1
|
Wu XH, Ke ZB, Chen ZJ, Liu WQ, Xue YT, Chen SH, Chen DN, Zheng QS, Xue XY, Wei Y, Xu N. Periprostatic fat magnetic resonance imaging based radiomics nomogram for predicting biochemical recurrence-free survival in patients with non-metastatic prostate cancer after radical prostatectomy. BMC Cancer 2024; 24:1459. [PMID: 39604917 PMCID: PMC11600801 DOI: 10.1186/s12885-024-13207-4] [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: 02/02/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVE To build and validate a periprostatic fat magnetic resonance imaging (MRI) based radiomics nomogram for prediction of biochemical recurrence-free survival (bRFS) of patients with non-metastatic prostate cancer (PCa) receiving radical prostatectomy (RP). METHODS A retrospective study was conducted on 356 patients with non-metastatic PCa who underwent preoperative mpMRI followed by RP treatment at our institution. Radiomic features were extracted from both intratumoral region and the periprostatic fat region, which were segmented on images obtained through T2-weighted imaging (T2WI) and apparent-diffusion coefficient (ADC) imaging. Three radiomics models were developed by applying the Least absolute shrinkage and selection operator (LASSO) Cox regression, followed by Cox risk regression to construct a combined radiomics-clinical model by integrating the optimal radiomics score and clinicopathological risk factors to draw a nomogram. The predictive performance was evaluated using receiver operating characteristic (ROC) curves, Kaplan-Meier analysis and calibration curves. RESULTS One hundred and twenty-one patients (33.98%) experienced biochemical recurrence. ROC analyses showed that the Area Under the Curve (AUC) of the periprostatic fat-intratumoral radiomics model demonstrated the highest AUC at 0.921 (95%CI, 0.857-0.981), 0.875 (95%CI, 0.763-0.950), 0.854 (95%CI, 0.706-0.923) for 1-year, 3-years and 5-years bRFS. Multivariate Cox regression analysis revealed that Pathological T stage, ISUP grading group and Positive surgical margin were independent prognostic factors for predicting bRFS. A radiomics-clinical nomogram based on these clinical predictors and periprostatic fat-intratumoral radiomics score was constructed. Kaplan-Meier analyses showed that radiomics-clinical nomogram was significantly related with survival of PCa (P < 0.001); and calibration curves revealed the predicted and observed survival probability of 1-year, 3-year and 5-year bRFS had high degree of consistency in the training and validation group. The radiomics-clinical nomogram showed a significant improvement than the clinical model for 1-year (AUC, 0.944; 95%CI, 0.912-0.990 vs. AUC, 0.839; 95%CI, 0.661-0.928; P = 0.009), 3-year (AUC, 0.864; 95%CI, 0.772-0.969 vs. AUC, 0.776; 95%CI, 0.602-0.872; P = 0.008), and 5-year bRFS (AUC, 0.907; 95%CI, 0.836-0.982 vs. AUC, 0.819; 95%CI, 0.687-0.915; P = 0.027). CONCLUSIONS This study developed and validated the radiomics-clinical nomogram for the prediction of bRFS in non-metastatic PCa patients underwent RP.
Collapse
Affiliation(s)
- Xiao-Hui Wu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Zhi-Bin Ke
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Ze-Jia Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Wen-Qi Liu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Yu-Ting Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Shao-Hao Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Dong-Ning Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Qing-Shui Zheng
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Region Medical centre, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
2
|
Fan S, Liu H, Hou J, Zheng G, Gu P, Liu X. Characterizing adipocytokine-related signatures for prognosis prediction in prostate cancer. Front Cell Dev Biol 2024; 12:1475980. [PMID: 39524226 PMCID: PMC11544632 DOI: 10.3389/fcell.2024.1475980] [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: 08/04/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Background Prostate cancer (PCa) is a prevalent malignant tumor in males, with a significant incidence of biochemical recurrence (BCR) despite advancements in treatment. Adipose tissue surrounding the prostate, known as periprostatic adipose tissue (PPAT), contributes to PCa invasion through adipocytokine production. However, the relationship between adipocytokine-related genes and PCa prognosis remains understudied. This study was conducted to provide a theoretical basis and serve as a reference for the use of adipocytokine-related genes as prognostic markers in PCa. Methods Transcriptome and survival data of PCa patients from The Cancer Genome Atlas (TCGA) database were analyzed. Differential gene expression analysis was conducted using the DESeq2 and limma packages. Prognostic genes were identified through univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression. A prognostic model was developed and validated utilizing receiver operating characteristic (ROC) and Kaplan-Meier (K-M) curves. Assessments of immune cell infiltration and drug sensitivity were also carried out. Subsequently, the function of BNIP3L gene in PCa was verified. Results A total of 47 adipocytokine-related differentially expressed genes (DEGs) were identified. Five genes (PPARGC1A, APOE, BNIP3L, STEAP4, and C1QTNF3) were selected as prognostic markers. The prognostic model demonstrated significant predictive accuracy in both training and validation cohorts. Patients with higher risk scores exhibited poorer survival outcomes. Immune cell infiltration analysis revealed that the high-risk group had increased immune and ESTIMATE scores, while the low-risk group had higher tumor purity. In vitro experiments confirmed the suppressive effects of BNIP3L on PCa cell proliferation, migration, and invasion. Conclusion The prognostic model independently predicts the survival of patients with PCa, aiding in prognostic prediction and therapeutic efficacy. It expands the study of adipocytokine-related genes in PCa, presenting novel targets for treatment.
Collapse
Affiliation(s)
- Shicheng Fan
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Haolin Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jian Hou
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Guiying Zheng
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Peng Gu
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xiaodong Liu
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
3
|
Wu X, Ruan Z, Ke Z, Lin F, Chen J, Xue Y, Lin B, Chen S, Chen D, Zheng Q, Xue X, Wei Y, Xu N. Magnetic resonance imaging-based radiomics nomogram for the evaluation of therapeutic responses to neoadjuvant chemohormonal therapy in high-risk non-metastatic prostate cancer. Cancer Med 2024; 13:e70001. [PMID: 39031016 PMCID: PMC11258568 DOI: 10.1002/cam4.70001] [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: 02/01/2024] [Revised: 05/30/2024] [Accepted: 06/30/2024] [Indexed: 07/22/2024] Open
Abstract
PURPOSE The aim of this study was to assess the potential application of a radiomics features-based nomogram for predicting therapeutic responses to neoadjuvant chemohormonal therapy (NCHT) in patients with high-risk non-metastatic prostate cancer (PCa). METHODS Clinicopathologic information was retrospectively collected from 162 patients with high-risk non-metastatic PCa receiving NCHT and radical prostatectomy at our center. The postoperative pathological findings were used as the gold standard for evaluating the efficacy of NCHT. The least absolute shrinkage and selection operator (LASSO) was conducted to develop radiomics signature. Multivariate logistic regression analyses were conducted to identify the predictors of a positive pathological response to NCHT, and a nomogram was constructed based on these predictors. RESULTS Sixty-three patients (38.89%) experienced positive pathological response to NCHT. Receiver operating characteristic analyses showed that the area under the curve (AUC) of periprostatic fat (PPF) radiomics signature was 0.835 (95% CI, 0.754-0.898), while the AUC of intratumoral radiomics signature was 0.822 (95% CI, 0.739-0.888). Multivariate logistic regression analysis revealed that PSA level, PPF radiomics signature and intratumoral radiomics signature were independent predictors of positive pathological response. A nomogram based on these three predictors was constructed. The AUC was 0.908 (95% CI, 0.839-0.954). The Hosmer-Lemeshow goodness-of-fit test showed that the nomogram was well calibrated. Decision curve analysis revealed the favorable clinical practicability of the nomogram. The nomogram was successfully validated in the validation cohort. Kaplan-Meier analyses showed that nomogram and positive pathological response were significantly related with survival of PCa. CONCLUSION The radiomics-clinical nomogram based on mpMRI radiomics features exhibited superior predictive ability for positive pathological response to NCHT in high-risk non-metastatic PCa.
Collapse
Affiliation(s)
- Xiao‐Hui Wu
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Zhong‐Tian Ruan
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Zhi‐Bin Ke
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Fei Lin
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Jia‐Yin Chen
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Yu‐Ting Xue
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Bin Lin
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Shao‐Hao Chen
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Dong‐Ning Chen
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Qing‐Shui Zheng
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Xue‐Yi Xue
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Yong Wei
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Ning Xu
- Department of Urology, Urology Research Institute, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Urology, National Region Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| |
Collapse
|
4
|
Pan Y, Xu Y, Fan C, Miao X, Shen Y, Wang Q, Wu J, Hu H, Wang H, Xiang M, Ye B. The role of neck adipose tissue in lymph node metastasis of head and neck cancer. Front Oncol 2024; 14:1390824. [PMID: 38800384 PMCID: PMC11116645 DOI: 10.3389/fonc.2024.1390824] [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: 02/24/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Previous studies indicated that adipose tissue significantly influences cancer invasion and lymphatic metastasis. However, the impact of neck adipose tissue (NAT) on lymph node metastasis associated with head and neck cancer remains ambiguous. Here, we systematically assess the classification and measurement criteria of NAT and evaluate the association of adipose tissue and cancer-associated adipocytes with head and neck cancer. We delve into the potential mechanisms by which NAT facilitate cervical lymph node metastasis in head and neck cancer, particularly through the secretion of adipokines such as leptin, adiponectin, and Interleukin-6. Our aim is to elucidate the role of NAT in the progression and metastasis of head and neck cancer, offering new insights into prevention and treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Mingliang Xiang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bin Ye
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Zheng X, Xie X, Wang W, Wang L, Tan B. Silencing of matrix metalloprotease-12 delays the progression of castration-resistant prostate cancer by regulating autophagy and lipolysis. Braz J Med Biol Res 2024; 57:e13351. [PMID: 38511770 PMCID: PMC10946229 DOI: 10.1590/1414-431x2024e13351] [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: 10/12/2023] [Accepted: 02/13/2024] [Indexed: 03/22/2024] Open
Abstract
The complex pathogenesis of castration-resistant prostate cancer (CRPC) makes it challenging to identify effective treatment methods. Matrix metalloproteinase (MMP)-12 can degrade elastin as well as various extracellular matrix (ECM) components, which is associated with cancer progression. However, the relationship between MMP-12 and CRPC progression is poorly understood. In this study, we observed the effect of MMP-12 on the progression of CRPC and further explored its potential mechanism of action. High levels of MMP-12 were observed in patients with CRPC. We therefore developed cell co-culture and mouse models to study the function of MMP-12. Silencing MMP-12 in CRPC cells disrupted lipid utilization and autophagy marker expression via the CD36/CPT1 and P62/LC3 pathways, respectively, leading to reduced CRPC cell migration and invasion. Moreover, animal experiments confirmed that MMP-12-knockdown CRPC xenograft tumors exhibited reduced tumor growth, and the mechanisms involved the promotion of cancer cell autophagy and the inhibition of lipid catabolism. According to our results, MMP-12 played important roles in the progression of CRPC by disrupting adipocyte maturation and regulating cancer migration and invasion via the modulation of autophagy and lipid catabolism pathways.
Collapse
Affiliation(s)
- Xiaoyu Zheng
- School of Clinical Medicine, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Xiaoqin Xie
- Department of Clinical Laboratory, Chongqing Blood Center, Chongqing, China
| | - Wei Wang
- Department of Orthopedics, The People's Hospital of Yubei District of Chongqing City, Chongqing, China
| | - Liang Wang
- Department of Dermatology, Daping Hospital, Army Medical University, Chongqing, China
| | - Bing Tan
- School of Clinical Medicine, Chongqing Medical and Pharmaceutical College, Chongqing, China
- Department of Urology and Medical Sciences Research Center, University-Town Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Cancel M, Crottes D, Bellanger D, Bruyère F, Mousset C, Pinault M, Mahéo K, Fromont G. Variable effects of periprostatic adipose tissue on prostate cancer cells: Role of adipose tissue lipid composition and cancer cells related factors. Prostate 2024; 84:358-367. [PMID: 38112233 DOI: 10.1002/pros.24655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 11/28/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Periprostatic adipose tissue (PPAT) is likely to modulate prostate cancer (PCa) progression. We analyzed the variations in the effect of PPAT on cancer cells, according to its fatty acid (FA) composition and tumor characteristics. METHODS The expression of markers of aggressiveness Ki67 and Zeb1, and epigenetic marks that could be modified during PCa progression, was analyzed by immunohistochemistry on a tissue-micro-array containing 59 pT3 PCa, including intra-prostatic areas and extra-prostatic foci in contact with PPAT belonging to the same tumor. In addition, we cocultivated PC3 and LNCaP cell lines with PPAT, which were then analyzed for FA composition. RESULTS Although the contact between PPAT and cancer cells led overall to an increase in Ki67 and Zeb1, and a decrease in the epigenetic marks 5MC, 5HMC, and H3K27ac, these effects were highly heterogeneous. Increased proliferation in extra-prostatic areas was associated with the international society of uropathology score. PC3 and LNCaP cocultures with PPAT led to increased Ki67, Zeb1 and H3K27me3, but only for PPAT associated with aggressive PCa. PC3 proliferation was correlated with high 20.2 n-6 and low 20.5n-3 in PPAT. CONCLUSIONS These results suggest that the effects of PPAT on cancer cells may depend on both PCa characteristics and PPAT composition, and could lead to propose nutritional supplementation.
Collapse
Affiliation(s)
- Mathilde Cancel
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
- Department of Medical Oncology, CHU Tours, Tours, France
| | - David Crottes
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
| | - Dorine Bellanger
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
| | | | - Coralie Mousset
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
- Department of Pathology, CHU Tours, Tours, France
| | - Michelle Pinault
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
| | - Karine Mahéo
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
| | - Gaëlle Fromont
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
- Department of Pathology, CHU Tours, Tours, France
| |
Collapse
|
7
|
Enke JS, Groß M, Grosser B, Sipos E, Steinestel J, Löhr P, Waidhauser J, Lapa C, Märkl B, Reitsam NG. SARIFA as a new histopathological biomarker is associated with adverse clinicopathological characteristics, tumor-promoting fatty-acid metabolism, and might predict a metastatic pattern in pT3a prostate cancer. BMC Cancer 2024; 24:65. [PMID: 38216952 PMCID: PMC10785487 DOI: 10.1186/s12885-023-11771-9] [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: 10/13/2023] [Accepted: 12/17/2023] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Recently, we introduced Stroma-AReactive-Invasion-Front-Areas (SARIFA) as a novel hematoxylin-eosin (H&E)-based histopathologic prognostic biomarker for various gastrointestinal cancers, closely related to lipid metabolism. To date, no studies on SARIFA, which is defined as direct tumor-adipocyte-interaction, beyond the alimentary tract exist. Hence, the objective of our current investigation was to study the significance of SARIFA in pT3a prostate cancer (PCa) and explore its association with lipid metabolism in PCa as lipid metabolism plays a key role in PCa development and progression. METHODS To this end, we evaluated SARIFA-status in 301 radical prostatectomy specimens and examined the relationship between SARIFA-status, clinicopathological characteristics, overall survival, and immunohistochemical expression of FABP4 and CD36 (proteins closely involved in fatty-acid metabolism). Additionally, we investigated the correlation between SARIFA and biochemical recurrence-free survival (BRFS) and PSMA-positive recurrences in PET/CT imaging in a patient subgroup. Moreover, a quantitative SARIFA cut-off was established to further understand the underlying tumor biology. RESULTS SARIFA positivity occurred in 59.1% (n = 178) of pT3a PCas. Our analysis demonstrated that SARIFA positivity is strongly associated with established high-risk features, such as R1 status, extraprostatic extension, and higher initial PSA values. Additionally, we observed an upregulation of immunohistochemical CD36 expression specifically at SARIFAs (p = 0.00014). Kaplan-Meier analyses revealed a trend toward poorer outcomes, particularly in terms of BRFS (p = 0.1). More extensive tumor-adipocyte interaction, assessed as quantity-dependent SARIFA-status on H&E slides, is also significantly associated with high-risk features, such as lymph node metastasis, and seems to be associated with worse survival outcomes (p = 0.16). Moreover, SARIFA positivity appeared to be linked to more distant lymph node and bone metastasis, although statistical significance was slightly not achieved (both p > 0.05). CONCLUSIONS This is the first study to introduce SARIFA as easy-and-fast-to-assess H&E-based biomarker in locally advanced PCa. SARIFA as the histopathologic correlate of a distinct tumor biology, closely related to lipid metabolism, could pave the way to a more detailed patient stratification and to the development of novel drugs targeting lipid metabolism in pT3a PCa. On the basis of this biomarker discovery study, further research efforts on the prognostic and predictive role of SARIFA in PCa can be designed.
Collapse
Affiliation(s)
- Johanna S Enke
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Matthias Groß
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Bianca Grosser
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Eva Sipos
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Julie Steinestel
- Urology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Phillip Löhr
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Johanna Waidhauser
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Bruno Märkl
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Nic G Reitsam
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany.
| |
Collapse
|
8
|
Oregel-Cortez MI, Frayde-Gómez H, Quintana-González G, García-González V, Vazquez-Jimenez JG, Galindo-Hernández O. Resistin Induces Migration and Invasion in PC3 Prostate Cancer Cells: Role of Extracellular Vesicles. Life (Basel) 2023; 13:2321. [PMID: 38137922 PMCID: PMC10744490 DOI: 10.3390/life13122321] [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: 05/15/2023] [Revised: 07/10/2023] [Accepted: 07/20/2023] [Indexed: 12/24/2023] Open
Abstract
Resistin is an adipokine with metabolic and inflammatory functions. Epidemiological and translational studies report that an increase in plasma levels and tissue expression of resistin increases the aggressiveness of prostate tumor cells. Extracellular vesicles (EVs) are secreted constitutively and induced by cytokines, growth factors, and calcium and are found in multiple biological fluids such as saliva, serum, semen, and urine. In particular, EVs have been shown to promote tumor progression through the induction of proliferation, growth, angiogenesis, resistance to chemotherapy, and metastasis. However, the role of resistin in the migration, invasion, and secretion of EVs in invasive prostate tumor cells remains to be studied. In the present study, we demonstrate that resistin induces increased migration and invasion in PC3 cells. In addition, these phenomena are accompanied by increased p-FAK levels and increased secretion of MMP-2 and MMP-9 in resistin-treated PC3 cells. Interestingly, EVs isolated from supernatants of PC3 cells treated with resistin induce an increase in migration and invasion accompanied by high MMP-2 and MMP-9 secretion in an autocrine stimulation model. In summary, our data for the first time demonstrate that resistin induces migration and invasion, partly through the secretion of EVs with pro-invasive characteristics in PC3 cells.
Collapse
Affiliation(s)
- Mario Israel Oregel-Cortez
- Departamento de Bioquimíca, Facultad de Medicina, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico; (M.I.O.-C.); (H.F.-G.); (G.Q.-G.); (V.G.-G.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico
- Facultad de Deportes, Universidad Autónoma de Baja California, Mexicali 21289, Baja California, Mexico
| | - Héctor Frayde-Gómez
- Departamento de Bioquimíca, Facultad de Medicina, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico; (M.I.O.-C.); (H.F.-G.); (G.Q.-G.); (V.G.-G.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico
- Hospital Regional de Especialidad No. 30, Instituto Mexicano del Seguro Social, Mexicali 21100, Baja California, Mexico
| | - Georgina Quintana-González
- Departamento de Bioquimíca, Facultad de Medicina, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico; (M.I.O.-C.); (H.F.-G.); (G.Q.-G.); (V.G.-G.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico
| | - Victor García-González
- Departamento de Bioquimíca, Facultad de Medicina, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico; (M.I.O.-C.); (H.F.-G.); (G.Q.-G.); (V.G.-G.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico
| | - Jose Gustavo Vazquez-Jimenez
- Laboratorio de Fisiología, Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico;
| | - Octavio Galindo-Hernández
- Departamento de Bioquimíca, Facultad de Medicina, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico; (M.I.O.-C.); (H.F.-G.); (G.Q.-G.); (V.G.-G.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico
| |
Collapse
|
9
|
Capuozzo M, Celotto V, Landi L, Ferrara F, Sabbatino F, Perri F, Cascella M, Granata V, Santorsola M, Ottaiano A. Beyond Body Size: Adiponectin as a Key Player in Obesity-Driven Cancers. Nutr Cancer 2023; 75:1848-1862. [PMID: 37873648 DOI: 10.1080/01635581.2023.2272343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 10/25/2023]
Abstract
Obesity, a complex and multifactorial disease influenced by genetic, environmental, and psychological factors, has reached epidemic proportions globally, posing a significant health challenge. In addition to its established association with cardiovascular disease and type II diabetes, obesity has been implicated as a risk factor for various cancers. However, the precise biological mechanisms linking obesity and cancer remain largely understood. Adipose tissue, an active endocrine organ, produces numerous hormones and bioactive molecules known as adipokines, which play a crucial role in metabolism, immune responses, and systemic inflammation. Notably, adiponectin (APN), the principal adipocyte secretory protein, exhibits reduced expression levels in obesity. In this scoping review, we explore and discuss the role of APN in influencing cancer in common malignancies, including lung, breast, colorectal, prostate, gastric, and endometrial cancers. Our review aims to emphasize the critical significance of investigating this field, as it holds great potential for the development of innovative treatment strategies that specifically target obesity-related malignancies. Furthermore, the implementation of more rigorous and comprehensive prevention and treatment policies for obesity is imperative in order to effectively mitigate the risk of associated diseases, such as cancer.
Collapse
Affiliation(s)
| | | | | | | | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
| | - Francesco Perri
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Naples, Italy
| | - Marco Cascella
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Naples, Italy
| | - Vincenza Granata
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Naples, Italy
| | | | | |
Collapse
|
10
|
Bocian-Jastrzębska A, Malczewska-Herman A, Kos-Kudła B. Role of Leptin and Adiponectin in Carcinogenesis. Cancers (Basel) 2023; 15:4250. [PMID: 37686525 PMCID: PMC10486522 DOI: 10.3390/cancers15174250] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Hormones produced by adipocytes, leptin and adiponectin, are associated with the process of carcinogenesis. Both of these adipokines have well-proven oncologic potential and can affect many aspects of tumorigenesis, from initiation and primary tumor growth to metastatic progression. Involvement in the formation of cancer includes interactions with the tumor microenvironment and its components, such as tumor-associated macrophages, cancer-associated fibroblasts, extracellular matrix and matrix metalloproteinases. Furthermore, these adipokines participate in the epithelial-mesenchymal transition and connect to angiogenesis, which is critical for cancer invasiveness and cancer cell migration. In addition, an enormous amount of evidence has demonstrated that altered concentrations of these adipocyte-derived hormones and the expression of their receptors in tumors are associated with poor prognosis in various types of cancer. Therefore, leptin and adiponectin dysfunction play a prominent role in cancer and impact tumor invasion and metastasis in different ways. This review clearly and comprehensively summarizes the recent findings and presents the role of leptin and adiponectin in cancer initiation, promotion and progression, focusing on associations with the tumor microenvironment and its components as well as roles in the epithelial-mesenchymal transition and angiogenesis.
Collapse
Affiliation(s)
- Agnes Bocian-Jastrzębska
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinogy, Medical University of Silesia, 40-514 Katowice, Poland; (A.M.-H.); (B.K.-K.)
| | | | | |
Collapse
|
11
|
Na H, Song Y, Lee HW. Emphasis on Adipocyte Transformation: Anti-Inflammatory Agents to Prevent the Development of Cancer-Associated Adipocytes. Cancers (Basel) 2023; 15:cancers15020502. [PMID: 36672449 PMCID: PMC9856688 DOI: 10.3390/cancers15020502] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Of the various cell types in the tumor microenvironment (TME), adipocytes undergo a dynamic transformation when activated by neighboring cancer cells. Although these adipocytes, known as cancer-associated adipocytes (CAAs), have been reported to play a crucial role in tumor progression, the factors that mediate their transformation remain elusive. In this review, we discuss the hypothesis that inflammatory signals involving NF-ĸB activation can induce lipolysis and adipocyte dedifferentiation. This provides a mechanistic understanding of CAA formation and introduces the concept of preventing adipocyte transformation via anti-inflammatory agents. Indeed, epidemiological studies indicate a higher efficacy of nonsteroidal anti-inflammatory drugs (NSAIDs) in obese patients with cancer, suggesting that NSAIDs can modulate the TME. Inhibition of cyclooxygenase-2 (COX-2) and prostaglandin production leads to the suppression of inflammatory signals such as NF-ĸB. Thus, we suggest the use of NSAIDs in cancer patients with metabolic disorders to prevent the transformation of TME components. Moreover, throughout this review, we attempt to expand our knowledge of CAA transformation to improve the clinical feasibility of targeting CAAs.
Collapse
Affiliation(s)
- Heeju Na
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Yaechan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Gemcro Corporation, Seoul 03722, Republic of Korea
- Correspondence: ; Tel.: +82-2-2123-7642
| |
Collapse
|