1
|
Hua F, Cai Y. PAX2 induces endometrial cancer by inhibiting mitochondrial function via the CD133-AKT1 pathway. Mol Cell Biochem 2025; 480:3765-3781. [PMID: 39891863 PMCID: PMC12095341 DOI: 10.1007/s11010-025-05216-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Endometrial cancer (EC) is a malignancy of the endometrial epithelium. The prevalence and mortality rates associated with the disease are on the rise globally. A total of 20 cases of type I EC tissues were collected for transcriptomic sequencing, our findings indicate that PAX2 is highly expressed in EC tissues and is closely related to the pathogenesis of EC. PAX2 is a member of the paired homeobox domain family and has been linked to the development of a number of different tumours. In normal endometrial tissue, PAX2 is methylated; however, in EC, it is demethylated. Nevertheless, few studies have focused on its role in EC. A protein-protein interaction (PPI) analysis revealed a regulatory relationship between PAX2 and CD133, which in turn affects the activity of AKT1. CD133 is a well-known marker of tumor stem cells and is involved in tumor initiation, metastasis, recurrence, and drug resistance; AKT1 promotes cell survival by inhibiting apoptosis and is considered a major promoter of many types of cancer. Nevertheless, further investigation is required to ascertain whether PAX2 affects the progression of EC by regulating the CD133-AKT1 pathway. The present study demonstrated that PAX2 promoted cell proliferation, migration, invasion and adhesion, and inhibited apoptosis. Its mechanism of action was found to be the inhibition of mitochondrial oxidative phosphorylation, promotion of glycolysis, increase in mitochondrial copy number, and increase in the levels of reactive oxygen species (ROS) and hexokinase, as well as the concentration of mitochondrial calcium ions. This was achieved through the promotion of CD133 expression and the phosphorylation of AKT1. In conjunction with the aforementioned regulatory pathways, the progression of EC is facilitated.
Collapse
Affiliation(s)
- Fu Hua
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao, Nanjing, 210009, China
- Department of Gynecology, the Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - YunLang Cai
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao, Nanjing, 210009, China.
| |
Collapse
|
2
|
Yuwen Z, Chen X, Chen K, Zou T, Mao G, Liu H, Zhang L. Enhancing clinical precision in lung cancer tissue biopsy through elevated response-threshold of an endoplasmic reticulum-targeted fluorogenic probe. Mater Today Bio 2025; 32:101654. [PMID: 40160246 PMCID: PMC11953968 DOI: 10.1016/j.mtbio.2025.101654] [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] [Received: 01/19/2025] [Revised: 02/19/2025] [Accepted: 03/09/2025] [Indexed: 04/02/2025] Open
Abstract
Lung carcinoma is the leading cause of mortality globally, posing a significant public health concern. Fluorescent-mediated tumor imaging is emerging as a novel diagnostic and therapeutic approach in clinical practice. Nevertheless, traditional probes lack accuracy in diagnosing tumors due to the overlap in baseline values of certain tumor biomarkers between normal and tumor cells as both exhibit turn-on fluorescence, rendering it impossible to distinguish tumor tissue from normal tissue with high resolution. We introduce a sensing strategy that constructs a probe with an elevated biomarker response-threshold and targeting ability for the endoplasmic reticulum (ER), enabling precise distinction between tumor and normal cells, and successfully develop such a probe. Elevating the response-threshold is advantageous in minimizing interference from baseline values of biomarkers in normal cells. Additionally, targeting the ER ensures that the probe's response range is consistent with the biomarker content in the ER, collectively enhancing differentiation between normal and cancer cells. Using this novel probe, a distinct bright fluorescence signal from tumors could be observed in confocal imaging of tumor tissues from tumor-bearing mice after intravenous injection, in stark contrast to the limited fluorescence emanating from normal tissues. Furthermore, this probe demonstrated exceptional precision in distinguishing clinical lung cancer tissue from para-cancer tissue. This work presents a more reliable tumor detection strategy, capable of accurate diagnosis even when the biomarker is highly expressed in both normal and tumor tissues. It promises to be a valuable tool for future clinical applications, particularly in intraoperative assisted resection.
Collapse
Affiliation(s)
- Zhiyang Yuwen
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, PR China
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Xinglong Chen
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, PR China
| | - Kexin Chen
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Tenglong Zou
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Guojiang Mao
- Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, 453007, PR China
| | - Hongwen Liu
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China
| | - Lemeng Zhang
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, PR China
| |
Collapse
|
3
|
Meixner E, Harrabi S, Seidensaal K, Koczur B, Tessonnier T, Lentz-Hommertgen A, Hoeltgen L, Hoegen-Saßmannshausen P, Weykamp F, Liermann J, Hörner-Rieber J, Debus J. Partial Breast Reirradiation for Breast Cancer Recurrences After Repeat Breast-Conserving Surgery with Proton Beam Therapy: The Prospective BREAST Trial (NCT06954623). J Clin Med 2025; 14:3416. [PMID: 40429411 PMCID: PMC12112130 DOI: 10.3390/jcm14103416] [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] [Received: 03/23/2025] [Revised: 04/18/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
(1) Background: The management of ipsilateral breast cancer recurrence depends on the extent of the tumor, and staging results, and mastectomy is currently the standard of care for previously irradiated patients. Studies are increasingly investigating suitable candidates for the repeated use of breast-conserving approaches as an alternative to mastectomy. But this includes the crucial necessity for curative reirradiation (Re-RT). The therapeutic challenge in reirradiation involves finding a balance between tumor control and the risk of severe toxicity from cumulative radiation doses in previously irradiated organs. Re-RT options include the use of brachytherapy, intraoperative radiotherapy, or external beam RT with photons or electrons. The application of particle therapy using proton beam therapy represents an innovative radiotherapeutic technique for breast cancer patients that might offer advantageous physical properties, a superior dose reduction to adjacent organs-at-risk, and effective target volume coverage with lower integral doses to the patient's whole body. In addition, this technique could potentially offer higher radiobiological effects and tumor responses. (2) Methods: The BREAST trial (NCT06954623) will be conducted as a prospective, single-arm, phase II study in 20 patients with histologically proven invasive breast cancer recurrences after repeat breast-conserving surgery and with an indication for local reirradiation. The patients will receive partial-breast re-RT with proton beam therapy in 15 once-daily fractions up to a total dose of 40.05 Gy(RBE), delivered with active raster scanning. The required time interval will be 1 year after previous RT to the ipsilateral breast. (3) Results: The following results will be reported: The primary endpoint is defined as the cumulative overall occurrence of (sub)acute skin toxicity of grade ≥ 3 within 6 months after the start of re-RT. Secondary outcome includes an analysis of the local, regional, and distant control, progression-free and overall survival, quality of life, and cosmesis. The explorative and translational objectives of this study include planning comparisons to other RT techniques and irradiation types, dosimetric evaluations, analyses of radiological imaging features, and translational assessments of cardiac toxicity biomarkers and tumor markers. (4) Conclusions: Overall, the aim of this study is to evaluate the potential of proton beam therapy for partial breast reirradiation and to establish the underlying data for a randomized trial.
Collapse
Affiliation(s)
- Eva Meixner
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (S.H.); (K.S.); (A.L.-H.); (L.H.); (P.H.-S.); (F.W.); (J.L.); (J.H.-R.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69210 Heidelberg, Germany
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany; (B.K.); (T.T.)
| | - Semi Harrabi
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (S.H.); (K.S.); (A.L.-H.); (L.H.); (P.H.-S.); (F.W.); (J.L.); (J.H.-R.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69210 Heidelberg, Germany
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany; (B.K.); (T.T.)
| | - Katharina Seidensaal
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (S.H.); (K.S.); (A.L.-H.); (L.H.); (P.H.-S.); (F.W.); (J.L.); (J.H.-R.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69210 Heidelberg, Germany
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany; (B.K.); (T.T.)
| | - Beata Koczur
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany; (B.K.); (T.T.)
| | - Thomas Tessonnier
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany; (B.K.); (T.T.)
| | - Adriane Lentz-Hommertgen
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (S.H.); (K.S.); (A.L.-H.); (L.H.); (P.H.-S.); (F.W.); (J.L.); (J.H.-R.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69210 Heidelberg, Germany
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany; (B.K.); (T.T.)
| | - Line Hoeltgen
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (S.H.); (K.S.); (A.L.-H.); (L.H.); (P.H.-S.); (F.W.); (J.L.); (J.H.-R.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69210 Heidelberg, Germany
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany; (B.K.); (T.T.)
| | - Philipp Hoegen-Saßmannshausen
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (S.H.); (K.S.); (A.L.-H.); (L.H.); (P.H.-S.); (F.W.); (J.L.); (J.H.-R.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69210 Heidelberg, Germany
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany; (B.K.); (T.T.)
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Radiation Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Fabian Weykamp
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (S.H.); (K.S.); (A.L.-H.); (L.H.); (P.H.-S.); (F.W.); (J.L.); (J.H.-R.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69210 Heidelberg, Germany
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany; (B.K.); (T.T.)
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Radiation Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Jakob Liermann
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (S.H.); (K.S.); (A.L.-H.); (L.H.); (P.H.-S.); (F.W.); (J.L.); (J.H.-R.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69210 Heidelberg, Germany
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany; (B.K.); (T.T.)
| | - Juliane Hörner-Rieber
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (S.H.); (K.S.); (A.L.-H.); (L.H.); (P.H.-S.); (F.W.); (J.L.); (J.H.-R.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69210 Heidelberg, Germany
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany; (B.K.); (T.T.)
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Radiation Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Department of Radiation Oncology, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (S.H.); (K.S.); (A.L.-H.); (L.H.); (P.H.-S.); (F.W.); (J.L.); (J.H.-R.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69210 Heidelberg, Germany
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany; (B.K.); (T.T.)
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Radiation Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
4
|
Shi Y, Guo Y, Dai J, Lu J, Huang S, Lu X. "Rebuilding Myself"- An intervention enhancing adaptability of cancer patients to return to work: a feasibility study. Rev Esc Enferm USP 2025; 59:e20240181. [PMID: 40354657 PMCID: PMC12070807 DOI: 10.1590/1980-220x-reeusp-2024-0181en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 01/22/2025] [Indexed: 05/14/2025] Open
Abstract
OBJECTIVE The aim of this research was to examine the feasibility and effects of the "Rebuilding Myself" intervention to enhance adaptability of cancer patients to return to work. METHODS A randomized controlled trial with a two-arm, single-blind design was employed. The control group received usual care, whereas the intervention group received "Rebuilding Myself" interventions. The effects were evaluated before the intervention, mid-intervention, and post-intervention. The outcomes were the adaptability to return to work, self-efficacy of returning to work, mental resilience, quality of life, and work ability. RESULTS The results showed a recruitment rate of 73.17%, a retention rate of 80%. Statistically significant differences were found between the two groups in cancer patients' adaptability to return to work, self-efficacy to return to work, mental resilience, and the dimension of bodily function, emotional function, fatigue, insomnia, and general health of quality of life. CONCLUSION "Rebuilding Myself" intervention was proven to be feasible and can initially improve cancer patients' adaptability to return to work. The intervention will help provide a new direction for clinicians and cancer patients to return to work.
Collapse
Affiliation(s)
- Yue Shi
- Nantong University, School of Nursing and Rehabilitation, Nantong,
China
| | - Yujie Guo
- Nantong University, School of Nursing and Rehabilitation, Nantong,
China
| | - Jingjing Dai
- Nantong University, Affiliated Hospital, Nantong, China
| | - Jianli Lu
- Nantong University, Affiliated Maternity and Child Health Care
Hospital, Nantong, China
| | - Suyun Huang
- Yangzhou University, Affiliated Taizhou Second People’s Hospital,
Taizhou, China
| | - Xiaopeng Lu
- Nantong Sixth People’s Hospital, Nantong, China
| |
Collapse
|
5
|
Majeed AJJ, Warrick J, Raman JD. The diagnostic dilemma of squamous differentiation in prostate cancer: A case report. Urol Case Rep 2025; 60:103034. [PMID: 40248516 PMCID: PMC12004368 DOI: 10.1016/j.eucr.2025.103034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025] Open
Abstract
Squamous differentiation in prostate cancer is rare, presenting unique diagnostic and therapeutic challenges. We report a case of high-grade prostatic adenocarcinoma with focal squamous differentiation in a 73-year-old man without prior radiation or androgen deprivation. Immunohistochemistry confirmed the squamous component arose from the adenocarcinoma, not a separate malignancy. This case highlights the importance of morphological assessment and strategic immunohistochemical evaluation to distinguish prostate cancer variants. A biopsy capturing only the squamous component could have led to misdiagnosis and inappropriate treatment. To improve diagnostic accuracy and management, we propose the term 'adenocarcinoma with focal squamous differentiation' over adenosquamous carcinoma.
Collapse
Affiliation(s)
- Abdul-Jawad J. Majeed
- Penn State College of Medicine, University Park Regional Campus, State College, PA, USA
| | - Joshua Warrick
- Department of Pathology and Laboratory Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Jay D. Raman
- Department of Urology, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
6
|
Wu Y, Zhao J, Zhao S, Li J, Luo J, Wang Y. PFKFB4 promotes endometrial cancer by regulating glycolysis through SRC‑3 phosphorylation. Oncol Rep 2025; 53:53. [PMID: 40116122 PMCID: PMC11948970 DOI: 10.3892/or.2025.8886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/06/2025] [Indexed: 03/23/2025] Open
Abstract
The present study aimed to investigate the role of 6‑phosphofructo‑2‑kinase/fructose‑2,6‑biphosphatase 4 (PFKFB4) in endometrial cancer cells and to explore its potential molecular mechanisms. PFKFB4 expression in endometrial cancer tissues was detected by immunohistochemistry. Cell Counting Kit‑8, Transwell assays and flow cytometry were used to detect cell proliferation, invasion and apoptosis in endometrial cancer cells after PFKFB4 knockdown. An enzyme‑linked immunosorbent assay was used to detect the glucose and lactic acid contents. Western blotting was performed to detect the levels of glycolysis‑related enzymes, steroid receptor coactivator‑3 (SRC‑3), and phosphorylated SRC‑3. In vivo experiments were performed to investigate the tumorigenic potential of PFKFB4. PFKFB4 expression was upregulated in endometrial cancer tissues compared with that in normal controls, and its upregulation was positively correlated with the depth of myometrial invasion, lymph node metastasis, surgical pathological stage and vascular invasion. PFKFB4 knockdown significantly inhibited proliferation and invasion, increased apoptosis, and decreased oxygen consumption and lactic acid production in endometrial cancer cells. PFKFB4 knockdown decreased SRC‑3 phosphorylation. After simultaneous PFKFB4 knockdown and SRC‑3 overexpression in cancer cells, oxygen consumption, lactic acid production, and glycolysis‑related protein expression were increased compared with those in control cells. PFKFB4 knockdown inhibited tumor proliferation, apoptosis and the expression of Ki‑67. PFKFB4 may regulate glycolysis in endometrial cancer cells by targeting SRC‑3, thus promoting endometrial cancer progression.
Collapse
Affiliation(s)
- Yaling Wu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
- Department of Gynecology and Obstetrics, People's Hospital of Shanxi, Taiyuan, Shanxi 030012, P.R. China
| | - Jianzhen Zhao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shuangshuang Zhao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jianfang Li
- Department of Gynecology and Obstetrics, People's Hospital of Shanxi, Taiyuan, Shanxi 030012, P.R. China
| | - Jin Luo
- Department of Pathology, People's Hospital of Shanxi, Taiyuan, Shanxi 030012, P.R. China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
7
|
Liang Y, Xu T, Xu S, Xu X, Wang L, Wang Z, Wang S. A HClO-activated BODIPY based ratiometric fluorescent probe with dual near-infrared channels for differentiating cancerous cells from normal cells and surgical guidance of tumor resection. Biosens Bioelectron 2025; 275:117247. [PMID: 39929086 DOI: 10.1016/j.bios.2025.117247] [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: 12/25/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/23/2025]
Abstract
Cancer (malignant tumor), a serious disease with a high mortality rate, early-stage diagnosis and treatment are very important for cancer therapy. The average concentration of reactive oxygen species (ROS) in cancer cells is ten times higher than in normal cells, and the overexpression of hypochlorous acid (HClO) is closely associated with cancer progression. Herein, a novel near-infrared (NIR) Golgi apparatus-targetable (GA) fluorescent probe GA-BOD-S was synthesized using the BODIPY and phenothiazine as the basic fluorescence framework for HClO monitoring. GA-BOD-S exhibited a colorimetric and ratiometric dual-mode recognition for HClO with the excellent merits including low detection limit (35 nM), fast response time (within 13 s), and exceptional GA targeting capability (Pearson's coefficient 0.98). Notably, both fluorescence emission wavelengths of GA-BOD-S before and after reaction with HClO are located in the NIR region, significantly enhancing the quality of fluorescence imaging in vivo. Cellular experiments revealed that GA-BOD-S can effectively visualize both endogenous and exogenous HClO within cells and living zebrafish. GA-BOD-S was successfully employed for high-contrast imaging of cancer cells and normal cells based on the differences in intracellular ROS levels, and achieved in-situ imaging of tumors in vivo. Furthermore, the precision of tumor resection surgery was significantly improved under the guidance of fluorescence probe GA-BOD-S, which provided a new perspective for the diagnosis and treatment of cancer disease.
Collapse
Affiliation(s)
- Yueyin Liang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Tongtong Xu
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Song Xu
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Xu Xu
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Liping Wang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Department of Biobank, Yangzhou, Jiangsu, China.
| | - Zhonglong Wang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China.
| | - Shifa Wang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China.
| |
Collapse
|
8
|
An Y, Feng Q, Jia L, Sha X, Zhang T, Lu L, Wang R, Bai B. Present progress in biomarker discovery of endometrial cancer by multi-omics approaches. Clin Proteomics 2025; 22:15. [PMID: 40281423 PMCID: PMC12032760 DOI: 10.1186/s12014-025-09528-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 01/14/2025] [Indexed: 04/29/2025] Open
Abstract
Endometrial cancer (EC), a prevalent and intricate disease, is associated with a poor prognosis among gynecological malignancies. Its incidence rising globally underscores the urgent need for biomarkers detection in both research and clinical settings. Over the past decade, we've witnessed rapid advancements in biological methodologies and techniques. A multitude of omics technologies, encompassing genomic/transcriptomic sequencing and proteomic/metabolomic mass spectrometry, have been extensively employed to analyze both tissue and liquid samples derived from EC patients. The integration of multi-omics data has not only broadened our understanding of the disease but also unearthed valuable biomarkers specific to EC. This review encapsulates the recent progress and future prospects in the application of multi-omics technologies in EC research, emphasizing the potential of multi-omics in uncovering novel biomarkers and enhancing clinical assessments.
Collapse
Affiliation(s)
- Yuhao An
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518020, China.
| | - Quanxin Feng
- Department of Gastrointestinal Surgery, Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi, 710032, China
| | - Li Jia
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Xinrui Sha
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518020, China
| | - Tuanjie Zhang
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518020, China
| | - Linlin Lu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Rui Wang
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518020, China.
| | - Bin Bai
- Department of Gastrointestinal Surgery, Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi, 710032, China.
| |
Collapse
|
9
|
Gofton C, Bartolomeo AD, Wijekulasuriya S, Cai S, Boutros R, Stafford-Bell F, Caldwell K, McCaughan G, Zekry A, Strasser SI, Levy M, Sheehan C, Goodall S, Davis JM, Sheahan L, Liu K, Greenaway S, Davison S, Huynh TD, Quadri Z, Zurynski YA, Agar M, George J. Palliative and supportive care in patients with hepatocellular carcinoma: a qualitative study on attitudes and perceptions of health professionals. Support Care Cancer 2025; 33:384. [PMID: 40237862 PMCID: PMC12003453 DOI: 10.1007/s00520-025-09403-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Integration of palliative and supportive care in cancer treatment pathways is becoming standardised. While there has been significant qualitative research in oncology on palliative and supportive care integration into clinical care, there is little evidence that focusses on clinicians who manage hepatocellular carcinoma (HCC) and their perceptions on palliative and supportive care. AIM To investigate the attitudes and perceptions regarding palliative and supportive care of healthcare professionals managing patients with HCC. DESIGN Qualitative study involving semi-structured individual interviews transcribed verbatim and analysed thematically. SETTING/PARTICIPANTS A total of 25 healthcare professionals including hepatologists, gastroenterology trainees, hepatology clinical nurse consultants, social workers, and palliative care specialists providing care to patients with HCC recruited at 4 tertiary hospitals via purposive sampling. RESULTS The following themes emerged: (1) availability of palliative care services, (2) need for clear referral pathways and processes, (3) patients' limited understanding of palliative care, (4) recognition of benefits of palliative care, and (5) the lack of training in hepatology services for palliative care provision. CONCLUSION Health professionals' perceptions of integration of palliative and supportive care in liver cancer care are hampered by multiple barriers. Opportunities to establish a more cohesive approach to care integration for patients with liver cancer have been identified. TRIAL REGISTRATION: ACTRN12623000010695 (date of registration 9/01/2023). What is already known about the topic? • Integration of palliative and supportive care for cancer can have profound benefits for patients' symptom burden and quality of life. • There is a lack of empirical studies examining the perspectives of health professionals who manage liver cancer on the integration of palliative and supportive care into the treatment pathways for patients. What this paper adds • This study identifies a number of barriers to the implementation of palliative and supportive care into liver cancer treatment algorithms. • The absence of sufficient evidence in clinical guidelines impairs the capacity of health professionals to improve the integration of palliative and supportive care for liver cancer patients. Implications for practice, theory, or policy. • As liver cancer prevalence increases, further effort is required to develop appropriate evidence-based clinical guidelines and referral pathways to support the integration of palliative and supportive care within existing liver cancer services.
Collapse
Affiliation(s)
- Cameron Gofton
- Storr Liver Centre, Westmead Hospital, WSLHD, Westmead Institute for Medical Research and University of Sydney, Sydney, Australia.
- Royal North Shore Hospital, St Leonards, Australia.
- Hepatology Services, NSLHD, St Leonards, Australia.
| | - Anna Di Bartolomeo
- Storr Liver Centre, Westmead Hospital, WSLHD, Westmead Institute for Medical Research and University of Sydney, Sydney, Australia
| | - Shalini Wijekulasuriya
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Shenghan Cai
- Centre for Healthcare Resilience and Implementation Science, Australian Institute of Health Innovation, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Rose Boutros
- Storr Liver Centre, Westmead Hospital, WSLHD, Westmead Institute for Medical Research and University of Sydney, Sydney, Australia
| | - Fiona Stafford-Bell
- Department of Palliative Care, Liverpool Hospital, SWSLHD, Liverpool, Australia
| | - Kim Caldwell
- Department of Palliative Care, St George Hospital, SESLHD, Sydney, Australia
| | - Geoffrey McCaughan
- A W Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital,, SLHD and University of Sydney, Camperdown, Australia
| | - Amany Zekry
- Department of Gastroenterology and Hepatology, St George Hospital, SESLHD, Sydney, Australia
| | - Simone I Strasser
- A W Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital,, SLHD and University of Sydney, Camperdown, Australia
| | - Miriam Levy
- Department of Gastroenterology and Hepatology, Liverpool Hospital, SWSLHD, Liverpool, Australia
| | - Caitlin Sheehan
- Department of Palliative Care, St George Hospital, SESLHD, Sydney, Australia
| | | | - Jan Maree Davis
- Palliative Care Service, SESLHD Southern Sector, Kogarah, Australia
| | - Linda Sheahan
- Department of Palliative Care, St George Hospital, SESLHD, Sydney, Australia
| | - Ken Liu
- A W Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital,, SLHD and University of Sydney, Camperdown, Australia
| | - Sally Greenaway
- Department of Palliative Care, Westmead Hospital, Westmead, Australia
| | - Scott Davison
- Department of Gastroenterology and Hepatology, Liverpool Hospital, SWSLHD, Liverpool, Australia
| | - Thang Du Huynh
- Department of Palliative Care, Liverpool Hospital, SWSLHD, Liverpool, Australia
| | - Zujaj Quadri
- Department of Palliative Care, Liverpool Hospital, SWSLHD, Liverpool, Australia
| | - Yvonne A Zurynski
- Australian Institute of Health Innovation, Macquarie University, Sydney, Australia
| | - Meera Agar
- University of Technology Sydney, Ultimo, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Hospital, WSLHD, Westmead Institute for Medical Research and University of Sydney, Sydney, Australia.
| |
Collapse
|
10
|
Zhang L, Li Q, Wu M, Feng X, Dai W, Chen P, Chen D, Zheng Z, Lin X, Wei G. TRIM22 governs tumorigenesis and protects against endometrial cancer-associated cachexia by inhibiting inflammatory response and adipose thermogenic activity. Cancer Metab 2025; 13:17. [PMID: 40200303 PMCID: PMC11980105 DOI: 10.1186/s40170-025-00386-2] [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: 12/03/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Endometrial cancer (EC) is one of the most common cancers in women, with a short overall survival and poor prognosis. Besides the biologically aggressive EC properties, Cancer-associated cachexia is the main factor. However, the detailed mechanism underlying EC-related cachexia and its harmful effects on EC progression and patient prognosis remains unclear. METHODS For clinical specimen and the vitro experiment, we detected TRIM22 expression level, EC patients' survival time, EC cell functional change, and adipose thermogenic changes to identify the function of TRIM22 in EC progression, EC-associated cachexia, and their molecular mechanisms. Then, for the vivo experiment, we exploited the xenografts in mice to identify the function of TRIM22 again, and to screen the drug therapeutic schedule. RESULTS Herein, we demonstrated that TRIM22 inhibited EC cell growth, invasion, and migration. Interleukin (IL)-6 mediated brown adipose tissue activation and white adipose tissue browning which induced EC-related cachexia. TRIM22 suppressed the EC cells' secretion of IL-6, and IL-6 mediated EC-related cachexia. Mechanistically, TRIM22 inhibited EC progression by suppressing the nucleotide-binding oligomerization domain 2(NOD2)/nuclear factor-kappaB (NF-κB) signaling pathway, with the purpose of impeding the production of IL-6. Moreover, we revealed that TRIM22 inhibited EC-associated cachexia by suppressing the IL-6/IL-6 receptor (IL-6R) signaling pathway. Therapeutically, we demonstrated that combination treatment with a TRIM22 inducer (progesterone) and a thermogenic inhibitor (IL-6R antibody) synergistically augmented the antitumor efficacy of carbotaxol (carboplatin and paclitaxel), in vivo. CONCLUSION Our data reveals that TRIM22-EC-IL-6-cachexia cross-communication has important clinical relevance and that the use of combined therapy holds great promise for enhancing the efficacy of anti-ECs. (Fig. graphical abstract).
Collapse
Affiliation(s)
- Liping Zhang
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Quanrong Li
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Meiting Wu
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Xiushan Feng
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Weichao Dai
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Peifang Chen
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Dezhao Chen
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Zhiqun Zheng
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Xiaoyan Lin
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.
| | - Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
11
|
Yildirim OS, Yildiz P, Karaer A, Calleja-Agius J, Ozcan S. Exploring the protein signature of endometrial cancer: A comprehensive review through diverse samples and mass spectrometry-based proteomics. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:108783. [PMID: 39488491 DOI: 10.1016/j.ejso.2024.108783] [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: 10/04/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024]
Abstract
Endometrial cancer (EC) is increasing incidence among women, and it constitutes a health problem for women globally. An important aspect of EC management involves the use of protein biomarkers for early detection and monitoring. Protein biomarkers allow the identification of high-risk patients, the detection of the disease in its early stages, and the assessment of treatment responses. Mass spectrometry (MS)-based proteomics offers robust analytical techniques and a comprehensive understanding of proteins. Proteomics methods allow scientists to investigate both the quantities and functions of proteins. Thus, it provides valuable insights into how proteins are altered under different conditions. This review summarizes recent advances in MS-based proteomic biomarker discovery for EC, focusing on different sample types and MS-based techniques used in clinical studies. The review emphasized in detail the most commonly used key sources such as blood, urine, vaginal fluids and tissue. Furthermore, MS-based proteomics techniques such as untargeted, targeted, sequential window acquisition of all theoretical mass spectra (SWATH-MS) and mass spectrometry imaging used in the discovery and validation/validation phases were evaluated. This review highlights the importance of biomarker discovery and clinical translation to improve diagnostic and therapeutic outcomes in EC. It aims to provide a comprehensive overview of MS-based proteomics in EC, guiding future research and clinical applications.
Collapse
Affiliation(s)
- Oyku Su Yildirim
- Department of Chemistry, Middle East Technical University (METU), 06800, Ankara, Turkiye
| | - Pelin Yildiz
- Department of Chemistry, Middle East Technical University (METU), 06800, Ankara, Turkiye
| | - Abdullah Karaer
- Reproductive Sciences & Advanced Bioinformatics Application & Research Center, Inonu University, 44280, Malatya, Turkiye; Department of Obstetrics and Gynecology, School of Medicine, Inonu University, 44280, Malatya, Turkiye
| | - Jean Calleja-Agius
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, Msida, MSD2080, Malta
| | - Sureyya Ozcan
- Department of Chemistry, Middle East Technical University (METU), 06800, Ankara, Turkiye; Cancer Systems Biology Laboratory (CanSyL), Middle East Technical University (METU), 06800, Ankara, Turkiye.
| |
Collapse
|
12
|
Kang OJ, Cho YJ, Lim MC, Lee YJ, Seo SS, Kang S, Park SY, Kim YS, Nam JH, Park JY. Docetaxel/Cisplatin Chemotherapy Followed by Pelvic Radiation Therapy in Patients With High-risk Endometrial Cancer After Staging Surgery: A Phase 2 Study. Int J Radiat Oncol Biol Phys 2025; 121:1229-1236. [PMID: 39557307 DOI: 10.1016/j.ijrobp.2024.11.071] [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: 05/09/2024] [Revised: 09/11/2024] [Accepted: 11/03/2024] [Indexed: 11/20/2024]
Abstract
PURPOSE To evaluate the efficacy and safety of docetaxel/cisplatin chemotherapy followed by pelvic radiation therapy after staging surgery in patients with high-risk endometrial cancer. METHODS AND MATERIALS In this open-label, single-arm, phase 2 trial conducted at 2 South Korean centers, we enrolled patients with histologically confirmed endometrial cancer who had undergone staging surgery. Inclusion criteria were based on International Federation of Gynecology and Obstetrics (FIGO) Staging 2009: stage I patients with ≥2 risk factors (grade 3, positive lymphovascular invasion, more than half of myometrium invasion); stage IB and II patients with clear cell or serous adenocarcinoma; stage II patients post-type 1 hysterectomy; and patients at stage III. Patients underwent 3 cycles of chemotherapy with docetaxel (70 mg/m2) and cisplatin (60 mg/m2) followed by pelvic radiation therapy ranging from 45 to 50.4 Gy. Disease status and adverse events were evaluated using RECIST 1.1 and Common Terminology Criteria for Adverse Events 4.0, respectively, with scheduled imaging and assessments throughout the study. RESULTS A total of 62 patients were included in this study and were followed for a median duration of 65 months (IQR, 48-86 months). The progression-free survival rates at 1, 3, and 5 years were 98.4%, 86.9%, and 79.1%, respectively. The overall survival rates at 1, 3, and 5 years were 98.4%, 96.4%, and 96.4%, respectively. After chemotherapy, 62.9% of patients experienced severe neutropenia, with 3.2% having grade 3 or 4 anemia. Common mild side effects included nausea (58.1%) and alopecia (38.7%). Postradiation, 16.7% experienced grade 3 neutropenia, and a few had grade 1 or 2 anemia (3.3%), with most other side effects being mild and no critical toxicities reported. CONCLUSIONS Patients with endometrial cancer with high-risk factors could benefit from adjuvant chemotherapy using docetaxel/cisplatin, followed by radiation therapy, with manageable toxicities.
Collapse
Affiliation(s)
- Ok-Ju Kang
- Department of Obstetrics and Gynecology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Yoon-Jung Cho
- Department of Obstetrics and Gynecology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Myong Cheol Lim
- Center for Gynecologic Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Yeon Jee Lee
- Center for Gynecologic Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Sang Soo Seo
- Center for Gynecologic Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Sokbom Kang
- Center for Gynecologic Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Sang-Yoon Park
- Center for Gynecologic Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Young Seok Kim
- Department of Radiation Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Joo-Hyun Nam
- Department of Obstetrics and Gynecology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jeong-Yeol Park
- Department of Obstetrics and Gynecology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Guo Y, Li L, Yang K, Song Y, Xu L, Zhao F, Li W. Enhancing prognostic insights: myometrial invasion patterns in endometrial carcinoma, with emphasis on MELF pattern-a comprehensive review and meta-analysis. Eur J Cancer Prev 2025; 34:173-184. [PMID: 38870068 DOI: 10.1097/cej.0000000000000903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
PURPOSE The microcystic, elongated, and fragmented (MELF) pattern, characterized by myxoid and inflamed stroma, is readily identifiable as a form of myometrial infiltration. This meta-analysis endeavors to assess the prognostic significance of MELF infiltration patterns in patients diagnosed with endometrial cancer. METHODS A comprehensive literature search, spanning until 11 October 2023, across PubMed , Embase , Cochrane , and Web of Science databases, identified 23 relevant studies involving 5199 patients. Data analysis was performed using Stata 16.0. RESULTS Analysis indicates that MELF infiltration predicts a higher risk of lymph node metastasis in endometrial cancer patients [hazard ratios (HR) = 5.05; 95% confidence interval (CI), 3.62-7.05; P < 0.05]. Notably, this association remains consistent across various patient demographics, analytical approaches, study designs, and treatment modalities. However, MELF infiltration does not significantly correlate with recurrence (HR = 1.05; 95% CI, 0.73-1.52; P > 0.05), overall survival (HR = 1.24; 95% CI, 0.91-1.68; P > 0.05), or disease-free survival (HR = 1.40; 95% CI, 0.85-2.28; P > 0.05). CONCLUSION While MELF infiltration heightens the risk of lymph node metastasis in endometrial cancer, its impact on recurrence, overall survival, and disease-free survival remains statistically insignificant.
Collapse
Affiliation(s)
| | - Luying Li
- School of Medicine, Shihezi University
| | | | | | - Lu Xu
- School of Medicine, Shihezi University
| | | | - Wenting Li
- School of Medicine, Shihezi University
- Department of Gynaecology, The First Affiliated Hospital of Shihezi University, Shihezi, China
| |
Collapse
|
14
|
Pang JHQ, Ng NZP. Limb Salvage in Patients with Concomitant Advanced Malignancy. J Clin Med 2025; 14:1642. [PMID: 40095581 PMCID: PMC11900209 DOI: 10.3390/jcm14051642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
Background: We are experiencing an increasing number of patients presenting with advanced malignancy who are also presenting with chronic limb threatening ischemia. Method: With advancements in medicine and surgery, we are suggesting a shift in mindset of how we treat this group of patients and proposing a more holistic approach for treatment options. Result: In this original article, we highlight our approach in managing such patients and suggest an algorithm. Conclusions: We hope that our article brings awareness and assists vascular surgeons in managing such cases, resulting in improved quality of life for these patients.
Collapse
Affiliation(s)
- Jolyn Hui Qing Pang
- Department of General Surgery, Singapore General Hospital, Outram Road, Singapore 169608, Singapore;
| | - Nick Zhi Peng Ng
- Department of Vascular Surgery, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
| |
Collapse
|
15
|
Ducas A, Mangano A, Borgioli L, Cassiani J, Lopez P, Giulianotti PC. The role of artificial intelligence in pancreatic surgery: Current and future perspectives. Ann Hepatobiliary Pancreat Surg 2025; 29:1-4. [PMID: 39228175 PMCID: PMC11830889 DOI: 10.14701/ahbps.24-130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024] Open
Affiliation(s)
- Alvaro Ducas
- Division of General, Minimally Invasive and Robotic Surgery, Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
| | - Alberto Mangano
- Division of General, Minimally Invasive and Robotic Surgery, Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
| | - Leonardo Borgioli
- Department of Electrical and Computer Engineering, College of Engineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Jessica Cassiani
- Division of General, Minimally Invasive and Robotic Surgery, Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
| | - Paula Lopez
- Division of General, Minimally Invasive and Robotic Surgery, Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
| | - Pier Cristoforo Giulianotti
- Division of General, Minimally Invasive and Robotic Surgery, Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
16
|
Zhang S, Jin J, Zheng Q, Wang Z. Building a cancer risk and survival prediction model based on social determinants of health combined with machine learning: A NHANES 1999 to 2018 retrospective cohort study. Medicine (Baltimore) 2025; 104:e41370. [PMID: 39928823 PMCID: PMC11813008 DOI: 10.1097/md.0000000000041370] [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: 09/12/2024] [Revised: 11/27/2024] [Accepted: 01/10/2025] [Indexed: 02/12/2025] Open
Abstract
The occurrence and progression of cancer is a significant focus of research worldwide, often accompanied by a prolonged disease course. Concurrently, researchers have identified that social determinants of health (SDOH) (employment status, family income and poverty ratio, food security, education level, access to healthcare services, health insurance, housing conditions, and marital status) are associated with the progression of many chronic diseases. However, there is a paucity of research examining the influence of SDOH on cancer incidence risk and the survival of cancer survivors. The aim of this study was to utilize SDOH as a primary predictive factor, integrated with machine learning models, to forecast both cancer risk and prognostic survival. This research is grounded in the SDOH data derived from the National Health and Nutrition Examination Survey dataset spanning 1999 to 2018. It employs methodologies including adaptive boosting, gradient boosting machine (GradientBoosting), random forest (RF), extreme gradient boosting, light gradient boosting machine, support vector machine, and logistic regression to develop models for predicting cancer risk and prognostic survival. The hyperparameters of these models-specifically, the number of estimators (100-200), maximum tree depth (10), learning rate (0.01-0.2), and regularization parameters-were optimized through grid search and cross-validation, followed by performance evaluation. Shapley Additive exPlanations plots were generated to visualize the influence of each feature. RF was the best model for predicting cancer risk (area under the curve: 0.92, accuracy: 0.84). Age, non-Hispanic White, sex, and housing status were the 4 most important characteristics of the RF model. Age, gender, employment status, and household income/poverty ratio were the 4 most important features in the gradient boosting machine model. The predictive models developed in this study exhibited strong performance in estimating cancer incidence risk and survival time, identifying several factors that significantly influence both cancer incidence risk and survival, thereby providing new evidence for cancer management. Despite the promising findings, this study acknowledges certain limitations, including the omission of risk factors in the cancer survivor survival model and potential biases inherent in the National Health and Nutrition Examination Survey dataset. Future research is warranted to further validate the model using external datasets.
Collapse
Affiliation(s)
- Shiqi Zhang
- Graduate School, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Jianan Jin
- Graduate School, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Qi Zheng
- State Key Laboratory of Infectious Disease Diagnosis and Treatment, The First Affiliated Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Infectious Disease Diagnosis and Treatment, Hangzhou, Zhejiang, PR China
| | - Zhenyu Wang
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China
| |
Collapse
|
17
|
Chu AK, Sehabi WY, Kearns E, Aubry T, Nissim R, Wheatley-Price P, Lebel S. Behind the label: a qualitative study of the self-perceptions of individuals with advanced lung cancer receiving immunotherapy or targeted therapy. Support Care Cancer 2025; 33:152. [PMID: 39907811 DOI: 10.1007/s00520-025-09152-y] [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: 05/23/2024] [Accepted: 01/03/2025] [Indexed: 02/06/2025]
Abstract
OBJECTIVES Individuals with lung cancer are living longer due to recent treatment advances such as immunotherapy and targeted therapy. The preferred label(s) of this new population are unknown. This is important as personal/advocacy benefits have been reported by using "survivor" or "thriver" by some groups; however, these same labels have been rejected by others. The goal of the present study was to explore the meaning of different labels and reasons given for preferred label(s) among people diagnosed with advanced lung cancer receiving these treatments. METHODS The study is part of a larger project conducted in partnership with Lung Cancer Canada to identify the supportive care needs of individuals with advanced lung cancer receiving immunotherapy or targeted therapy. Participants (n = 24) were recruited across Canada. In-depth qualitative interviews were coded using reflexive thematic analysis. RESULTS Labels are important and serve four functions: meaning-making, acceptance, maintaining a positive outlook, and shaping how people in participants' circle understand and react to them. Participants felt their experience did not fit traditional labels. While "survivor" was associated with maintaining a positive outlook by some, it was strongly rejected by others as it implied a cure. Some felt "patient" conveyed realism, but others found it deprived them of empowerment. "Person living with cancer" was perceived as reflecting the ongoing nature of the disease while not overly centring cancer within a person's identity. CONCLUSION Participants identified with various labels and emphasized how the label reflected their identity and psychological experiences of the disease. Thus, it is important that clinicians elicit and use individuals' preferred terms, and default to person-first language if unknown.
Collapse
Affiliation(s)
- Alanna K Chu
- School of Psychology, University of Ottawa, 136 Jean Jacques Lussier, Room 3087, Ottawa, ON, K1N6N5, Canada.
| | - Wardat Yasmine Sehabi
- School of Psychology, University of Ottawa, 136 Jean Jacques Lussier, Room 3087, Ottawa, ON, K1N6N5, Canada
| | - Emma Kearns
- School of Psychology, University of Ottawa, 136 Jean Jacques Lussier, Room 3087, Ottawa, ON, K1N6N5, Canada
| | - Tim Aubry
- School of Psychology, University of Ottawa, 136 Jean Jacques Lussier, Room 3087, Ottawa, ON, K1N6N5, Canada
| | - Rinat Nissim
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Paul Wheatley-Price
- The Ottawa Hospital, Ottawa, ON, Canada
- The Lung Cancer Canada, Ottawa, ON, Canada
| | - Sophie Lebel
- School of Psychology, University of Ottawa, 136 Jean Jacques Lussier, Room 3087, Ottawa, ON, K1N6N5, Canada
| |
Collapse
|
18
|
Zhou T, Li X, Zhao F, Zhou J, Sun B. Lactamase β reprograms lipid metabolism to inhibit the progression of endometrial cancer through attenuating MDM2-mediated p53 ubiquitination and degradation. Arch Biochem Biophys 2025; 764:110287. [PMID: 39746389 DOI: 10.1016/j.abb.2024.110287] [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: 05/30/2024] [Revised: 12/09/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Lactamase β (LACTB) inhibits the metastasis and progression of multiple malignant tumors. However, little is known about its role in endometrial cancer (EC). Our study aimed to investigate the function and potential molecular mechanism of LACTB in modulating EC progression. METHODS LACTB expression was measured via immunohistochemistry staining, Western blot and qRT-PCR. The role of LACTB in EC was investigated both in vivo and in vitro by employing xenograft mice models and using colony formation, EdU, and Transwell assays, along with flow cytometric analysis. In addition, to assess LACTB function on lipid metabolism, lipid droplets in EC cells were labeled with Nile red. Western blot, immunofluorescence staining, co-immunoprecipitation, ubiquitination assay, and cycloheximide chase assay and rescue experiments were performed to confirm the interaction between LACTB, p53, and MDM2 in EC. RESULTS LACTB expression was downregulated in EC. LACTB inhibited the malignant phenotypes and reprogramed lipid metabolism in EC cells. Moreover, LACTB significantly upregulated p53 by attenuating the MDM2-mediated ubiquitination and degradation of p53. Besides, LACTB silencing facilitated the malignant phenotypes and reprogramed lipid metabolism in EC cells; this was reversed with p53 overexpression. LACTB knockdown facilitated EC progression via downregulating p53 in vivo. CONCLUSION LACTB repressed EC cell proliferation and metastasis, and reprogramed lipid metabolism via attenuating the MDM2-mediated ubiquitination and degradation of p53.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Laboratory of Laparoscopic Technology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, Shandong, China
| | - Xiaorong Li
- Department of Obstetrics and Gynecology, Weihaiwei People's Hospital, Weihai, 264299, Shandong, China
| | - Fangfang Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Laboratory of Laparoscopic Technology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, Shandong, China
| | - Jing Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Laboratory of Laparoscopic Technology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, Shandong, China
| | - Binghui Sun
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Laboratory of Laparoscopic Technology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, Shandong, China.
| |
Collapse
|
19
|
Silk T, Hacker KE, Growdon W, Pothuri B. The advent of immune checkpoint inhibition for the treatment of patients with primary advanced or recurrent dMMR/MSI high endometrial cancer in 2025. Curr Opin Obstet Gynecol 2025; 37:22-29. [PMID: 39611619 DOI: 10.1097/gco.0000000000001004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
PURPOSE OF REVIEW The Cancer Genome Atlas identified four distinct molecular subtypes of endometrial cancer (EC): POLE mutated, mismatch repair deficient (dMMR), copy number low, and copy number high. The goal of this review is to summarize the profound clinical implications of molecular subtyping, particularly in guiding treatment decisions for dMMR and microsatellite instability high (MSI-H) EC. RECENT FINDINGS Clinical trials have demonstrated the remarkable efficacy of immunotherapy in dMMR/MSI-H EC tumors. Trials including GARNET, KEYNOTE-158, NRG GY-018, and RUBY have shown significant improvements in clinical outcomes for patients with advanced and recurrent disease, leading to FDA approvals for immunotherapy in both frontline and recurrent EC treatment settings.Building on these successes, recent studies, including DUO-E, are exploring combination therapies to enhance the efficacy of immunotherapy in EC. Simultaneously, trials including NRG GY-020, are investigating the potential benefits of immunotherapy in early-stage disease. SUMMARY Immunotherapy therapy has revolutionized the treatment of endometrial cancer in both upfront and recurrent settings, with molecular subtyping identifying patients most likely to benefit, especially those with dMMR/MSI-H tumors.
Collapse
Affiliation(s)
- Tarik Silk
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | | | | | | |
Collapse
|
20
|
Xi X, Zhang X, Tang J, Fan X, Du J. HLA-DMB correlates with antitumor immunity and an improved prognosis in endometrial carcinoma tumors. Front Oncol 2025; 14:1525601. [PMID: 39917362 PMCID: PMC11798792 DOI: 10.3389/fonc.2024.1525601] [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: 11/10/2024] [Accepted: 12/20/2024] [Indexed: 02/09/2025] Open
Abstract
Introduction Endometrial Carcinoma (UCEC) is a prevalent malignant tumor within the female reproductive system. HLA-DMB, the beta chain of the non-classical MHC class II protein HLA-DM, has been implicated in the progression of various cancers. However, its role in the development of endometrial carcinoma remains unclear. Therefore, we conducted a preliminary exploration of the prognostic value and potential mechanisms of HLA-DMB in uterine corpus endometrial carcinoma (UCEC). Methods The differential expression of HLA-DMB was analyzed in 554 tumor samples and 35 normal samples obtained from the TCGA database. The differential expression of HLA-DMB across various cancers, along with immune infiltration analysis, was conducted using the TIMER2.0 database. Additionally, the expression of HLA-DMB in endometrial carcinoma was examined in the GEPIA2 database, along with its relationship to prognosis. Furthermore, TISIDB was utilized to predict the relationships between HLA-DMB and various immune enhancement factors as well as immunosuppressive factors. Gene Ontology (GO) analysis and Gene Set Enrichment Analysis (GSEA) were employed to explore the signaling pathways associated with HLA-DMB in endometrial cancer. Univariate COX regression analysis was performed to identify prognostic factors for endometrial carcinoma (EC), and a multivariate COX proportional hazards regression model was used to confirm that HLA-DMB can serve as an independent prognostic factor for EC. The protein interaction network of HLA-DMB was constructed using the STRING database, and the chemical drugs related to HLA-DMB were predicted through the CTD database. Finally, the expression of HLA-DMB was validated by qPCR and immunohistochemistry. Results The expression of HLA-DMB at both mRNA and protein levels is significantly higher in UCEC tissues compared to normal tissues. Prognostic analyses indicate that increased expression of HLA-DMB correlates with improved patient prognosis, suggesting its potential as an independent prognostic factor for UCEC. Furthermore, in endometrial cancer, elevated levels of HLA-DMB are associated with higher immune infiltration scores and are closely related to various immune-enhancing factors. Mechanistically, HLA-DMB primarily participates in CD22-mediated regulation of B cell receptors (BCR), leading to BCR antigen activation and the production of second messengers. In our drug analysis, we identified several chemical agents associated with HLA-DMB, including cisplatin, dexamethasone, and ethinylestradiol. Discussion This study elucidates the function and underlying mechanisms of HLA-DMB in UCEC, providing a potential biomarker and target for immunotherapy in this disease.
Collapse
Affiliation(s)
- Xiaoyu Xi
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaona Zhang
- Department of Gynecology, Hengshui Fifth People’s Hospital, Hengshui, Hebei, China
| | - Jianxin Tang
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiumei Fan
- Department of Obstetrics and Gynecology, Xingtai Third Hospital, Xingtai, Hebei, China
| | - Jiexian Du
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
21
|
Zahergivar A, Golagha M, Stoddard G, Anderson PS, Woods L, Newman A, Carter MR, Wang L, Ibrahim M, Chamberlin J, Auffermann WF, Kabakus I, Burt JR. Prognostic value of coronary artery calcium scoring in patients with non-small cell lung cancer using initial staging computed tomography. BMC Med Imaging 2024; 24:350. [PMID: 39731094 DOI: 10.1186/s12880-024-01544-6] [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: 07/11/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Lung cancer is a leading cause of cancer-related mortality worldwide, with non-small cell lung cancer (NSCLC) comprising 85% of cases. Due to the lack of early clinical signs, metastasis often occurs before diagnosis, impacting treatment and prognosis. Cardiovascular disease (CVD) is a common comorbidity in lung cancer patients, with shared risk factors exacerbating outcomes. METHODS This study investigates the association between coronary artery calcium (CAC) scores, major adverse cardiovascular events (MACE), and survival outcomes in NSCLC patients, utilizing positron emission tomography-computed tomography (PET-CT) for CAC scoring. A retrospective cohort study of 154 NSCLC patients (mean age 66.3 years, 52% women) at the University of Utah (2005-2022) was conducted. Baseline PET-CT or CT imaging was used to quantify CAC scores, categorized into five risk levels. Cox proportional hazards and logistic regression analyses assessed the impact of CAC scores on survival and cardiovascular events, adjusting for confounders such as age, gender, and smoking status. RESULTS Higher CAC scores were significantly associated with increased MACE, acute myocardial infarction (MI), and poorer overall survival. The severe risk CAC score group had significantly lower survival (p = 0.022). Logistic regression revealed a strong association between higher CAC scores and MI incidence (moderate: OR = 13.8, severe: OR = 21.2) and MACE (severe: OR = 10.2). Smoking history was a significant predictor of overall survival (p = 0.006). CONCLUSION CAC scoring via PET-CT provides valuable prognostic insights in NSCLC patients, highlighting the need for integrated cardiovascular risk management in this population. Further research and advanced technologies like machine learning could enhance CAC scoring application in clinical practice. TRIAL REGISTRATION Retrospectively registered.
Collapse
Affiliation(s)
- Aryan Zahergivar
- Internal Medicine Department, District of Columbia, George Washington University, District of Columbia, Washington, USA
| | - Mahshid Golagha
- Urology Oncology Branch, National Cancer Institutes, National Institutes of Health, Bethesda, MD, USA
| | - Greg Stoddard
- Biomedical Informatics, University of Utah, Salt Lake City, Utah, USA
| | - Parker Sage Anderson
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Lacey Woods
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Anna Newman
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Malorie R Carter
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Libo Wang
- Department of Internal Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, USA
| | - Mark Ibrahim
- Department of Internal Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, USA
| | - Jordan Chamberlin
- Department of Radiology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - William F Auffermann
- Department of Radiology, Cardiothoracic Imaging, University of Utah, 30 N 1900 E #1A71, Salt Lake City, Utah, 84132, USA
| | - Ismail Kabakus
- Department of Radiology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jeremy R Burt
- Department of Radiology, Cardiothoracic Imaging, University of Utah, 30 N 1900 E #1A71, Salt Lake City, Utah, 84132, USA.
| |
Collapse
|
22
|
Berzins NJ, Orsega-Smith E, Mackenzie M, Galantino ML, Culos-Reed N, Leonard T, Narducci E. What Do We Know About Group Health Coaching and Cancer Survivorship? A Scoping Review. Am J Lifestyle Med 2024:15598276241303444. [PMID: 39659619 PMCID: PMC11626552 DOI: 10.1177/15598276241303444] [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] [Indexed: 12/12/2024] Open
Abstract
Group health coaching (GHC) may be a suitable method for supporting healthy lifestyle behaviors in cancer patients and survivors. The aim of this scoping review was to explore GHC interventions targeting this population, specifically examining program composition and measured outcomes. A systematic search strategy was used to identify intervention studies focused on GHC with cancer patients and survivors. Seven studies met the criteria. Studies focused on physical activity, diet, weight loss, or some combination thereof utilizing GHC by itself or as one component of an exercise and/or diet intervention. There was a wide range of measured outcomes, grouped into: feasibility/acceptability; physical activity/exercise; body composition and biomarkers; diet; distress, quality of life, fatigue; and other. Overall, studies were found to be feasible and showed positive results for weight loss, diet, and quality of life. Findings for changes in physical activity, distress, and fatigue were mixed. Additionally, variability was found in many of the GHC components. This review suggests GHC for cancer patients and survivors is still in the nascent stages. However, these studies were deemed feasible and satisfactory to participants, with positive outcomes noted. While still in the early stages, GHC appears promising for supporting positive lifestyle behaviors in this population.
Collapse
Affiliation(s)
| | | | | | - Mary Lou Galantino
- Stockton University, Galloway, NJ, USA (MLG)
- University of Witwatersrand Johannesburg, Johannesburg, South Africa (MLG)
| | | | - Tara Leonard
- University of Delaware, Newark, DE, USA (NJB, EO, MM, TL)
| | - Erika Narducci
- Cancer Support Community Delaware, Wilmington, DE, USA (EN)
| |
Collapse
|
23
|
Song Y, Zhang J, Li Y, Cheng L, Song H, Zhang Y, Du G, Yu S, Zou Y, Xu Q. Exploring Bioinformatics Tools to Analyze the Role of CDC6 in the Progression of Polycystic Ovary Syndrome to Endometrial Cancer by Promoting Immune Infiltration. Int J Mol Sci 2024; 25:12974. [PMID: 39684684 DOI: 10.3390/ijms252312974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Cell division cycle 6 (CDC6) is essential for the initiation of DNA replication in eukaryotic cells and contributes to the development of various human tumors. Polycystic ovarian syndrome (PCOS) is a reproductive endocrine disease in women of childbearing age, with a significant risk of endometrial cancer (EC). However, the role of CDC6 in the progression of PCOS to EC is unclear. Therefore, we examined CDC6 expression in patients with PCOS and EC. We evaluated the relationship between CDC6 expression and its prognostic value, potential biological functions, and immune infiltrates in patients with EC. In vitro analyses were performed to investigate the effects of CDC6 knockdown on EC proliferation, migration, invasion, and apoptosis. CDC6 expression was significantly upregulated in patients with PCOS and EC. Moreover, this protein caused EC by promoting the aberrant infiltration of macrophages into the immune microenvironment in patients with PCOS. A functional enrichment analysis revealed that CDC6 exerted its pro-cancer and pro-immune cell infiltration functions via the PI3K-AKT pathway. Moreover, it promoted EC proliferation, migration, and invasion but inhibited apoptosis. This protein significantly reduced EC survival when mutated. These findings demonstrate that CDC6 regulates the progression of PCOS to EC and promotes immune infiltration.
Collapse
Affiliation(s)
- Yuhang Song
- School of Basic Medicine, Xinjiang Medical University, Urumqi 830054, China
- School of Clinical Medicine, Xinjiang Medical University, Urumqi 830054, China
| | - Jing Zhang
- Department of Immunology, School of Basic Medicine, Central South University, Changsha 410017, China
| | - Yao Li
- School of Basic Medicine, Xinjiang Medical University, Urumqi 830054, China
| | - Lufeng Cheng
- Basic Medical College, Xinjiang Medical University, Urumqi 830054, China
| | - Hua Song
- School of Clinical Medicine, Xinjiang Medical University, Urumqi 830054, China
| | - Yuhang Zhang
- School of Clinical Medicine, Xinjiang Medical University, Urumqi 830054, China
| | - Guoqing Du
- School of Basic Medicine, Xinjiang Medical University, Urumqi 830054, China
| | - Sunyue Yu
- School of Clinical Medicine, Xinjiang Medical University, Urumqi 830054, China
| | - Yizhou Zou
- Department of Immunology, School of Basic Medicine, Central South University, Changsha 410017, China
| | - Qi Xu
- School of Basic Medicine, Xinjiang Medical University, Urumqi 830054, China
| |
Collapse
|
24
|
Peiretti M, Altieri A, Candotti G, Fais G, Ungredda A, Mais V, Fanni D, Angioni S. Evaluation of Different Risk Factors for Metastatic Sentinel Lymph Nodes in Endometrial Cancer. Cancers (Basel) 2024; 16:4035. [PMID: 39682221 PMCID: PMC11640191 DOI: 10.3390/cancers16234035] [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: 10/10/2024] [Revised: 11/07/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: This study investigates which demographic, clinical and pathological factors of women with early-stage presurgical EC could be considered risk factors for the presence of different subtypes of metastases in sentinel lymph nodes (SLNs). Methods: This is a retrospective single-center study that collected data between December 2015 and April 2024. EC patients who underwent total hysterectomy with salpingo-oophorectomy and SLN mapping with indocyanine green (ICG) were recorded. Results: Data from 98 women with EC were analyzed. The endometrioid histotype was present in 85 (86%) women, and the non-endometrioid histotype was present in 13 (13%) women. High-grade EC (G3) was present in 21 (21.4%) patients, and low-grade EC (G1-G2) was present in 77 (78.6%) patients. The total number of women with SLN metastasis was 21/98 (21%). Of 21 women, 5 had MAC, 6 had MIC and 10 had ITCs. Conclusions: Preliminary analysis identified three risk factors for nodal involvement: age greater than 67 years, high-grade endometrial carcinomas and myometrial invasion greater than or equal to 50%. Lymphovascular space invasion, histotype 2 and p53 mutation showed a slight, but not statistically significant, tendency to be risk factors for SLN positivity. A deeper analysis with univariate uninominal logistic regression showed that high-grade EC is related to a greater probability of MACs, as shown in other studies, and that low-grade EC (grades 1 and 2) had a strong relationship with low-volume metastasis (LVM); further studies are needed to confirm these results.
Collapse
Affiliation(s)
- Michele Peiretti
- Department of Surgical Sciences, Division of Gynaecology and Obstetrics, University of Cagliari, 09042 Cagliari, Italy; (M.P.); (G.F.); (A.U.); (V.M.); (S.A.)
| | - Alfonso Altieri
- Department of Surgical Sciences, Division of Gynaecology and Obstetrics, University of Cagliari, 09042 Cagliari, Italy; (M.P.); (G.F.); (A.U.); (V.M.); (S.A.)
| | - Giorgio Candotti
- Department of Obstetrics and Gynecology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
| | - Giuseppina Fais
- Department of Surgical Sciences, Division of Gynaecology and Obstetrics, University of Cagliari, 09042 Cagliari, Italy; (M.P.); (G.F.); (A.U.); (V.M.); (S.A.)
| | - Andrea Ungredda
- Department of Surgical Sciences, Division of Gynaecology and Obstetrics, University of Cagliari, 09042 Cagliari, Italy; (M.P.); (G.F.); (A.U.); (V.M.); (S.A.)
| | - Valerio Mais
- Department of Surgical Sciences, Division of Gynaecology and Obstetrics, University of Cagliari, 09042 Cagliari, Italy; (M.P.); (G.F.); (A.U.); (V.M.); (S.A.)
| | - Daniela Fanni
- Department of Medical Sciences, Pathology Unit, University of Cagliari, 09124 Cagliari, Italy;
| | - Stefano Angioni
- Department of Surgical Sciences, Division of Gynaecology and Obstetrics, University of Cagliari, 09042 Cagliari, Italy; (M.P.); (G.F.); (A.U.); (V.M.); (S.A.)
| |
Collapse
|
25
|
Elagawany M, Abdel Ghany LMA, Ibrahim TS, Alharbi AS, Abdel-Aziz MS, El-labbad EM, Ryad N. Development of certain benzylidene coumarin derivatives as anti-prostate cancer agents targeting EGFR and PI3Kβ kinases. J Enzyme Inhib Med Chem 2024; 39:2311157. [PMID: 38348846 PMCID: PMC10866054 DOI: 10.1080/14756366.2024.2311157] [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: 12/04/2023] [Accepted: 01/22/2024] [Indexed: 02/15/2024] Open
Abstract
Novel coumarin derivatives were synthesised and tested for their cytotoxicity against human cancer cells (PC-3 and MDA-MB-231). Compounds 5, 4b, and 4a possessed potent cytotoxic activity against PC-3 cells with IC50 3.56, 8.99, and 10.22 µM, respectively. Compound 4c displayed cytotoxicity more than erlotinib in the MDA-MB-231 cells with IC50 8.5 µM. Moreover, compound 5 exhibited potent inhibitory activity on EFGR with IC50 0.1812 µM, as well as PI3Kβ inhibitory activity that was twofold higher than LY294002, suggesting that this compound has a dual EGFR and PI3Kβ inhibiting activity. Docking aligns with the in vitro results and sheds light on the molecular mechanisms underlying dual targeting. Furthermore, compound 5 decreased AKT and m-TOR expression in PC-3 cells, showing that it specifically targets these cells via the EGFR/PI3K/Akt/m-TOR signalling pathway. Simultaneously, compound 5 caused cell cycle arrest at S phase and induced activation of both intrinsic and extrinsic apoptotic pathways.
Collapse
Affiliation(s)
- Mohamed Elagawany
- Department of Pharmaceutical Chemistry, Damanhour University, Damanhour, Buhaira, Egypt
| | - Lina M. A. Abdel Ghany
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Giza, Egypt
| | - Tarek S. Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrhman S. Alharbi
- Department of Chemistry, College of Science and Arts, Shaqra University, Sajir, Shaqra, Saudi Arabia
| | - Mohamed S. Abdel-Aziz
- Microbial Chemistry Department, Biotechnology Research Institute, National Research Centre, Cairo, Egypt
| | - Eman M. El-labbad
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman, United Arab Emirates
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| | - Noha Ryad
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Giza, Egypt
| |
Collapse
|
26
|
Byers S, Song X. Nonparametric Biomarker Based Treatment Selection With Reproducibility Data. Stat Med 2024; 43:5077-5087. [PMID: 39291682 PMCID: PMC11583955 DOI: 10.1002/sim.10218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/17/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024]
Abstract
We consider evaluating biomarkers for treatment selection under assay modification. Survival outcome, treatment, and Affymetrix gene expression data were attained from cancer patients. Consider migrating a gene expression biomarker to the Illumina platform. A recent novel approach allows a quick evaluation of the migrated biomarker with only a reproducibility study needed to compare the two platforms, achieved by treating the original biomarker as an error-contaminated observation of the migrated biomarker. However, its assumptions of a classical measurement error model and a linear predictor for the outcome may not hold. Ignoring such model deviations may lead to sub-optimal treatment selection or failure to identify effective biomarkers. To overcome such limitations, we adopt a nonparametric logistic regression to model the relationship between the event rate and the biomarker, and the deduced marker-based treatment selection is optimal. We further assume a nonparametric relationship between the migrated and original biomarkers and show that the error-contaminated biomarker leads to sub-optimal treatment selection compared to the error-free biomarker. We obtain the estimation via B-spline approximation. The approach is assessed by simulation studies and demonstrated through application to lung cancer data.
Collapse
Affiliation(s)
- Sara Byers
- Department of Epidemiology and Biostatistics, College of Public HealthUniversity of GeorgiaAthensGeorgiaUSA
| | - Xiao Song
- Department of Epidemiology and Biostatistics, College of Public HealthUniversity of GeorgiaAthensGeorgiaUSA
| |
Collapse
|
27
|
Albertí-Valls M, Olave S, Olomí A, Macià A, Eritja N. Advances in Immunotherapy for Endometrial Cancer: Insights into MMR Status and Tumor Microenvironment. Cancers (Basel) 2024; 16:3918. [PMID: 39682106 DOI: 10.3390/cancers16233918] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Endometrial cancer is one of the most common gynecological malignancies, and while early-stage cases are highly treatable, recurrent or advanced EC remains challenging to manage. Immunotherapy, particularly immune checkpoint inhibitors, has revolutionized treatment approaches in oncology, and its application in EC has shown promising results. Key to immunotherapy efficacy in EC is the tumor's mismatch repair status, with MMR-deficient tumors demonstrating a higher tumor mutational burden and increased PD-L1 expression, making them more susceptible to immune checkpoint inhibitors (ICIs) such as pembrolizumab, durvalumab, and dostarlimab. However, not all mismatch repair-deficient (MMRd) tumors respond to ICIs, particularly those with a "cold" tumor microenvironment (TME) characterized by poor immune infiltration. In contrast, some MMR-proficient tumors with a "hot" TME respond well to ICIs, underscoring the complex interplay between MMR status, tumor mutational burden (TMB), and TME. To overcome resistance in cold tumors, novel therapies, including Chimeric Antigen Receptor (CAR) T cells and tumor-infiltrating lymphocytes are being explored, offering targeted immune-based strategies to enhance treatment efficacy. This review discusses the current understanding of immunotherapy in EC, emphasizing the prognostic and therapeutic implications of MMR status, TME composition, and emerging cell-based therapies.
Collapse
Affiliation(s)
- Manel Albertí-Valls
- Oncologic Pathology Group, Biomedical Research Institute of Lleida (IRBLleida), University of Lleida (UdL), Av. Rovira Roure 80, 25198 Lleida, Spain
| | - Sara Olave
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Anna Olomí
- Developmental and Oncogenic Signaling, Biomedical Research Institute of Lleida (IRBLleida), University of Lleida (UdL), Av. Rovira Roure 80, 25198 Lleida, Spain
| | - Anna Macià
- Oncologic Pathology Group, Biomedical Research Institute of Lleida (IRBLleida), University of Lleida (UdL), Av. Rovira Roure 80, 25198 Lleida, Spain
| | - Núria Eritja
- Oncologic Pathology Group, Biomedical Research Institute of Lleida (IRBLleida), University of Lleida (UdL), Av. Rovira Roure 80, 25198 Lleida, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
28
|
Ahad A, Ejaz A, Fatima E, Sultan W, Ahmad O, Jamil A, Iqbal J, Ullah I, Alraies MC, Minhas AMK. Rising mortality related to cardiovascular disease and prostate cancer amongst older men across the United States. Curr Probl Cardiol 2024; 49:102785. [PMID: 39127433 DOI: 10.1016/j.cpcardiol.2024.102785] [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: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVE There is a significant association between cardiovascular diseases (CVD) and prostate cancer (PCa), leading to high mortality. This study evaluates the trends in mortality associated with CVDs and PCa among older (≥ 65 years) men in the United States (US). METHODS This analysis utilized the Centers for Disease Control and Prevention's Wide-Ranging Online Data for Epidemiologic Research (CDC WONDER). The analysis of Multiple Cause of Death Files was carried out from 1999 to 2019 to identify fatalities with CVD and PCa listed as either contributory or underlying causes of death. Crude and age-adjusted mortality rates (AAMRs) per 100,000 populations for variables such as year, race and ethnicity, and geographic regions were determined. To assess annual percent change (APC), a Joinpoint regression program was employed. RESULTS Overall AAMR was 54.3 in 1999 and 34.6 in 2019. After a decline in AAMR from 1999 to 2015, an alarming rise in mortality was observed until 2019. Mortality rates were highest among Non-Hispanic (NH) Black and African American men (74.9). Geographically, the highest mortalities were witnessed in the West (46.4) and non-metropolitan areas (44.6). States with AAMRs ranking in the 90th percentile were Nebraska, California, North Dakota, the District of Columbia, and Mississippi. CONCLUSION After decreasing death rates associated with CVD and PCa from 1999 to 2015, a reversal in the trend was observed from 2015 to 2019. Addressing this increase in death rates, especially among the vulnerable population, requires focused attention and targeted strategies to implement necessary safeguards in the upcoming years.
Collapse
Affiliation(s)
- Abdul Ahad
- Department of Physiology, Khyber Medical College, Peshawar, Pakistan
| | - Arooba Ejaz
- Department of Medicine, Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Wania Sultan
- Department of Medicine, Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Owais Ahmad
- Department of Medicine, Islamic International Medical College, Riphah International University, Islamabad, Pakistan
| | - Adeena Jamil
- Department of Medicine, Dow International Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Javed Iqbal
- Nursing Department, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar.
| | - Irfan Ullah
- Institute of Public Health and Social Sciences, Khyber Medical University, Peshawar, Pakistan; Department of Internal Medicine, Khyber Teaching Hospital, Peshawar, Pakistan
| | - M Chadi Alraies
- Department of Cardiology, Detroit Medical Center, Detroit, MI, USA
| | | |
Collapse
|
29
|
Holland AM, Wilson HH, Gambill BC, Lorenz WR, Salvino MJ, Rose ML, Brown KS, Tawkaliyar R, Scarola GT, Patel V, Terejanu GA, Matulay JT. The Influence of Disparities on Prostate Cancer at Diagnosis in the Charlotte Metropolitan Area. Ann Surg Oncol 2024; 31:8394-8404. [PMID: 39080130 DOI: 10.1245/s10434-024-15675-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/11/2024] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Prostate cancer (PCa) is the most diagnosed noncutaneous malignancy and second leading-cause of cancer death in men, yet screening is decreasing. As PCa screening has become controversial, socioeconomic disparities in PCa diagnosis and outcomes widen. This study was designed to determine the current disparities influencing PCa diagnosis in Charlotte, NC. METHODS The Levine Cancer Institute database was queried for patients with PCa, living in metropolitan Charlotte. Socioeconomic status (SES) was determined by the Area Deprivation Index (ADI); higher ADI indicated lower SES. Patients were compared by their National Comprehensive Cancer Network risk stratification. Artificial intelligence predictive models were trained and heatmaps were created, demonstrating the geographic and socioeconomic disparities in late-stage PCa. RESULTS Of the 802 patients assessed, 202 (25.2%) with high-risk PCa at diagnosis were compared with 198 (24.7%) with low-risk PCa. High-risk PCa patients were older (69.8 ± 9.0 vs. 64.0 ± 7.9 years; p < 0.001) with lower SES (ADI block: 98.4 ± 20.9 vs. 92.1 ± 19.8; p = 0.004) and more commonly African-American (White: 66.2% vs. 78.3%, African-American: 31.3% vs. 20.7%; p = 0.009). On regression, ADI block was an independent predictor (odds ratio [OR] = 1.013, 95% confidence interval [CI] 1.002-1.024; p = 0.024) of high-risk PCa at diagnosis, whereas race was not (OR = 1.312, 95% CI 0.782-2.201; p = 0.848). A separate regression demonstrated higher ADI (OR = 1.016, 95% CI 1.004-1.027; p = 0.006) and older age (OR = 1.083, 95% CI 1.054-1.114; p < 0.001) were independent predictors for high-risk PCa. Findings, depicted in heatmaps, demonstrated the geographic locations where men with PCa were predicted to have high-risk disease based on their age and SES. CONCLUSIONS Socioeconomic status was more closely associated with high-risk PCa at diagnosis than race. Although, of any variable, age was most predictive. The heatmaps identified areas that would benefit from increased awareness, education, and screening to facilitate an earlier PCa diagnosis.
Collapse
Affiliation(s)
- Alexis M Holland
- Division of Gastrointestinal and Minimally Invasive Surgery, Department of Surgery, Atrium Health Carolinas Medical Center, Charlotte, NC, USA
| | - Hadley H Wilson
- Division of Gastrointestinal and Minimally Invasive Surgery, Department of Surgery, Atrium Health Carolinas Medical Center, Charlotte, NC, USA
| | - Benjamin C Gambill
- Department of Computer Science, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - William R Lorenz
- Division of Gastrointestinal and Minimally Invasive Surgery, Department of Surgery, Atrium Health Carolinas Medical Center, Charlotte, NC, USA
| | | | - Mikayla L Rose
- Division of Gastrointestinal and Minimally Invasive Surgery, Department of Surgery, Atrium Health Carolinas Medical Center, Charlotte, NC, USA
| | - Kiara S Brown
- Division of Gastrointestinal and Minimally Invasive Surgery, Department of Surgery, Atrium Health Carolinas Medical Center, Charlotte, NC, USA
| | - Rahmatulla Tawkaliyar
- Division of Gastrointestinal and Minimally Invasive Surgery, Department of Surgery, Atrium Health Carolinas Medical Center, Charlotte, NC, USA
| | - Gregory T Scarola
- Division of Gastrointestinal and Minimally Invasive Surgery, Department of Surgery, Atrium Health Carolinas Medical Center, Charlotte, NC, USA
| | - Vipul Patel
- Division of Urologic Oncology, Department of Urology, Advent Health Orlando and Advent Health Cancer Institute, Celebration, FL, USA
| | - Gabriel A Terejanu
- Department of Computer Science, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Justin T Matulay
- Division of Urologic Oncology, Department of Urology, Atrium Health Carolinas Medical Center and Levine Cancer Institute, Charlotte, NC, USA.
| |
Collapse
|
30
|
Seo J, Ha G, Lee G, Nasiri R, Lee J. Modeling tumor-immune interactions using hybrid spheroids and microfluidic platforms for studying tumor-associated macrophage polarization in melanoma. Acta Biomater 2024:S1742-7061(24)00629-9. [PMID: 39461691 DOI: 10.1016/j.actbio.2024.10.036] [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: 07/16/2024] [Revised: 10/19/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
Tumor-associated macrophages (TAMs), as key components of tumor microenvironment (TME), exhibit phenotypic plasticity in response to environmental cues, causing polarization into either pro-inflammatory M1 phenotypes or immunosuppressive M2 phenotypes. Although TAM has been widely studied for its crucial involvement in the initiation, progression, metastasis, and immune regulation of cancer cells, there have been limited attempts to understand how the metastatic potentials of cancer cells influence TAM polarization within TME. Here, we developed a miniaturized TME model using a 3D hybrid system composed of murine melanoma cells and macrophages, aiming to investigate interactions between cancer cells exhibiting various metastatic potentials and macrophages within TME. The increase in spheroid size within this model was associated with a reduction in cancer cell viability. Examining macrophage surface marker expression and cytokine secretion indicated the development of diverse TMEs influenced by both spheroid size and the metastatic potential of cancer cells. Furthermore, a high-throughput microfluidic platform equipped with trapping systems and hybrid spheroids was employed to simulate the tumor-immune system of complex TMEs and for comparative analysis with traditional 3D culture models. This study provides insight into TAM polarization in melanoma with different heterogeneities by modeling cancer-immune systems, which can be potentially employed for immune-oncology research, drug screening, and personalized therapy. STATEMENT OF SIGNIFICANCE: This study presents the development of a 3D hybrid spheroid system designed to model tumor-immune interactions, providing a detailed analysis of how melanoma cell metastatic potential influences tumor-associated macrophage (TAM) polarization. By utilizing a microfluidic platform, we are able to replicate and investigate the complex tumor-immune system of the tumor microenvironments (TMEs) under continuous flow conditions. Our model holds significant potential for high-throughput drug screening and personalized medicine applications, offering a versatile tool for advancing cancer research and treatment strategies.
Collapse
Affiliation(s)
- Junki Seo
- Division of Interdisciplinary Bioscience & Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Giheon Ha
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Geonho Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Rohollah Nasiri
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, United States
| | - Junmin Lee
- Division of Interdisciplinary Bioscience & Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea; Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Incheon 21983, Republic of Korea.
| |
Collapse
|
31
|
Chen X, Li Y, Su J, Zhang L, Liu H. Progression in Near-Infrared Fluorescence Imaging Technology for Lung Cancer Management. BIOSENSORS 2024; 14:501. [PMID: 39451714 PMCID: PMC11506746 DOI: 10.3390/bios14100501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/01/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024]
Abstract
Lung cancer is a major threat to human health and a leading cause of death. Accurate localization of tumors in vivo is crucial for subsequent treatment. In recent years, fluorescent imaging technology has become a focal point in tumor diagnosis and treatment due to its high sensitivity, strong selectivity, non-invasiveness, and multifunctionality. Molecular probes-based fluorescent imaging not only enables real-time in vivo imaging through fluorescence signals but also integrates therapeutic functions, drug screening, and efficacy monitoring to facilitate comprehensive diagnosis and treatment. Among them, near-infrared (NIR) fluorescence imaging is particularly prominent due to its improved in vivo imaging effect. This trend toward multifunctionality is a significant aspect of the future advancement of fluorescent imaging technology. In the past years, great progress has been made in the field of NIR fluorescence imaging for lung cancer management, as well as the emergence of new problems and challenges. This paper generally summarizes the application of NIR fluorescence imaging technology in these areas in the past five years, including the design, detection principles, and clinical applications, with the aim of advancing more efficient NIR fluorescence imaging technologies to enhance the accuracy of tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Xinglong Chen
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China; (X.C.); (Y.L.); (J.S.)
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Yuning Li
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China; (X.C.); (Y.L.); (J.S.)
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Jialin Su
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China; (X.C.); (Y.L.); (J.S.)
| | - Lemeng Zhang
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China; (X.C.); (Y.L.); (J.S.)
| | - Hongwen Liu
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China;
| |
Collapse
|
32
|
Rowlands CE, Folberg AM, Beickman ZK, Devor EJ, Leslie KK, Givens BE. Particles and Prejudice: Nanomedicine Approaches to Reducing Health Disparities in Endometrial Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300096. [PMID: 37312613 PMCID: PMC10716380 DOI: 10.1002/smll.202300096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/25/2023] [Indexed: 06/15/2023]
Abstract
Endometrial cancer is the most common gynecological malignancy worldwide and unfortunately has a much higher mortality rate in Black women compared with White women. Many potential factors contribute to these mortality rates, including the underlying effects of systemic and interpersonal racism. Furthermore, other trends in medicine have potential links to these rates including participation in clinical trials, hormone therapy, and pre-existing health conditions. Addressing the high incidence and disparate mortality rates in endometrial cancer requires novel methods, such as nanoparticle-based therapeutics. These therapeutics have been growing in increasing prevalence in pre-clinical development and have far-reaching implications in cancer therapy. The rigor of pre-clinical studies is enhanced by the likeness of the model to the human body. In systems for 3D cell culture, for example, the extracellular matrix mimics the tumor more closely. The increasing emphasis on precision medicine can be applied to cancer using nanoparticle-based methods and applied to pre-clinical models by using patient-derived model data. This review highlights the intersections of nanomedicine, precision medicine, and racial disparities within endometrial cancer and provides insights into reducing health disparities using recent scientific advances on the nanoscale.
Collapse
Affiliation(s)
- Claire E Rowlands
- Department of Chemical and Materials Engineering, University of Kentucky, 512 Administration Drive, Lexington, KY, 40506, USA
| | - Abigail M Folberg
- Department of Psychology, University of Nebraska at Omaha, 6100 W. Dodge Road, ASH 347E, Omaha, NE, 68182, USA
| | - Zachary K Beickman
- Department of Chemical Engineering, Purdue University, 480 Stadium Mall Drive, West Lafayette, IN, 47907, USA
| | - Eric J Devor
- Department of Obstetrics and Gynecology, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Kimberly K Leslie
- Division of Molecular Medicine, Department of Internal Medicine, Department of Obstetrics and Gynecology, The University of New Mexico Comprehensive Cancer Center | The University of New Mexico Health Sciences Center, 1021 Medical Arts Ave NE, Albuquerque, NM, 87131, USA
| | - Brittany E Givens
- Department of Chemical and Materials Engineering, University of Kentucky, 512 Administration Drive, Lexington, KY, 40506, USA
| |
Collapse
|
33
|
He W, Liu W, Liu X, Tan W. The mechanism of L1 cell adhesion molecule interacting with protein tyrosine kinase 2 to regulate the focal adhesion kinase-growth factor receptor-bound protein 2-son of sevenless-rat sarcoma pathway in the identification and treatment of type I high-risk endometrial cancer. Cytojournal 2024; 21:34. [PMID: 39563667 PMCID: PMC11574687 DOI: 10.25259/cytojournal_50_2024] [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: 04/24/2024] [Accepted: 09/06/2024] [Indexed: 11/21/2024] Open
Abstract
Objective The objective of this study was to investigate how L1 cell adhesion molecule (L1CAM) interacting with protein tyrosine kinase 2 (PTK2) affects endometrial cancer (EC) progression and determine its association with the focal adhesion kinase (FAK)-growth factor receptor-bound protein 2 (GRB2)-son of sevenless (SOS)-rat sarcoma (RAS) pathway. EC is a female cancer of major concern in the world, and its incidence has increased rapidly in recent years. L1CAM is considered a reliable marker of poor prognosis in patients with EC. Material and Methods A single-center and prospective study was conducted using data from the Cancer Genome Atlas and samples from normal and EC tissues to explore the differential expression of L1CAM. Additional experimental models included human immortalized endometrial epithelium cells (hEECs) and EC cell lines such as KLE, RL95-2, and Ishikawa. L1CAM expression was regulated using lentiviruses designed for either overexpression or interference, and PTK2/focal adhesion kinase (FAK) signaling was inhibited with PF431396. Transfected KLE cells were injected into mice, and tumor growth was monitored over 14 days. Cellular proliferation and survival were assessed using cell counting kit, colony formation, and terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate (dUTP) nick-end labeling assays. Metastatic behavior was evaluated through Transwell assays for cell migration and invasion. The expression levels of matrix metallopeptidase (MMP) 2 and MMP9 were determined by Western blot. In addition, the activation of the FAK-GRB2-SOS-RAS pathway was examined by assessing the protein levels of FAK, GRB2, SOS, and RAS. Results There was a significant difference in L1CAM expression between EC tumor tissues and normal tissues, and L1CAM messenger RNA (1.85-fold) and L1CAM protein (2.59-fold) were significantly more expressed in EC tissues (P < 0.01) than in normal tissues. The tumor growth of L1CAM overexpressing EC cells was faster than that of negative control EC cells (6.43 fold; P < 0.001). L1CAM promoted the expression of FAK (1.43-2.72-fold; P < 0.001); enhanced EC cell proliferation (P < 0.01), survival and motility (P < 0.001), migration (P < 0.001), and invasion (P < 0.001); and activated the FAK-GRB2-SOS-RAS pathway, all of which were reversed when FAK expression was not upregulated (P < 0.001). Conclusion By upregulating PTK2 and its encoded protein FAK, L1CAM was found to promote tumor progression and increase the activation of the FAK-GRB2-SOS-RAS pathway. These findings establish L1CAM and PTK2 as reference genes for poor prognostic prediction in EC and as targets for EC therapy, providing a valuable basis for distinguishing between benign and malignant endometrial conditions and justifying the necessity of targeted therapeutic approaches.
Collapse
Affiliation(s)
- Wei He
- Department of Gynaecology and Obstetrics, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Liu
- Department of Gynaecology and Obstetrics, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiumei Liu
- Department of Gynecology and Oncology, Maternal and Child Care Health Hospital of Qinhuangdao, Qinhuangdao, China
| | - Wenhua Tan
- Department of Gynaecology and Obstetrics, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
34
|
Gao T, Xiang C, Ding X, Xie M. Dual-locked fluorescent probes for precise diagnosis and targeted treatment of tumors. Heliyon 2024; 10:e38174. [PMID: 39381214 PMCID: PMC11458960 DOI: 10.1016/j.heliyon.2024.e38174] [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] [Received: 07/02/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
Cancer continues to pose a significant threat to global health, with its high mortality rates largely attributable to delayed diagnosis and non-specific treatments. Early and accurate diagnosis is crucial, yet it remains challenging due to the subtle and often undetectable early molecular changes. Traditional single-target fluorescent probes often fail to accurately identify cancer cells, relying solely on single biomarkers and consequently leading to high rates of false positives and inadequate specificity. In contrast, dual-locked fluorescent probes represent a breakthrough, designed to enhance diagnostic precision. By requiring the simultaneous presence of two specific tumor-associated biomarkers or microenvironmental conditions, these probes significantly reduce non-specific activations typical of conventional single-analyte probes. This review discusses the structural designs, response mechanisms, and biological applications of dual-locked probes, highlighting their potential in tumor imaging and treatment. Importantly, the review addresses the challenges, and perspectives in this field, offering a comprehensive look at the current state and future potential of dual-locked fluorescent probes in oncology.
Collapse
Affiliation(s)
- Tang Gao
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Can Xiang
- Department of Scientific Management, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xintao Ding
- Department of Biomedical Informatics, Columbia University Graduate School of Arts and Sciences, New York, NY, United States
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
35
|
Li J, Zhou M, Xie J, Chen J, Yang M, Ye C, Cheng S, Liu M, Li R, Tan R. Organoid modeling meets cancers of female reproductive tract. Cell Death Discov 2024; 10:410. [PMID: 39333482 PMCID: PMC11437045 DOI: 10.1038/s41420-024-02186-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
Diseases of the female reproductive system, especially malignant tumors, pose a serious threat to women's health worldwide. One of the key factors limiting research progress in this area is the lack of representative models. Organoid technology, especially tumor organoids, has been increasingly applied in the study of female reproductive system tumors due to their high heterogeneity, close resemblance to the physiological state, easy acquisition and cultivation advantages. They play a significant role in understanding the origin and causes of tumors, drug screening, and personalized treatment and more. This article reviews the organoid models for the female reproductive system, focusing on the cancer research advancements. It discusses the methods for constructing tumor organoids of the female reproductive tract and summarizes the limitations of current research. The aim is to offer a reference for future development and application of these organoid models, contributing to the advancement of anti-tumor drugs and treatment strategies for female reproductive tract cancer patients.
Collapse
Affiliation(s)
- Jiao Li
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Mengting Zhou
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Xie
- Information Technology Center, West China Hospital of Sichuan University, Sichuan University, Chengdu, China
| | - Jiani Chen
- Chongqing Medical University, Chongqing, China
| | - Mengni Yang
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Changjun Ye
- Rehabilitation Department, Changgeng Yining Hospital, Wenzhou, China
| | - Shihu Cheng
- Geriatric Department, Changgeng Yining Hospital, Wenzhou, China
| | - Miao Liu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| |
Collapse
|
36
|
Yan B, Zhao T, Deng Y, Zhang Y. Preoperative prediction of lymph node metastasis in endometrial cancer patients via an intratumoral and peritumoral multiparameter MRI radiomics nomogram. Front Oncol 2024; 14:1472892. [PMID: 39364314 PMCID: PMC11446724 DOI: 10.3389/fonc.2024.1472892] [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: 07/30/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024] Open
Abstract
Introduction While lymph node metastasis (LNM) plays a critical role in determining treatment options for endometrial cancer (EC) patients, the existing preoperative methods for evaluating the lymph node state are not always satisfactory. This study aimed to develop and validate a nomogram based on intra- and peritumoral radiomics features and multiparameter magnetic resonance imaging (MRI) features to preoperatively predict LNM in EC patients. Methods Three hundred and seventy-four women with histologically confirmed EC were divided into training (n = 220), test (n = 94), and independent validation (n = 60) cohorts. Radiomic features were extracted from intra- and peritumoral regions via axial T2-weighted imaging (T2WI) and apparent diffusion coefficient (ADC) mapping. A radiomics model (annotated as the Radscore) was established using the selected features from different regions. The clinical parameters were statistically analyzed. A nomogram was developed by combining the Radscore and the most predictive clinical parameters. Decision curve analysis (DCA) and the net reclassification index (NRI) were used to assess the clinical benefit of using the nomogram. Results Nine radiomics features were ultimately selected from the intra- and peritumoral regions via ADC mapping and T2WI. The nomogram combining the Radscore, serum CA125 level, and tumor area ratio achieved the highest AUCs in the training, test and independent validation sets (nomogram vs. Radscore vs. clinical model: 0.878 vs. 0.850 vs. 0.674 (training), 0.877 vs. 0.838 vs. 0.668 (test), and 0.864 vs. 0.836 vs. 0.618 (independent validation)). The DCA and NRI results revealed the nomogram had greater diagnostic performance and net clinical benefits than the Radscore alone. Conclusion The combined intra- and peritumoral region multiparameter MRI radiomics nomogram showed good diagnostic performance and could be used to preoperatively predict LNM in patients with EC.
Collapse
Affiliation(s)
- Bin Yan
- Department of Radiology, Shaanxi Provincial Tumor Hospital, Xi’an, China
| | - Tingting Zhao
- Department of Medical Imaging, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Ying Deng
- Department of Radiology, Shaanxi Provincial Tumor Hospital, Xi’an, China
| | - Yili Zhang
- Department of Medical Oncology, Shaanxi Provincial Tumor Hospital, Xi’an, China
| |
Collapse
|
37
|
Gong X, Wang Z, You J, Gao J, Chen K, Chu J, Sui X, Dang J, Liu X. Pyroptosis-associated genes and tumor immune response in endometrial cancer. Discov Oncol 2024; 15:433. [PMID: 39264524 PMCID: PMC11393226 DOI: 10.1007/s12672-024-01315-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024] Open
Abstract
The occurrence and progression of tumors are linked to the process of pyroptosis. However, the precise involvement of pyroptosis-associated genes (PRGs) in endometrial cancer (EC) remains uncertain. 29 PRGs were identified as being either up-regulated or down-regulated in EC. PRGs subgroup analysis demonstrated distinct survival outcomes and diverse responses to chemotherapy and immune checkpoint blockade therapy. A higher expression of GPX4 and NOD2, coupled with lower levels of CASP6, PRKACA, and NLRP2, were found to be significantly associated with higher overall survival (OS) rates (p < 0.05). Conversely, lower expression of NOD2 was linked to lower progression-free survival (p = 0.021) and advanced tumor stage(p = 0.0024). NOD2, NLRP2, and TNM stages were identified as independent prognostic factors (p < 0.001). The LASSO prognostic model exhibited a notable decrease in OS among EC patients in the high-risk score group (ROC-AUC10-years: 0.799, p = 0.00644). Furthermore, NOD2 displayed a positive correlation with the infiltration of immune cells and the expression of immune checkpoints (p < 0.001). GPX4 and CASP6 are significantly associated with TMB and MSI (RTMB = 0.39; RMSI = 0.23). Additionally, a substantial upregulation of NOD2 was confirmed in both EC cells and tissue, indicating a positive relationship between advanced TNM stage (p < 0.0001) and infiltration of M1 phenotype macrophages. Nonetheless, its impact on patient OS did not reach statistical significance (p = 0.141). Our findings have contributed to the advancement of a prognostic model for EC patients. NOD2 receptor-mediated pyroptosis mechanism potentially regulates tumor immunity and promotes the transformation of macrophages from the M2 phenotype to the M1 phenotype, which significantly impacts the progression of EC.
Collapse
Affiliation(s)
- Xiaodi Gong
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| | - Zhifeng Wang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Jiahao You
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Jinghai Gao
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Kun Chen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Jing Chu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Xiaoxin Sui
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Jianhong Dang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| | - Xiaojun Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| |
Collapse
|
38
|
Nassief G, Anaeme A, Moussa K, Mansour AN, Ansstas G. Recent Advancements in Cell-Based Therapies in Melanoma. Int J Mol Sci 2024; 25:9848. [PMID: 39337333 PMCID: PMC11432154 DOI: 10.3390/ijms25189848] [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: 08/19/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Malignant melanoma outcomes have drastically changed in recent years due to the introduction of immune checkpoint inhibitors (ICIs). However, many patients still experience intolerable side effects, therapy resistance, and disease progression on ICI therapy. Therefore, there remains a need for novel therapeutics that address this gap in treatment options. Cell-based therapies have gained wide attention as a therapeutic option that could address this gap in treatment options for advanced melanoma. These therapies work by extracting certain cell types produced in the human body such as T-cells, modifying them based on a specific target, and transfusing them back into the patient. In the realm of cancer therapy, cell-based therapies utilize immune cells to target tumor cells while sparing healthy cells. Recently, the Food and Drug Administration (FDA) has approved the usage of lifileucel, a tumor-infiltrating lymphocyte (TIL) therapy, in advanced melanoma. This came following recent results from the C-144-01 study (NCT02360579), which demonstrated the efficacy and safety of TILs in metastatic melanoma patients who otherwise failed on standard ICI/targeted therapy. Thus, the results of this trial as well as the recent FDA approval have proven the viability of utilizing cell-based therapies to fill the gap in treatment options for patients with advanced melanoma. This review aims to provide a comprehensive overview of major cell-based therapies that have been utilized in melanoma by delineating results of the most recent multi-center phase II/ III clinical trials that evaluate the efficacy and safety of major cell-based therapies in melanoma. Additionally, we provide a summary of current limitations in each cell-based therapeutic option as well as a future direction of how to further extrapolate these cell-based therapies in advanced melanoma.
Collapse
Affiliation(s)
- George Nassief
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63110, USA
| | - Angela Anaeme
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63110, USA
| | - Karen Moussa
- UMKC School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Abdallah N Mansour
- Department of Medicine, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - George Ansstas
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63110, USA
| |
Collapse
|
39
|
Gomez D, Feng JJ, Cheok S, Shah I, Dicharry H, Cote DJ, Briggs RG, Guerra GA, Peterson R, Salhia B, Neman J, Attenello F, Chow F, Musabelliu EK, Zada G. Incidence of brain metastasis according to patient race and primary cancer origin: a systematic review. J Neurooncol 2024; 169:457-467. [PMID: 38896356 PMCID: PMC11341633 DOI: 10.1007/s11060-024-04748-6] [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: 05/20/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE A systematic review was conducted to investigate differences in incidence and primary origin of synchronous brain metastasis (sBM) in varying racial groups with different primary cancers. METHODS Adhering to PRISMA 2020 guidelines a search was conducted using PubMed and Ovid databases for publications from January 2000 to January 2023, with search terms including combinations of "brain metastasis," "race," "ethnicity," and "incidence." Three independent reviewers screened for inclusion criteria encompassing studies clearly reporting primary cancer sites, patient demographics including race, and synchronous BM (sBM) incidence. RESULTS Of 806 articles, 10 studies comprised of mainly adult patients from the United States met final inclusion for data analysis. Higher sBM incidence proportions were observed in American Indian/Alaska native patients for primary breast (p < 0.001), colorectal (p = 0.015), and esophageal cancers (p = 0.024) as well as in Asian or Pacific islanders for primary stomach (p < 0.001), thyroid (p = 0.006), and lung/bronchus cancers (p < 0.001) yet higher proportions in White patients for malignant melanoma (p < 0.001). Compared to White patients, Black patients had higher sBM incidence likelihood in breast cancer (OR = 1.27, p = 0.01) but lower likelihood in renal (OR = 0.46, p < 0.001) and esophageal cancers (OR = 0.31, p = 0.005). American Indian/Alaska native patients had a higher sBM likelihood (OR = 3.78, p = 0.004) relative to White patients in esophageal cancer. CONCLUSIONS These findings reveal several comparative racial differences in sBM incidence arising from different primary cancer origins, underscoring a need for further research to explain these variations. Identifying the factors contributing to these disparities holds the potential to promote greater equity in oncological care according to cancer type.
Collapse
Affiliation(s)
- David Gomez
- Department of Neurosurgery Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| | - Jeffrey J Feng
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Stephanie Cheok
- Department of Neurological Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ishan Shah
- Department of Neurosurgery Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Holly Dicharry
- LSU Health Shreveport School of Medicine, Louisiana State University, Shreveport, LA, 71103, USA
| | - David J Cote
- Department of Neurosurgery Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Robert G Briggs
- Department of Neurosurgery Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Gage A Guerra
- Department of Neurosurgery Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Racheal Peterson
- Department of Neurosurgery Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Bodour Salhia
- Department of Neurosurgery Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
| | - Josh Neman
- Department of Neurosurgery Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
| | - Frank Attenello
- Department of Neurosurgery Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Frances Chow
- Department of Neurosurgery Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Neurology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Erion K Musabelliu
- Krembil Research Institute, University Health Network, and University of Toronto, Toronto, ON, Canada
| | - Gabriel Zada
- Department of Neurosurgery Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| |
Collapse
|
40
|
Itani MI, Farah B, Wasvary M, Wadehra A, Wilson T, Rutledge B, Naylor P, Beal EW, Mutchnick M. Impact of DAA Treatment for HCV on Hepatocellular Carcinoma in a Predominately African American Population. J Gastrointest Cancer 2024; 55:1324-1332. [PMID: 38972941 DOI: 10.1007/s12029-024-01076-w] [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] [Accepted: 05/27/2024] [Indexed: 07/09/2024]
Abstract
PURPOSE This study tested the hypothesis that our predominately AA medical center population would demonstrate a decline in HCV-driven HCC diagnosis following the initiation of DAA treatment in 2014. Also evaluated was whether achieving an SVR prior to diagnosis of HCC improved outcomes in patients who had an HCV diagnosis after completion of treatment. METHODS All patients with HCC seen at the Detroit Medical Center from 2009 to 2021 were identified using ICD-10 codes, and medical records were evaluated. Outcomes were evaluated as either alive or death/hospice as of December of 2022. RESULTS There were 461 patients with HCC of whom 433 (94%) had racial information in the database (AA = 351; non-AA = 82). HCC incidence regardless of race peaked in 2017, with a subsequent decline through 2021. HCV as a risk factor was higher in AA as compared to non-AA (85% vs. 53% p = 0.0001). Outcome (alive vs. death/hospice) was better for SVR patients compared to untreated patients (54% vs. 19%; p = 0.0009). HCC patients who achieved SVR also had better liver function at diagnosis as defined by Child-Pugh score (74% vs. 49% Class A p = 0.04) at the time of diagnosis. CONCLUSIONS Racial disparity in HCC etiology was confirmed with AA more likely to have HCV than non-AA. The reduction in HCC patients with HCV confirms the impact of DAA treatment and prior successful treatment of HCV yields better outcomes. Increasing HCV treatment rates especially in AA patients will have a major impact on HCC development and treatment outcomes. WHAT IS KNOWN • African Americans are more likely to have HCV infection as compared to non-AA. • Hepatocellular carcinoma is increasing in incidence in the US. • The role of HCV in the development of HCC remains to be further investigated. WHAT IS NEW • HCC diagnosis in a single urban medical center study increased from 2009 as a result of HCV as a risk factor. • HCC declined post 2018 due primarily to a reduction in HCV infection as the risk factor. • African Americans were more likely to have HCV as the risk factor as compared to non-AA patients who were more likely to have no known risk factor on record (i.e., cryptogenic).
Collapse
Affiliation(s)
- Mohamad I Itani
- Department of Internal Medicine, Division of Gastroenterology & Hepatology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Bassem Farah
- Department of Internal Medicine, Division of Gastroenterology & Hepatology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Margaret Wasvary
- Department of Internal Medicine, Division of Gastroenterology & Hepatology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Anshu Wadehra
- Department of Internal Medicine, Division of Gastroenterology & Hepatology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Tj Wilson
- Department of Internal Medicine, Division of Gastroenterology & Hepatology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Brian Rutledge
- Department of Internal Medicine, Division of Gastroenterology & Hepatology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Paul Naylor
- Department of Internal Medicine, Division of Gastroenterology & Hepatology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| | - Eliza W Beal
- Departments of Surgery and Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Milton Mutchnick
- Department of Internal Medicine, Division of Gastroenterology & Hepatology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
41
|
Truesdell P, Chang J, Coto Villa D, Dai M, Zhao Y, McIlwain R, Young S, Hiley S, Craig AW, Babak T. Pharmacogenomic discovery of genetically targeted cancer therapies optimized against clinical outcomes. NPJ Precis Oncol 2024; 8:186. [PMID: 39198692 PMCID: PMC11358483 DOI: 10.1038/s41698-024-00673-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Despite the clinical success of dozens of genetically targeted cancer therapies, the vast majority of patients with tumors caused by loss-of-function (LoF) mutations do not have access to these treatments. This is primarily due to the challenge of developing a drug that treats a disease caused by the absence of a protein target. The success of PARP inhibitors has solidified synthetic lethality (SL) as a means to overcome this obstacle. Recent mapping of SL networks using pooled CRISPR-Cas9 screens is a promising approach for expanding this concept to treating cancers driven by additional LoF drivers. In practice, however, translating signals from cell lines, where these screens are typically conducted, to patient outcomes remains a challenge. We developed a pharmacogenomic (PGx) approach called "Clinically Optimized Driver Associated-PGx" (CODA-PGX) that accurately predicts genetically targeted therapies with clinical-stage efficacy in specific LoF driver contexts. Using approved targeted therapies and cancer drugs with available real-world evidence and molecular data from hundreds of patients, we discovered and optimized the key screening principles predictive of efficacy and overall patient survival. In addition to establishing basic technical conventions, such as drug concentration and screening kinetics, we found that replicating the driver perturbation in the right context, as well as selecting patients where those drivers are genuine founder mutations, were key to accurate translation. We used CODA-PGX to screen a diverse collection of clinical stage drugs and report dozens of novel LoF genetically targeted opportunities; many validated in xenografts and by real-world evidence. Notable examples include treating STAG2-mutant tumors with Carboplatin, SMARCB1-mutant tumors with Oxaliplatin, and TP53BP1-mutant tumors with Etoposide or Bleomycin.
Collapse
Affiliation(s)
- Peter Truesdell
- Leapfrog Bio, San Mateo, USA
- Cancer Biology & Genetics, Queen's Cancer Research Institute; Queen's University, Kingston, Canada
| | | | | | - Meiou Dai
- Leapfrog Bio, San Mateo, USA
- Cancer Biology & Genetics, Queen's Cancer Research Institute; Queen's University, Kingston, Canada
| | - Yulei Zhao
- Cancer Biology & Genetics, Queen's Cancer Research Institute; Queen's University, Kingston, Canada
- Department of Cellular and Genetic Medicine, Frontier innovation center, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Robin McIlwain
- Leapfrog Bio, San Mateo, USA
- Cancer Biology & Genetics, Queen's Cancer Research Institute; Queen's University, Kingston, Canada
| | - Stephanie Young
- Cancer Biology & Genetics, Queen's Cancer Research Institute; Queen's University, Kingston, Canada
| | - Shawna Hiley
- Third Degree Science Communication, Edmonton, Canada
| | - Andrew W Craig
- Cancer Biology & Genetics, Queen's Cancer Research Institute; Queen's University, Kingston, Canada
| | - Tomas Babak
- Leapfrog Bio, San Mateo, USA.
- Department of Biology; Queen's University, Kingston, Canada.
| |
Collapse
|
42
|
González-Martínez S, Pérez-Mies B, Cortés J, Palacios J. Single-cell RNA sequencing in endometrial cancer: exploring the epithelial cells and the microenvironment landscape. Front Immunol 2024; 15:1425212. [PMID: 39229264 PMCID: PMC11368840 DOI: 10.3389/fimmu.2024.1425212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/06/2024] [Indexed: 09/05/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) technology has emerged as a powerful tool for dissecting cellular heterogeneity and understanding the intricate biology of diseases, including cancer. Endometrial cancer (EC) stands out as the most prevalent gynecological malignancy in Europe and the second most diagnosed worldwide, yet its cellular complexity remains poorly understood. In this review, we explore the contributions of scRNA-seq studies to shed light on the tumor cells and cellular landscape of EC. We discuss the diverse tumoral and microenvironmental populations identified through scRNA-seq, highlighting the implications for understanding disease progression. Furthermore, we address potential limitations inherent in scRNA-seq studies, such as technical biases and sample size constraints, emphasizing the need for larger-scale research encompassing a broader spectrum of EC histological subtypes. Notably, a significant proportion of scRNA-seq analyses have focused on primary endometrioid carcinoma tumors, underscoring the need to incorporate additional histological and aggressive types to comprehensively capture the heterogeneity of EC. By critically evaluating the current state of scRNA-seq research in EC, this review underscores the importance of advancing towards more comprehensive studies to accelerate our understanding of this complex disease.
Collapse
Affiliation(s)
- Silvia González-Martínez
- “Contigo Contra el Cáncer de la Mujer” Foundation, Madrid, Spain
- Molecular Pathology of Cancer Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- Centre for Biomedical Research in Cancer Networks (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Belén Pérez-Mies
- Molecular Pathology of Cancer Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- Centre for Biomedical Research in Cancer Networks (CIBERONC), Carlos III Health Institute, Madrid, Spain
- Department of Pathology, Ramón y Cajal University Hospital, Madrid, Spain
- Faculty of Medicine, University of Alcalá, Madrid, Spain
| | - Javier Cortés
- “Contigo Contra el Cáncer de la Mujer” Foundation, Madrid, Spain
- Centre for Biomedical Research in Cancer Networks (CIBERONC), Carlos III Health Institute, Madrid, Spain
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron-salud Group, Barcelona, Spain
- Medica Scientia Innovation Research, Barcelona, Spain
- Medica Scientia Innovation Research, Ridgewood, NJ, United States
- Department of Medicine, Faculty of Biomedical and Health Sciences, European University of Madrid, Madrid, Spain
- IOB Institute of Oncology Madrid, Hospital Beata María Ana de Jesús, Madrid, Spain
| | - José Palacios
- Molecular Pathology of Cancer Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- Centre for Biomedical Research in Cancer Networks (CIBERONC), Carlos III Health Institute, Madrid, Spain
- Department of Pathology, Ramón y Cajal University Hospital, Madrid, Spain
- Faculty of Medicine, University of Alcalá, Madrid, Spain
| |
Collapse
|
43
|
Murakami K, Ganguly S. The Nectin family ligands, PVRL2 and PVR, in cancer immunology and immunotherapy. Front Immunol 2024; 15:1441730. [PMID: 39156900 PMCID: PMC11327090 DOI: 10.3389/fimmu.2024.1441730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
In recent years, immunotherapy has emerged as a crucial component of cancer treatment. However, its efficacy remains limited across various cancer types, highlighting unmet needs. Poliovirus receptor-related 2 (PVRL2) and Poliovirus receptor (PVR) are members of the Nectin and Nectin-like Molecules family, known for their role as cell-cell adhesion molecules. With the development of immunotherapy, their involvement in tumor immune mechanisms as immune checkpoint factors has garnered significant attention. PVRL2 and PVR are predominantly expressed on tumor cells and antigen-presenting cells, binding to PVRIG and TIGIT, respectively, which are primarily found on T and NK cells, thereby suppressing antitumor immunity. Notably, gynecological cancers such as ovarian and endometrial cancers exhibit high expression levels of PVRL2 and PVR, with similar trends observed in various other solid and hematologic tumors. Targeting these immune checkpoint pathways offers a promising therapeutic avenue, potentially in combination with existing treatments. However, the immunomodulatory mechanism involving these bindings, known as the DNAM-1 axis, is complex, underscoring the importance of understanding it for developing novel therapies. This article comprehensively reviews the immunomodulatory mechanisms centered on PVRL2 and PVR, elucidating their implications for various cancer types.
Collapse
Affiliation(s)
| | - Sudipto Ganguly
- The Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
44
|
Xu J, Liao J, Yan Q, Jiao J, Hu N, Zhang W, Shi L, Deng M, Huang S, Tang X. Trends analysis of cancer incidence, mortality, and survival for the elderly in the United States, 1975-2020. Cancer Med 2024; 13:e70062. [PMID: 39082934 PMCID: PMC11289898 DOI: 10.1002/cam4.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 07/02/2024] [Accepted: 07/20/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND Cancer burden from the elderly has been rising largely due to the aging population. However, research on the long-term epidemiological trends in cancer of the elderly is lacking. METHODS Registry data of this population-based cross-sectional study were from the Surveillance, Epidemiology, and End Results (SEER) database. The study population aged 65 years or more, from geographically distinct regions. Joinpoint regression and JP Surv method were used to analyze cancer trends and survival. RESULTS Mortality rate during 1975-2020 decreased from 995.20 to 824.99 per 100,000 elderly persons, with an average annual decrease of 0.421% (95% CI, 0.378-0.464). While overall incidence increased with no significance. Prostate (29%) and breast (26%) cancer were the most common malignancies, respectively, in elderly males and females, and the mortality for both of the two (prostate 15%, breast 14%) ranked just behind lung and bronchus cancer, which had the highest mortality rates in males (29%) and females (23%). Many cancers showed adverse trends in the latest follow-up periods (the last period calculated by the Joinpoint method). For intrahepatic cholangiocarcinoma, incidence (male Annual Percentage Change [APC] = 7.4*; female APC = 6.7*) and mortality (male APC = 3.0*; female APC = 3.3*) increased relatively fast, and its survival was also terrible (3-year survival only 10%). Other cancers with recent increasing mortality included cancer of anus, anal canal and anorectum, retroperitoneum, pleura, peritoneum, etc. Most cancers had favorable trends of survival during the nearest follow-up period. CONCLUSION Against the background of overall improvement, many cancers showed adverse trends. Further research for the underlying mechanisms and targeted implements towards adverse trends is also urgent.
Collapse
Affiliation(s)
- Jia Xu
- Department of GastroenterologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan ProvinceLuzhouChina
| | - Jingyuan Liao
- Department of GastroenterologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan ProvinceLuzhouChina
| | - Qiong Yan
- Department of GastroenterologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan ProvinceLuzhouChina
| | - Jiang Jiao
- Department of GastroenterologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan ProvinceLuzhouChina
| | - Nan Hu
- Department of GastroenterologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan ProvinceLuzhouChina
| | - Wei Zhang
- Department of GastroenterologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan ProvinceLuzhouChina
| | - Lei Shi
- Department of GastroenterologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan ProvinceLuzhouChina
| | - Mingming Deng
- Department of GastroenterologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan ProvinceLuzhouChina
| | - Shu Huang
- Department of GastroenterologyLianshui County People' HospitalHuaianChina
- Department of GastroenterologyLianshui People' Hospital of Kangda College Affiliated to Nanjing Medical UniversityHuaianChina
| | - Xiaowei Tang
- Department of GastroenterologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan ProvinceLuzhouChina
| |
Collapse
|
45
|
Wang J, Chen Z, Lai Y, Ma Z, Wang L, Fiori PL, Carru C, Capobianco G, Zhou L. Insights into inflammation and implications for the pathogenesis and long-term outcomes of endometrial cancer: genome-wide surveys and a clinical cohort study. BMC Cancer 2024; 24:846. [PMID: 39020272 PMCID: PMC11253470 DOI: 10.1186/s12885-024-12630-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Despite evidence showing a connection between inflammation and endometrial cancer (EC) risk, the surveys on genetic correlation and cohort studies investigating the impact on long-term outcomes have yet to be refined. We aimed to address the impact of inflammation factors on the pathogenesis, progression and consequences of EC. METHODS For the genetic correlation analyses, a two-sample of Mendelian randomization (MR) study was applied to investigate inflammation-related single-nucleotide polymorphisms involved with endometrial cancer from GWAS databases. The observational retrospective study included consecutive patients diagnosed with EC (stage I to IV) with surgeries between January 2010 and October 2020 at the Cancer Hospital of Shantou University Medical College. RESULTS The 2-sample MR surveys indicated no causal relationship between inflammatory cytokines and endometrial cancer. 780 cases (median age, 55.0 years ) diagnosed with EC were included in the cohort and followed up for an average of 6.8 years. Increased inflammatory parameters at baseline were associated with a higher FIGO stage and invasive EC risk (odds ratios [OR] 1.01 to 4.20). Multivariate-cox regression suggested that multiple inflammatory indicators were significantly associated with overall survival (OS) and progression-free survival (PFS) (P < 0.05). Nomogram models based on inflammatory risk and clinical factors were developed for OS and PFS with C-index of 0.811 and 0.789, respectively. LASSO regression for the validation supported the predictive efficacy of inflammatory and clinical factors on the long-term outcomes of EC. CONCLUSIONS Despite the fact that the genetic surveys did not show a detrimental impact of inflammatory cytokines on the endometrial cancer risk, our cohort study suggested that inflammatory level was associated with the progression and long-term outcomes of EC. This evidence may contribute to new strategies targeted at decreasing inflammation levels during EC therapy.
Collapse
Affiliation(s)
- Jing Wang
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zhichao Chen
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yaozhen Lai
- Department of Gynecologic Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Zebiao Ma
- Department of Gynecologic Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Luanhong Wang
- Department of Gynecologic Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Pier Luigi Fiori
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Giampiero Capobianco
- Gynecologic and Obstetric Clinic, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Li Zhou
- Department of Gynecologic Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|
46
|
Tang M, Dang P, Liu T, Yang K, Wang Y, Tse G, Liu H, Liu Y, Chan JSK, Liu C, Li G. Risk factors and outcomes of pericardial effusion in cancer patients receiving PD-1 inhibitors. Int J Cardiol 2024; 407:132029. [PMID: 38583590 DOI: 10.1016/j.ijcard.2024.132029] [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: 06/06/2023] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Programmed cell death 1 (PD-1) inhibitors can induce various adverse reactions associated with immunity, of which cardiotoxicity is a serious complication. Limited research exists on the link between PD-1 inhibitor use and pericardial effusion (PE) occurrence and outcomes. METHODS We conducted a retrospective study at the First Affiliated Hospital of Xi'an Jiaotong University from 2017 to 2019, comparing cancer patients who developed PE within 2 years after PD-1 inhibitor therapy to those who did not. Our primary outcome was the all-cause mortality rate at one year. We applied the Kaplan-Meier method for survival analysis. Multivariate logistic regression was utilized to identify PE risk factors, adjusting for potential confounders. RESULTS A total of 91 patients were finally included, of whom 39 patients had PE. Compared to non-PE group, one-year all-cause mortality was nearly 5 times higher in PE group (64.10% vs. 13.46%, P < 0.001). Patients who developed PE within 2 years of taking PD-1 inhibitors were significantly associated with increased all-cause mortality compared with those who did not (HR: 6.26, 95%CI: 2.70-14.53, P < 0.001). Multivariable logistic regression showed that use of sintilimab (OR: 14.568, 95%CI: 3.431-61.857, P < 0.001), history of lung cancer (OR: 15.360, 95%CI: 3.276-72.017, P = 0.001), and history of hypocalcemia (OR: 7.076, 95%CI: 1.879-26.649, P = 0.004) were independent risk factors of PE development in patients received PD-1 inhibitors therapy. CONCLUSIONS In cancer patients receiving PD-1 inhibitors, PE was associated with higher one-year mortality. Use of sintilimab, and history of lung cancer or hypocalcemia were linked to PE occurrence.
Collapse
Affiliation(s)
- Manyun Tang
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, China; Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Peizhu Dang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tong Liu
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, China; Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Kun Yang
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, China; Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Yifei Wang
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, China; Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China; Epidemiology Research Unit, Cardiovascular Analytics Group, PowerHealth Limited, Hong Kong, China; School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China
| | - Hui Liu
- Biobank of The First Affiliated Hospital of Xian Jiaotong University, Xi'an, China
| | - Yufeng Liu
- Biobank of The First Affiliated Hospital of Xian Jiaotong University, Xi'an, China
| | - Jeffrey Shi Kai Chan
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, PowerHealth Research Institute, Hong Kong, China
| | - Chang Liu
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, China; Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, China.
| | - Guoliang Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
47
|
Cheney L, Barbaro JM, McDermott G, Berman JW. Antiretroviral Drugs Impact Autophagy: Opportunities for Drug Repurposing. FRONT BIOSCI-LANDMRK 2024; 29:242. [PMID: 39082334 PMCID: PMC11837255 DOI: 10.31083/j.fbl2907242] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/19/2024] [Accepted: 04/30/2024] [Indexed: 01/04/2025]
Abstract
Autophagy is an evolutionarily conserved process in which intracellular macromolecules are degraded in a lysosomal-dependent manner. It is central to cellular energy homeostasis and to quality control of intracellular components. A decline in autophagic activity is associated with aging, and contributes to the development of various age-associated pathologies, including cancer. There is an ongoing need to develop chemotherapeutic agents to improve morbidity and mortality for those diagnosed with cancer, as well as to decrease the cost of cancer care. Autophagic programs are altered in cancer cells to support survival in genetically and metabolically unstable environments, making autophagy an attractive target for new chemotherapy. Antiretroviral drugs, which have dramatically increased the life- and health spans of people with human immunodeficiency virus (HIV) (PWH), have offered promise in the treatment of cancer. One mechanism underlying the antineoplastic effects of antiretroviral drugs is the alteration of cancer cell autophagy that can potentiate cell death. Antiretroviral drugs could be repurposed into the cancer chemotherapy arsenal. A more complete understanding of the impact of antiretroviral drugs on autophagy is essential for effective repurposing. This review summarizes our knowledge of the effects of antiretroviral drugs on autophagy as potential adjunctive chemotherapeutic agents, and highlights gaps to be addressed to reposition antiretroviral drugs into the antineoplastic arsenal successfully.
Collapse
Affiliation(s)
- Laura Cheney
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John M. Barbaro
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Joan W. Berman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
48
|
Berzins NJ, Mackenzie M, Galantino ML, Pickles N, Hebbel S, Leonard T, Beneck D, Peterson M. Preliminary Effectiveness Study of a Community-Based Wellness Coaching for Cancer Survivors Program. Am J Lifestyle Med 2024; 18:465-474. [PMID: 39262887 PMCID: PMC11384845 DOI: 10.1177/15598276221076040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/20/2021] [Accepted: 01/10/2022] [Indexed: 09/13/2024] Open
Abstract
PURPOSE Substantial cancer burden may be prevented through lifestyle modifications. The purpose of this study was to determine the preliminary effectiveness of health coaching for the improvement of health, fitness, and overall well-being of cancer survivors in a community setting. METHODS Participants were recruited from Cancer Support Community Delaware locations. Health coaching was provided to people diagnosed with cancer anywhere along the survivorship continuum. Coaches provided 6 individual sessions. Surveys were sent pre- and post-intervention on topics including fitness, eating habits, sleep, perceived stress, anxiety, depression, and quality of life. Results were analyzed using multilevel modeling. RESULTS 48 participants completed an average of 85% of health coaching sessions. Coaching participants noted improvements in weekly physical activity frequency, including moderate-vigorous physical activity. Increases were found in healthy eating behavior. Participants reported improvements in the quality of their sleep, including changes in sleep duration and sleep efficiency. Significant reductions were found in perceived stress, anxiety, and depression. Importantly, participants reported improved quality of life, particularly in areas of physical and emotional well-being, as well as functional and total well-being. CONCLUSION Preliminary findings indicate significant behavior change in measured outcomes and suggest health coaching may be an important tool for cancer survivorship.
Collapse
Affiliation(s)
| | | | - Mary Lou Galantino
- Stockton University, Galloway, NJ, USA
- University of Witwatersrand, Johannesburg, South Africa
| | | | - Sean Hebbel
- Cancer Support Community Delaware, Wilmington, DE, USA
| | | | | | | |
Collapse
|
49
|
Naciri I, Andrade-Ludena MD, Yang Y, Kong M, Sun S. An emerging link between lncRNAs and cancer sex dimorphism. Hum Genet 2024; 143:831-842. [PMID: 38095719 PMCID: PMC11176266 DOI: 10.1007/s00439-023-02620-7] [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: 05/25/2023] [Accepted: 11/05/2023] [Indexed: 06/15/2024]
Abstract
The prevalence and progression of cancer differ in males and females, and thus, sexual dimorphism in tumor development directly impacts clinical research and medicine. Long non-coding RNAs (lncRNAs) are increasingly recognized as important players in gene expression and various cellular processes, including cancer development and progression. In recent years, lncRNAs have been implicated in the differences observed in cancer incidence, progression, and treatment responses between men and women. Here, we present a brief overview of the current knowledge regarding the role of lncRNAs in cancer sex dimorphism, focusing on how they affect epigenetic processes in male and female mammalian cells. We discuss the potential mechanisms by which lncRNAs may contribute to sex differences in cancer, including transcriptional control of sex chromosomes, hormonal signaling pathways, and immune responses. We also propose strategies for studying lncRNA functions in cancer sex dimorphism. Furthermore, we emphasize the importance of considering sex as a biological variable in cancer research and the need to investigate the role lncRNAs play in mediating these sex differences. In summary, we highlight the emerging link between lncRNAs and cancer sex dimorphism and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Ikrame Naciri
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA, 92697, USA
| | - Maria D Andrade-Ludena
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA, 92697, USA
| | - Ying Yang
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA, 92697, USA
| | - Mei Kong
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA, 92697, USA.
| | - Sha Sun
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
50
|
Paulino E, Marquarte Santana L, Gomes de Mesquita G, de Melo AC. Is p53 immunohistochemistry alone useful for delineating adjuvant endometrial treatment in low-middle-income countries? Eur J Obstet Gynecol Reprod Biol 2024; 298:192-196. [PMID: 38788535 DOI: 10.1016/j.ejogrb.2024.05.023] [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: 03/25/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
OBJECTIVE Endometrial cancer (EC) treatment changed substantially with the introduction of molecular classification. Low-middle income (LMIC) countries will face barriers to including molecular classification to guide treatment. This study aims to analyse the value of p53 immunohistochemistry to delineate adjuvant treatment in FIGO stages I and II. METHODS Patients with EC treated between 2010 and 2016 were retrospectively evaluated. Patients included in this analysis must have reviewed FIGO stage I/II high-grade histologies (endometrioid grade 3, serous, clear cell, carcinosarcoma, mixed and undifferentiated). Samples were subjected to p53 immunohistochemistry. Recurrence-free and overall survival were analysed using the Kaplan-Meier method and log-rank test. Cox proportional hazards regression was performed for multivariable analysis. RESULTS From 2010 to 2016, 265 patients met the inclusion criteria. Patients with aberrant p53 (71.4 %) were associated with older age (59.7 % vs 77.8 % with more than 60 years), relapse (12.5 vs 29.6 %) and death (22.2 vs 46.7 %). The pattern of relapse was not different, with most being at extrapelvic sites (55.5 % vs 62.3 % for p53 wild type and aberrant, respectively). The median overall survival was not reached versus 92.2 months for p53 wild type and aberrant, respectively (p = 0.003). In multivariate analysis, chemotherapy decreased death (p = 0.014) in p53 aberrant tumours, a benefit not seen in the wild-type cohort (p = 0.22). CONCLUSION This retrospective analysis corroborates the finding of worse outcomes for p53 aberrant tumours in stage I/II EC and the benefit of more aggressive adjuvant treatment (systemic therapy and radiotherapy). Although not ideal as a sole molecular marker, p53 immunohistochemistry could complement the classical anatomopathological features and be part of the decision-making process with patients in LMIC.
Collapse
Affiliation(s)
- Eduardo Paulino
- Brazilian National Cancer Institute, Rio de Janeiro, Brazil; Oncologia D'OR, Rio de Janeiro, Brazil
| | - Luana Marquarte Santana
- Brazilian National Cancer Institute, Rio de Janeiro, Brazil; Oncologia D'OR, Rio de Janeiro, Brazil
| | | | - Andreia Cristina de Melo
- Brazilian National Cancer Institute, Rio de Janeiro, Brazil; Grupo Oncoclinicas, Rio de Janeiro, Brazil.
| |
Collapse
|