Luthringer DJ, Bhowmick NA, Pei R, You S, Di Vizio D, Tseng HR, Chen JF, Zhu Y, Posadas EM. Prostate cancer extracellular vesicle digital scoring assay - a rapid noninvasive approach for quantification of disease-relevant mRNAs. NANO TODAY 2023; 48:101746. [PMID: 36711067 PMCID: PMC9879227 DOI: 10.1016/j.nantod.2022.101746] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Optimizing outcomes in prostate cancer (PCa) requires precision in characterization of disease status. This effort was directed at developing a PCa extracellular vesicle (EV) Digital Scoring Assay (DSA) for detecting metastasis and monitoring progression of PCa. PCa EV DSA is comprised of an EV purification device (i.e., EV Click Chip) and reverse-transcription droplet digital PCR that quantifies 11 PCa-relevant mRNA in purified PCa-derived EVs. A Met score was computed for each plasma sample based on the expression of the 11-gene panel using the weighted Z score method. Under optimized conditions, the EV Click Chips outperformed the ultracentrifugation or precipitation method of purifying PCa-derived EVs from artificial plasma samples. Using PCa EV DSA, the Met score distinguished metastatic (n = 20) from localized PCa (n = 20) with an area under the receiver operating characteristic curve of 0.88 (95% CI:0.78-0.98). Furthermore, longitudinal analysis of three PCa patients showed the dynamics of the Met scores reflected clinical behavior even when disease was undetectable by imaging. Overall, a sensitive PCa EV DSA was developed to identify metastatic PCa and reveal dynamic disease states noninvasively. This assay may complement current imaging tools and blood-based tests for timely detection of metastatic progression that can improve care for PCa patients.
Collapse
Affiliation(s)
- Jasmine J. Wang
- Division of Medical Oncology, Department of Medicine,
Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
| | - Na Sun
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of
Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese
Academy of Sciences, Suzhou, PR China
| | - Yi-Te Lee
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
| | - Minhyung Kim
- Department of Biomedical Sciences, Cedars-Sinai Medical
Center, Los Angeles, CA, USA
| | - Tatyana Vagner
- Department of Surgery, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
| | | | - Zhili Wang
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of
Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese
Academy of Sciences, Suzhou, PR China
| | - Zijing Chen
- Division of Medical Oncology, Department of Medicine,
Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ryan Y. Zhang
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
| | - Junseok Lee
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
| | - Ceng Zhang
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
| | - Hubert Tang
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
| | - Josephine Widjaja
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
| | - Tiffany X. Zhang
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
| | - Dongping Qi
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
| | - Pai-Chi Teng
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
| | - Yu Jen Jan
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
| | - Kuan-Chu Hou
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
| | - Candace Hamann
- Division of Medical Oncology, Department of Medicine,
Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Howard M. Sandler
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
- Department of Radiation Oncology, Cedars-Sinai Medical
Center, Los Angeles, CA, USA
| | - Timothy J. Daskivich
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
- Division of Urology, Department of Surgery, Cedars-Sinai
Medical Center, Los Angeles, CA, USA
| | - Daniel J. Luthringer
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
- Department of Pathology and Laboratory Medicine,
Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Neil A. Bhowmick
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical
Center, Los Angeles, CA, USA
- Department of Medicine, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
| | - Renjun Pei
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of
Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese
Academy of Sciences, Suzhou, PR China
| | - Sungyong You
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical
Center, Los Angeles, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
| | - Dolores Di Vizio
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical
Center, Los Angeles, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
- Department of Pathology and Laboratory Medicine,
Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, David Geffen School
of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jie-Fu Chen
- Department of Pathology, Memorial Sloan Kettering Cancer
Center, New York, NY, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for
Molecular Imaging, Department of Molecular and Medical Pharmacology, University of
California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, David Geffen School
of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Edwin M. Posadas
- Division of Medical Oncology, Department of Medicine,
Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los
Angeles, CA, USA
| |
Collapse
|
8
|
The prognosis of lipid reprogramming with the HMG-CoA reductase inhibitor, rosuvastatin, in castrated Egyptian prostate cancer patients: Randomized trial. PLoS One 2022; 17:e0278282. [PMID: 36480560 PMCID: PMC9731457 DOI: 10.1371/journal.pone.0278282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 10/24/2022] [Indexed: 12/13/2022] Open
Abstract
AIM The role of surgical castration and rosuvastatin treatment on lipid profile and lipid metabolism related markers was evaluated for their prognostic significance in metastatic prostate cancer (mPC) patients. METHODS A total of 84 newly diagnosed castrated mPC patients treated with castration were recruited and divided into two groups: Group I served as control (statin non-users) while group II treated with Rosuvastatin (20 mg/day) for 6 months and served as statin users. Prostate specific antigen (PSA), epidermal growth factor receptor (EGFR), Caveolin-1 (CAV1), lipid profile (LDL, HDL, triglycerides (TG) and total cholesterol (TC)) and lipid metabolism related markers (aldoketoreductase (AKR1C4), HMG-CoA reductase (HMGCR), ATP-binding cassette transporter A1 (ABCA1), and soluble low density lipoprotein receptor related protein 1 (SLDLRP1)) were measured at baseline, after 3 and 6 months. Overall survival (OS) was analyzed by Kaplan-Meier and COX regression for prognostic significance. RESULTS Before castration, HMG-CoA reductase was elevated in patients <65 years (P = 0.009). Bone metastasis was associated with high PSA level (P = 0.013), but low HMGCR (P = 0.004). Patients with positive family history for prostate cancer showed high levels of EGFR, TG, TC, LDL, alkaline phosphatase (ALP), but low AKR1C4, SLDLRP1, CAV1 and ABCA-1 levels. Smokers had high CAV1 level (P = 0.017). After 6 months of castration and rosuvastatin administration, PSA, TG, LDL and TC were significantly reduced, while AKR1C4, HMGCR, SLDLRP1, CAV1 and ABCA-1 were significantly increased. Overall survival was reduced in patients with high baseline of SLDLRP1 (>3385 pg/ml, P = 0.001), PSA (>40 ng/ml, P = 0.003) and CAV1 (>4955 pg/ml, P = 0.021). CONCLUSION Results of the current study suggest that the peripheral lipidogenic effects of rosuvastatin may have an impact on the treatment outcome and survival of castrated mPC patients. TRAIL REGISTRATION This trial was registered at the Pan African Clinical Trial Registry with identification number PACTR202102664354163 and at ClinicalTrials.gov with identification number NCT04776889.
Collapse
|
9
|
Yang M, Zhu X, Shen Y, He Q, Qin Y, Shao Y, Yuan L, Ye H. GPX2 predicts recurrence-free survival and triggers the Wnt/β-catenin/EMT pathway in prostate cancer. PeerJ 2022; 10:e14263. [PMID: 36312753 PMCID: PMC9615941 DOI: 10.7717/peerj.14263] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/27/2022] [Indexed: 01/24/2023] Open
Abstract
Objective This study aimed to establish a prognostic model related to prostate cancer (PCa) recurrence-free survival (RFS) and identify biomarkers. Methods The RFS prognostic model and key genes associated with PCa were established using Least Absolute Shrinkage and Selection Operator (LASSO) and Cox regression from the Cancer Genome Atlas (TCGA)-PRAD and the Gene Expression Omnibus (GEO) GSE46602 datasets. The weighted gene co-expression network (WGCNA) was used to analyze the obtained key modules and genes, and gene set enrichment analysis (GSEA) was performed. The phenotype and mechanism were verified in vitro. Results A total of 18 genes were obtained by LASSO regression, and an RFS model was established and verified (TCGA, AUC: 0.774; GSE70768, AUC: 0.759). Three key genes were obtained using multivariate Cox regression. WGCNA analysis obtained the blue module closely related to the Gleason score (cor = -0.22, P = 3.3e - 05) and the unique gene glutathione peroxidase 2 (GPX2). Immunohistochemical analysis showed that the expression of GPX2 was significantly higher in patients with PCa than in patients with benign prostatic hyperplasia (P < 0.05), but there was no significant correlation with the Gleason score (GSE46602 and GSE6919 verified), which was also verified in the GSE46602 and GSE6919 datasets. The GSEA results showed that GPX2 expression was mainly related to the epithelial-mesenchymal transition (EMT) and Wnt pathways. Additionally, GPX2 expression significantly correlated with eight kinds of immune cells. In human PCa cell lines LNCaP and 22RV1, si-GPX2 inhibited proliferation and invasion, and induced apoptosis when compared with si-NC. The protein expression of Wnt3a, glycogen synthase kinase 3β (GSK3β), phosphorylated (p)-GSK3β, β-catenin, p-β-catenin, c-myc, cyclin D1, and vimentin decreased; the expression of E-cadherin increased; and the results for over-GPX2 were opposite to those for over-NC. The protein expression of GPX2 decreased, and β-catenin was unchanged in the si-GPX2+ SKL2001 group compared with the si-NC group. Conclusion We successfully constructed the PCa RFS prognostic model, obtained RFS-related biomarker GPX2, and found that GPX2 regulated PCa progression and triggered Wnt/β-catenin/EMT pathway molecular changes.
Collapse
Affiliation(s)
- Ming Yang
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xudong Zhu
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Shen
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qi He
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuan Qin
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yiqun Shao
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lin Yuan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hesong Ye
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
10
|
Shropshire DB, Acosta FM, Fang K, Benavides J, Sun LZ, Jin VX, Jiang JX. Association of adenosine signaling gene signature with estrogen receptor-positive breast and prostate cancer bone metastasis. Front Med (Lausanne) 2022; 9:965429. [PMID: 36186774 PMCID: PMC9520286 DOI: 10.3389/fmed.2022.965429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/23/2022] [Indexed: 11/20/2022] Open
Abstract
Bone metastasis is a common and devastating consequence of several major cancer types, including breast and prostate. Osteocytes are the predominant bone cell, and through connexin (Cx) 43 hemichannels release ATP to the bone microenvironment that can be hydrolyzed to adenosine. Here, we investigated how genes related to ATP paracrine signaling are involved in two common bone-metastasizing malignancies, estrogen receptor positive (ER+) breast and prostate cancers. Compared to other sites, bone metastases of both cancer types expressed higher levels of ENTPD1 and NT5E, which encode CD39 and CD73, respectively, and hydrolyze ATP to adenosine. ADORA3, encoding the adenosine A3 receptor, had a similar expression pattern. In primary ER+ breast cancer, high levels of the triplet ENTPD1/NT5E/ADORA3 expression signature was correlated with lower overall, distant metastasis-free, and progression-free survival. In ER+ bone metastasis biopsies, this expression signature is associated with lower survival. This expression signature was also higher in bone-metastasizing primary prostate cancers than in those that caused other tumor events or did not lead to progressive disease. In 3D culture, a non-hydrolyzable ATP analog inhibited the growth of breast and prostate cancer cell lines more than ATP did. A3 inhibition also reduced spheroid growth. Large-scale screens by the Drug Repurposing Hub found ER+ breast cancer cell lines were uniquely sensitive to adenosine receptor antagonists. Together, these data suggest a vital role for extracellular ATP degradation and adenosine receptor signaling in cancer bone metastasis, and this study provides potential diagnostic means for bone metastasis and specific targets for treatment and prevention.
Collapse
Affiliation(s)
- Daniel Brian Shropshire
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| | - Francisca M. Acosta
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| | - Kun Fang
- Division of Biostatistics and MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jaime Benavides
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Lu-Zhe Sun
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, United States
| | - Victor X. Jin
- Division of Biostatistics and MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jean X. Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
- *Correspondence: Jean X. Jiang,
| |
Collapse
|
11
|
Elevated IsoPSA Selects for Clinically Significant Prostate Cancer Without a Preference for Any Particular Adverse Histopathologic or Radiographic Feature. Urology 2022; 168:150-155. [DOI: 10.1016/j.urology.2022.05.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/26/2022] [Indexed: 01/04/2023]
|
12
|
Rode MP, Silva AH, Cisilotto J, Rosolen D, Creczynski-Pasa TB. miR-425-5p as an exosomal biomarker for metastatic prostate cancer. Cell Signal 2021; 87:110113. [PMID: 34371055 DOI: 10.1016/j.cellsig.2021.110113] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/21/2022]
Abstract
Prostate cancer-related deaths are mostly caused by metastasis, which indicates the importance of identifying clinical prognostic biomarkers. In this study, we evaluated the expression profile of exosomal microRNAs (miRNAs) derived from metastatic prostate cancer (mPCa) cell lines (LNCaP and PC-3). miRNA signatures in exosomes and cells were evaluated by miRNA microarray analysis. Fourteen miRNAs were identified as candidates for specific noninvasive biomarkers. The expression of five miRNAs was validated using RT-qPCR, which confirmed that miR-205-5p, miR-148a-3p, miR-125b-5p, miR-183-5p, and miR-425-5p were differentially expressed in mPCa exosomes. Bioinformatic analyses showed that miR-425-5p was associated with residual tumor, pathologic T and N stages, and TP53 status in PCa samples. Gene ontology analysis of negatively correlated and predicted targeted genes showed enrichment of genes related to bone development pathways. The LinkedOmics database indicated that the potential target HSPB8 has a significant negative correlation with miR-425-5p. In conclusion, this study identified a panel of exosomal miRNAs with potential value as prognostic biomarkers for prostate cancer.
Collapse
Affiliation(s)
- Michele Patrícia Rode
- Department of Pharmaceutical Sciences, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | - Adny Henrique Silva
- Department of Pharmaceutical Sciences, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | - Júlia Cisilotto
- Department of Pharmaceutical Sciences, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | - Daiane Rosolen
- Department of Pharmaceutical Sciences, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | | |
Collapse
|
13
|
Brady-Nicholls R, Zhang J, Zhang T, Wang AZ, Butler R, Gatenby RA, Enderling H. Predicting patient-specific response to adaptive therapy in metastatic castration-resistant prostate cancer using prostate-specific antigen dynamics. Neoplasia 2021; 23:851-858. [PMID: 34298234 PMCID: PMC8322456 DOI: 10.1016/j.neo.2021.06.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 11/27/2022] Open
Abstract
Abiraterone acetate (AA) has been proven effective for metastatic castration-resistant prostate cancer (mCRPC), and it has been proposed that adaptive AA may reduce toxicity and prolong time to progression, when compared to continuous AA. We developed a simple quantitative model of prostate-specific antigen (PSA) dynamics to evaluate prostate cancer (PCa) stem cell enrichment as a plausible driver of AA treatment resistance. The model incorporated PCa stem cells, non-stem PCa cells and PSA dynamics during adaptive therapy. A leave-one-out analysis was used to calibrate and validate the model against longitudinal PSA data from 16 mCRPC patients receiving adaptive AA in a pilot clinical study. Early PSA treatment response dynamics were used to predict patient response to subsequent treatment. We extended the model to incorporate metastatic burden and also investigated the survival benefit of adding concurrent chemotherapy for patients predicted to become resistant. Model simulations demonstrated PCa stem cell self-renewal as a plausible driver of resistance to adaptive therapy. Evolutionary dynamics from individual treatment cycles combined with metastatic burden measurements predicted patient response with 81% accuracy (specificity=92%, sensitivity=50%). In those patients predicted to progress, simulations of the addition of concurrent chemotherapy suggest a benefit between 1% and 11% reduction in probability of progression when compared to adaptive AA alone. This study developed the first mCRPC patient-specific mathematical model to use early PSA treatment response dynamics to predict subsequent responses to adaptive AA, demonstrating the putative value of integrating mathematical modeling into clinical decision making.
Collapse
Affiliation(s)
- Renee Brady-Nicholls
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Jingsong Zhang
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Tian Zhang
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, 20 Duke Medicine Cir, Durham, NC, USA
| | - Andrew Z Wang
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Robert Butler
- Physical Sciences Oncology Center, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, , USA
| | - Robert A Gatenby
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Heiko Enderling
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
14
|
Kabunda J, Gabela L, Kalinda C, Aldous C, Pillay V, Nyakale N. Comparing 99mTc-PSMA to 99mTc-MDP in Prostate Cancer Staging of the Skeletal System. Clin Nucl Med 2021; 46:562-568. [PMID: 34028421 PMCID: PMC8174142 DOI: 10.1097/rlu.0000000000003702] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/23/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE This prospective study was aimed at assessing the ability of 99mTc-PSMA scan to detect bone metastases in prostate cancer (PCa) against 99mTc-MDP scan as a standard and assess the correlation of these modalities in PCa staging of bone involvement. PATIENTS AND METHODS Forty-one patients (41) with histologically confirmed PCa were scanned using both methods. Planar imaging was performed with additional regional SPECT/CT 3 to 4 hours posttracer injection. Scans were reported as positive, negative, or equivocal. In the case of positive scans, lesions were quantified by each of the 3 reporters separately. Planar and SPECT/CT images were reported together to obtain the final report on each scan. RESULTS Our preliminary results showed no significant difference in the detection of bone metastases between the 2 scans. 99mTc-PSMA detected 52 of the 55 bone lesions detected on 99mTc-MDP. However, 99mTc-PSMA provided extra information by reporting lymph nodal metastases in 7 patients and residual disease in the prostate in 2 patients with biochemical progression after radical therapy. In 1 patient, the PSMA scan resulted in change in management with patient now on 177Lu-PSMA radioligand therapy. Equivocal findings were reported in 4 patients on 99mTc-MDP and none on 99mTc-PSMA. CONCLUSIONS 99mTc-PSMA was comparable to 99mTc-MDP in detection of bone metastases and demonstrated an additional benefit of providing information on visceral disease. 99mTc-PSMA may be a better alternative to 99mTc-MDP in staging, restaging, and assessment of patients with biochemical progression after radical therapy of PCa in a resource-limited setup like ours while also assisting to detect patients eligible for PSMA-labeled radioligand therapy.
Collapse
Affiliation(s)
- Joseph Kabunda
- From the University of KwaZulu-Natal, Durban, South Africa
| | - Lerato Gabela
- From the University of KwaZulu-Natal, Durban, South Africa
| | | | - Colleen Aldous
- From the University of KwaZulu-Natal, Durban, South Africa
| | | | | |
Collapse
|
15
|
Ozdemir S, Ersay AR, Koc Ozturk F, Ozdemir BS. Predictive value of standard serum markers for bone metastases in prostate cancer. AFRICAN JOURNAL OF UROLOGY 2021. [DOI: 10.1186/s12301-021-00170-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Abstract
Background
The early detection of bone metastases is very important in prostate cancer follow-up. This study aimed to compare conventional tumor markers, namely free prostate-specific antigen (free PSA), total prostate-specific antigen (total PSA), free PSA/total PSA ratio, alkaline phosphatase (ALP) values, Gleason scores and 99 m Tc-MDP bone scintigraphy findings in the prediction of bone metastases in prostate cancer.
Methods
In total, 175 patients with prostate cancer who underwent whole-body bone scintigraphy were included in the study. All selected scintigraphic studies were reprocessed. Free PSA, total PSA, free PSA/total PSA ratio, alkaline phosphatase (ALP) values and Gleason scores of patients were recorded.
Results
The results of our study show that the presence of bone metastasis correlates very weakly with free PSA/total PSA ratio (rho = 0.179), weakly with total PSA (rho = 0.318) and Gleason score (rho = 0.382), moderately with ALP (rho = 0.539), free PSA (0.416). Only ALP variable had a diagnostic value and ALP cutoff value was 76.50 IU/L, with 80% sensitivity and 82.1% specificity.
Conclusion
According to the results of our study; the free PSA, total PSA, free PSA/total PSA ratio and Gleason score values were not considered as a reliable parameter in the prostate cancer cases follow-up for bone metastasis development. Only ALP had a diagnostic value and ALP cutoff value was 76.50 IU / L with 80% sensitivity and 82.1% specificity in predicting bone metastases in prostate cancer.
Collapse
|
16
|
Tarabaih M, Degheili JA, Nasser M. Isolated Solitary Lung Nodule in a Patient With Idiopathic Pulmonary Fibrosis and Concomitant Prostate Cancer: A Challenging Diagnosis. Cureus 2021; 13:e14218. [PMID: 33948408 PMCID: PMC8086736 DOI: 10.7759/cureus.14218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Prostate cancer is the most commonly diagnosed malignancy and the second most common cause of death in men after lung cancer. Isolated pulmonary metastasis from prostate cancer, without bone or lymph node involvement, is rare and accounts for less than 1% of cases. The diagnosis of solitary lung metastasis is even more challenging in patients with concomitant pulmonary disease and often mandates tissue biopsy from the lung nodule. We herein present a case of an elderly man with idiopathic pulmonary fibrosis who presented with a solitary lung nodule three years after a laparoscopic radical prostatectomy for localized prostate cancer. Initially thought as a primary lung lesion secondary to his pulmonary fibrosis, further workup and ultimately a lung segmentectomy proved a metastatic prostatic adenocarcinoma. The serum prostatic specific antigen dropped to nadir following resection, and he remained stable six months later.
Collapse
Affiliation(s)
- Mohamad Tarabaih
- Department of Oncology, Hôpital Lyon Sud, Institut de Carcinologie des Hospices Civils de Lyon (ICHCL), Lyon, FRA
| | - Jad A Degheili
- Division of Pediatric Urology, Department of Surgery, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, CAN.,Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, LBN
| | - Mouhamad Nasser
- Department of Respiratory Medicine, National Coordinating Reference Centre for Rare Pulmonary Diseases, Louis Pradel Hospital, University Hospital of Lyon, Lyon, FRA
| |
Collapse
|
17
|
Huang CY, Chen CH. Clinical characteristics and survival outcomes in patients with a high PSA and non-metastatic prostate cancer. J Formos Med Assoc 2021; 121:181-186. [PMID: 33715926 DOI: 10.1016/j.jfma.2021.02.015] [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: 12/20/2020] [Revised: 02/01/2021] [Accepted: 02/21/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND/PURPOSE To investigate the clinical presentation and survival outcomes of patients with both a high prostate-specific antigen (PSA) value and non-metastatic prostate cancer (PC). METHODS In total, 2053 PC patients were managed in our institute between January 2008 and December 2014. A total of 343 (16.7%) patients who presented with PSA values > 100 ng/mL were enrolled. Non-metastatic and metastatic PC were identified in 67 (group 1) and 276 (group 2) patients, respectively. Furthermore, 75 metastatic PC patients with PSA values < 20 ng/mL were included (group 3) for comparison. All demographics and survival outcomes were retrospectively reviewed by a questionnaire. RESULTS Group 2 patients had a higher PSA level than did group 1 (median: 1095 vs. 283 ng/mL, p < 0.001), and a higher Gleason grade than did groups 1 and 3 (grade group 4 plus 5: 60%, 77%, and 56%, for groups 1, 2, and 3, respectively; p < 0.001). Other demographics were similar among groups. Group 1 patients survived significantly longer than group 2 and 3 in terms of overall and cancer-specific survival rates (5-year overall survival rates: 87.5%, 46.3%, and 66.9%; 5-year cancer-specific survival rates: 94.7%, 52.7%, and 68.7% for groups 1, 2, and 3, respectively). Group 1 patients receiving local definitive treatments, such as radiation therapy or cryoablation, received survival and metastasis-free benefits compared to those without local treatment. CONCLUSION Patients with a high PSA value were not destined to have metastatic PC. Non-metastatic PC patients with a high PSA level obtained a survival benefit from local prostate-definitive treatments.
Collapse
Affiliation(s)
- Cheng-Yu Huang
- Department of Urology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Hsin Chen
- Department of Urology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
18
|
Westerberg M, Larsson R, Holmberg L, Stattin P, Garmo H. Simulation model of disease incidence driven by diagnostic activity. Stat Med 2020; 40:1172-1188. [PMID: 33241594 PMCID: PMC7894333 DOI: 10.1002/sim.8833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 01/24/2023]
Abstract
It is imperative to understand the effects of early detection and treatment of chronic diseases, such as prostate cancer, regarding incidence, overtreatment and mortality. Previous simulation models have emulated clinical trials, and relied on extensive assumptions on the natural history of the disease. In addition, model parameters were typically calibrated to a variety of data sources. We propose a model designed to emulate real‐life scenarios of chronic disease using a proxy for the diagnostic activity without explicitly modeling the natural history of the disease and properties of clinical tests. Our model was applied to Swedish nation‐wide population‐based prostate cancer data, and demonstrated good performance in terms of reconstructing observed incidence and mortality. The model was used to predict the number of prostate cancer diagnoses with a high or limited diagnostic activity between 2017 and 2060. In the long term, high diagnostic activity resulted in a substantial increase in the number of men diagnosed with lower risk disease, fewer men with metastatic disease, and decreased prostate cancer mortality. The model can be used for prediction of outcome, to guide decision‐making, and to evaluate diagnostic activity in real‐life settings with respect to overdiagnosis and prostate cancer mortality.
Collapse
Affiliation(s)
- Marcus Westerberg
- Department of Mathematics, Uppsala University, Uppsala, Sweden.,Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Rolf Larsson
- Department of Mathematics, Uppsala University, Uppsala, Sweden
| | - Lars Holmberg
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.,Translational Oncology & Urology Research (TOUR), School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Pär Stattin
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Hans Garmo
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
19
|
Nilbert M, Thomsen LA, Winther Jensen J, Møller H, Borre M, Widenlou Nordmark A, Lambe M, Brändström H, Kørner H, Møller B, Ursin G. The power of empirical data; lessons from the clinical registry initiatives in Scandinavian cancer care. Acta Oncol 2020; 59:1343-1356. [PMID: 32981417 DOI: 10.1080/0284186x.2020.1820573] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND In Scandinavia, there is a strong tradition for research and quality monitoring based on registry data. In Denmark, Norway and Sweden, 63 clinical registries collect data on disease characteristics, treatment and outcome of various cancer diagnoses and groups based on process-related and outcome-related variables. AIM We describe the cancer-related clinical registries, compare organizational structures and quality indicators and provide examples of how these registries have been used to monitor clinical performance, develop prediction models, assess outcome and provide quality benchmarks. Further, we define unmet needs such as inclusion of patient-reported outcome variables, harmonization of variables and barriers for data sharing. RESULTS AND CONCLUSIONS The clinical registry framework provides an empirical basis for evidence-based development of high-quality and equitable cancer care. The registries can be used to follow implementation of new treatment principles and monitor patterns of care across geographical areas and patient groups. At the same time, the lessons learnt suggest that further developments and coordination are needed to utilize the full potential of the registry initiative in cancer care.
Collapse
Affiliation(s)
- Mef Nilbert
- Department of Oncology, Lund University, Lund, Sweden
- The Danish Cancer Society Research Center, Copenhagen, Denmark
- Clinical Research Department, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| | | | - Jens Winther Jensen
- The Danish Clinical Quality Program and Clinical Registries, Aarhus, Denmark
| | - Henrik Møller
- The Danish Clinical Quality Program and Clinical Registries, Aarhus, Denmark
| | - Michael Borre
- The Association of Danish Multidisciplinary Cancer Groups, Aarhus, Denmark
| | | | - Mats Lambe
- The Federation of Regional Cancer Centers, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Hartwig Kørner
- Institute of Surgical Sciences, University of Bergen, Bergen, Norway
| | | | | |
Collapse
|