1
|
Filho ODS, Rondinelli R, Acetta AC, Bertolace S, Palladino A, Guedes CD, Vieira CCR, Thuler LCS, Diniz C, dos Santos EC, Abdelhay E. Impact of CD44/326 peritoneal cells as response indicators in advanced gastric cancer patients receiving repeated intraperitoneal perfusion normothermic chemotherapy. Ther Adv Med Oncol 2025; 17:17588359251337480. [PMID: 40351323 PMCID: PMC12064912 DOI: 10.1177/17588359251337480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Background In gastric cancer (GC), the dissemination of neoplastic cells (NCs) in the peritoneal cavity is related to disease progression and poor prognosis. Elimination of NC through chemotherapy is needed to achieve better outcomes before conversion surgery. Objectives The objective of this study was to evaluate the impact of NC CD44+/CD326+ levels through flow cytometry (FC) on peritoneal lavage (PL) fluid as a response indicator for conversion surgery. Methods Patients with GC and NCs in the peritoneal cavity with or without peritoneal carcinomatosis (PC) and ascites were evaluated via minimally invasive staging. The PLs of patients were analyzed by FC to quantify NCs. All patients were treated with repeated intraperitoneal perfusion normothermic chemotherapy (RIPPENC). Patients who had negative NCs or reduced NCs were referred for conversion surgery. Results Thirty patients were enrolled in this study and divided into three groups. In the first group, 20 patients with positive cytology (C+) and/or PC with a PC index (PCI) ⩽6 were treated with RIPPENC. Otherwise, six patients with C+ and PC with a PCI >7 and four patients with C+, ascites, and a PCI ranging from 15 to 22 were treated with palliative RIPPENC. The percentage of CD44+/CD326+ cells was correlated with the number of RIPPENC cycles and resections. The median follow-up time was 14.8 months. The overall median survival since the first RIPPENC was 14.6 months among those who did not undergo resection and 22.6 months among those who underwent resection (p = 0.001). Moreover, we observed a correlation between the percentage of CD44+/CD326+ cells in the PL region and patient survival. Conclusion The use of FC to identify PL CD44+/CD326+ cell levels may be an important innovative biomarker for determining the presence of NCs, directly affecting the success of RIPPENC for conversion surgery.
Collapse
Affiliation(s)
- Odilon de Souza Filho
- Abdominopelvic Surgery Service, Division of Oncological Surgery, National Cancer Institute, Rio de Janeiro, Brazil
| | - Reinaldo Rondinelli
- Abdominopelvic Surgery Service, Division of Oncological Surgery, National Cancer Institute, Rio de Janeiro, Brazil
| | - Antônio Carlos Acetta
- Abdominopelvic Surgery Service, Division of Oncological Surgery, National Cancer Institute, Rio de Janeiro, Brazil
| | - Sérgio Bertolace
- Abdominopelvic Surgery Service, Division of Oncological Surgery, National Cancer Institute, Rio de Janeiro, Brazil
| | - Alexandre Palladino
- Division of Clinical Oncology, National Cancer Institute, Rio de Janeiro, Brazil
| | | | | | | | - Claudia Diniz
- Immunology Laboratory, Division of Specialized Laboratories, National Cancer Institute, Rio de Janeiro, Brazil
| | - Everton Cruz dos Santos
- Immunology Laboratory, Division of Specialized Laboratories, National Cancer Institute, Rio de Janeiro, Brazil
| | - Eliana Abdelhay
- Immunology Laboratory, Division of Specialized Laboratories, National Cancer Institute, Praça Cruz Vermelha, 23-Centro, Rio de Janeiro 20230-130, Brazil
| |
Collapse
|
2
|
Tsai YT, Wen YW, Tsai TY, Lin CY, Ng SH, Ku HY, Lou PJ, Wang CP, Lin JC, Hua CH, Lee SR, Fan KH, Chen WC, Lee LY, Chien CY, Chen TM, Terng SD, Tsai CY, Wang HM, Hsieh CH, Yeh CH, Lin CH, Tsao CK, Cheng NM, Kang CJ, Fang TJ, Huang SF, Lee LA, Fang KH, Wang YC, Lin WN, Hsin LJ, Yen TC, Liao CT. Pathological primary tumor status, rather than adjuvant therapy, predicts survival outcomes in pT1-3N1M0 oral cavity squamous cell carcinoma: A nationwide cohort study. Oral Oncol 2025; 164:107289. [PMID: 40184884 DOI: 10.1016/j.oraloncology.2025.107289] [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: 12/09/2024] [Revised: 02/24/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Theeighth edition oftheAmerican Joint Committee onCancer (AJCC) Staging Manualclassifies pT1-3N1M0 oralcavity squamous cellcarcinoma (OCSCC) asp-Stage III. However, the prognosis within this group is heterogeneous, and the clinical benefit of adjuvant therapy for patients with a single nodal metastasis remains unclear.Here, we analyzed nationwide data from Taiwan to assess survival outcomes and examine the role of adjuvant therapy in this population. METHODS A retrospective analysis of 1324 pT1-3N1M0 OCSCC patients who underwent surgical resection between 2011 and 2021 was conducted using data from the Taiwan Cancer Registry and National Health Insurance Research Database. Cox proportional hazards models were applied to identify independent prognostic factors for disease-specific survival (DSS) and overall survival (OS). RESULTS Among the cohort, 247 patients (18.7 %) had pT1N1, 699 (52.8 %) had pT2N1, and 378 (28.5 %) had pT3N1 disease. The 5-year DSS rates for pT1N1, pT2N1, and pT3N1 were 82 %, 79 %, and 69 %, respectively, while OS rates were 73 %, 70 %, and 60 % (both p < 0.0001). No significant differences in DSS or OS were observed between surgery alone and adjuvant therapy. In multivariable analysis, pT3N1 disease was independently associated with worse survival outcomes (HR: 1.76,p = 0.0011 for DSS; HR: 1.63,p = 0.0005 for OS), whereas adjuvant therapies were not independent prognostic factors. CONCLUSION Among patients with pT1-3N1M0 OCSCC, those presenting with pT3N1 disease demonstrated significantly poorer DSS and OS. Notably, in patients lacking additional adverse pathological features, the omission of adjuvant therapy did not adversely impact survival endpoints.
Collapse
Affiliation(s)
- Yao-Te Tsai
- Department of Otorhinolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan, ROC
| | - Yu-Wen Wen
- Clinical Informatics and Medical Statistics Research Center, Chang Gung University, Taoyuan, Taiwan, ROC; Division of Thoracic Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Tsung-You Tsai
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chien-Yu Lin
- Department of Radiation Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Shu-Hang Ng
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Hsiu-Ying Ku
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan, ROC
| | - Pei-Jen Lou
- Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Cheng Ping Wang
- Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Jin-Ching Lin
- Department of Radiation Oncology, Changhua Christian Hospital, Changhua, Taiwan, ROC
| | - Chun-Hung Hua
- Department of Otorhinolaryngology, China Medical University Hospital, Taichung, Taiwan, ROC
| | - Shu-Ru Lee
- Research Service Center for Health Information, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Kang-Hsing Fan
- Department of Radiation Oncology, New Taipei Municipal TuCheng Hospital, Taiwan, ROC
| | - Wen-Cheng Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Chiayi and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chih-Yen Chien
- Kaohsiung Chang Gung Memorial Hospital, Doctoral Program of Clinical and Experimental Medicine, National Sun Yat-sen University, Taiwan, ROC
| | - Tsung-Ming Chen
- Department of Otolaryngology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan, ROC
| | - Shyuang-Der Terng
- Department of Head and Neck Surgery, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan, ROC
| | - Chi-Ying Tsai
- Department of Oral and Maxillofacial Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Hung-Ming Wang
- Department of Medical Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chia-Hsun Hsieh
- Department of Medical Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chih-Hua Yeh
- Department of Radiation Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chih-Hung Lin
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chung-Kan Tsao
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Nai-Ming Cheng
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chung-Jan Kang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Tuan-Jen Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Shiang-Fu Huang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Li-Ang Lee
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Ku-Hao Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Yu-Chien Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Wan-Ni Lin
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Li-Jen Hsin
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Tzu-Chen Yen
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chun-Ta Liao
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC.
| |
Collapse
|
3
|
Wankhede D, Halama N, Kloor M, Edelmann D, Brenner H, Hoffmeister M. Prognostic Value of CD8+ T Cells at the Invasive Margin Is Comparable to the Immune Score in Nonmetastatic Colorectal Cancer: A Prospective Multicentric Cohort Study. Clin Cancer Res 2025; 31:1711-1718. [PMID: 40293274 DOI: 10.1158/1078-0432.ccr-24-3275] [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: 10/02/2024] [Revised: 11/23/2024] [Accepted: 02/19/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE The Immunoscore predicts colorectal cancer prognosis but faces adoption barriers because of complex software and reimbursement issues. This study used open-source methods to explore a simplified prognostic model in nonmetastatic colorectal cancer by focusing on single T-cell markers. EXPERIMENTAL DESIGN A multicentric prospective cohort study in patients with nonmetastatic colorectal cancer assessed CD3+ and CD8+ tumor-infiltrating lymphocytes (TIL) in the invasive margin (IM) and tumor core (TC) using QuPath. An immune cell score (ICS), based on TIL densities (CD3-IM, CD8-IM, CD3-TC, and CD8-TC), was calculated similarly to the Immunoscore. A split sample approach (70:30) estimated adjusted HRs for cancer-specific survival in training and validation sets. Classification and regression tree analysis identified the most prognostic TIL, and its model was compared with an ICS model for performance (Brier score) and discrimination (concordance probability estimate). RESULTS Over a median follow-up of 9.0 years, 203 colorectal cancer-specific deaths occurred among 1,260 patients. Classification and regression tree-selected CD8-IM was the most prognostic TIL at a cutoff of 231 cells/mm2. Patients with high CD8-IM had better cancer-specific survival than low CD8-IM in both training (HR 0.58, 95% confidence interval, 0.40-0.84) and validation sets (HR 0.35, 95% confidence interval, 0.21-0.60). Brier scores of CD8-IM and ICS survival models were comparable in both training and validation cohorts, whereas the survival discrimination of CD8-IM slightly outperformed the ICS in the validation set (concordance probability estimate: CD8-IM: 0.748; ICS: 0.730). CONCLUSIONS CD8-IM alone provided prognostic information comparable with the ICS. Simplified, cost-effective TIL assessments could improve clinical translation and guide adjuvant therapy in early-stage colorectal cancer.
Collapse
Affiliation(s)
- Durgesh Wankhede
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | - Niels Halama
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Department of Translational Immunotherapy (D240), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Helmholtz Institute for Translational Oncology, Mainz, Germany
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Cooperation Unit Applied Tumor Biology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Dominic Edelmann
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
4
|
Guan L, Torres-Saavedra PA, Zhao X, Major MB, Holmes BJ, Nguyen NK, Kumaravelu P, Hodge T, Diehn M, Zevallos J, Christopher Holsinger F, Emami B, Jordan RC, Hayward MC, Sagar SM, Morrison W, Schultz C, Caudell JJ, Jones CU, Bratman SV, Galloway TJ, Ma DJ, Yom SS, Kudrimoti M, Kim HE, Harris J, Le QT, Neil Hayes D. Association between Locoregional Failure and NFE2L2/KEAP1/CUL3 Mutations in NRG/RTOG 9512: A Randomized Trial of Radiation Fractionation in T2N0 Glottic Cancer. Clin Cancer Res 2025; 31:1615-1624. [PMID: 39656603 PMCID: PMC12045734 DOI: 10.1158/1078-0432.ccr-24-2334] [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: 07/19/2024] [Revised: 10/10/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE NFE2L2/KEAP1/CUL3 mutations have been validated for radioresistance in cell-based assays and animal models. However, clinical validation of these biomarkers has been challenging because of multimodality treatment regimens. This study aims to investigate the association between NFE2L2/KEAP1/CUL3 mutations and patient outcomes, including local failure, locoregional failure, disease-free survival (DFS), and overall survival, using samples from a phase III trial in which patients were treated with radiation monotherapy at two controlled doses. PATIENTS AND METHODS We investigated NFE2L2/KEAP1/CUL3 mutations in 250 randomized patients with T2N0 glottic squamous cell carcinoma receiving definitive radiotherapy in the NRG/RTOG 9512 trial. A total of 119 patients had available biospecimens that were subjected to amplicon-based next-generation sequencing to assess for the presence of NFE2L2/KEAP1/CUL3 mutations without regard to outcomes. Mutations in NFE2L2/KEAP1/CUL3 were assessed blinded to clinical outcomes. Cox models (two-sided α = 0.05) were used to evaluate the association with clinical outcomes, performed by an independent statistical team. RESULTS Nineteen of 119 patients (16.0%) had NFE2L2/KEAP1/CUL3 mutations. Patient, treatment, and tumor characteristics were similar between those with and without mutations. Patients with mutation compared with those without had significantly more local failure [HR = 3.50; 95% confidence interval (CI), 1.56-7.89; P = 0.0025] and locoregional failure (HR = 3.80; 95% CI, 1.80-8.03; P = 0.0005). DFS was significantly worse for the mutated compared with the nonmutated group in the first 2 years (HR = 2.88; 95% CI, 1.46-5.66; P = 0.0022). The median DFS was shorter in the mutation group (10.3 months) versus those with intact NFE2L2/KEAP1/CUL3 (4.2 years). CONCLUSIONS NFE2L2/KEAP1/CUL3 mutations may predict radiation treatment failure in T2N0 glottic cancer. See related commentary by Rao, p. 1563.
Collapse
MESH Headings
- Adult
- Aged
- Female
- Humans
- Male
- Middle Aged
- Biomarkers, Tumor/genetics
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/mortality
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/radiotherapy
- Cullin Proteins/genetics
- Disease-Free Survival
- Dose Fractionation, Radiation
- Glottis/pathology
- Glottis/radiation effects
- Kelch-Like ECH-Associated Protein 1/genetics
- Laryngeal Neoplasms/radiotherapy
- Laryngeal Neoplasms/genetics
- Laryngeal Neoplasms/pathology
- Laryngeal Neoplasms/mortality
- Mutation
- Neoplasm Recurrence, Local/epidemiology
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/prevention & control
- Neoplasm Staging
- NF-E2-Related Factor 2/genetics
- Prognosis
Collapse
Affiliation(s)
- Li Guan
- Stanford University, School of Medicine, Stanford, CA
| | | | - Xiaobei Zhao
- The University of Tennessee Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN
| | - Michael B. Major
- Washington University School of Medicine in St. Louis, St. Louis, MO
| | | | - Ngan K. Nguyen
- The University of Tennessee Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN
| | - Parasakthy Kumaravelu
- The University of Tennessee Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN
| | - Tim Hodge
- The University of Tennessee Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN
| | | | - Jose Zevallos
- Washington University School of Medicine in St. Louis, St. Louis, MO
| | | | | | | | | | - Stephen M. Sagar
- Juravinski Cancer Center at Hamilton Health Sciences, Hamilton, ON
| | | | | | | | | | - Scott V. Bratman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON
| | | | | | - Sue S. Yom
- UCSF Medical Center-Mount Zion, San Francisco, CA
| | | | - Harold E. Kim
- Wayne State University/Karmanos Cancer Institute, Detroit, MI
| | - Jonathan Harris
- NRG Oncology Statistics and Data Management Center, American College of Radiology, Philadelphia, PA
| | - Quynh-Thu Le
- Stanford University, School of Medicine, Stanford, CA
| | - D. Neil Hayes
- The University of Tennessee Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN
| |
Collapse
|
5
|
Mi SY, Liao YJ, Kang D, Tang X, Chen G, Pan ZZ, Zhan J, Zhang RX. Decreasing Use of CT in Favor of Colonoscopy: A Retrospective Analysis of Post-Operative Imaging in Nonmetastatic dMMR/MSI-H Sporadic Colorectal Cancer. Ann Surg Oncol 2025; 32:3814-3827. [PMID: 39948309 DOI: 10.1245/s10434-025-16989-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/23/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND There is no significant difference in postoperative follow-up imaging examinations for varying mismatch repair (MMR)/microsatellite instability (MSI) statuses for sporadic colorectal cancer while facing different prognoses. METHODS We conducted a retrospective study, using Kaplan-Meier curve to compare survival/progression status by varying imaging examination and frequency, using Cox regression to analyze tumor characteristic impact on survival, and monitoring polyp detection time in each colonoscopy screening. RESULTS A total of 282 deficient MMR (dMMR)/MSI-high (MSI-H) patients were included. After a 7 year follow-up, all patients-including 143 examined by CT and colonoscopy, 86 singularly examined (CT/colonoscopy/ultrasound), and 53 received no imaging examination-demonstrated a favorable overall survival (87.97%, 95% CI 75.99-89.19) and disease-free survival (81.92%, 95% CI 70.38-89.30), which had no significant difference between imaging methods. Cox regression proved that different imaging methods had no influence on the prognosis. Stage III patients experienced a deterioration in disease-free survival but not differed between varying imaging methods. Different CT follow-up interval exhibited no difference in survival and progression. Polyps were detected in 39.75% (64/161) of patients during follow-up; 32.81% (21/64) were detected in colonoscopy conducted 0-6 months after surgery, reaching the highest. For multiple polyps, the median of detection interval was 13-24 months (first and second polyps), 7-12 months (second and third polyps). CONCLUSIONS For nonmetastatic dMMR/MSI-H sporadic colorectal cancer patients, less CT scans (interval > 1 year) are tolerable because of good prognosis, whereas more colonoscopy screenings (0-6 months after surgery first year; 13-24 months after first polyps detection; 7-12 months after second polyps detection) is considerable.
Collapse
Affiliation(s)
- Si-Yuan Mi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yi-Jun Liao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Da Kang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Xin Tang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Gong Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Zhi-Zhong Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jianhua Zhan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.
| | - Rong-Xin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.
| |
Collapse
|
6
|
Torok Z, Garai K, Bovari-Biri J, Adam Z, Miskei JA, Kajtar B, Sarosi V, Pongracz JE. Serum and exosome WNT5A levels as biomarkers in non-small cell lung cancer. Respir Res 2025; 26:141. [PMID: 40223089 PMCID: PMC11995597 DOI: 10.1186/s12931-025-03216-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 04/01/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Despite significant advances in the treatment of lung cancer (LC), there are no reliable biomarkers to effectively predict therapy response and overall survival (O/S) in non-small cell lung cancer (NSCLC) subtypes. While targeted therapies have improved survival rates in lung adenocarcinoma (LUAD), effective treatment options for lung squamous cell carcinoma (LUSC) are still limited. Recent evidence indicates that exosome-bound WNT5A may significantly contribute to disease progression. Our study assessed the WNT5A protein as a potential biomarker for diagnosing patients and predicting prognosis to assist in therapy selection. METHODS Primary tumor tissue and serum samples were collected from a cohort of 60 patients with histologically confirmed NSCLC before therapy. Healthy serum donors served as controls. Exosomes were isolated, then exosome number and size were measured, and WNT5A protein levels were identified in tissue and in vesicle-free, vesicle-bound fractions of the serum by ELISA. RESULTS Extensive statistical analysis (ROC, AUC, Cox, etc.) revealed that elevated WNT5A levels on the serum-exosome surface correlated with distant metastasis, advanced disease stage, and lymph node involvement in LUSC but not in LUAD patients. Moreover, a high WNT5A exosome surface expression was associated with a poor response to therapy and shorter O/S in LUSC patients. Additionally, serum-exosome surface + cargo WNT5A content distinguished LUAD and LUSC subtypes. CONCLUSIONS WNT5A, particularly its serum exosome-bound form, may serve as a valuable biomarker after further validation for differentiating NSCLC subtypes and predicting disease progression. Importantly, the information can become available from a simple serum sample at the time of diagnosis.
Collapse
Affiliation(s)
- Zsofia Torok
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, Pecs, H-7624, Hungary
- Department of Pulmonology, 1st Internal Medicine, The Medical School and Clinical Centre, University of Pecs, 12 Szigeti Str, Pecs, H-7624, Hungary
| | - Kitti Garai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, Pecs, H-7624, Hungary
| | - Judit Bovari-Biri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, Pecs, H-7624, Hungary
| | - Zoltan Adam
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, Pecs, H-7624, Hungary
| | - Judith A Miskei
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, Pecs, H-7624, Hungary
| | - Bela Kajtar
- Department of Pathology, The Medical School and Clinical Centre, University of Pecs, 12 Szigeti Str, Pecs, H-7624, Hungary
| | - Veronika Sarosi
- Department of Pulmonology, 1st Internal Medicine, The Medical School and Clinical Centre, University of Pecs, 12 Szigeti Str, Pecs, H-7624, Hungary
| | - Judit E Pongracz
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, Pecs, H-7624, Hungary.
| |
Collapse
|
7
|
Rajput SK, Minhas K, Azam I, Habib S, Shaikh U, Lalani EN. Prognostic implications of MUC1 and XBP1 concordant expression in multiple myeloma: A retrospective study. PLoS One 2025; 20:e0320934. [PMID: 40179083 PMCID: PMC11967961 DOI: 10.1371/journal.pone.0320934] [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: 11/20/2023] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Multiple myeloma (MM) is a disease of malignant plasma cells (PC) with poor survival. Disease progression and treatment relapse are attributed to MM cancer stem cells (CSCs) and signaling molecules such as MUC1 and XBP1. The study aimed to determine the prognostic value of expression of CSC-associated biomarkers, MUC1 and XBP1 in MM, which has not been explored previously. In this study, we determined the immunohistochemical expression of CSC markers (ALDH1, CD117, and CD34), MUC1, and XBP1 in 128 MM formalin-fixed paraffin-embedded bone marrow archival blocks. The expression of biomarkers was assessed for association with clinicopathological variables and patient survival. Descriptive analysis, survival plots and crude association between outcome and independent variables were assessed using Kaplan Meier and Log rank test. Univariate and multivariable analyses were performed using simple and multiple Cox regression models. The results are reported as crude and adjusted hazard ratios with 95% confidence intervals. Expression of ALDH1 and CD117 was found in 51% and 48% of the tumors, respectively. ALDH1 expression was associated with 1.83 years of reduced survival for patients with CD56-negative tumors. MUC1 expression was observed in 62%, whereas XBP1 was expressed in 48% of tumors. Combinatorial group analysis of XBP1 and MUC1 stratified patients into two prognostic groups. Cases with tumors negative for expression of MUC1 and XBP1 (XBP1-/ MUC1-) were categorized as a good prognostic group with increased survival of 3.42 years compared to cases with tumors expressing both (Worst prognosis, XBP1 + /MUC1+). Concordant expression of MUC1 and XBP1 in MM defines a subset of patients with adverse outcomes. The adjusted hazard ratio showed a four-fold increased risk of mortality associated with the concordant expression of MUC1 and XBP1 in patients > 65 years of age.
Collapse
Affiliation(s)
- Sheerien Kareem Rajput
- Centre for Regenerative Medicine and Stem Cell Research, The Aga Khan University, Karachi, Pakistan
| | - Khurram Minhas
- Department of Pathology and Laboratory Medicine, The Aga Khan University, Karachi, Pakistan
| | - Iqbal Azam
- Department of Community Health Sciences, The Aga Khan University, Karachi, Pakistan
| | - Sadia Habib
- Centre for Regenerative Medicine and Stem Cell Research, The Aga Khan University, Karachi, Pakistan
| | - Usman Shaikh
- Department of Pathology and Laboratory Medicine, The Aga Khan University, Karachi, Pakistan
| | - El-Nasir Lalani
- Centre for Regenerative Medicine and Stem Cell Research, The Aga Khan University, Karachi, Pakistan
| |
Collapse
|
8
|
Kariri YA, Alsaleem M, Al-Kawaz A, Alhatlani BY, Mongan NP, Green AR, Rakha EA. Cell division cycle 6 is an independent prognostic biomarker in breast cancer. Pathology 2025; 57:297-304. [PMID: 39668074 DOI: 10.1016/j.pathol.2024.09.006] [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: 04/18/2024] [Revised: 09/01/2024] [Accepted: 09/16/2024] [Indexed: 12/14/2024]
Abstract
Cell division cycle 6 (CDC6) is a cell cycle protein involved in cell cycle control, DNA replication and cancer cell apoptosis. This study investigated the prognostic value of CDC6 in breast cancer (BC) utilising large well-characterised cohorts of early-stage BC. CDC6 messenger RNA (mRNA) was assessed using the Molecular Taxonomy of the Breast Cancer International Consortium (n=1980), the Cancer Genome Atlas (n=854) and Kaplan-Meier plotter (n=4,929) cohorts. CDC6 protein expression was evaluated using immunohistochemistry in a large (n=951) well-characterised Nottingham BC cohort. The associations between CDC6, clinicopathological parameters, molecular features and patient outcomes were assessed. High CDC6 expression positively correlated with dysregulation of key BC-related genes, including gene involved in cell cycle, DNA damage repair, epithelial cell migration, and tumour microenvironment control, as well as with markers characteristic of the basal-like phenotype (CK5, CK14 and CK17). High CDC6 mRNA and protein expression were associated with clinicopathological parameters characteristic of aggressive behaviour, including high tumour grade, large tumour size, the presence of lymphovascular invasion and hormone receptor negativity. High CDC6 protein expression was an independent predictor of poor outcome [p=0.007; hazard ratio (HR)=1.3; 95% confidence interval (CI) 1.2-1.9). This study indicates that CDC6 is an independent prognostic biomarker in BC. These results warrant further functional validation for CDC6 as a potential therapeutic target in BC.
Collapse
Affiliation(s)
- Yousif A Kariri
- Department of Clinical Laboratory Science, College of Applied Medical Science, Shaqra University, Shaqra, Saudi Arabia
| | - Mansour Alsaleem
- Unit of Scientific Research, Applied College, Qassim University, Saudi Arabia
| | - Abdulbaqi Al-Kawaz
- Department of Oral Pathology, College of Dentistry, Mustansiriyah University, Baghdad, Iraq
| | - Bader Y Alhatlani
- Unit of Scientific Research, Applied College, Qassim University, Saudi Arabia
| | - Nigel P Mongan
- Biodiscovery Institute, Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, UK; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK; Pathology Department, Nottingham University Hospitals NHS Trust, Nottingham, UK; Pathology Department, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
9
|
de Jong JJ, Proudfoot JA, Daneshmand S, Svatek RS, Narayan V, Gibb EA, Davicioni E, Joshi S, Dahmen A, Li R, Inman BA, Shah P, Chaplin I, Wright J, Lotan Y. A luminal non-coding RNA-based genomic classifier confirms favourable outcomes in patients with clinically organ-confined bladder cancer treated with radical cystectomy. BJU Int 2025; 135:648-656. [PMID: 39485082 PMCID: PMC11913596 DOI: 10.1111/bju.16572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
OBJECTIVE To further evaluate a genomic classifier (GC) in a cohort of patients undergoing radical cystectomy (RC), as long non-coding RNA (lncRNA)-based genomic profiling has suggested utility in identifying a distinct tumour subgroup corresponding to a favourable prognosis in patients with bladder cancer. PATIENTS AND METHODS Transcriptome-wide expression profiling using Decipher Bladder was performed on transurethral resection of bladder tumour samples from a cohort of patients with high-grade, clinically organ-confined (cTa-T2N0M0) urothelial carcinoma (UC) who subsequently underwent RC without any neoadjuvant therapy (n = 226). The lncRNA-based luminal favourable status was determined using a previously developed GC. The primary endpoint was overall survival (OS) after RC. Secondary endpoints included cancer-specific mortality and upstaging at RC. RESULTS In the study, 134 patients were clinical non-muscle-invasive bladder cancer (cTa/Tis/T1) and 92 patients were cT2. We identified 60 patients with luminal favourable subtype, all of which showed robust gene expression patterns associated with less aggressive bladder cancer biology. On multivariate analysis, patients with the luminal favourable subtype (vs without) were significantly associated with lower odds of upstaging to pathological (p)T3+ disease (odds ratio [OR] 0.32, 95% confidence interval [CI] 0.12-0.82; P = 0.02), any upstaging (OR 0.41, 95% CI 0.20-0.83; P = 0.01), and any upstaging and/or pN+ (OR 0.50, 95% CI 0.25-1.00; P = 0.05). Luminal favourable bladder cancer was significantly associated with better OS (hazard ratio 0.33, 95% CI 0.15-0.74; P = 0.007). CONCLUSIONS This study validates the performance of the GC for identifying UCs with a luminal favourable subtype, harbouring less aggressive tumour biology.
Collapse
Affiliation(s)
| | | | - Siamak Daneshmand
- Department of Urology, USC/Norris Comprehensive Cancer CenterUniversity of Southern CaliforniaLos AngelesCAUSA
| | | | | | | | | | | | | | | | | | | | | | | | - Yair Lotan
- University of Texas SouthwesternDallasTXUSA
| |
Collapse
|
10
|
Petruk N, Wood SL, Gregory W, Lopez-Guajardo A, Oliva M, Mella M, Sandholm J, Jukkola A, Brown JE, Selander KS. Increased primary breast tumor expression of CD73 is associated with development of bone metastases and is a potential biomarker for adjuvant bisphosphonate use. Sci Rep 2025; 15:9449. [PMID: 40108234 PMCID: PMC11923362 DOI: 10.1038/s41598-025-92841-9] [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: 11/25/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025] Open
Abstract
PURPOSE Increased CD73 expression has been associated with progression in various cancer types. Results of the AZURE and other trials suggest that, in postmenopausal breast cancer patients, adjuvant bisphosphonates inhibit bone relapses and prolong overall survival. Based on these findings, adjuvant bisphosphonates (typically zoledronic acid) are standard-of-care in postmenopausal patients with high-risk early breast cancer. However, biomarkers are needed for improved patient selection. The aim of this study was to investigate the association of primary tumor CD73 expression with later development of bone metastases. METHODS To determine whether CD73 levels correlated with tumor parameters (hormone receptor status, tumor stage and grade), patient outcomes (bone metastases and survival) or other patient characteristics (menopausal status, chemotherapy or statin use), we analyzed primary breast tumor CD73 expression immunohistochemically in tumor microarray samples from the AZURE (BIG01/04) trial. RESULTS In the AZURE control arm, high CD73 score are significantly prognostic for overall survival (p-value = 0.03, HR = 1.87, 95% CI = 1.06-3.29), disease-free survival (p-value = 0.06, HR = 1.66, 95% CI = 0.982-2.8) and time to first metastasis to bone (p-value = 0.04, HR = 2.23, 95% CI = 1.04-4.81), as compared with low CD73 scores. However, high CD73 score did not display an association with time to non-bone metastasis or first recurrence to a non-skeletal site. In the zoledronate arm, high CD73 score did not have association with patient outcomes, first metastasis to bone, nor with bone recurrence at any time (distant recurrence, including skeletal) or first non-skeletal recurrence. In multivariate testing, CD73 had no significant association with age, ER status, tumor stage, histological grade, menopausal status, chemotherapy or statin use in either arm. CONCLUSIONS High CD73 expression is associated with development of bone metastases. Zoledronate counteracts this effect. These results suggest that CD73 expression might serve as a biomarker for adjuvant zoledronic acid use.
Collapse
Affiliation(s)
- Nataliia Petruk
- Institute of Biomedicine, University of Turku, Turku, Finland
- Western Cancer Centre FICAN West, Turku, Finland
| | - Steven L Wood
- Division of Clinical Medicine, Medical School, Sheffield, UK
| | - Walter Gregory
- Division of Clinical Medicine, Medical School, Sheffield, UK
| | | | - Maria Oliva
- Division of Clinical Medicine, Medical School, Sheffield, UK
| | - Mikko Mella
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Oncology and Radiation Therapy, Oulu University Hospital, Oulu, Finland
| | - Jouko Sandholm
- Turku Bioscience Centre, University of Turku, Åbo Akademi University, Turku, Finland
| | - Arja Jukkola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Janet E Brown
- Division of Clinical Medicine, Medical School, Sheffield, UK
| | - Katri S Selander
- Department of Oncology and Radiation Therapy, Oulu University Hospital, Oulu, Finland.
- Department of Translational Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
11
|
Isebia KT, de Jong AC, van Dessel LF, de Weerd V, Beaufort C, Helmijr J, Nakauma-González JA, van Riet J, Hamberg P, Vis D, van der Heijden MS, Beije N, Lolkema MP, Deger T, Wilting SM, de Wit R, Jansen MPHM, Martens JWM. Cell-free DNA aneuploidy score as a dynamic early response marker in prostate cancer. Mol Oncol 2025. [PMID: 40084488 DOI: 10.1002/1878-0261.13797] [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: 07/25/2024] [Revised: 12/03/2024] [Accepted: 12/24/2024] [Indexed: 03/16/2025] Open
Abstract
Cell-free circulating tumor DNA (ctDNA) has emerged as a promising biomarker for response evaluation in metastatic castration-resistant prostate cancer (mCRPC). The current study evaluated the modified fast aneuploidy screening test-sequencing system (mFast-SeqS), a quick, tumor-agnostic and affordable ctDNA assay that requires a small input of DNA, to generate a genome-wide aneuploidy (GWA) score in mCRPC patients, and correlated this to matched metastatic tumor biopsies. In this prospective multicenter study, GWA scores were evaluated from blood samples of 196 mCRPC patients prior to treatment (baseline) with taxanes (docetaxel and cabazitaxel) and androgen receptor signaling inhibitors (ARSI; abiraterone and enzalutamide), and from 74 mCRPC patients at an early timepoint during treatment (early timepoint; median 21 days). Z-scores per chromosome arm were tested for their association with tumor tissue genomic alterations. We found that a high tumor load in blood (GWAhigh) at baseline was associated with poor response to ARSI [HR: 2.63 (95% CI: 1.86-3.72) P < 0.001] but not to taxanes. Interestingly, GWAhigh score at the early timepoint was associated with poor response to both ARSIs [HR: 6.73 (95% CI: 2.60-17.42) P < 0.001] and taxanes [2.79 (95% CI: 1.34-5.78) P = 0.006]. A significant interaction in Cox proportional hazards analyses was seen when combining GWA status and type of treatment (at baseline P = 0.008; early timepoint P = 0.018). In summary, detection of ctDNA in blood by mFast-SeqS is cheap, fast and feasible, and could be used at different timepoints as a potential predictor for outcome to ARSI and taxane treatment in mCRPC.
Collapse
Affiliation(s)
- Khrystany T Isebia
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Anouk C de Jong
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Lisanne F van Dessel
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Vanja de Weerd
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Corine Beaufort
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Jean Helmijr
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | | | - Job van Riet
- Division of AI in Oncology, German Cancer Research Center (DFKZ), Heidelberg, Germany
| | - Paul Hamberg
- Department of Internal Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Daniel Vis
- Department of Medical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Michiel S van der Heijden
- Department of Medical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Nick Beije
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Martijn P Lolkema
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Teoman Deger
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Saskia M Wilting
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Ronald de Wit
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Maurice P H M Jansen
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| |
Collapse
|
12
|
Cottrell TR, Lotze MT, Ali A, Bifulco CB, Capitini CM, Chow LQM, Cillo AR, Collyar D, Cope L, Deutsch JS, Dubrovsky G, Gnjatic S, Goh D, Halabi S, Kohanbash G, Maecker HT, Maleki Vareki S, Mullin S, Seliger B, Taube J, Vos W, Yeong J, Anderson KG, Bruno TC, Chiuzan C, Diaz-Padilla I, Garrett-Mayer E, Glitza Oliva IC, Grandi P, Hill EG, Hobbs BP, Najjar YG, Pettit Nassi P, Simons VH, Subudhi SK, Sullivan RJ, Takimoto CH. Society for Immunotherapy of Cancer (SITC) consensus statement on essential biomarkers for immunotherapy clinical protocols. J Immunother Cancer 2025; 13:e010928. [PMID: 40054999 PMCID: PMC11891540 DOI: 10.1136/jitc-2024-010928] [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: 10/28/2024] [Accepted: 02/05/2025] [Indexed: 03/12/2025] Open
Abstract
Immunotherapy of cancer is now an essential pillar of treatment for patients with many individual tumor types. Novel immune targets and technical advances are driving a rapid exploration of new treatment strategies incorporating immune agents in cancer clinical practice. Immunotherapies perturb a complex system of interactions among genomically unstable tumor cells, diverse cells within the tumor microenvironment including the systemic adaptive and innate immune cells. The drive to develop increasingly effective immunotherapy regimens is tempered by the risk of immune-related adverse events. Evidence-based biomarkers that measure the potential for therapeutic response and/or toxicity are critical to guide optimal patient care and contextualize the results of immunotherapy clinical trials. Responding to the lack of guidance on biomarker testing in early-phase immunotherapy clinical trials, we propose a definition and listing of essential biomarkers recommended for inclusion in all such protocols. These recommendations are based on consensus provided by the Society for Immunotherapy of Cancer (SITC) Clinical Immuno-Oncology Network (SCION) faculty with input from the SITC Pathology and Biomarker Committees and the Journal for ImmunoTherapy of Cancer readership. A consensus-based selection of essential biomarkers was conducted using a Delphi survey of SCION faculty. Regular updates to these recommendations are planned. The inaugural list of essential biomarkers includes complete blood count with differential to generate a neutrophil-to-lymphocyte ratio or systemic immune-inflammation index, serum lactate dehydrogenase and albumin, programmed death-ligand 1 immunohistochemistry, microsatellite stability assessment, and tumor mutational burden. Inclusion of these biomarkers across early-phase immunotherapy clinical trials will capture variation among trials, provide deeper insight into the novel and established therapies, and support improved patient selection and stratification for later-phase clinical trials.
Collapse
Affiliation(s)
- Tricia R Cottrell
- Queen's University Sinclair Cancer Research Institute, Kingston, Ontario, Canada
| | | | - Alaa Ali
- Stem Cell Transplant and Cellular Immunotherapy Program, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, Washington, DC, USA
| | - Carlo B Bifulco
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Christian M Capitini
- University of Wisconsin School of Medicine and Public Health and Carbone Cancer Center, Madison, Wisconsin, USA
| | | | - Anthony R Cillo
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Deborah Collyar
- Patient Advocates In Research (PAIR), Danville, California, USA
| | - Leslie Cope
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | | | - Sacha Gnjatic
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Susan Halabi
- Duke School of Medicine and Duke Cancer Institute, Durham, North Carolina, USA
| | - Gary Kohanbash
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Holden T Maecker
- Stanford University School of Medicine, Stanford, California, USA
| | - Saman Maleki Vareki
- Department of Oncology and Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Sarah Mullin
- Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Barbara Seliger
- Campus Brandenburg an der Havel, Brandenburg Medical School, Halle, Germany
| | - Janis Taube
- Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Wim Vos
- Radiomics.bio, Liège, Belgium
| | - Joe Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Kristin G Anderson
- Department of Microbiology, Immunology and Cancer Biology, Department of Obstetrics and Gynecology, Beirne B. Carter Center for Immunology Research and the University of Virginia Comprehensive Cancer Center, University of Virginia, Charlottesville, Virginia, USA
| | - Tullia C Bruno
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Codruta Chiuzan
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | | | | | | | | | - Elizabeth G Hill
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Brian P Hobbs
- Dell Medical School, The University of Texas, Austin, Texas, USA
| | - Yana G Najjar
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | | | - Sumit K Subudhi
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ryan J Sullivan
- Massachusetts General Hospital, Harvard Medical School, Needham, Massachusetts, USA
| | | |
Collapse
|
13
|
Hua CH, Lin JC, Wen YW, Kang CJ, Lin CY, Ku HY, Fan KH, Ng SH, Lee SR, Tsai YT, Chen WC, Lee LY, Chien CY, Wang CP, Che TM, Terng SD, Tsai CY, Wang HM, Hsieh CH, Yeh CH, Lin CH, Tsao CK, Cheng NM, Fang TJ, Huang SF, Lee LA, Fang KH, Wang YC, Lin WN, Hsin LJ, Yen TC, Liao CT. Is postoperative adjuvant therapy necessary for pT3N0 oral cavity squamous cell carcinoma? Oral Oncol 2025; 162:107198. [PMID: 39879895 DOI: 10.1016/j.oraloncology.2025.107198] [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: 11/10/2024] [Revised: 01/05/2025] [Accepted: 01/20/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND The current NCCN guidelines advocate for the use of adjuvant radiotherapy (RT) or chemoradiotherapy (CRT) in pT3N0 oral cavity squamous cell carcinoma (OCSCC). Here, we sought to evaluate whether postoperative RT/CRT may confer a survival advantage in pT3N0 patients who lack adverse pathological features. METHODS A dataset of 852 pT3N0 OCSCC patients treated between 2018 and 2021 was analyzed. Patients with adverse pathological features (i.e., positive margins, margins <5 mm, lymphovascular invasion, or perineural invasion) were excluded, leaving 235 eligible patients. The cohort was further refined by omitting 12 patients who received surgery plus chemotherapy and two cases who received RT doses <5000 cGy, resulting in two study groups: surgery alone (n = 131) and surgery plus RT/CRT (n = 90). RESULTS Baseline characteristics were comparable between groups, except for a higher proportion of buccal subsite (p = 0.0063) and female patients (p = 0.0495) in the surgery alone group. Kaplan-Meier analyses revealed no significant differences in 4-year disease-specific survival (DSS) (84.2 % versus 85.9 %, p = 0.8834) or overall survival (OS) (75.6 % versus 84.8 %, p = 0.2376). These findings were confirmed by multivariable analyses and remained consistent after propensity score matching (n = 74 per group), with similar survival outcomes between groups (4-year DSS, 90.7 % versus 83.2 %, p = 0.4020; 4-year OS, 87.0 % versus 81.9 %, p = 0.6121) CONCLUSIONS: For patients with pT3N0 OCSCC lacking adverse pathological features, the addition of adjuvant RT/CRT does not appear to confer a survival benefit over surgery alone.
Collapse
Affiliation(s)
- Chun-Hung Hua
- Department of Otorhinolaryngology, China Medical University Hospital Taichung Taiwan Republic of China
| | - Jin-Ching Lin
- Department of Radiation Oncology Changhua Christian Hospital Changhua Taiwan Republic of China
| | - Yu-Wen Wen
- Clinical Informatics and Medical Statistics Research Center, Chang Gung University Taoyuan Taiwan Republic of China; Division of Thoracic Surgery, Chang Gung Memorial Hospital Taoyuan Taiwan Republic of China
| | - Chung-Jan Kang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Chien-Yu Lin
- Department of Radiation Oncology, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Hsiu-Ying Ku
- National Institute of Cancer Research, National Health Research Institutes Miaoli Taiwan Republic of China
| | - Kang-Hsing Fan
- Department of Radiation Oncology, New Taipei Municipal TuCheng Hospital Taiwan Republic of China
| | - Shu-Hang Ng
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Shu-Ru Lee
- Research Service Center for Health Information, Chang Gung University Taoyuan Taiwan Republic of China
| | - Yao-Te Tsai
- Department of Otorhinolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital Chiayi Taiwan Republic of China
| | - Wen-Cheng Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Chiayi and Chang Gung University Taoyuan Taiwan Republic of China
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Chih-Yen Chien
- Kaohsiung Chang Gung Memorial Hospital, Doctoral Program of Clinical and Experimental Medicine, National Sun Yat-sen University Taiwan Republic of China
| | - Cheng Ping Wang
- Department of Otolaryngology, National Taiwan University Hospital and College of Medicine Taipei Taiwan Republic of China
| | - Tsung-Ming Che
- Department of Otolaryngology, Shuang Ho Hospital, Taipei Medical University New Taipei City Taiwan Republic of China
| | - Shyuang-Der Terng
- Department of Head and Neck Surgery, Koo Foundation Sun Yat-Sen Cancer Center Taipei Taiwan Republic of China
| | - Chi-Ying Tsai
- Department of Oral and Maxillofacial Surgery, Chang Gung Memorial Hospital, Chang Gung University Taoyuan Taiwan Republic of China
| | - Hung-Ming Wang
- Department of Medical Oncology, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Chia-Hsun Hsieh
- Department of Medical Oncology, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Chih-Hua Yeh
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Chih-Hung Lin
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Chung-Kan Tsao
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Nai-Ming Cheng
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Tuan-Jen Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Shiang-Fu Huang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Li-Ang Lee
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Ku-Hao Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Yu-Chien Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Wan-Ni Lin
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Li-Jen Hsin
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Tzu-Chen Yen
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China
| | - Chun-Ta Liao
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University Taoyuan Taiwan Republic of China.
| |
Collapse
|
14
|
Wong FC, Merker SR, Bauer L, Han Y, Le VMH, Wenzel C, Böthig L, Heiduk M, Drobisch P, Rao VS, Malekian F, Mansourkiaei A, Sperling C, Polster H, Pecqueux M, Istvanffy R, Ye L, Kong B, Aust DE, Baretton G, Seifert L, Seifert AM, Weitz J, Demir IE, Kahlert C. Extracellular vesicles from pancreatic cancer and its tumour microenvironment promote increased Schwann cell migration. Br J Cancer 2025; 132:326-339. [PMID: 39863771 PMCID: PMC11832759 DOI: 10.1038/s41416-024-02915-0] [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: 03/24/2024] [Revised: 10/27/2024] [Accepted: 11/19/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) exhibits a high frequency of neural invasion (NI). Schwann cells (SCs) have been shown to be reprogrammed to facilitate cancer cell migration and invasion into nerves. Since extracellular vesicles (EVs) affect the tumour microenvironment and promote metastasis, the present study analysed the involvement of EVs from pancreatic cancer cells and their microenvironment in altering SC phenotype as part of the early events in the process of NI. METHODS EVs were isolated from human/murine PDAC cells, pancreatic stellate cells (PSCs), human tissues and plasma to perform a novel 3D migration assay, qRT-PCR and western blot. Kaplan-Meier and Cox regression analyses were employed to evaluate the clinical potential of plasma EV-derived candidate from 165 PDAC patients. RESULTS The EVs from PDAC cells, PSCs derived from human tumour tissues, other cell types in the tumour microenvironment from tumour tissues and circulating plasma act as drivers of a pro-migratory phenotype of SCs by inducing dedifferentiation in SCs. Notably, p75NTR expression was upregulated in the plasma-derived EVs from patients with NI (Pn1) relative to those without NI (Pn0). High expression of plasma-derived EV p75NTR correlated with reduced overall survival and was identified as an independent prognostic factor. CONCLUSIONS These findings suggest that EV-mediated SC migration underlies the interactions contributing to PDAC-associated NI with implications for improved outcome and therapeutic strategy.
Collapse
Affiliation(s)
- Fang Cheng Wong
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| | - Sebastian R Merker
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Lisa Bauer
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Yi Han
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Van Manh Hung Le
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Carina Wenzel
- Institute for Pathology, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Lukas Böthig
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Max Heiduk
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Pascal Drobisch
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Venkatesh Sadananda Rao
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Farzaneh Malekian
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Ana Mansourkiaei
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Christian Sperling
- Institute for Pathology, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Heike Polster
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mathieu Pecqueux
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rouzanna Istvanffy
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany
| | - Linhan Ye
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany
| | - Bo Kong
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniela E Aust
- Institute for Pathology, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
- Tumour and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Gustavo Baretton
- Institute for Pathology, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
- Tumour and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Lena Seifert
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Else Kröner Clinician Scientist Professor for "Translational Tumor Immunological Research", Dresden, Germany
| | - Adrian M Seifert
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
- Else Kröner Clinician Scientist Professor for "Translational Pancreatic Surgery", Munich, Germany
| | - Christoph Kahlert
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany.
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany.
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
15
|
Cabezas-Camarero S, Pérez-Alfayate R, García-Barberán V, Gandía-González ML, García-Feijóo P, López-Cade I, Lorca V, Casado-Fariñas I, Cerón MA, Paz-Cabezas M, Sotelo MJ, García Conde M, Roldán Delgado H, Sánchez Medina Y, Díaz-Millán I, Pérez-Segura P. ctDNA detection in cerebrospinal fluid and plasma and mutational concordance with the primary tumor in a multicenter prospective study of patients with glioma. Ann Oncol 2025:S0923-7534(25)00064-X. [PMID: 39978637 DOI: 10.1016/j.annonc.2025.02.005] [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: 09/12/2024] [Revised: 01/18/2025] [Accepted: 02/03/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Cerebrospinal fluid (CSF) stands as an easily accessible reservoir for circulating tumor DNA (ctDNA) analysis in patients with central nervous system (CNS) tumors, although evidence is still limited. Our aim was to prospectively evaluate the feasibility of detecting ctDNA for mutational analysis in CSF and plasma in patients with glioma. METHODS This was a prospective study of patients with glioma diagnosed at four third-level hospitals in Spain. A customized next-generation sequencing (NGS) eight-gene panel (IDH1, IDH2, ATRX, TP53, PTEN, PIK3CA, EGFR, BRAF) was used in paired CSF, plasma and tumor samples. Mutation concordance occurred when the same pathogenic gene variant was detected in tumor and ctDNA. The prognostic value of ctDNA and that of its median variant allele frequency (mVAF) were analyzed. RESULTS Between February 2017 and March 2020, 37 patients with glioma were enrolled. The 32 patients with analyzable CSF samples comprised patients with new diagnosis (n = 23) and relapse (n = 9); World Health Organization fifth Edition types: IDH-mutant astrocytoma (n = 10), IDH-mutant oligodendroglioma (n = 6) and IDH-wildtype glioblastoma (n = 16); CSF-ctDNA-positive: 19/32 (59%); and CSF-ctDNA-negative: 13/32 (41%). CSF mutation numbers were 1 (10/19), 2 (7/19) and 3 (2/19). Frequencies of CSF-ctDNA-mutated genes were EGFR (8/19, 42%), PTEN (7/19, 37%), TP53 (6/19, 32%), IDH1 (5/19, 26%) and PIK3CA (4/19, 21%). Tumor-CSF mutation concordance was found in 16/19 (84%). Progression-free and overall survival were significantly shorter in ctDNA-positive patients with VAF equal to or greater than the mVAF compared with ctDNA-positive patients with VAF lower than the mVAF. No association was found between ctDNA in CSF and distance to closest CSF reservoir, tumor size or IDH status. ctDNA was detected in 2 of 14 (14%) individual plasma samples, in both cases concordant with the primary tumor. CONCLUSION CSF is a reliable reservoir for ctDNA analyses in patients with glioma. ctDNA is detectable in plasma although at a lower rate. Larger, prospective studies should be conducted to refine the potential role of liquid biopsy in this disease.
Collapse
Affiliation(s)
- S Cabezas-Camarero
- Department of Medical Oncology, Hospital Clínico Universitario San Carlos, IdISSC, Madrid, Spain; Department of Medical Oncology Department, IOB Institute of Oncology-Madrid, Madrid, Spain.
| | - R Pérez-Alfayate
- Department of Neurosurgery, Hospital Clínico Universitario San Carlos, IdISSC, Madrid, Spain
| | - V García-Barberán
- Molecular Oncology Laboratory, Hospital Clínico Universitario San Carlos, IdISSC, Madrid, Spain
| | - M L Gandía-González
- Department of Neurosurgery, Hospital Universitario La Paz, IdIPaz, Madrid, Spain
| | - P García-Feijóo
- Department of Neurosurgery, Hospital Universitario La Paz, IdIPaz, Madrid, Spain
| | - I López-Cade
- Experimental Therapeutics Unit, Department of Medical Oncology, Hospital Clínico Universitario San Carlos, IdISSC, Madrid, Spain
| | - V Lorca
- Molecular Oncology Laboratory, Hospital Clínico Universitario San Carlos, IdISSC, Madrid, Spain
| | - I Casado-Fariñas
- Pathology Department, Hospital Clínico Universitario San Carlos, Madrid, Spain
| | - M A Cerón
- Experimental Therapeutics Unit, Department of Medical Oncology, Hospital Clínico Universitario San Carlos, IdISSC, Madrid, Spain
| | - M Paz-Cabezas
- Department of Medical Oncology, Hospital Clínico Universitario San Carlos, IdISSC, Madrid, Spain; Molecular Oncology Laboratory, Hospital Clínico Universitario San Carlos, IdISSC, Madrid, Spain
| | - M J Sotelo
- Department of Medical Oncology, Aliada Cancer Center, Lima, Spain; Department of Medical Oncology, Clínica San Felipe, Lima, Spain; Department of Medical Oncology, Hospital María Auxiliadora, Lima, Perú, Spain
| | - M García Conde
- Department of Neurosurgery, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife
| | - H Roldán Delgado
- Department of Neurosurgery, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife
| | - Y Sánchez Medina
- Department of Neurosurgery, Hospital Nuestra Señora de la Candelaria, Santa Cruz de Tenerife, Spain
| | - I Díaz-Millán
- Department of Medical Oncology, Hospital Clínico Universitario San Carlos, IdISSC, Madrid, Spain
| | - P Pérez-Segura
- Department of Medical Oncology, Hospital Clínico Universitario San Carlos, IdISSC, Madrid, Spain; Department of Medical Oncology Department, IOB Institute of Oncology-Madrid, Madrid, Spain
| |
Collapse
|
16
|
Elias M, Bouchal J, Kral M, Kurfurstova D. Contemporary review of prognostic markers of prostate cancer from a pathologist perspective. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2025. [PMID: 39907090 DOI: 10.5507/bp.2025.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025] Open
Abstract
Prostate cancer is the most frequently diagnosed malignant tumour in men worldwide. To treat this condition, prognostic markers to distinguish indolent from aggressive disease, and biomarkers for metastatic forms are needed. From a pathologist's perspective, despite the plethora of emerging biomarkers, none to date has made its way into clinical practice. The need for prognostic and predictive markers following histological evaluation remains. This overview of some putative immunohistochemical and genetic markers reveals the pitfalls of biomarker research, notably verifiability, validity and interlaboratory comparison. Meta-analyses and extensive cooperation between pathology departments are a sine qua non. Codes of Best Practice such as the REMARK guidelines have been advocated as a path forward. Currently, the most widely used and validated prognostic marker remains the Gleason score. Ki67 along with PTEN are the most promising prognostic markers.
Collapse
Affiliation(s)
- Martin Elias
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Milan Kral
- Department of Urology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Daniela Kurfurstova
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| |
Collapse
|
17
|
Koch A, Reinhardt P, Elicin O, Aebersold DM, Schanne DH. Predictive biomarkers of radiotherapy- related dermatitis, xerostomia, mucositis and dysphagia in head and neck cancer: A systematic review. Radiother Oncol 2025; 203:110689. [PMID: 39706342 DOI: 10.1016/j.radonc.2024.110689] [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: 08/20/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Radiotherapy is essential for treating head and neck cancer but often leads to severe toxicity. Traditional predictors include anatomical location, tumor extent, and dosimetric data. Recently, biomarkers have been explored to better predict and understand toxicity. This review aims to summarize the current literature, assess data quality, and guide future research. METHODS Two reviewers independently screened EMBASE and PubMed for studies published between 2010 and 2023. Endpoints were dermatitis, mucositis, sticky saliva/xerostomia, and dysphagia. Statistical analysis was performed using R, and bias assessed via a modified QUIPS questionnaire. Pathway analysis was conducted using gProfiler. The study adhered to PRISMA and COSMOS-E guidelines and was registered in the PROSPERO database (#CRD42023361245). RESULTS Of 2,550 abstracts, 69 publications met the inclusion criteria. These studies involved a median of 81 patients, primarily male (75 %), with common primary tumors in the nasopharynx (32 %) and oropharynx (27 %). Most patients (84 %) had advanced disease (stage III/IV). The most frequently studied biomarkers were DNA-based single-nucleotide polymorphisms (SNPs, 59 %), salivary proteins (13 %), and bacteria (10 %). Ten statistically-significant biomarkers (all SNPs) in low-bias publications were identified, particularly in DNA repair and cell detoxification pathways. Data quality was often poor and few validation studies were present in the dataset. CONCLUSION This review provides an overview of the research landscape, highlights research gaps and provides recommendations for future research directions. We identified several potential biomarkers, particularly in DNA repair pathways, that align with current understanding of radiation-induced cell damage. However, the overall data quality was poor, with key clinical variables often missing. Overall, rigorous standardization of reporting, validation studies and multi-center collaborations to increase study power and sample sizes are necessary to build high-level evidence for clinical application.
Collapse
Affiliation(s)
- Alexander Koch
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Philipp Reinhardt
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Olgun Elicin
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Daniel M Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Daniel H Schanne
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland; Graduate School for Health Sciences, University of Bern, Switzerland.
| |
Collapse
|
18
|
Nie F, Guo J, Pan J, Guo Z, Wang C, Yan J, Ma W. Effects of antibiotics on the anti-tumor efficacy of immune checkpoint inhibitor therapy. Clin Transl Oncol 2025; 27:790-799. [PMID: 39046682 DOI: 10.1007/s12094-024-03615-0] [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: 05/05/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024]
Abstract
PURPOSE Immunotherapy using immune checkpoint inhibitors (ICIs) has shown several benefits over traditional therapies. However, the eligible population remains small. Antibiotic (ATB) use might reduce immunotherapy efficacy by disrupting the gut microbiota. However, in China, ATB effect on ICI therapy efficacy remains unelucidated. We aimed to assess the effects of ATBs on the anti-tumor efficacy of ICIs to provide a reference for clinical use. METHODS We included 134 patients with advanced tumors undergoing ICI therapy at Shanghai Jiading District Central Hospital from January 1, 2021, to October 1, 2023. They were divided into Non-ATB and ATB groups based on ATB use within 30 days before and after ICI administration. Moreover, we compared progression-free (PFS) and overall (OS) survival between the groups. RESULTS Median PFS and OS were lower in the ATB than in the Non-ATB group (PFS: 4.0 vs. 5.5 months; OS: 5.4 vs. 6.5 months). Univariate analysis revealed that ATB use significantly affected PFS (hazard ratio [HR] = 2.318, 95% confidence interval [CI] = 1.281-4.194, P = 0.005) and OS (HR = 2.115, 95% CI = 1.161-3.850, P = 0.014). Moreover, multivariate analysis revealed poor PFS (HR = 2.573, 95% CI = 1.373-4.826, P = 0.003) and OS (HR = 2.452, 95% CI = 1.298-4.632, P = 0.006) in patients who received ATBs during ICI therapy. CONCLUSIONS ATB use is negatively correlated with ICI therapy efficacy, leading to reduced PFS and OS in patients undergoing such treatment. Owing to the significant impact of ATBs on the human gut microbiome, regulation of the gut microbiome may emerge as a novel therapeutic target that can enhance the clinical activity of ICIs.
Collapse
Affiliation(s)
- Fangfang Nie
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - JunGang Guo
- Department of Thoracic Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - JiaYi Pan
- Department of Pharmacy, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, No.1 Cheng Bei Road, Jia ding District, Shanghai, 201800, China
| | - ZhaoJiao Guo
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - Chun Wang
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - Jun Yan
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China.
| | - WeiNa Ma
- Department of Pharmacy, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, No.1 Cheng Bei Road, Jia ding District, Shanghai, 201800, China.
| |
Collapse
|
19
|
Emanuelle Pereira Santos V, Luiz de França Neto P, Eda de Oliveira Isídio B, Henrique Bezerra Fontes P, Andrêssa de Moura I, Isabel Santos Cruz B, Máyra Gois de Sousa M, Luana Dos Santos D, de França São Marcos B, Sousa de Pinho S, Mendonça Alves Bandeira B, Loureiro Leão S, de Almeida Lima T, da Conceição Viana Invenção M, Rosa Sales Leal L, Cristofer Flores Espinoza B, Silva de Macêdo L, do Nascimento Carvalho M, Jéssica Duarte Silva A, Carlos de Freitas A. An overview about biomarkers in breast cancer: Insights into the diagnostic and prognostic significance. Clin Chim Acta 2025; 567:120030. [PMID: 39515632 DOI: 10.1016/j.cca.2024.120030] [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: 04/30/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Breast cancer (BC) is one of the most significant neoplasms globally due to its high incidence and mortality, particularly among females. As a highly heterogeneous pathology, biomarkers are essential for characterizing specific tumors. Currently, several biological processes are well-described in the context of this neoplasm, such as alterations in BRCA1/2, HER, and pathways involving estrogen and progesterone hormone receptors. These studies have enabled the use of these findings as more precise methods for diagnosis, prognosis, and treatment. However, beyond patients who do not exhibit these classic markers, some individuals within the same risk group respond differently to treatment. Therefore, the search for biological markers that can improve diagnosis, aid in stratification, or serve as therapeutic targets is continuous and urgent. Genetic signatures have led to molecular tests currently used in clinical practice, though certain limitations persist. Understanding genetic and epigenetic mechanisms facilitates the identification of potential biomarkers. Biomarker targets must undergo experimental and clinical trials on samples of significant size before reaching clinical utility. In this review, we compile the classical markers and describe the potential use of other markers associated with the biological processes of this neoplasm.
Collapse
Affiliation(s)
- Vanessa Emanuelle Pereira Santos
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Pedro Luiz de França Neto
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Beatriz Eda de Oliveira Isídio
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Pedro Henrique Bezerra Fontes
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Ingrid Andrêssa de Moura
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Bruna Isabel Santos Cruz
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Mylenna Máyra Gois de Sousa
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Daffany Luana Dos Santos
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Bianca de França São Marcos
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Samara Sousa de Pinho
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Beatriz Mendonça Alves Bandeira
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Stephanie Loureiro Leão
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Thainá de Almeida Lima
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Maria da Conceição Viana Invenção
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Lígia Rosa Sales Leal
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Benigno Cristofer Flores Espinoza
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Larissa Silva de Macêdo
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Matheus do Nascimento Carvalho
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Anna Jéssica Duarte Silva
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| |
Collapse
|
20
|
Kang CJ, Wen YW, Lin CY, Ng SH, Tsai YT, Ku HY, Lou PJ, Wang CP, Lin JC, Hua CH, Lee SR, Fan KH, Chen WC, Lee LY, Chien CY, Chen TM, Terng SD, Tsai CY, Wang HM, Hsieh CH, Yeh CH, Lin CH, Tsao CK, Cheng NM, Fang TJ, Huang SF, Lee LA, Fang KH, Wang YC, Lin WN, Hsin LJ, Yen TC, Liao CT. Prognostic significance of diagnosis-to-surgery interval in oral cavity squamous cell carcinoma: A nationwide study. Oral Oncol 2025; 161:107196. [PMID: 39842233 DOI: 10.1016/j.oraloncology.2025.107196] [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: 11/29/2024] [Revised: 12/18/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND The question as to whether prolonged diagnosis-to-surgery intervals (DSIs) may compromise survival outcomes in patients with oral cavity squamous cell carcinoma (OCSCC) remains unanswered. This nationwide study was designed to address this issue. METHODS We analyzed data from 26,214 patients with first primary OCSCC identified in the Taiwanese Cancer Registry Database between 2011 and 2021. The optimal DSI cutoff was determined based on 5-year disease-specific survival (DSS) and overall survival (OS) rates using Cox regression analysis. Patients were categorized into three distinct DSI groups: ≤20 days (47 %), 21-31 days (31 %), and > 31 days (22 %). RESULTS The 5-year DSS and OS rates for the ≤20/21-31/>31 days groups were 81 %/78 %/77 % and 73 %/70 %/68 %, respectively (both p < 0.0001). Patients in the ≤20 days group had a higher prevalence of pathological stages I-II. After adjustment for potential confounders in multivariable analysis, a DSI > 31 days (versus ≤ 20 days) retained independent associations with adverse outcomes at 5 years, with hazard ratios of 1.07 for both DSS and OS. Propensity score matching and multivariable analysis comparing DSI ≤ 20 days to DSI > 31 days stratified by pathological stage III-IV showed that higher DSS and OS rates were observed in patients with DSI ≤ 20 days than DSI > 31 days (68 %/66 %, p = 0.0586; 60 %/57 %, p = 0.0228, respectively), with hazard ratios of 1.09 for both DSS and OS. CONCLUSIONS Our findings indicate that DSI is an independent predictor of 5-year DSS and OS in patients with OCSCC. A DSI exceeding 31 days, or even 21 days, may potentially decrease survival outcomes.
Collapse
Affiliation(s)
- Chung-Jan Kang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Yu-Wen Wen
- Clinical Informatics and Medical Statistics Research Center, Chang Gung University, Taoyuan, Taiwan, ROC; Division of Thoracic Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Chien-Yu Lin
- Department of Radiation Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Shu-Hang Ng
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Yao-Te Tsai
- Department of Otorhinolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan, ROC
| | - Hsiu-Ying Ku
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan, ROC
| | - Pei-Jen Lou
- Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Cheng Ping Wang
- Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Jin-Ching Lin
- Department of Radiation Oncology, Changhua Christian Hospital, Changhua, Taiwan, ROC
| | - Chun-Hung Hua
- Department of Otorhinolaryngology, China Medical University Hospital, Taichung, Taiwan, ROC
| | - Shu-Ru Lee
- Research Service Center for Health Information, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Kang-Hsing Fan
- Department of Radiation Oncology, New Taipei Municipal TuCheng Hospital, Taiwan, ROC
| | - Wen-Cheng Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Chiayi and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chih-Yen Chien
- Kaohsiung Chang Gung Memorial Hospital, Doctoral Program of Clinical and Experimental Medicine, National Sun Yat-sen University, Taiwan, ROC
| | - Tsung-Ming Chen
- Department of Otolaryngology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan, ROC
| | - Shyuang-Der Terng
- Department of Head and Neck Surgery, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan, ROC
| | - Chi-Ying Tsai
- Department of Oral and Maxillofacial Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Hung-Ming Wang
- Department of Medical Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chia-Hsun Hsieh
- Department of Medical Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chih-Hua Yeh
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chih-Hung Lin
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chung-Kan Tsao
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Nai-Ming Cheng
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Tuan-Jen Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Shiang-Fu Huang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Li-Ang Lee
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Ku-Hao Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Yu-Chien Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Wan-Ni Lin
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Li-Jen Hsin
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Tzu-Chen Yen
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chun-Ta Liao
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC.
| |
Collapse
|
21
|
Öztürk SD, Turan G, Gezer Ş. Prognostic value of circulating and tumor microenvironmental biomarkers in endometrial cancer. Pathol Res Pract 2025; 266:155765. [PMID: 39721096 DOI: 10.1016/j.prp.2024.155765] [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: 09/05/2024] [Revised: 11/23/2024] [Accepted: 12/08/2024] [Indexed: 12/28/2024]
Abstract
AIM Endometrial cancer (EC) is one of the three most common gynecological malignancies. Thus, it is estimated that the mortality rates due to this disease will increase. Our aim is to study the immune microenvironment in all ECs together with Hematoxylin Eosin (H&E), immunohistochemical (IHC) and Biochemical parameters (NLR) and to evaluate their contribution to prognosis by comparing them with each other. MATERIALS AND METHODS A total of 340 EC patients were included in the study. PDL-1 immunohistochemical stain was applied to blocks prepared from representative tumor tissues by Tissue Microarray method. Tumor- infiltrating lymphocytes and neutrophils were also scored with Hematoxylin Eosin stain. The NLR values calculated from peripheral blood. The effects of all parameters on survival, recurrence/metastasis were statistically evaluated. RESULTS Regarding TIL Scores, patients with higher TIL scores were found to have longer disease-free survival and total survival. In patients with high TIN scores, disease-free survival and overall survival were shorter. (p < 0.001) The overall survival of patients with PDL-1 negative was found to be longer. (p: 0.07). In terms of the effect on OS, an NLR cut-off value of 2.72 was determined (specificity 38.89 %, sensitivity 82.57 %, AUC 0.65). OS was 77.11 months ( ± 0.04, 95 % CI: 71.8-81.9) in the patients with NLR value slower than 2.72 and 76.5 months ( ± 1.3, 95 % CI: 36.3-63.7) in those with NLR ≥ 2.72 (p: 0.001). CONCLUSION In this study, all patients of EC with high TIL scores, low TIN scores, and with PD-L1 negativity exhibited significantly longer survival times. TIN appears to be a highly valuable parameter in terms of both OS and PFS, particularly compared to TIL and PD-L1. This research represent a pioneering study for future studies involving larger samples in the context of being able to use different parameters in predicting prognosis and treatment in patients with EC.
Collapse
Affiliation(s)
- Seda Duman Öztürk
- Department of Pathology, Kocaeli University School of Medicine, Kocaeli, Turkey.
| | - Gupse Turan
- Department of Pathology, Kocaeli University School of Medicine, Kocaeli, Turkey
| | - Şener Gezer
- Department of Obstetrics and Gynecology, Kocaeli University School of Medicine, Kocaeli, Turkey
| |
Collapse
|
22
|
Griffiths-King DJ, Delivett C, Peet A, Waite J, Novak J. Limited research investigating the value of MRI in predicting future cognitive morbidity in survivors of paediatric brain tumours: A systematic-review and call to action for clinical neuroimaging researchers. PLoS One 2025; 20:e0314721. [PMID: 39883618 PMCID: PMC11781722 DOI: 10.1371/journal.pone.0314721] [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: 07/22/2024] [Accepted: 11/14/2024] [Indexed: 02/01/2025] Open
Abstract
Survivors of pediatric brain tumours are at a high risk of cognitive morbidity. Reliable individual-level predictions regarding the likelihood, degree, and affected domains of cognitive impairment would be clinically beneficial. While established risk factors exist, quantitative MRI analysis may enhance predictive value, above and beyond current clinical risk models. This systematic review addresses the question: "Do MRI markers predict future cognitive functioning in pediatric brain tumour survivors?" We conducted a comprehensive search for studies published up to March 2024 that assessed MRI variables as predictors of later neuropsychological outcomes in pediatric brain tumour patients. Only studies that acquired MRI scans at an earlier timepoint to predict subsequent cognitive test performance were included. Surprisingly, few studies met these criteria, with identified research focusing primarily on MRI measures of cerebellar and white matter damage as features in predicting cognitive outcomes. Ultimately, this review reveals a limited literature, characterized by small sample sizes and poor-quality studies, placing findings at high risk of bias. Consequently, the quality and conclusions drawn from the existing research are constrained, especially in the context of prediction studies. Given the significant implications for this clinical population, this review highlights the urgent need for further investigation and a 'call to action' for medical imaging researchers in pediatric neuro-oncology.
Collapse
Affiliation(s)
| | - Christopher Delivett
- Aston Institute of Health and Neurodevelopment, Aston University, Birmingham, United Kingdom
| | - Andrew Peet
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Birmingham Women’s and Children’s Hospital NHS Foundation Trust, Institute of Child Health, Birmingham, United Kingdom
| | - Jane Waite
- Aston Institute of Health and Neurodevelopment, Aston University, Birmingham, United Kingdom
| | - Jan Novak
- Aston Institute of Health and Neurodevelopment, Aston University, Birmingham, United Kingdom
| |
Collapse
|
23
|
Shenasa E, He Y, Wang Z, Tu D, Gao D, Kos Z, Thornton S, Nielsen TO. Digital Profiling of Immune Biomarkers in Breast Cancer: Relation to Anthracycline Benefit. Mod Pathol 2025; 38:100718. [PMID: 39863112 DOI: 10.1016/j.modpat.2025.100718] [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: 08/13/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Assessment of the tumor-immune microenvironment can be used as a prognostic tool for improved survival and as a predictive biomarker for treatment benefit, particularly from immune-modulating treatments including cytotoxic chemotherapy. Using digital spatial profiling (DSP), we studied the tumor-immune microenvironment of 522 breast cancer cases by quantifying 35 immune biomarkers on tissue microarrays from the MA.5 phase III clinical trial. In this trial, node-positive breast cancer patients were randomized to receive either non-anthracycline chemotherapy (cyclophosphamide, methotrexate, 5'-fluorouracil [CMF]) or anthracycline-containing cytotoxic chemotherapy (CEF). Donor block hematoxylin and eosin (H&E)-stained sections were scored for the level of stromal tumor-infiltrating lymphocytes (sTILs), according to the international guidelines. We hypothesized that patients with higher levels of tumor-immune infiltration, assessed by either DSP or H&E staining, would benefit from CEF (relative to CMF) more than patients with lower immune infiltration. Unsupervised hierarchical clustering of digitally scored biomarkers revealed 2 patient clusters: immune infiltrated versus ignored. Following a prespecified statistical plan crafted to meet REMARK (REporting recommendations for tumor MARKer prognostic studies) guidelines, we found that the DSP-derived Immune Cluster assignment did not predict an improved 10-year relapse-free survival for patients receiving CEF compared with CMF. However, a secondary hypothesis revealed a significant predictive value for H&E sTILs assessed on full-faced sections for CEF benefit over CMF in the entire cohort and the human epidermal growth factor receptor 2-enriched subset. As exploratory analyses, supervised clustering of DSP-scored biomarkers suggested that low levels of T-cell immunoglobulin and mucin domain 3 TIM-3 and high levels of human leukocyte antigen HLA-DR and programmed cell death protein ligand PD-L-1 are associated with sensitivity to CEF. Although novel high-plex techniques provide a detailed insight into the tumor microenvironment, conventional H&E staining remains a powerful tool that can be applied to full-faced sections to assess the value of the immune microenvironment, particularly sTILs, in predicting benefits from immunogenic chemotherapies.
Collapse
Affiliation(s)
- Elahe Shenasa
- Interdisciplinary Oncology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ye He
- Visual Computing and Virtual Reality Key Laboratory of Sichuan Province, Sichuan Normal University, Sichuan, China
| | - Zehui Wang
- Mathematics and Statistics, Queen's University, Kingston, Ontario, Canada
| | - Dongsheng Tu
- Community Health & Epidemiology, Queen's University, Kingston, Ontario, Canada
| | - Dongxia Gao
- MAPcore, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zuzana Kos
- Pathology, BC Cancer Vancouver Centre, Vancouver, British Columbia, Canada
| | - Shelby Thornton
- MAPcore, University of British Columbia, Vancouver, British Columbia, Canada
| | - Torsten O Nielsen
- Interdisciplinary Oncology, University of British Columbia, Vancouver, British Columbia, Canada; MAPcore, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
24
|
Li F, Liang J, Wei X. Epigenetic modification of Castor zinc finger 1 (CASZ1) is associated with tumor microenvironments and prognosis of clear cell renal cell carcinoma. Int J Surg 2025; 111:988-997. [PMID: 39235847 PMCID: PMC11745609 DOI: 10.1097/js9.0000000000002070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) represents the predominant and remarkably diverse form of renal cell carcinoma. The involvement of the Castor zinc finger 1 (CASZ1) gene in adverse prognostic outcomes has been observed across different cancer types. Nevertheless, the specific altered activities and associated multi-omics characteristics of CASZ1 in ccRCC remain unelucidated. METHOD In order to explore the expression of CASZ1, evaluate its prognostic significance, and aid in the therapeutic decision-making process for patients with ccRCC, The Cancer Genome Atlas (TCGA), Gene expression omnibus (GEO), and The Human Protein Atlas (HPA) databases were utilized to gather data on clinicopathological data, prognostic information, genomic, methylomic and immunomic data. Additionally, the Genomics of Drug Sensitivity in Cancer (GDSC) database provided information on drug sensitivity. RESULTS CASZ1 expression was found to be significantly reduced in ccRCC and was associated with unfavorable pathological characteristics and a bleak prognosis. Diminished CASZ1 mRNA levels were notably correlated with heightened cytosine-phosphate-guanine (CpG) methylation, indicating a poorer prognosis for patients with increased methylation. Examination of RNA-seq data from TCGA indicated that the CASZ1-high expression subgroup displayed heightened immune cell infiltration and increased expression of immune checkpoint markers, potentially suggesting a more favorable response to immunotherapy. Furthermore, data from the GDSC database indicated that the CASZ1-low expression subgroup might exhibit greater sensitivity to anti-angiogenetic treatments, such as Sunitinib and Axitinib. CONCLUSIONS These results indicate that CASZ1 may function as a biomarker for distinguishing various tumor microenvironment phenotypes, predicting prognosis, and assisting in treatment decisions for individuals with ccRCC.
Collapse
Affiliation(s)
| | | | - Xin Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu City, Sichuan Province, People’s Republic of China
| |
Collapse
|
25
|
Song Y, Peng Y, Qin C, Jiang S, Lin J, Lai S, Wu J, Ding M, Du Y, Yu L, Xu T. Antibiotic use attenuates response to immune checkpoint blockade in urothelial carcinoma via inhibiting CD74-MIF/COPA: revealing cross-talk between anti-bacterial immunity and ant-itumor immunity. Int J Surg 2025; 111:972-987. [PMID: 38995167 PMCID: PMC11745717 DOI: 10.1097/js9.0000000000001901] [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: 12/17/2023] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Immune checkpoint blockade (ICB) has emerged as a promising therapy for both resectable urothelial carcinoma (UC) patients preparing for radical surgery and unresectable UC patients, whereas the objective response rate of ICB remains unsatisfactory due to various factors. Antibiotic (ATB) use can influence intratumoral bacteria, which may further reduce ICB efficacy. The study aims to evaluate the effects of ATB use on prognosis and response in UC patients undergoing ICB, and explore potential molecular mechanisms of ATBs and intratumoral bacteria impacting UC immune microenvironment. MATERIALS AND METHODS Pooled analyses, synthesizing evidence from 3496 UC patients with ICB treatment, were conducted. In addition, single-cell RNA and single-cell microbiome data were analyzed based on eight UC samples and 63 185 single cells. Bulk RNA sequencing and clinical data from a single-arm, multicenter, atezolizumab-treated, phase 2 trial, IMvigor210, were used for validation. RESULTS ATB use exhibited worse overall survival (HR=1.46, 95% CI=[1.20-1.77], P <0.001 and lower objective response (OR=0.43, 95% CI=[0.27-0.68], P <0.001 in UC patients receiving ICB. Single-cell transcriptome and single-cell microbiome analyses identified the presence of intratumoral bacteria was obviously related to elevated antibacterial immune functions; and antibacterial immunity was positively correlated to antitumor immunity in UC immune microenvironment. Intratumoral bacteria could up-regulate CD74-MIF/COPA signaling of immune cells and activation of CD74-MIF/COPA mediated the promotion of T cell antitumor function induced by antibacterial immune cells. UC patients with higher CD74-MIF/COPA signaling carried better overall survival (HR=1.60, 95% CI=[1.19-2.15], P =0.002) in immunotherapy cohort. CONCLUSION ATB use reduces overall survival and objective response to ICB in UC patients. Antibacterial immune cell functions induced by intracellular bacteria in the UC microenvironment might up-regulate the function of antitumor T immune cells via activating CD74-MIF/COPA , whereas ATB could inhibit the above process through killing intracellular bacteria and result in poorer clinical benefit of ICB. The use of ATB should be considered carefully during the neoadjuvant immunotherapy period for resectable UC patients preparing for radical surgery and during the immunotherapy period for unresectable UC patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yiqing Du
- Department of Urology, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Luping Yu
- Department of Urology, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Tao Xu
- Department of Urology, Peking University People’s Hospital, Beijing, People’s Republic of China
| |
Collapse
|
26
|
Sinn K, Elbeialy A, Mosleh B, Aigner C, Schelch K, Laszlo V, Dome B, Hoda MA, Grusch M. High circulating activin A plasma levels are associated with tumour stage and poor survival in treatment-naive lung squamous cell cancer patients. Transl Oncol 2025; 51:102153. [PMID: 39405924 PMCID: PMC11525229 DOI: 10.1016/j.tranon.2024.102153] [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: 07/01/2024] [Revised: 09/09/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVES Lung squamous cell carcinoma (LUSC) is associated with a poor prognosis and a lack of specific treatment options. The dysregulation of activin A (ActA) has been reported in various malignancies. Herein, we investigated the diagnostic and prognostic significance of ActA in LUSC. MATERIALS AND METHODS ActA concentrations were measured using ELISA in plasma samples of 128 LUSC patients (stage I-IV) and 73 controls, and correlated those values with clinicopathological parameters and survival. RESULTS ActA plasma levels were significantly higher in therapy-naive LUSC patients compared to controls (444.1 ± 310.9 pg/mL vs 338.9 ± 145.5 pg/mL, p = 0.010). ActA levels significantly correlated with advanced stage as well as with T and N factors. High circulating ActA levels were significantly increased in metastatic disease patients compared to M0 disease. Further, patients with ActA levels above a computationally established optimal cut-off value of 443.0 pg/mL had a significantly worse median overall (OS, 17.63 vs 64.77 months, HR 0.391, 95 % CI 0.200-0.762, p < 0.001) and median disease-/progression-free survival (DFS/PFS; 11.57 vs 30.20 months, HR 0.502, 95 % CI 0.248-1.019, p = 0.020). Multivariate analysis revealed that high ActA levels were an independent prognostic factor for shorter OS (p = 0.001) and DFS/PFS (p = 0.018). A newly developed score combining CRP and ActA levels was also an independent prognostic factor for OS and DFS/PFS. CONCLUSION Measurement of circulating ActA levels may help identify advanced-stage LUSC patients, and this value could serve as a prognostic parameter in LUSC. Thus, ActA may be a novel blood-based biomarker for identifying LUSC patients with distant metastasis.
Collapse
Affiliation(s)
- Katharina Sinn
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Ahmed Elbeialy
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Berta Mosleh
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Clemens Aigner
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Karin Schelch
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria; Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Viktoria Laszlo
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Balazs Dome
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria; Department of Tumour Biology, National Korányi Institute of Pulmonology, Budapest, Hungary; Department of Thoracic Surgery, National Institute of Oncology, Semmelweis University, Budapest, Hungary; Department of Translational Medicine, Lund University, Lund, Sweden
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Michael Grusch
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
27
|
Gopaul VL, Winstone L, Gatien BG, Nault BD, Maiti S, Opperman RM, Majumder M. A Prospective Tumour Marker for Breast Cancer: YWHAB and Its Role in Promoting Oncogenic Phenotypes. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:935-956. [PMID: 39703345 PMCID: PMC11656333 DOI: 10.2147/bctt.s479384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024]
Abstract
Background YWHAB (14-3-3 Beta) was found in the secretome of miR-526b and miR-655 overexpressed breast cancer (BRCA) cell lines. The potential of YWHAB as a therapeutic target or biomarker for BRCA is investigated here. Methods After YWHAB was knocked down with siRNA, BRCA cell lines were used for in vitro assays (proliferation, migration, epithelial-to-mesenchymal transition). In silico analysis and in situ validation with BRCA plasma and biopsy tissues were used to test YWHAB's biomarker potential. Results YWHAB RNA and protein expression are elevated in aggressive BRCA cell lines, and the knockdown of YWHAB inhibited cell migration, proliferation, and EMT in all subtypes of tumour cell lines. YWHAB expression is significantly higher in BRCA biopsy tissue and blood plasma compared to control tissues and benign plasmas. YWHAB is expressed in all hormonal subtypes of BRCA tumours and has shown increased expression in advanced tumour stages. Its high expression is linked to poor patient survival. YWHAB is a sensitivity tumour marker (AUC of 0.7340, p = 0.0012) but is not a promising blood biomarker. Nevertheless, combined with pri-miR-526b, YWHAB mRNA expression shows potential as a BRCA blood biomarker (AUC of 0.711, p = 0.032), which must be validated in a larger sample set. Conclusion We elucidate the novel role of YWHAB as a therapeutic target in BRCA, given that its inhibition mitigated aggressive phenotypes across all tumour subtypes, including triple-negative breast cancer. Furthermore, YWHAB emerges as a potential tumour marker, exhibiting high expression in metastatic BRCA and correlating with poor patient survival; however, it is not a sensitive blood biomarker.
Collapse
Affiliation(s)
| | - Lacey Winstone
- Department of Biology, Brandon University, Brandon, MB, Canada
| | | | - Braydon D Nault
- Department of Biology, Brandon University, Brandon, MB, Canada
| | - Sujit Maiti
- Department of Biology, Brandon University, Brandon, MB, Canada
| | - Reid M Opperman
- Department of Biology, Brandon University, Brandon, MB, Canada
| | | |
Collapse
|
28
|
Vieira E Silva FF, Caponio VCA, Ballini A, Chamorro-Petronacci CM, Lourenzo-Pouso AI, García-García A, Di Domenico M, Suaréz-Peñaranda JM, Pérez-Sayáns M, Padín-Iruegas ME. Smac/DIABLO protein acts as an independent prognostic factor in oral squamous cell carcinoma. Sci Rep 2024; 14:30065. [PMID: 39627250 PMCID: PMC11614858 DOI: 10.1038/s41598-024-76962-1] [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: 06/04/2024] [Accepted: 10/18/2024] [Indexed: 12/06/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) poses significant health risks with increasing incidence and mortality rates. In this context, there is an urgent need to explore novel biomarkers to enhance therapeutic strategies and improve survival. Understanding apoptotic evasion in cancer pathogenesis, this pioneering study aims to investigate the correlation between a pro-apoptotic protein Smac/DIABLO and patient prognosis within the OSCC cohort. Immunohistochemistry (IHC) was employed to analyze Smac/DIABLO protein expression and correlate with clinicopathological and prognostic factors during a long-term follow-up. Smac/DIABLO low expression was associated with worse overall survival (OS), relapse-free survival (RFS), disease-specific survival (DSS), and an increase in risk of lymph node metastasis (LNM) in univariate analyses. Furthermore, multivariate analyses confirmed Smac/DIABLO as an independent prognostic factor, predicting poorer OS [Hazard Ratio (HR) = 3.6 (95% CI 1.7-7.6), p < 0.001], RFS [HR = 2.9 (95% CI 1.4-5.6), p = 0.003], DSS [HR = 6.7 (95% CI 2.7-16.7), p < 0.001], and increased likelihood of LNM [Odds Ratio (OR) = 4.8 (95% CI 1.4-15.9), p = 0.011]. Patients with positive Smac/DIABLO expression exhibited three times higher survival probability. Low proapoptotic protein Smac/DIABLO expression significantly influences prognostic predictions and strongly correlates with poor OSCC outcomes. Future studies involving Smac-mimetic drugs in OSCC are needed to evaluate their pro-apoptotic potential in cancer cells.
Collapse
Affiliation(s)
- Fábio França Vieira E Silva
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, University of Santiago de Compostela, San Francisco Street, s/n, Santiago de Compostela, 15782, Spain
- Health Research Institute of Santiago de Compostela (FIDIS), ORALRES Group, Santiago de Compostela University Clinical Hospital, University of Santiago de Compostela, Choupana Street, s/n, Santiago de Compostela, 15706, Spain
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via Abramo Lincoln, 5, Caserta, 81100, Italy
| | - Vito Carlo Alberto Caponio
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, Foggia, 71122, Italy
| | - Andrea Ballini
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via Abramo Lincoln, 5, Caserta, 81100, Italy.
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, Foggia, 71122, Italy.
| | - Cintia Micaela Chamorro-Petronacci
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, University of Santiago de Compostela, San Francisco Street, s/n, Santiago de Compostela, 15782, Spain
- Health Research Institute of Santiago de Compostela (FIDIS), ORALRES Group, Santiago de Compostela University Clinical Hospital, University of Santiago de Compostela, Choupana Street, s/n, Santiago de Compostela, 15706, Spain
| | - Alejandro Ismael Lourenzo-Pouso
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, University of Santiago de Compostela, San Francisco Street, s/n, Santiago de Compostela, 15782, Spain
| | - Abel García-García
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, University of Santiago de Compostela, San Francisco Street, s/n, Santiago de Compostela, 15782, Spain
- Health Research Institute of Santiago de Compostela (FIDIS), ORALRES Group, Santiago de Compostela University Clinical Hospital, University of Santiago de Compostela, Choupana Street, s/n, Santiago de Compostela, 15706, Spain
| | - Marina Di Domenico
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via Abramo Lincoln, 5, Caserta, 81100, Italy
| | - José Manuel Suaréz-Peñaranda
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, University of Santiago de Compostela, San Francisco Street, s/n, Santiago de Compostela, 15782, Spain
- Health Research Institute of Santiago de Compostela (FIDIS), ORALRES Group, Santiago de Compostela University Clinical Hospital, University of Santiago de Compostela, Choupana Street, s/n, Santiago de Compostela, 15706, Spain
| | - Mario Pérez-Sayáns
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, University of Santiago de Compostela, San Francisco Street, s/n, Santiago de Compostela, 15782, Spain
- Health Research Institute of Santiago de Compostela (FIDIS), ORALRES Group, Santiago de Compostela University Clinical Hospital, University of Santiago de Compostela, Choupana Street, s/n, Santiago de Compostela, 15706, Spain
- Materials Institute of Santiago de Compostela (iMATUS), Avenida do Mestre Mateo, 25, Santiago de Compostela, 15782, Spain
| | - María Elena Padín-Iruegas
- Human Anatomy and Embryology Area, Department of Functional Biology and Health Sciences, University of Vigo, Lagoas-Marcosende, s/n, Vigo, 36310, Spain
| |
Collapse
|
29
|
Mezher MA, Din S, Ilyas M, Bayat O, Abbasi QH, Ashraf I. Data Transmission Enhancement Using Optimal Coding Technique Over In Vivo Channel for Interbody Communication. BIG DATA 2024; 12:556-568. [PMID: 35377193 DOI: 10.1089/big.2021.0224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Wireless in vivo actuators and sensors are examples of sophisticated technologies. Another breakthrough is the use of in vivo wireless medical devices, which provide scalable and cost-effective solutions for wearable device integration. In vivo wireless body area networks devices reduce surgery invasiveness and provide continuous health monitoring. Also, patient data may be collected over a long period of time. Given the large fading in in vivo channels due to the signal path going through flesh, bones, skins, and blood, channel coding is considered a solution for increasing the efficiency and overcoming inter-symbol interference in wireless communications. Simulations are performed by using 50 MHz bandwidth at Ultra-Wideband frequencies (3.10-10.60 GHz). Optimal channel coding (Turbo codes, Convolutional codes, with the help of polar codes) improves data transmission performance over the in vivo channel in this research. Moreover, the results reveal that turbo codes outperform polar and convolutional codes in terms of bit error rate. Other approaches perform similarly when the information block length is increased. The simulation in this work indicates that the in vivo channel shows less performance than the Rayleigh channel due to the dense structure of the human body (flesh, skins, blood, bones, muscles, and fat).
Collapse
Affiliation(s)
- Mohanad Ahmed Mezher
- Department of Electrical and Computer Engineering, Institute of Graduate Studies, Altinbas University, Istanbul, Turkey
| | - Sadia Din
- Department of Information and Communication Engineering, Yeungnam University, Gyeongsan-si, Republic of Korea
| | - Muhammad Ilyas
- Department of Electrical and Computer Engineering, Institute of Graduate Studies, Altinbas University, Istanbul, Turkey
| | - Oguz Bayat
- School of Engineering and Natural Sciences, Altinbas University, Istanbul, Turkey
| | | | - Imran Ashraf
- Department of Information and Communication Engineering, Yeungnam University, Gyeongsan-si, Republic of Korea
| |
Collapse
|
30
|
Klibngern H, Kang CJ, Lee LY, Ng SH, Lin CY, Fan KH, Chen WC, Lin JC, Tsai YT, Lee SR, Chien CY, Hua CH, Wang CP, Chen TM, Terng SD, Tsai CY, Wang HM, Hsieh CH, Yeh CH, Lin CH, Tsao CK, Cheng NM, Fang TJ, Huang SF, Lee LA, Fang KH, Wang YC, Lin WN, Hsin LJ, Yen TC, Wen YW, Liao CT. Margin-to-depth ratio as an independent prognostic factor in resected oral cavity squamous cell carcinoma: A nationwide cohort study. Oral Oncol 2024; 159:107102. [PMID: 39488902 DOI: 10.1016/j.oraloncology.2024.107102] [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: 04/29/2024] [Revised: 07/31/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND The prognostic significance of margin-to-depth ratio (MDR) in oral cavity squamous cell carcinoma (OCSCC) remains unclear, particularly in comparison to traditional margin status. We aimed to examine the association between MDR and clinical outcomes in a large Taiwanese cohort. METHODS A total of 18,324 patients with first primary OCSCC were categorized by margin status: positive (1013), <5 mm (8371), and ≥ 5 mm (8940). Disease-specific survival (DSS) and overall survival (OS) served as the main outcome measures. RESULTS After excluding patients with positive margins (MDR = 0), the optimal MDR cutoff value for DSS and OS was 0.6. Patients with MDR > 0.6 showed significantly better 5-year DSS and OS rates (87 %, 81 %) compared to those with MDR ≤ 0.6 (71 %, 63 %) and MDR = 0 (53 %, 43 %). Multivariable analysis identified MDR ≤ 0.6 as independently associated with both DSS and OS in the entire cohort (hazard ratio [HR] = 1.34/1.32). This finding was consistent in the subgroups with surgical margins < 5 mm (HR = 1.39 for DSS and 1.38 for OS) and margins ≥ 5 mm (HR = 1.21 for both DSS and OS). In subgroups with surgical margins < 5 mm and ≥ 5 mm, an MDR > 0.6 was associated with better survival outcomes. CONCLUSIONS An MDR (cutoff: 0.6) is independently associated with prognosis in OCSCC, offering improved risk stratification compared to margin status alone. While MDR may guide surgical margin modification, further research is needed to determine whether MDR could serve as a postoperative indicator for adjuvant therapy in patients with close or clear margins.
Collapse
Affiliation(s)
- Hanpon Klibngern
- Department of Otolaryngology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chung-Jan Kang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Shu-Hang Ng
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chien-Yu Lin
- Department of Radiation Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Kang-Hsing Fan
- Department of Radiation Oncology, New Taipei Municipal TuCheng Hospital, Taiwan, ROC
| | - Wen-Cheng Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Chiayi, Taiwan, ROC
| | - Jin-Ching Lin
- Department of Radiation Oncology, Changhua Christian Hospital, Changhua, Taiwan, ROC
| | - Yao-Te Tsai
- Department of Otorhinolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan, ROC
| | - Shu-Ru Lee
- Research Service Center for Health Information, Chang Gung University, Taoyuan Taiwan, ROC
| | - Chih-Yen Chien
- Department of Otolaryngology, Chang Gung Memorial Hospital Kaohsiung Medical Center, Chang Gung University College of Medicine, Taiwan, ROC
| | - Chun-Hung Hua
- Department of Otorhinolaryngology, China Medical University Hospital, Taichung, Taiwan, ROC
| | - Cheng-Ping Wang
- Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan, ROC
| | - Tsung-Ming Chen
- Department of Otolaryngology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan, ROC
| | - Shyuang-Der Terng
- Department of Head and Neck Surgery, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan, ROC
| | - Chi-Ying Tsai
- Department of Oral and Maxillofacial Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Hung-Ming Wang
- Department of Medical Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chia-Hsun Hsieh
- Department of Medical Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chih-Hua Yeh
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chih-Hung Lin
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chung-Kan Tsao
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Nai-Ming Cheng
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Tuan-Jen Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Shiang-Fu Huang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Li-Ang Lee
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Ku-Hao Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Yu-Chien Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Wan-Ni Lin
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Li-Jen Hsin
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Tzu-Chen Yen
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Yu-Wen Wen
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC; Division of Thoracic Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Chun-Ta Liao
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC.
| |
Collapse
|
31
|
Montoyo-Pujol YG, Ponce JJ, Delgado-García S, Martín TA, Ballester H, Castellón-Molla E, Ramos-Montoya A, Lozano-Cubo I, Sempere-Ortells JM, Peiró G. High CTLA-4 gene expression is an independent good prognosis factor in breast cancer patients, especially in the HER2-enriched subtype. Cancer Cell Int 2024; 24:371. [PMID: 39523362 PMCID: PMC11552348 DOI: 10.1186/s12935-024-03554-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is the most common cancer in women and the leading cause of cancer-related death worldwide. This heterogeneous disease has been historically considered a non-immunogenic type of cancer. However, recent advances in immunotherapy have increased the interest in knowing the role of the immune checkpoints (IC) and other immune regulation pathways in this neoplasia. METHODS In this retrospective study, we evaluated the correlation of mRNA expression of CTLA-4, PDCD1 (PD1), CD274 (PD-L1), PDCD1LG2 (PD-L2), CD276 (B7-H3), JAK2, and FOXO1 with clinicopathological factors and BC patient's outcome by real-time quantitative polymerase chain reaction (qPCR). RESULTS Our results showed that immunoregulatory gene expression depends on BC immunophenotype being CTLA-4 and PDCD1 (PD1) overexpressed on triple-negative/basal-like (TN/BL) and luminal B/HER2-positive phenotypes, respectively, and CD276 (B7-H3), JAK2 and FOXO1 associated with both luminal A and luminal B/HER2-negative tumors. In addition, we found that these genes can also be related to aggressive and non-aggressive clinicopathological characteristics in BC. Finally, survival analysis showed that CTLA-4 expression levels emerge as a significant independent factor of good prognosis in BC patients, especially in the HER2-enriched subtype. CONCLUSION Considering all these data, we can conclude that the expression of immunoregulatory genes depends on tumor phenotype and has potential clinical implications in BC patients.
Collapse
Affiliation(s)
- Yoel G Montoyo-Pujol
- Research Unit, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain.
- Medical Oncology Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain.
| | - José J Ponce
- Medical Oncology Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - Silvia Delgado-García
- Gynecology and Obstetrics Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - Tina A Martín
- Gynecology and Obstetrics Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - Hortensia Ballester
- Gynecology and Obstetrics Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - Elena Castellón-Molla
- Pathology Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - Angela Ramos-Montoya
- Gynecology and Obstetrics Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - Inmaculada Lozano-Cubo
- Medical Oncology Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
| | - J Miguel Sempere-Ortells
- Research Unit, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain
- Biotechnology Department, Immunology Division, University of Alicante, Ctra San Vicente s/n. 03080-San Vicente del Raspeig, Alicante, 03010, Spain
| | - Gloria Peiró
- Research Unit, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain.
- Pathology Department, Dr Balmis University General Hospital, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante, 03010, Spain.
- Biotechnology Department, Immunology Division, University of Alicante, Ctra San Vicente s/n. 03080-San Vicente del Raspeig, Alicante, 03010, Spain.
| |
Collapse
|
32
|
Tsai YT, Chen WC, Wen YW, Lin CY, Fan KH, Lin JC, Ng SH, Lee SR, Kang CJ, Lee LY, Chien CY, Hua CH, Wang CP, Chen TM, Terng SD, Tsai CY, Wang HM, Hsieh CH, Yeh CH, Lin CH, Tsao CK, Cheng NM, Fang TJ, Huang SF, Lee LA, Fang KH, Wang YC, Lin WN, Hsin LJ, Yen TC, Liao CT. Prognostic significance of adjuvant therapy and specific radiation dosages in Taiwanese patients with oral cavity cancer and extra-nodal extension: a nationwide cohort study. BMC Cancer 2024; 24:1320. [PMID: 39456017 PMCID: PMC11515095 DOI: 10.1186/s12885-024-13048-1] [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: 06/28/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND The evidence for adjuvant chemoradiotherapy (CRT) of oral cavity squamous cell carcinoma (OCSCC) with extra-nodal extension (ENE) in National Comprehensive Cancer Network (NCCN) guidelines is derived from patients with head and neck cancer. The guidelines further suggest a radiation dose ranging from 6000 to 6600 cGy. In this nationwide study, we sought to evaluate the prognostic significance of adjuvant therapy and the specific radiation dosage in Taiwanese patients with pure OCSCC and ENE. METHODS A retrospective analysis of 1577 OCSCC patients with ENE who underwent resection and received adjuvant CRT or radiotherapy (RT) between January 2011 and December 2020 was conducted. RESULTS Multivariable analysis revealed that adjuvant RT, more than four pathologically positive nodes, and radiation dosage below 6000 cGy were independent risk factors for unfavorable 5-year disease-specific survival (DSS) and overall survival (OS). Comparing patients who received CRT (n = 1453) to those treated with RT (n = 124) before and after propensity score (PS) matching, the 5-year outcomes were as follows: before PS matching, DSS (54% versus 30%, p < 0.0001), OS (42% versus 18%, p < 0.0001); after PS matching (n = 111 in each group), DSS (52% versus 30%, p = 0.0016), OS (38% versus 21%, p = 0.0019). For patients who underwent CRT, the 5-year outcomes for different radiation dose groups (6600 - 7000 cGy, n = 1155 versus 6000 - 6500 cGy, n = 199) were as follows: before PS matching, DSS (52% versus 54%, p = 0.1904), OS (43% versus 46%, p = 0.1610); after PS matching (n = 199 in each group), DSS (55% versus 54%, p = 0.8374), OS (46.5% versus 46.3%, p = 0.7578). CONCLUSIONS For OCSCC patients with ENE, our study shows CRT improved survivals than RT alone, underscoring the clinical significance of chemotherapy. Patients undergoing CRT with irradiation doses ranging from 6000 to 6500 cGy exhibited comparable survival outcomes to those receiving doses of 6600-7000 cGy. This observation suggests that irradiation doses exceeding the 6600 cGy may not confer the survival advantage in these patients. Further research is needed to confirm our results and explore the optimal irradiation dose for managing these patients.
Collapse
Affiliation(s)
- Yao-Te Tsai
- Department of Otorhinolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan, ROC
| | - Wen-Cheng Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Chiayi and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Yu-Wen Wen
- Clinical Informatics and Medical Statistics Research Center, Chang Gung University, Taoyuan, Taiwan, ROC
- Division of Thoracic Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Chien-Yu Lin
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Kang-Hsing Fan
- Department of Radiation Oncology, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan, ROC
| | - Jin-Ching Lin
- Department of Radiation Oncology, Changhua Christian Hospital, Changhua, Taiwan, ROC
| | - Shu-Hang Ng
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Shu-Ru Lee
- Research Service Center for Health Information, Chang Gung University, Taoyuan Taiwan, ROC
| | - Chung-Jan Kang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chih-Yen Chien
- Doctoral Program of Clinical and Experimental Medicine, Kaohsiung Chang Gung Memorial Hospital, National Sun Yat-sen University, Taiwan, ROC
| | - Chun-Hung Hua
- Department of Otorhinolaryngology, China Medical University Hospital, Taichung, Taiwan, ROC
| | - Cheng Ping Wang
- Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan, ROC
| | - Tsung-Ming Chen
- Department of Otolaryngology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan, ROC
| | - Shyuang-Der Terng
- Department of Head and Neck Surgery, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan, ROC
| | - Chi-Ying Tsai
- Department of Oral and Maxillofacial Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Hung-Ming Wang
- Department of Medical Oncology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chia-Hsun Hsieh
- Department of Medical Oncology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chih-Hua Yeh
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chih-Hung Lin
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chung-Kan Tsao
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, ROC
| | - Nai-Ming Cheng
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Tuan-Jen Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Shiang-Fu Huang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Li-Ang Lee
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Ku-Hao Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Yu-Chien Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Wan-Ni Lin
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Li-Jen Hsin
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Tzu-Chen Yen
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chun-Ta Liao
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan, ROC.
- Department of Otorhinolaryngology, Head and Neck Surgery, Linkou Chang Gung Memorial Hospital, Chang Gung University, Linkou, No. 5, Fu-Hsing ST., Kwei-Shan, Taoyuan, Taiwan.
| |
Collapse
|
33
|
Saastad SA, Skjervold AH, Ytterhus B, Engstrøm MJ, Bofin AM. PD-L1 protein expression in breast cancer. J Clin Pathol 2024; 77:730-736. [PMID: 37553245 DOI: 10.1136/jcp-2023-208942] [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: 04/25/2023] [Accepted: 07/26/2023] [Indexed: 08/10/2023]
Abstract
AIMS The immune checkpoint marker, Programmed cell death-ligand 1 (PD-L1), is expressed by both cancer epithelial cells and tumour-infiltrating immune cells (TICs) thus constituting a potential target for immunotherapy. This is of particular interest in triple negative breast cancer. In this study, we assessed the prognostic value of PD-L1 expression in tumour epithelial cells and TICs in a series of patients with breast cancer with long-term follow-up, and associations between PD-L1 expression and histopathological type and grade, proliferation and molecular subtype. METHODS Using immunohistochemistry for PD-L1 in tissue microarrays, we assessed PD-L1 expression in 821 tumours. Expression of PD-L1 was assessed separately in the epithelial and stromal compartments and classified as <1%, ≥1% to <10% or ≥10% positive staining cells. We correlated PD-L1 expression in tumour epithelial cells and TICs with tumour characteristics using Pearson's χ2 test, and prognosis by cumulative incidence of death from breast cancer and Cox regression analyses. RESULTS We found membranous staining in ≥1% of tumour epithelial cells in 53/821 cases (6.5%). Of these, 21 (2.6%) were ≥10%. Among TICs, staining (≥1%) was seen in 144/821 cases (17.6%). Of these, 62 were ≥10% (7.6%). PD-L1 was associated with high histopathological grade and proliferation, and the medullary and metaplastic patterns. In TICs, PD-L1 ≥1% found in 22/34 (34.4%) human epidermal growth factor receptor 2 type and 29/58 (50%) basal phenotype. An independent association between PD-L1 expression and prognosis was not observed. CONCLUSIONS PD-L1 is expressed more frequently in TICs than tumour epithelial cells. Expression in TICs is associated with aggressive tumour characteristics and non-luminal tumours but not with prognosis.
Collapse
Affiliation(s)
- Sigurd A Saastad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anette H Skjervold
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Borgny Ytterhus
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Monica Jernberg Engstrøm
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Sugrery, St. Olav's Hospital Trondheim University Hospital, Trondheim, Norway
| | - Anna M Bofin
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
34
|
He Q, Xiao B, Tan Y, Wang J, Tan H, Peng C, Liang B, Cao Y, Xiao M. Integrated multicenter deep learning system for prognostic prediction in bladder cancer. NPJ Precis Oncol 2024; 8:233. [PMID: 39414931 PMCID: PMC11484793 DOI: 10.1038/s41698-024-00731-6] [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: 06/10/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024] Open
Abstract
Precise survival risk stratification is crucial for personalized therapy in bladder cancer (BCa). This study developed and validated an end-to-end deep learning system using histological slides to predict overall survival (OS) risk in BCa patients. We employed the BlaPaSeg tile classifier to generate tissue probability heatmaps and segmentation maps, trained two prognostic networks, MacroVisionNet and UniVisionNet, and explored six potential BCa prognostic biomarkers. Across all cohorts, the AUC for BlaPaSeg ranged from 0.9906 to 0.9945, while the C-index varied from 0.655 to 0.834 for MacroVisionNet and 0.661 to 0.853 for UniVisionNet. After covariate adjustment, the hazard ratio (HR) values for high-risk groups were 1.97 to 5.06 in MacroVisionNet and 2.13 to 4.01 in UniVisionNet. The high-risk Coloc (Tumor Co-localization score) and IMTS (Integrated Muscle Tumor Score) groups illustrated a higher death risk with HR values from 1.41 to 10.16. The system improves BCa survival prediction and supports refined patient management.
Collapse
Affiliation(s)
- Quanhao He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Bangxin Xiao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yiwen Tan
- Department of Pathology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Jun Wang
- Department of Nursing, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- School of Nursing, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Hao Tan
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Canjie Peng
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Bing Liang
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong Distinct, Chongqing, 400016, P. R. China.
- Department of Clinical Pathololgy Laboratory of Pathology Diagnostic Center, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong Distinct, Chongqing, 400016, P. R. China.
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong Distinct, Chongqing, 400016, P. R. China.
| | - Youde Cao
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong Distinct, Chongqing, 400016, P. R. China.
- Department of Clinical Pathololgy Laboratory of Pathology Diagnostic Center, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong Distinct, Chongqing, 400016, P. R. China.
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong Distinct, Chongqing, 400016, P. R. China.
| | - Mingzhao Xiao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China.
| |
Collapse
|
35
|
Saini KS, Somara S, Ko HC, Thatai P, Quintana A, Wallen ZD, Green MF, Mehrotra R, McGuigan S, Pang L, Das S, Yadav K, Neric D, Cantini L, Joshi C, Iwamoto K, Dubbewar S, Vidal L, Chico I, Severson E, Lorini L, Badve S, Bossi P. Biomarkers in head and neck squamous cell carcinoma: unraveling the path to precision immunotherapy. Front Oncol 2024; 14:1473706. [PMID: 39439946 PMCID: PMC11493772 DOI: 10.3389/fonc.2024.1473706] [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: 07/31/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
Recent strides in understanding the molecular underpinnings of head and neck cancers have sparked considerable interest in identifying precise biomarkers that can enhance prognostication and enable personalized treatment strategies. Immunotherapy has particularly revolutionized the therapeutic landscape for head and neck squamous cell carcinoma, offering new avenues for treatment. This review comprehensively examines the application and limitations of the established and emerging/novel biomarkers for head and neck squamous cell carcinoma. Established biomarkers, including well-characterized genetic mutations, protein expressions, and clinical factors, have been extensively studied and validated in clinical practice. Novel biomarkers identified through molecular analyses, including novel genetic alterations, immune-related markers, and molecular signatures, are currently being investigated and validated in preclinical and clinical settings. Biomarkers hold the potential to deepen our understanding of head and neck squamous cell carcinoma biology and guide therapeutic strategies. The evolving paradigm of predictive biomarkers facilitates the study of individual responses to specific treatments, including targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Kamal S. Saini
- Fortrea Inc., Durham, NC, United States
- Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | | | | | | | | | | | | | | | | | | | - Soma Das
- Fortrea Inc., Durham, NC, United States
| | - Kavita Yadav
- George Institute for Global Health, New Delhi, India
| | | | | | | | | | | | | | | | | | - Luigi Lorini
- Medical Oncology and Hematology Unit, IRCCS Humanitas Cancer Centre, Milan, Italy
| | - Sunil Badve
- Emory University, Atlanta, GA, United States
| | - Paolo Bossi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Cancer Centre, Milan, Italy
- Università degli Studi di Brescia, Brescia, Italy
| |
Collapse
|
36
|
Chen Y, Sun Z, Yin J, Ahmad MU, Zhou Z, Feng W, Yang F, Zhou K, Xie J, Bie C, Chen H, Jiang Y. Digital assessment of tertiary lymphoid structures and therapeutic responses in gastric cancer: a multicentric retrospective study. Int J Surg 2024; 110:6732-6747. [PMID: 38884256 PMCID: PMC11486929 DOI: 10.1097/js9.0000000000001834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Tertiary lymphoid structures (TLSs) are associated with favorable prognosis and enhanced response to anticancer therapy. A digital assessment of TLSs could provide an objective alternative that mitigates variability inherent in manual evaluation. This study aimed to develop and validate a digital gene panel based on biological prior knowledge for assessment of TLSs, and further investigate its associations with survival and multiple anticancer therapies. MATERIALS AND METHODS The present study involved 1704 patients with gastric cancer from seven cancer centers. TLSs were identified morphologically through hematoxylin-and-eosin staining. The authors further developed a digital score based on targeted gene expression profiling to assess TLSs status, recorded as gene signature of tertiary lymphoid structures (gsTLS). For enhanced interpretability, we employed the SHapley Additive exPlanation (SHAP) analysis to elucidate its contribution to the prediction. The authors next evaluated the signature's associations with prognosis, and investigated its predictive accuracy for multiple anticancer therapies, including adjuvant chemotherapy and immunotherapy. RESULTS The gsTLS panel with nine gene features achieved high accuracies in predicting TLSs status in the training, internal, and external validation cohorts (area under the curve, range: 0.729-0.791). In multivariable analysis, gsTLS remained an independent predictor of disease-free and overall survival (hazard ratio, range: 0.346-0.743, all P <0.05) after adjusting for other clinicopathological variables. SHAP analysis highlighted gsTLS as the strongest predictor of TLSs status compared with clinical features. Importantly, patients with high gsTLS (but not those with low gsTLS) exhibited substantial benefits from adjuvant chemotherapy ( P <0.05). Furthermore, the authors found that the objective response rate to antiprogrammed cell death protein 1 (anti-PD-1) immunotherapy was significantly higher in the high-gsTLS group (40.7%) versus the low-gsTLS group (5.6%, P =0.036), and the diagnosis was independent from Epstein-Barr virus, tumor mutation burden, and programmed cell death-ligand 1 (PD-L1) expression. CONCLUSION The gsTLS digital panel enables accurate assessment of TLSs status, and provides information regarding prognosis and responses to multiple therapies for gastric cancer.
Collapse
Affiliation(s)
- Yan Chen
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Zepang Sun
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junmei Yin
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - M. Usman Ahmad
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zixia Zhou
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Wanying Feng
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Fan Yang
- Department of Computer Science, Wake Forest University, Winston Salem
| | - Kangneng Zhou
- College of Computer Science, Nankai University, Tianjin, People’s Republic of China
| | - Jingjing Xie
- Graduate Group of Epidemiology, University of California Davis, Davis, California USA
| | - Caiqun Bie
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Hongzhuan Chen
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Yuming Jiang
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| |
Collapse
|
37
|
Prost D, Iseas S, Gatineau M, Adam J, Cavalieri S, Bergamini C, Licitra L, Raymond É. Systemic treatments in recurrent or metastatic salivary gland cancer: a systematic review. ESMO Open 2024; 9:103722. [PMID: 39368417 PMCID: PMC11490782 DOI: 10.1016/j.esmoop.2024.103722] [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: 04/29/2024] [Revised: 07/20/2024] [Accepted: 08/12/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Salivary gland cancers are infrequent and pose a challenge owing to their histological diversity and varied clinical behavior, making the selection of optimal systemic treatments for advanced or recurrent stages difficult. This systematic review aims to assess overall survival outcomes and systemic treatment responses across four types of salivary cancers. METHODS A PubMed and Google Scholar search identified studies involving initially advanced or relapsed cases undergoing systemic treatment. Studies with clear, individualized data on treatment responses and outcomes were selected based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) checklist. Of the 723 studies screened, 44 met our inclusion criteria. RESULTS A total of 426 cases of recurrent/metastatic salivary gland cancer, mostly salivary duct carcinoma (SDC; n = 219) and adenoid cyst carcinoma (ACC; n = 167), were included. Histomolecular markers were heavily associated with histology, with HER2 overexpression and androgen receptor nuclear expression typically found in SDC and adenocarcinoma not otherwise specified cases and KIT overexpression only in ACC. The response rates were associated with specific receptor blockage, with trastuzumab plus chemotherapy, and bicalutamide being the most effective (overall response rate 80% and 42.8%, respectively). Moreover, the response to treatment positively influenced overall survival (responders 38 versus non-responders 18.7 median months; P < 0.001). In this retrospective analysis of a particular cohort, survival outcomes per histology types showed that anti-human epidermal growth factor receptor 2 therapy was more effective for SDC, while chemotherapy was more effective for ACC. CONCLUSION Systemic treatments contribute to the survival of patients with salivary gland cancer at relapsed or newly advanced stages. The response to treatment is heavily influenced by histological subtype and treatment specificity.
Collapse
Affiliation(s)
- D Prost
- Oncology Department, Hôpitaux Paris, Saint-Joseph Marie-Lannelongue, Paris; Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle Epinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2 Institute de Neurologie, Paris.
| | - S Iseas
- Oncology Department, Hôpitaux Paris, Saint-Joseph Marie-Lannelongue, Paris
| | - M Gatineau
- Oncology Department, Hôpitaux Paris, Saint-Joseph Marie-Lannelongue, Paris
| | - J Adam
- Pathology Department, Hôpitaux Paris Saint-Joseph Marie-Lannelongue, Paris, France
| | - S Cavalieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori di Milano, Milan; Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - C Bergamini
- Head and Neck Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori di Milano, Milan
| | - L Licitra
- Head and Neck Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori di Milano, Milan; Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - É Raymond
- Oncology Department, Hôpitaux Paris, Saint-Joseph Marie-Lannelongue, Paris
| |
Collapse
|
38
|
Orive D, Echepare M, Bernasconi-Bisio F, Sanmamed MF, Pineda-Lucena A, de la Calle-Arroyo C, Detterbeck F, Hung RJ, Johansson M, Robbins HA, Seijo LM, Montuenga LM, Valencia K. Protein Biomarkers in Lung Cancer Screening: Technical Considerations and Feasibility Assessment. Arch Bronconeumol 2024; 60 Suppl 2:S67-S76. [PMID: 39079848 DOI: 10.1016/j.arbres.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/28/2024] [Accepted: 07/12/2024] [Indexed: 08/25/2024]
Abstract
Lung cancer remains the leading cause of cancer-related deaths worldwide, mainly due to late diagnosis and the presence of metastases. Several countries around the world have adopted nation-wide LDCT-based lung cancer screening that will benefit patients, shifting the stage at diagnosis to earlier stages with more therapeutic options. Biomarkers can help to optimize the screening process, as well as refine the TNM stratification of lung cancer patients, providing information regarding prognostics and recommending management strategies. Moreover, novel adjuvant strategies will clearly benefit from previous knowledge of the potential aggressiveness and biological traits of a given early-stage surgically resected tumor. This review focuses on proteins as promising biomarkers in the context of lung cancer screening. Despite great efforts, there are still no successful examples of biomarkers in lung cancer that have reached the clinics to be used in early detection and early management. Thus, the field of biomarkers in early lung cancer remains an evident unmet need. A more specific objective of this review is to present an up-to-date technical assessment of the potential use of protein biomarkers in early lung cancer detection and management. We provide an overview regarding the benefits, challenges, pitfalls and constraints in the development process of protein-based biomarkers. Additionally, we examine how a number of emerging protein analytical technologies may contribute to the optimization of novel robust biomarkers for screening and effective management of lung cancer.
Collapse
Affiliation(s)
- Daniel Orive
- Solid Tumors Program, CIMA-University of Navarra, Pamplona, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain; Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Mirari Echepare
- Solid Tumors Program, CIMA-University of Navarra, Pamplona, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain; Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Franco Bernasconi-Bisio
- Molecular Therapeutics Program, CIMA-University of Navarra, Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Miguel Fernández Sanmamed
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Program of Immunology and Immunotherapy, CIMA-University of Navarra, Pamplona, Spain; Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Antonio Pineda-Lucena
- Navarra Health Research Institute (IDISNA), Pamplona, Spain; Molecular Therapeutics Program, CIMA-University of Navarra, Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Carlos de la Calle-Arroyo
- Instituto de Ciencia de los Datos e Inteligencia Artificial (DATAI), Universidad de Navarra, Pamplona, Spain
| | - Frank Detterbeck
- Division of Thoracic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | | | | | - Luis M Seijo
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain; Pulmonary Department, Clínica Universidad de Navarra, Madrid, Spain
| | - Luis M Montuenga
- Solid Tumors Program, CIMA-University of Navarra, Pamplona, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain; Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Navarra Health Research Institute (IDISNA), Pamplona, Spain.
| | - Karmele Valencia
- Solid Tumors Program, CIMA-University of Navarra, Pamplona, Spain; Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Navarra Health Research Institute (IDISNA), Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.
| |
Collapse
|
39
|
Galbraith NJ, Quinn JA, Al-Badran SS, Pennel KAF, Hillson LVS, Hatthakarnkul P, McKenzie M, Maka N, Loi L, Frixou M, Steele CW, Roxburgh CS, Horgan PG, McMillan DC, Edwards J. TAK1 expression is associated with increased PD-L1 and decreased cancer-specific survival in microsatellite-stable colorectal cancer. Transl Oncol 2024; 48:102064. [PMID: 39068768 PMCID: PMC11338118 DOI: 10.1016/j.tranon.2024.102064] [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: 03/14/2024] [Revised: 05/27/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Transforming growth factor β-activated protein kinase-1 (TAK1) plays an important role in MAPK and NFκB pathways and has been associated with colorectal cancer. The aim of this study was to determine how cytoplasmic and juxtanuclear punctate staining of TAK1 relates to immune checkpoint expression and cancer specific survival in colorectal cancer. METHODS Protein expression was assessed by immunohistochemistry on tissue microarrays from primary curative colorectal cancer resected specimens. Expression levels of cytoplasmic TAK1 by QuPath digital quantification and punctate TAK1 staining was scored using a manual point scoring technique and correlated with clinicopathological features, immune checkpoint expression and cancer-specific survival. Bulk RNA sequencing was performed in specimens to determine mutational profiles and differentially expressed genes. RESULTS A cohort of 875 patients who had undergone colorectal cancer resection were assessed for TAK1 expression. Higher levels of cytoplasmic TAK1 expression correlated with elevated PD1 and PD-L1 expression (p < 0.010). High punctate TAK1 expression was more commonly identified in poorly differentiated colorectal cancers (p = 0.036), had dysregulated mutational and transcriptional profiles with decreased insulin-like growth factor 2(IGF2) expression (p < 0.010), and independently predicted poor cancer-specific survival (HR 2.690, 95% CI 1.419-5.100, p = 0.002). The association of punctate TAK1 expression and recurrence remained after subgroup analysis for microsatellite-stable colorectal cancer (p = 0.028). DISCUSSION Punctate TAK1 expression is associated with worse cancer specific survival. TAK1 signalling may be an important pathway to investigate underlying mechanisms for recurrence in microsatellite-stable colorectal cancer.
Collapse
Affiliation(s)
- Norman J Galbraith
- School of Cancer Sciences, Wolfson-Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom; Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom.
| | - Jean A Quinn
- School of Cancer Sciences, Wolfson-Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Sara Sf Al-Badran
- School of Cancer Sciences, Wolfson-Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Kathryn A F Pennel
- School of Cancer Sciences, Wolfson-Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Lily V S Hillson
- School of Cancer Sciences, Wolfson-Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Phimmada Hatthakarnkul
- School of Cancer Sciences, Wolfson-Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Molly McKenzie
- School of Cancer Sciences, Wolfson-Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Noori Maka
- Department of Pathology, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Lynette Loi
- School of Cancer Sciences, Wolfson-Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Mikaela Frixou
- School of Cancer Sciences, Wolfson-Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Colin W Steele
- School of Cancer Sciences, Wolfson-Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom; Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Campbell S Roxburgh
- School of Cancer Sciences, Wolfson-Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom; Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Paul G Horgan
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Donald C McMillan
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Joanne Edwards
- School of Cancer Sciences, Wolfson-Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
40
|
Chen H, Wu M, Chen M. Prognostic Significance of Albumin-Globulin Ratio in Urachal Carcinoma. Urol Int 2024; 109:8-17. [PMID: 39342932 DOI: 10.1159/000540665] [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: 03/23/2024] [Accepted: 07/30/2024] [Indexed: 10/01/2024]
Abstract
INTRODUCTION Although albumin-globulin ratio (AGR) has been used in the prognostic assessment of multiple solid malignancies, so far no research has confirmed the prognostic significance of AGR as a biomarker for urachal carcinoma. We analyzed the relationship between AGR and prognosis in urachal carcinoma, aiming to identify a promising prognostic biomarker for urachal carcinoma. METHODS We reviewed the clinical data of 25 patients diagnosed with urachal carcinoma in the Xiangya Hospital, Central South University, from January 2008 to October 2021. The best cut-off value of preoperative AGR was determined according to the receiver operator characteristic curve. The Kaplan-Meier curve was used to analyze the effect of preoperative AGR on the overall survival (OS) and relapse-free survival (RFS) of patients. Cox proportional hazards model was used to analyze prognostic factors including preoperative AGR. RESULTS The best cut-off value of preoperative AGR in urachal carcinoma patients is 1.45. Low preoperative AGR is significantly associated with worse OS and RFS. Univariate analysis and multivariate analysis indicated that low preoperative AGR is an independent and reliable factor to predict poor prognosis, OS, and RFS in urachal carcinoma patients. CONCLUSION Urachal carcinoma patients with low preoperative AGR have worse prognosis, and preoperative AGR is a valuable prognostic indicator for urachal carcinoma research.
Collapse
Affiliation(s)
- Hengxin Chen
- Department of Urology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China,
| | - Menghai Wu
- Department of Urology, Xiangya Hospital Central South University, Changsha, China
| | - Minfeng Chen
- Department of Urology, Xiangya Hospital Central South University, Changsha, China
| |
Collapse
|
41
|
Saleh HA, Nabil G, Badawy SAMM. Comparative immunohistochemical expression of Beta catenin and CD163 between dysplastic / non-dysplastic oral lichen planus and lichenoid lesions (EX-VIVO STUDY). BMC Oral Health 2024; 24:1122. [PMID: 39327577 PMCID: PMC11430547 DOI: 10.1186/s12903-024-04822-5] [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: 06/28/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Oral lichen planus is a well-known chronic inflammatory mucocutaneous disorder, which has clinical and histological presentation that mimics oral lichenoid reaction. According to the fifth edition of WHO, both conditions are considered as oral potentially malignant disorders. Recent studies on oral potential disorders documented deregulation of some signaling molecules related to the Wnt/β-catenin pathway. Therefore this study aimed to compare the immune expression of β-catenin & CD163 in dysplastic /non-dysplastic cases of Oral lichen planus & oral lichenoid lesion. In addition, a statistical correlation between both immune markers was done regardless of the type of the study group. METHODS Four study groups were designated as 2 groups of Oral lichen planus (one dysplastic & one non -dysplastic) and the other 2 groups were oral lichenoid lesions (one dysplastic & one non -dysplastic). Ten cases in each group were collected and investigated by immunohistochemistry. The area percent of beta catenin and also counting of m2 macrophages expressing + CD163 marker was calculated in the study groups. RESULTS The Statistical analysis highlighted a statistically significant difference between the studied groups. Moreover, Pearson correlation test reported a significant moderate positive correlation between beta catenin & CD163 expression in the studied cases. CONCLUSION Our findings supported new perceptions of the mechanism by which tumor associated macrophage specific β-catenin signaling promotes the aggressive behavior of oral potential malignant disorders. CLINICAL RELEVANCE Evidence of the relationship between beta catenin and M2 macrophages (+ CD163) may enhance the development of macrophage-based strategies for treatment and improve the prognosis of such cases.
Collapse
Affiliation(s)
- Heba Ahmed Saleh
- Oral & Maxillofacial Pathology, Faculty of Dentistry, Cairo University, Cairo, Egypt.
- , 11 el Saraya Street, Manyal, Cairo, Egypt.
| | - Ghada Nabil
- Oral Medicine, Faculty of Dentistry, Cairo University, Cairo, Egypt
| | | |
Collapse
|
42
|
Cho D, Lord SJ, Ward R, IJzerman M, Mitchell A, Thomas DM, Cheyne S, Martin A, Morton RL, Simes J, Lee CK. Criteria for assessing evidence for biomarker-targeted therapies in rare cancers-an extrapolation framework. Ther Adv Med Oncol 2024; 16:17588359241273062. [PMID: 39229469 PMCID: PMC11369883 DOI: 10.1177/17588359241273062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/09/2024] [Indexed: 09/05/2024] Open
Abstract
Background Advances in targeted therapy development and tumor sequencing technology are reclassifying cancers into smaller biomarker-defined diseases. Randomized controlled trials (RCTs) are often impractical in rare diseases, leading to calls for single-arm studies to be sufficient to inform clinical practice based on a strong biological rationale. However, without RCTs, favorable outcomes are often attributed to therapy but may be due to a more indolent disease course or other biases. When the clinical benefit of targeted therapy in a common cancer is established in RCTs, this benefit may extend to rarer cancers sharing the same biomarker. However, careful consideration of the appropriateness of extending the existing trial evidence beyond specific cancer types is required. A framework for extrapolating evidence for biomarker-targeted therapies to rare cancers is needed to support transparent decision-making. Objectives To construct a framework outlining the breadth of criteria essential for extrapolating evidence for a biomarker-targeted therapy generated from RCTs in common cancers to different rare cancers sharing the same biomarker. Design A series of questions articulating essential criteria for extrapolation. Methods The framework was developed from the core topics for extrapolation identified from a previous scoping review of methodological guidance. Principles for extrapolation outlined in guidance documents from the European Medicines Agency, the US Food and Drug Administration, and Australia's Medical Services Advisory Committee were incorporated. Results We propose a framework for assessing key assumptions of similarity of the disease and treatment outcomes between the common and rare cancer for five essential components: prognosis of the biomarker-defined cancer, biomarker test analytical validity, biomarker actionability, treatment efficacy, and safety. Knowledge gaps identified can be used to prioritize future studies. Conclusion This framework will allow systematic assessment, standardize regulatory, reimbursement and clinical decision-making, and facilitate transparent discussions between key stakeholders in drug assessment for rare biomarker-defined cancers.
Collapse
Affiliation(s)
- Doah Cho
- National Health and Medical Research Council Clinical Trials Centre, Faculty of Medicine and Health, University of Sydney, Australia
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Locked Bag 77, Camperdown, NSW 1450, Australia
| | - Sarah J. Lord
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Robyn Ward
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Maarten IJzerman
- Faculty of Medicine, Dentistry and Health Sciences, Centre for Health Policy, University of Melbourne Centre for Cancer Research, Parkville, VIC, Australia
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Andrew Mitchell
- Department of Health Economics Wellbeing and Society, The Australian National University, Canberra, ACT, Australia
| | - David M. Thomas
- Centre for Molecular Oncology, University of New South Wales, Sydney, NSW, Australia
| | - Saskia Cheyne
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Andrew Martin
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- Centre for Clinical Research, University of Queensland, St Lucia, QLD, Australia
| | - Rachael L. Morton
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - John Simes
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Chee Khoon Lee
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
43
|
Alshamsan B, Elshenawy MA, Aseafan M, Fahmy N, Badran A, Elhassan T, Alsayed A, Suleman K, Al-Tweigeri T. Prognostic significance of the neutrophil to lymphocyte ratio in locally advanced breast cancer. Oncol Lett 2024; 28:429. [PMID: 39049989 PMCID: PMC11268088 DOI: 10.3892/ol.2024.14562] [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: 01/12/2024] [Accepted: 05/28/2024] [Indexed: 07/27/2024] Open
Abstract
The present study aimed to clarify the prognostic role of the pre-treatment neutrophil-to-lymphocyte ratio (NLR) for the response to neoadjuvant chemotherapy (NAC) in locally advanced breast cancer (LABC). Due to conflicting results in currently available data, the specific focus of the present study was on evaluating the associations between the pre-treatment NLR and the rate of achieving a pathological complete response (pCR) and survival outcomes. For the present study, data from a cohort of 465 consecutive patients with LABC who underwent NAC at King Feisal Specialist Hospital and Research Center (Riyadh, Saudi Arabia) between 2005 and 2014 were obtained from a prospective BC database and analyzed. Patients were stratified into two groups based on an optimal NLR cut-off determined using the receiver operating characteristic curve. Logistic regression analyses were conducted to assess variables associated with pCR, and Cox regression analyses were used to assess variables associated with survival outcomes. The low pre-treatment NLR group (≤2.2) was found to exhibit a higher likelihood of achieving a pCR (odds ratio, 2.59; 95% CI, 1.52-4.38; P<0.001), along with higher 5-year disease-free survival (DFS) [75.8 vs. 64.9%; hazard ratio (HR), 0.69; 95% CI, 0.50-0.94; P=0.02] and 5-year overall survival (OS; 90.3 vs. 81.9; HR, 0.62; 95% CI, 0.39-0.98; P=0.04) rates compared with those in the high NLR group (>2.2). Sub-group analysis revealed that the observed significance in survival outcomes was driven by the triple-negative BC (TNBC) subgroup. Patients with residual TNBC disease and a high pre-treatment NLR were observed to have lower 5-year DFS (44.4 vs. 75.0%; P=0.02) and 5-year OS (55.9 vs. 84.5%; P=0.055) rates compared with those with residual TNBC disease and a low NLR. To conclude, data from the present study suggest that the pre-treatment NLR can serve as a viable independent prognostic factor for pCR following NAC in patients with LABC and for survival outcomes, particularly for patients with TNBC.
Collapse
Affiliation(s)
- Bader Alshamsan
- Department of Medicine, College of Medicine, Qassim University, Buraydah, Qassim 52571, Kingdom of Saudi Arabia
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
| | - Mahmoud A. Elshenawy
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
- Department of Clinical Oncology, Faculty of Medicine, Menoufia University, Shebeen El-Kom 32511, Egypt
| | - Mohamed Aseafan
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
- Department of Internal Medicine, Section of Oncology, Security Forces Hospital, Riyadh 11481, Kingdom of Saudi Arabia
| | - Nermin Fahmy
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
- Department of Clinical Oncology and Nuclear Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ahmed Badran
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
- Department of Clinical Oncology, Ain Shams University Hospitals, Ain Shams 11517, Egypt
| | - Tusneem Elhassan
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
| | - Adher Alsayed
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
| | - Kausar Suleman
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
| | - Taher Al-Tweigeri
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
| |
Collapse
|
44
|
Cyll K, Skaaheim Haug E, Pradhan M, Vlatkovic L, Carlsen B, Löffeler S, Kildal W, Skogstad K, Hauge Torkelsen F, Syvertsen RA, Askautrud HA, Liestøl K, Kleppe A, Danielsen HE. DNA ploidy and PTEN as biomarkers for predicting aggressive disease in prostate cancer patients under active surveillance. Br J Cancer 2024; 131:895-904. [PMID: 38961192 PMCID: PMC11368925 DOI: 10.1038/s41416-024-02780-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: 10/20/2023] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Current risk stratification tools for prostate cancer patients under active surveillance (AS) may inadequately identify those needing treatment. We investigated DNA ploidy and PTEN as potential biomarkers to predict aggressive disease in AS patients. METHODS We assessed DNA ploidy by image cytometry and PTEN protein expression by immunohistochemistry in 3197 tumour-containing tissue blocks from 558 patients followed in AS at a Norwegian local hospital. The primary endpoint was treatment, with treatment failure (biochemical recurrence or initiation of salvage therapy) as the secondary endpoint. RESULTS The combined DNA ploidy and PTEN (DPP) status at diagnosis was associated with treatment-free survival in univariable- and multivariable analysis, with a HR for DPP-aberrant vs. DPP-normal tumours of 2.12 (p < 0.0001) and 1.94 (p < 0.0001), respectively. Integration of DNA ploidy and PTEN status with the Cancer of the Prostate Risk Assessment (CAPRA) score improved risk stratification (c-index difference = 0.025; p = 0.0033). Among the treated patients, those with DPP-aberrant tumours exhibited a significantly higher likelihood of treatment failure (HR 2.01; p = 0.027). CONCLUSIONS DNA ploidy and PTEN could serve as additional biomarkers to identify AS patients at increased risk of developing aggressive disease, enabling earlier intervention for nearly 50% of the patients that will eventually receive treatment with current protocol.
Collapse
Affiliation(s)
- Karolina Cyll
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway.
- Department of Urology, Vestfold Hospital Trust, 3103, Tønsberg, Norway.
| | - Erik Skaaheim Haug
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
- Department of Urology, Vestfold Hospital Trust, 3103, Tønsberg, Norway
| | - Manohar Pradhan
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
| | - Ljiljana Vlatkovic
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
| | - Birgitte Carlsen
- Department of Pathology, Vestfold Hospital Trust, 3103, Tønsberg, Norway
| | - Sven Löffeler
- Department of Urology, Vestfold Hospital Trust, 3103, Tønsberg, Norway
| | - Wanja Kildal
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
| | - Karin Skogstad
- Department of Urology, Vestfold Hospital Trust, 3103, Tønsberg, Norway
| | - Frida Hauge Torkelsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
| | - Rolf Anders Syvertsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
| | - Hanne A Askautrud
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
| | - Knut Liestøl
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
- Department of Informatics, University of Oslo, 0316, Oslo, Norway
| | - Andreas Kleppe
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
- Department of Informatics, University of Oslo, 0316, Oslo, Norway
- Centre for Research-based Innovation Visual Intelligence, UiT The Arctic University of Norway, Tromsø, Norway
| | - Håvard E Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, OX3 9DU, UK
| |
Collapse
|
45
|
Bertram CA, Donovan TA, Bartel A. Mitotic activity: A systematic literature review of the assessment methodology and prognostic value in feline tumors. Vet Pathol 2024; 61:743-751. [PMID: 38533803 PMCID: PMC11370206 DOI: 10.1177/03009858241239566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Increased proliferation is a driver of tumorigenesis, and quantification of mitotic activity is a standard task for prognostication. This systematic review is an analysis of all available references on mitotic activity in feline tumors to provide an overview of the assessment methods and prognostic value. A systematic literature search in PubMed and Scopus and a nonsystematic search in Google Scholar were conducted. All articles on feline tumors that correlated mitotic activity with patient outcome were identified. Data analysis revealed that of the 42 eligible articles, mitotic count (MC, mitotic figures/tumor area) was evaluated in 39 studies, and mitotic index (MI, mitotic figures/tumor cells) in 3 studies. The risk of bias was considered high for most studies (26/42, 62%) based on small study populations, insufficient details of the MC/MI methods, and lack of statistical measures for diagnostic accuracy or effect on outcome. The MC/MI methods varied between studies. A significant association of MC with survival was determined in 20 of 28 (71%) studies (10 studies evaluated other outcome metrics or provided individual patient data), while 1 study found an inverse effect. Three tumor types had at least 4 studies, and a prognostic association with survival was found in 5 of 6 studies on mast cell tumors, 5 of 5 on mammary tumors, and 3 of 4 on soft-tissue sarcomas. MI was shown to correlate with survival for mammary tumors by 2 research groups; however, comparisons to MC were not conducted. Further studies with standardized mitotic activity methods and appropriate statistical analysis for discriminant ability of patient outcome are needed to infer the prognostic value of MC and MI.
Collapse
|
46
|
Thomas C, Faghy MA, Chidley C, Phillips BE, Bewick T, Ashton RE. Blood Biomarkers of Long COVID: A Systematic Review. Mol Diagn Ther 2024; 28:537-574. [PMID: 39103645 DOI: 10.1007/s40291-024-00731-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Long coronavirus disease (COVID; LC) affects millions of people worldwide. The exact mechanisms which result in a broad, undulating and detrimental symptom profile remain unknown. Blood biomarkers associated with LC have been described; however, consensus on these remains elusive, in part due to a lack of continuity between studies on a universally accepted definition of LC. This systematic review aimed to consolidate current knowledge of blood biomarkers associated with the prevalence of LC on the basis of the World Health Organisation (WHO) clinical definition of this condition. ELIGIBILITY CRITERIA FOR SELECTING STUDIES Observational, cross-sectional, and randomised control studies published in the English language that studied blood biomarkers associated with the WHO definition of LC. All studies included participants who were ≥ 18 years old and group sizes ≥ 10 participants, and were compared against a control group without any known co-morbidities. METHODS A systematic literature search was conducted according to Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines and prospectively registered on Prospero (ID: CRD42022373121). The Cochrane, Embase, PubMed and Web of Science databases were searched from inception to January 2024. Search results were gathered using Rayyan software and data extracted using Microsoft Excel. The reporting recommendations for tumour markers prognostic studies (REMARK) questionnaire was used to assess the quality of the included studies. RESULTS A total of 45 observational and one interventional study comprising 4415 participants were included in this review which identified 525 blood biomarkers thought to be associated with LC. Three blood biomarker subtypes were associated with the development of LC: (1) immunological and inflammatory dysfunction, (2) endothelial/vascular dysfunction and (3) metabolic and clotting abnormalities. DISCUSSION AND CONCLUSIONS Our data are consistent with previous findings; however, no single biomarker was sufficiently associated with LC prevalence and instead a profile of biomarkers across various physiological systems may be more clinically useful. In all, 196 studies were excluded due to a lack of an adequately healthy comparator group and/or failure to meet the WHO LC definition. This demonstrates a need for further research incorporating a universal LC definition across all disease severity groups and symptom profiles, and longitudinal data reflecting the relapsing and remitting nature of this condition. Further investigation into blood biomarkers of LC, including clear reporting of healthy comparator groups and the investigation of acute and chronic biomarker changes, within the context of medical practice, may support the development of curative/restorative approaches.
Collapse
Affiliation(s)
- Callum Thomas
- Biomedical and Clinical Science Research Theme, School of Human Sciences, University of Derby, Derby, UK.
- Healthy Living for Pandemic Event Protection (HL-PIVOT) Network, Chicago, IL, USA.
| | - Mark A Faghy
- Biomedical and Clinical Science Research Theme, School of Human Sciences, University of Derby, Derby, UK
- Healthy Living for Pandemic Event Protection (HL-PIVOT) Network, Chicago, IL, USA
- Department of Physical Therapy, College of Applied Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Corinna Chidley
- Biomedical and Clinical Science Research Theme, School of Human Sciences, University of Derby, Derby, UK
| | - Bethan E Phillips
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and NIHR Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Thomas Bewick
- Department of Respiratory Medicine, University Hospitals of Derby and Burton NHS Foundation Trust, Derby, UK
| | - Ruth E Ashton
- Biomedical and Clinical Science Research Theme, School of Human Sciences, University of Derby, Derby, UK
- Healthy Living for Pandemic Event Protection (HL-PIVOT) Network, Chicago, IL, USA
- Research Centre for Physical Activity, Sport and Exercise Sciences, Coventry University, Coventry, UK
| |
Collapse
|
47
|
Ivanova D, Fakis G, Boukouvala S. Differential expression of NAT1 pharmacogene in hormone receptor positive vs. negative female breast tumors may affect drug treatment. Pharmacogenet Genomics 2024; 34:246-251. [PMID: 38842463 DOI: 10.1097/fpc.0000000000000540] [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: 06/07/2024]
Abstract
Studies have reported overexpression of NAT1 gene for xenobiotic metabolizing arylamine N -acetyltransferase type 1 in estrogen receptor positive breast tumors, and this association has been linked to patient chemoresistance and response to tamoxifen. We probed the expression of NAT1 , using quantitative reverse transcription PCR to screen clinically characterized breast cancer tissue cDNA arrays. Primers detecting all NAT1 alternative transcripts were used, and the protocol and results are reported according to consensus guidelines. The clinical information about 166 tumor samples screened is provided, including tumor stage, estrogen and progesterone receptor status and HER2 expression. NAT1 was found to be significantly ( P < 0.001) upregulated in hormone receptor positive vs. negative tumors. No correlation was apparent between NAT1 and tumor stage or HER2 expression. Our findings demonstrate a strong correlation between the expression of NAT1 and steroid hormone receptors in breast tumors, supporting its possible utility as a pharmacogenetic biomarker or drug target. Of the two polymorphic NAT genes, NAT1 is the one primarily expressed in breast tissue, and is subjected to regulation by two differential promoters and more than one polyadenylation signal. Hormonal factors may enhance NAT1 gene expression at the transcriptional or epigenetic level, and tamoxifen has additionally been shown to inhibit NAT1 enzymatic activity. The outcome of tamoxifen treatment is also more favorable in patients with NAT1 overexpressing tumors. The study adds to the growing body of evidence implicating NAT1 in breast cancer and its pharmacological treatment.
Collapse
Affiliation(s)
- Desislava Ivanova
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | | | | |
Collapse
|
48
|
Hsu C, Wen Y, Wang H, Hsieh C, Liao C, Lee L, Ng S, Lin C, Chen W, Lin J, Tsai Y, Lee S, Chien C, Hua C, Wang CP, Chen T, Terng S, Tsai C, Fan K, Yeh C, Lin C, Tsao C, Cheng N, Fang T, Huang S, Kang C, Lee L, Fang K, Wang Y, Lin W, Hsin L, Yen T, Liao C. Prognostic impact of bridge or neoadjuvant induction chemotherapy in patients with resected oral cavity cancer: A nationwide cohort study. Cancer Med 2024; 13:e70061. [PMID: 39101462 PMCID: PMC11299076 DOI: 10.1002/cam4.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/18/2024] [Accepted: 07/20/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND While surgery remains the primary treatment for oral squamous cell carcinoma (OCSCC), induction chemotherapy (IC) can be used as a bridging or neoadjuvant therapy. This nationwide study in Taiwan examines the survival outcomes of OCSCC patients who received IC before surgery. METHODS We analyzed data from 29,891 patients with OCSCC. Of these, 29,058 initially underwent surgery (OP group), whereas 833 received IC before surgery (IC + OP group). A propensity score (PS)-matched analysis (4, 1 ratio, 3260 vs. 815 patients) was performed considering tumor subsite, sex, age, Charlson comorbidity index, clinical T1-T4b tumors, clinical N0-3 disease, and clinical stage I-IV. RESULTS In the PS-matched cohort, the 5-year disease-specific survival (DSS) and overall survival (OS) rates were 65% and 57%, respectively. When comparing the OP and IC + OP groups, the 5-year DSS rates were 66% and 62%, respectively (p = 0.1162). Additionally, the 5-year OS rates were 57% and 56%, respectively (p = 0.9917). No significant intergroup differences in survival were observed for specific subgroups with cT4a tumors, cT4b tumors, cN3 disease, pT4b tumors, and pN3 disease. However, for patients with pT4a tumors, the OP group demonstrated superior 5-year outcomes compared to the IC + OP group, with a DSS of 62% versus 52% (p = 0.0006) and an OS of 53% versus 44% (p = 0.0060). Notably, patients with cT2-3, cN1, and c-Stage II disease in the IC + OP group were significantly more likely to achieve pT0-1 status (p < 0.05). CONCLUSIONS Following PS matching, the IC + OP group generally exhibited similar prognosis to the OP group. However, for pT4a tumors, the OP group showed superior 5-year outcomes. While IC may not universally improve survival, it could be advantageous for patients who respond positively to the treatment.
Collapse
Affiliation(s)
- Cheng‐Lung Hsu
- Department of Medical OncologyChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Yu‐Wen Wen
- Clinical Informatics and Medical Statistics Research CenterChang Gung UniversityTaoyuanTaiwan
- Division of Thoracic SurgeryChang Gung Memorial HospitalTaoyuanTaiwan
| | - Hung‐Ming Wang
- Department of Medical OncologyChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Chia‐Hsun Hsieh
- Department of Medical OncologyChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Chi‐Ting Liao
- Department of Medical OncologyChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Li‐Yu Lee
- Department of PathologyChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Shu‐Hang Ng
- Department of Diagnostic RadiologyChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Chien‐Yu Lin
- Department of Radiation OncologyChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Wen‐Cheng Chen
- Department of Radiation OncologyChang Gung Memorial HospitalChiayiTaiwan
| | - Jin‐Ching Lin
- Department of Radiation OncologyChanghua Christian HospitalChanghuaTaiwan
| | - Yao‐Te Tsai
- Department of Otorhinolaryngology‐Head and Neck SurgeryChang Gung Memorial HospitalChiayiTaiwan
| | - Shu‐Ru Lee
- Research Service Center for Health InformationChang Gung UniversityTaoyuanTaiwan
| | - Chih‐Yen Chien
- Department of Otolaryngology, Chang Gung Memorial Hospital Kaohsiung Medical CenterChang Gung University College of MedicineKaohsiungTaiwan
| | - Chun‐Hung Hua
- Department of OtorhinolaryngologyChina Medical University HospitalTaichungTaiwan
| | - Cheng Ping Wang
- Department of OtolaryngologyNational Taiwan University Hospital and College of MedicineTaipeiTaiwan
| | - Tsung‐Ming Chen
- Department of OtolaryngologyShuang Ho Hospital, Taipei Medical UniversityNew Taipei CityTaiwan
| | - Shyuang‐Der Terng
- Department of Head and Neck SurgeryKoo Foundation Sun Yat‐Sen Cancer CenterTaipeiTaiwan
| | - Chi‐Ying Tsai
- Department of Oral and Maxillofacial SurgeryChang Gung Memorial Hospital, Chang Gung UniversityTaoyuanTaiwan
| | - Kang‐Hsing Fan
- Department of Radiation OncologyNew Taipei Municipal TuCheng HospitalNew Taipei CityTaiwan
| | - Chih‐Hua Yeh
- Department of Diagnostic RadiologyChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Chih‐Hung Lin
- Department of Plastic and Reconstructive SurgeryChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Chung‐Kan Tsao
- Department of Plastic and Reconstructive SurgeryChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Nai‐Ming Cheng
- Department of Nuclear Medicine and Molecular Imaging CenterChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Tuan‐Jen Fang
- Department of Otorhinolaryngology, Head and Neck SurgeryChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Shiang‐Fu Huang
- Department of Otorhinolaryngology, Head and Neck SurgeryChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Chung‐Jan Kang
- Department of Otorhinolaryngology, Head and Neck SurgeryChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Li‐Ang Lee
- Department of Otorhinolaryngology, Head and Neck SurgeryChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Ku‐Hao Fang
- Department of Otorhinolaryngology, Head and Neck SurgeryChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Yu‐Chien Wang
- Department of Otorhinolaryngology, Head and Neck SurgeryChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Wan‐Ni Lin
- Department of Otorhinolaryngology, Head and Neck SurgeryChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Li‐Jen Hsin
- Department of Otorhinolaryngology, Head and Neck SurgeryChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Tzu‐Chen Yen
- Department of Nuclear Medicine and Molecular Imaging CenterChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| | - Chun‐Ta Liao
- Department of Otorhinolaryngology, Head and Neck SurgeryChang Gung Memorial Hospital and Chang Gung UniversityTaoyuanTaiwan
| |
Collapse
|
49
|
Li H, Aggarwal A, Toro P, Fu P, Badve SS, Cuzick J, Madabhushi A, Thorat MA. A prognostic and predictive computational pathology immune signature for ductal carcinoma in situ: retrospective results from a cohort within the UK/ANZ DCIS trial. Lancet Digit Health 2024; 6:e562-e569. [PMID: 38987116 DOI: 10.1016/s2589-7500(24)00116-x] [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: 01/14/2024] [Revised: 04/21/2024] [Accepted: 05/23/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND The density of tumour-infiltrating lymphocytes (TILs) could be prognostic in ductal carcinoma in situ (DCIS). However, manual TIL quantification is time-consuming and suffers from interobserver and intraobserver variability. In this study, we developed a TIL-based computational pathology biomarker and evaluated its association with the risk of recurrence and benefit of adjuvant treatment in a clinical trial cohort. METHODS In this retrospective cohort study, a computational pathology pipeline was developed to generate a TIL-based biomarker (CPath TIL categories). Subsequently, the signature underwent a masked independent validation on H&E-stained whole-section images of 755 patients with DCIS from the UK/ANZ DCIS randomised controlled trial. Specifically, continuous biomarker CPath TIL score was calculated as the average TIL density in the DCIS microenvironment and dichotomised into binary biomarker CPath TIL categories (CPath TIL-high vs CPath TIL-low) using the median value as a cutoff. The primary outcome was ipsilateral breast event (IBE; either recurrence of DCIS [DCIS-IBE] or invasive progression [I-IBE]). The Cox proportional hazards model was used to estimate the hazard ratio (HR). FINDINGS CPath TIL-score was evaluable in 718 (95%) of 755 patients (151 IBEs). Patients with CPath TIL-high DCIS had a greater risk of IBE than those with CPath TIL-low DCIS (HR 2·10 [95% CI 1·39-3·18]; p=0·0004). The risk of I-IBE was greater in patients with CPath TIL-high DCIS than those with CPath TIL-low DCIS (3·09 [1·56-6·14]; p=0·0013), and the risk of DCIS-IBE was non-significantly higher in those with CPath TIL-high DCIS (1·61 [0·95-2·72]; p=0·077). A significant interaction (pinteraction=0·025) between CPath TIL categories and radiotherapy was observed with a greater magnitude of radiotherapy benefit in preventing IBE in CPath TIL-high DCIS (0·32 [0·19-0·54]) than CPath TIL-low DCIS (0·40 [0·20-0·81]). INTERPRETATION High TIL density is associated with higher recurrence risk-particularly of invasive recurrence-and greater radiotherapy benefit in patients with DCIS. Our TIL-based computational pathology signature has a prognostic and predictive role in DCIS. FUNDING National Cancer Institute under award number U01CA269181, Cancer Research UK (C569/A12061; C569/A16891), and the Breast Cancer Research Foundation, New York (NY, USA).
Collapse
Affiliation(s)
- Haojia Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Arpit Aggarwal
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Paula Toro
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Sunil S Badve
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA; Winship Cancer Institute, Atlanta, GA, USA
| | - Jack Cuzick
- Centre for Cancer Screening, Prevention and Early Diagnosis, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Anant Madabhushi
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA; Joseph Maxwell Cleland Atlanta VA Medical Center, Atlanta, GA, USA.
| | - Mangesh A Thorat
- Centre for Cancer Screening, Prevention and Early Diagnosis, Wolfson Institute of Population Health, Queen Mary University of London, London, UK; School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK; Breast Surgery, Homerton University Hospital, London, UK; Breast Surgery, Guy's Hospital, Great Maze Pond, London, UK.
| |
Collapse
|
50
|
do Nascimento RG, da Conceição MPF, de Bastos DR, de Toledo Osorio CAB, López RVM, Reis EM, Cerqueira OLD. Prognostic value of Maspin protein level in patients with triple negative breast cancer. Sci Rep 2024; 14:15982. [PMID: 38987610 PMCID: PMC11237076 DOI: 10.1038/s41598-024-53870-y] [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: 10/13/2022] [Accepted: 02/06/2024] [Indexed: 07/12/2024] Open
Abstract
The search for prognostic markers in breast cancer has bumped into a typical feature of these tumors, intra and intertumoral heterogeneity. Changes in the expression profile, localization of these proteins or shedding to the surrounding stroma can be useful in the search for new markers. In this context, classification by molecular subtypes can bring perspectives for both diagnosis and screening for appropriate treatments. However, the Triple Negative (TN) subtype, which is already the one with the worst prognosis, lacks appropriate and consistent molecular markers. In this work, we analyzed 346 human breast cancer samples in tissue microarrays (TMA) from cases diagnosed with invasive breast carcinoma to assess the expression and localization pattern of Maspin and their correlation with clinical parameters. To complement our findings, we also used TCGA data to analyze the mRNA levels of these respective genes. Our data suggests that the TN subtype demonstrates a higher level of cytoplasmic Maspin compared to the other subtypes. Maspin transcript levels follow the same trend. However, TN patients with lower Maspin expression tend to have worse overall survival and free-survival metastasis rates. Finally, we used Maspin expression data to verify possible relationships with the clinicopathological information of our cohort. Our univariate analyses indicate that Maspin is related to the expression of estrogen receptor (ER) and progesterone receptor (PR). Furthermore, Maspin expression levels also showed correlation with Scarff-Bloom-Richardson (SBR) parameter, and stromal Maspin showed a relationship with lymph node involvement. Our data is not consistently robust enough to categorize Maspin as a prognostic marker. However, it does indicate a change in the expression profile within the TN subtype.
Collapse
Affiliation(s)
- Renan Gomes do Nascimento
- Center for Translational Research in Oncology, Cancer Institute of the State of São Paulo (ICESP), Clinical Hospital Faculty of Medicine, University of São Paulo (HCFMUSP), São Paulo, SP, 01246-000, Brazil
- Department of Clinical Pharmacy and Oncology, Hospital São Camilo (HSC), São Paulo, SP, 02401-300, Brazil
| | - Mércia Patrícia Ferreira da Conceição
- Center for Translational Research in Oncology, Cancer Institute of the State of São Paulo (ICESP), Clinical Hospital Faculty of Medicine, University of São Paulo (HCFMUSP), São Paulo, SP, 01246-000, Brazil
| | - Daniel Rodrigues de Bastos
- Center for Translational Research in Oncology, Cancer Institute of the State of São Paulo (ICESP), Clinical Hospital Faculty of Medicine, University of São Paulo (HCFMUSP), São Paulo, SP, 01246-000, Brazil
| | | | - Rossana Verónica Mendoza López
- Center for Translational Research in Oncology, Cancer Institute of the State of São Paulo (ICESP), Clinical Hospital Faculty of Medicine, University of São Paulo (HCFMUSP), São Paulo, SP, 01246-000, Brazil
| | - Eduardo Moraes Reis
- Departmento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Otto Luiz Dutra Cerqueira
- Center for Translational Research in Oncology, Cancer Institute of the State of São Paulo (ICESP), Clinical Hospital Faculty of Medicine, University of São Paulo (HCFMUSP), São Paulo, SP, 01246-000, Brazil.
- Departmento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil.
| |
Collapse
|