1
|
Kong XL, Lu XW, Dong SQ, Liu J, Zheng L, Chen LL, An ZH, Gao LM, Cao JL. Descending duodenal adenocarcinoma treated with pembrolizumab resulting in complete clinical response: A case report and literature review. World J Gastrointest Oncol 2025; 17:107568. [DOI: 10.4251/wjgo.v17.i6.107568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/16/2025] [Accepted: 05/21/2025] [Indexed: 06/13/2025] Open
Abstract
BACKGROUND Descending duodenal adenocarcinoma (DDA) is a rare malignancy of the digestive system, typically characterized by microsatellite instability-high (MSI-H). Pembrolizumab is a monoclonal antibody that has been approved for the treatment of MSI-H solid tumors in China.
CASE SUMMARY We present the case of a 55-year-old female patient diagnosed with DDA. Biopsy findings indicated MSI-H status with high expression of programmed cell death-ligand 1 (PD-L1). The patient was unable to undergo immediate surgery due to multiple metastatic lymph nodes in the retroperitoneum. After one cycle of the SOX (S-1 + oxaliplatin) chemotherapy regimen, the patient’s performance status significantly declined, and she experienced active gastrointestinal bleeding. Following active communication with the patient's family, pembrolizumab treatment was initiated. After two cycles of treatment, the disease was assessed as a partial response. A positron emission tomography/computed tomography scan performed after two years of treatment indicated a clinical complete response (CCR). The patient maintained this CCR for four years. She has now discontinued pembrolizumab for over one year, and no disease recurrence has been observed during re-examination.
CONCLUSION Patients with MSI-H DDA exhibiting high PD-L1 expression who are treated with pembrolizumab can achieve sustained CCR.
Collapse
Affiliation(s)
- Xiang-Lei Kong
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao 066000, Hebei Province, China
| | - Xi-Wei Lu
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao 066000, Hebei Province, China
| | - Si-Qi Dong
- Department of Orthopedics, The First Hospital of Qinhuangdao, Qinhuangdao 066000, Hebei Province, China
| | - Jiao Liu
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao 066000, Hebei Province, China
| | - Lei Zheng
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao 066000, Hebei Province, China
| | - Lan-Lan Chen
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao 066000, Hebei Province, China
| | - Zi-Hui An
- Department of Oncology, The First Hospital of Qinhuangdao, The Affiliated Qinhuangdao First Hospital of Hebei Medical University, Qinhuangdao 066000, Hebei Province, China
| | - Li-Ming Gao
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao 066000, Hebei Province, China
| | - Jun-Li Cao
- Department of Oncology, The First Hospital of Qinhuangdao, The Affiliated Qinhuangdao First Hospital of Hebei Medical University, Qinhuangdao 066000, Hebei Province, China
| |
Collapse
|
2
|
Gao Y, Peng Y, Zhou Y, Zhu J, Fu S, Chen Y, Cai C, Han Y, Shen H, Zeng S, Mao L, Xiao Z. Mitochondrial gene SLC25A24 regulated anti-tumor immunity and inhibited the proliferation and metastasis of colorectal cancer by PKG1-dependent cGMP/PKG1 pathway. Int Immunopharmacol 2025; 157:114664. [PMID: 40334626 DOI: 10.1016/j.intimp.2025.114664] [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: 01/29/2025] [Revised: 03/30/2025] [Accepted: 04/09/2025] [Indexed: 05/09/2025]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality globally, with metastasis playing a key role in its unfavorable prognosis. Emerging research has emphasized the pivotal role of mitochondria in tumor immune regulation. Nevertheless, the clinical relevance and functional role of SLC25A24, a mitochondrial solute carrier, in CRC remain largely unexplored. Through bioinformatics analyses and validation in clinical cohorts, this study identifies SLC25A24 as an independent prognostic marker in CRC, significantly linked to immune infiltration in CRC tissues. Our findings demonstrated that SLC25A24 expression is markedly reduced in CRC cell lines and tissues. Kaplan-Meier survival analysis revealed that lower SLC25A24 expression is associated with worse overall survival and progression-free survival in CRC patients. Interestingly, SLC25A24 expression was higher in microsatellite instability (MSI) CRC, which shows greater responsiveness to immune checkpoint inhibitors (ICIs). Functional experiments indicated that SLC25A24 overexpression suppresses CRC cell proliferation, migration, and invasion. Mechanistic studies revealed that SLC25A24 positively regulates the cGMP/PKG1 signaling pathway in CRC, influencing mitochondrial potential, apoptosis, and proliferation-related markers. This research highlights the SLC25A24-PKG1 axis as a potential therapeutic target to bolster anti-tumor immunity and curb CRC progression.
Collapse
Affiliation(s)
- Yan Gao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Yinghui Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yulai Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Jiang Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shenao Fu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Lei Mao
- Department of Oncology, Yueyang People's Hospital, Yueyang Hospital Afliated to Hunan Normal University, Yueyang 414000, Hunan, China.
| | - Zemin Xiao
- Department of Oncology, Changde Hospital, Xiangya School of Medicine, Central South University 818 Renmin Rd, Wuling District, Changde, Hunan 415000, China.
| |
Collapse
|
3
|
Cannarozzi AL, Biscaglia G, Parente P, Latiano TP, Gentile A, Ciardiello D, Massimino L, Di Brina ALP, Guerra M, Tavano F, Ungaro F, Bossa F, Perri F, Latiano A, Palmieri O. Artificial intelligence and whole slide imaging, a new tool for the microsatellite instability prediction in colorectal cancer: Friend or foe? Crit Rev Oncol Hematol 2025; 210:104694. [PMID: 40064251 DOI: 10.1016/j.critrevonc.2025.104694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common and second most deadly cancer worldwide. Despite advances in screening and treatment, CRC is heterogeneous and the response to therapy varies significantly, limiting personalized treatment options. Certain molecular biomarkers, including microsatellite instability (MSI), are critical in planning personalized treatment, although only a subset of patients may benefit. Currently, the primary methods for assessing MSI status include immunohistochemistry (IHC) for DNA mismatch repair proteins (MMRs), polymerase chain reaction (PCR)-based molecular testing, or next-generation sequencing (NGS). However, these techniques have limitations, are expensive and time-consuming, and often result in inter-method inconsistencies. Deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H) are critical predictive biomarkers of response to immune checkpoint inhibitor (ICI) therapy and MSI testing is recommended to identify patients who may benefit. There is a pressing need for a more robust, reliable, and cost-effective approach that accurately assesses MSI status. Recent advances in computational pathology, in particular the development of technologies that digitally scan whole slide images (WSI) at high resolution, as well as new approaches to artificial intelligence (AI) in medicine, are increasingly gaining ground. This review aims to provide an overview of the latest findings on WSI and advances in AI methods for predicting MSI status, summarize their applications in CRC, and discuss their strengths and limitations in daily clinical practice.
Collapse
Affiliation(s)
- Anna Lucia Cannarozzi
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Giuseppe Biscaglia
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo 71013, Italy.
| | - Tiziana Pia Latiano
- Oncology Unit, Fondazione Casa Sollievo della Sofferenza IRCCS, San Giovanni Rotondo 71013, Italy.
| | - Annamaria Gentile
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Davide Ciardiello
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan.
| | - Luca Massimino
- Gastroenterology and Digestive Endoscopy Department, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Anna Laura Pia Di Brina
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Maria Guerra
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Francesca Tavano
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Federica Ungaro
- Gastroenterology and Digestive Endoscopy Department, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Fabrizio Bossa
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Francesco Perri
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Anna Latiano
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Orazio Palmieri
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| |
Collapse
|
4
|
Wang F, Wang Y, Ran C, Liang J, Qi L, Zhang C, Ye Z. ZOOMit diffusion kurtosis imaging combined with diffusion weighted imaging for the assessment of microsatellite instability in endometrial cancer. Abdom Radiol (NY) 2025; 50:2720-2731. [PMID: 39641783 DOI: 10.1007/s00261-024-04720-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE Detecting microsatellite instability (MSI) plays a key role in the management of endometrial cancer (EC), as it is a critical predictive biomarker for Lynch syndrome or immunotherapy response. A pressing need exists for cost-efficient, broadly accessible tools to aid patient for universal testing. Herein, we investigate the value of ZOOMit diffusion kurtosis imaging (DKI) and diffusion weighted imaging (DWI) based on preoperative pelvic magnetic resonance imaging (MRI) images in assessing MSI in EC. METHODS Preoperative MRI examination including ZOOMit DKI and DWI of 81 EC patients were retrospectively analyzed. The apparent diffusion coefficient (ADC), mean kurtosis (MK), mean diffusivity (MD) and the largest tumor size based on MRI images, as well as patients' clinicopathological features were compared and analyzed according to different microsatellite statuses. RESULTS Of the 81 patients, 59 (72.8%) who were microsatellite stability (MSS) and 22 (27.2%) who were MSI. Interobserver agreement for the quantitative parameter measurements was excellent (ICC 0.78-0.98). The ADC and MD values were significantly lower, while Ki-67 proliferation level and MK values were significantly higher in the MSI group compared to those of the MSS group. The parameters of MD and MK were independent predictors for determining MSI, and their combination showed better diagnostic efficacy with an area under the receiver operating characteristic curve (AUROC) of 0.860 (95% confidence interval, 0.765, 0.927), although there was no significant difference compared to each individual parameter. CONCLUSION The microstructural heterogeneity assessment of ZOOMit DKI allowed for characterizing MSI status in EC. Within the current universal MSI testing paradigm, DKI may provide added value as a potential noninvasive imaging biomarker for preoperative assessment of MSI tumors, thereby facilitating clinical decision-making.
Collapse
Affiliation(s)
- Fang Wang
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Xuzhou Maternity and Child Health Care Hospital, Xvzhou, China
| | - Yafei Wang
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chenjiao Ran
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jing Liang
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lisha Qi
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | | | - Zhaoxiang Ye
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| |
Collapse
|
5
|
Rajoua N, Daunay A, Triki W, Baraket O, Bouchoucha S, Maghrebi H, Mabrouk A, Deleuze JF, How-Kit A, Kharrat M. HSP110 T17 marker matches the pentaplex panel and outperforms CAT-25 for detecting microsatellite instability in sporadic colorectal cancer. Cancer Genet 2025; 294-295:21-26. [PMID: 40064067 DOI: 10.1016/j.cancergen.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 06/01/2025]
Abstract
Microsatellite instability (MSI) is an important biomarker in colorectal cancer (CRC), influencing prognosis and treatment decisions. While conventional MSI detection typically relies on the pentaplex panel, newer markers like HSP110 T17 (HT-17) and CAT-25 may offer simpler, more cost-effective alternatives. This study aimed to assess the effectiveness of HT-17 and CAT-25 for detecting MSI in sporadic CRC and to explore any links between MSI status and clinicopathological features. A total of 96 Tunisian sporadic CRC patients were included, with MSI status evaluated using HT-17, CAT-25, and the refined mononucleotide repeat pentaplex panel through microsatellite genotyping. Clinicopathological data, such as tumor location and age at diagnosis, were also analyzed for associations with MSI. Among the 96 patients, 9 (9.38%) showed MSI, while 87 were microsatellite stable (MSS). HT-17 demonstrated 100% accuracy and sensitivity, matching the pentaplex panel's performance, while CAT-25 showed limited detection ability. MSI status was significantly linked to tumors in the proximal colon and, unexpectedly, to younger patients (<50 years old). HT-17 proved to be a reliable MSI marker in CRC, offering equivalent performance to the pentaplex panel, with the added advantages of simplicity and cost efficiency. The associations between MSI, tumor location, and younger age at diagnosis may provide valuable insights into CRC biology and clinical management. Further studies with larger cohorts are needed to validate HT-17' s clinical potential, with the goal of improving personalized treatment strategies and prognostic accuracy for CRC patients.
Collapse
Affiliation(s)
- Nasreddine Rajoua
- Human Genetics Laboratory (LR99ES10), Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Hassouna Ben Ayed street, La Rabta, 1007, Tunis, Tunisia; Laboratory of Research in Visceral Surgery and Digestive Pathology (LR12ES01), Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Hassouna Ben Ayed street, La Rabta, 1007, Tunis, Tunisia.
| | - Antoine Daunay
- Laboratory for Genomics, Jean Dausset Foundation - CEPH (Centre d'Etude du Polymorphisme Humain), 27 Juliette Dodu Street, 75010, Paris, France.
| | - Wissem Triki
- Department of Surgery, Habib Bougatfa Hospital, Faculty of Medicine of Tunis, University of Tunis El Manar, Hédi Saïdi Street, 7000, Bizerte, Tunisia.
| | - Oussema Baraket
- Laboratory of Research in Visceral Surgery and Digestive Pathology (LR12ES01), Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Hassouna Ben Ayed street, La Rabta, 1007, Tunis, Tunisia; Department of Surgery, Habib Bougatfa Hospital, Faculty of Medicine of Tunis, University of Tunis El Manar, Hédi Saïdi Street, 7000, Bizerte, Tunisia.
| | - Sami Bouchoucha
- Laboratory of Research in Visceral Surgery and Digestive Pathology (LR12ES01), Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Hassouna Ben Ayed street, La Rabta, 1007, Tunis, Tunisia; Department of Surgery, Habib Bougatfa Hospital, Faculty of Medicine of Tunis, University of Tunis El Manar, Hédi Saïdi Street, 7000, Bizerte, Tunisia.
| | - Houcine Maghrebi
- Laboratory of Research in Visceral Surgery and Digestive Pathology (LR12ES01), Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Hassouna Ben Ayed street, La Rabta, 1007, Tunis, Tunisia; Department of Surgery A, La Rabta Hospital, Faculty of Medicine of Tunis, University of Tunis El Manar, 1007, Tunis, Tunisia.
| | - Aymen Mabrouk
- Laboratory of Research in Visceral Surgery and Digestive Pathology (LR12ES01), Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Hassouna Ben Ayed street, La Rabta, 1007, Tunis, Tunisia; Department of Surgery A21, Charles Nicolle Hospital, Faculty of Medicine of Tunis, University of Tunis El Manar, 9-Avril 1938 Boulevard, 1006, Tunis, Tunisia.
| | - Jean-François Deleuze
- Laboratory for Genomics, Jean Dausset Foundation - CEPH (Centre d'Etude du Polymorphisme Humain), 27 Juliette Dodu Street, 75010, Paris, France; Centre National de Recherche en Génomique Humaine, CEA-Institut François Jacob, 2 Rue Gaston Crémieux, 91000, Évry-Courcouronnes, France.
| | - Alexandre How-Kit
- Laboratory for Genomics, Jean Dausset Foundation - CEPH (Centre d'Etude du Polymorphisme Humain), 27 Juliette Dodu Street, 75010, Paris, France.
| | - Maher Kharrat
- Human Genetics Laboratory (LR99ES10), Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Hassouna Ben Ayed street, La Rabta, 1007, Tunis, Tunisia.
| |
Collapse
|
6
|
Peng L, Ma W, Zhang X, Zhang F, Ma F, Ai K, Ma X, Jia Y, Ou-Yang H, Pei S, Wang T, Zhu Y, Wang L. Predictive value of combined DCE-MRI perfusion parameters and clinical features nomogram for microsatellite instability in colorectal cancer. Discov Oncol 2025; 16:892. [PMID: 40410525 PMCID: PMC12102045 DOI: 10.1007/s12672-025-02705-x] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/13/2025] [Indexed: 05/25/2025] Open
Abstract
OBJECTIVES To develop a nomogram that combines dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) perfusion parameters, ADC values and clinical features to preoperatively identify microsatellite instability (MSI) in patients with colorectal cancer (CRC). METHODS This retrospective study included 63 CRC patients who underwent preoperative DCE-MRI and had immunohistochemistry results available. Two radiologists, in a double-blind manner, placed two circular regions of interests in the area with the highest perfusion intensity on the DCE-MRI perfusion map and the corresponding area on the ADC map. Perfusion parameters and ADC values were measured, and the average values from both radiologists were used for subsequent analysis. Univariate analysis was performed to identify independent risk factors for MSI. A nomogram was then constructed by combining the most significant clinical risk factors with DCE-MRI perfusion parameters. The model's performance was evaluated using receiver operating characteristic (ROC) curves. Calibration curves, decision curve analysis (DCA), and clinical impact curves (CIC) were used to assess the nomogram's clinical utility and net benefit. RESULTS The nomogram prediction model, which combined PLT, LNM, Ktrans, Kep, iAUC, and ADC, demonstrated good predictive performance. The combined model had an AUC of 0.951 (95% CI: 0.903-0.998), an accuracy of 0.873, a sensitivity of 1.000, and a specificity of 0.818. Both the DCA and CIC demonstrated good clinical applicability and net benefit. CONCLUSION The nomogram method demonstrated good potential in the preoperative individualized identification of MSI status in CRC patients. This tool can assist clinicians in adopting appropriate treatment strategies and optimizing personalized stratification for CRC patients.
Collapse
Affiliation(s)
- Leping Peng
- Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Wenting Ma
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
| | - Xiuling Zhang
- Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Fan Zhang
- Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Fang Ma
- Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Kai Ai
- Department of Clinical and Technical Support, Philips Healthcare, Xi'an, 710065, Shanxi, China
| | - Xiaomei Ma
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
| | - Yingmei Jia
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
| | - Hong Ou-Yang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
| | - Shengting Pei
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
| | - Tao Wang
- Department of Colorectal Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
| | - Yuanhui Zhu
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China.
| | - Lili Wang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
7
|
Gill GS, Kharb S, Goyal G, Das P, Kurdia KC, Dhar R, Karmakar S. Immune Checkpoint Inhibitors and Immunosuppressive Tumor Microenvironment: Current Challenges and Strategies to Overcome Resistance. Immunopharmacol Immunotoxicol 2025:1-45. [PMID: 40376861 DOI: 10.1080/08923973.2025.2504906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025]
Abstract
Immune checkpoint inhibitors (ICIs) are shown to improve cancer treatment effectiveness by boosting the immune system of the patient. Nevertheless, the unique and highly suppressive TME poses a significant challenge, causing heterogeneity of response or resistance in a considerable number of patients. This review focuses on the evasive attributes of the TME. Immune evasion mechanism in TME include immunosuppressive cells, cytokine and chemokine signaling, metabolic alterations and overexpression of immune checkpoint molecules such as PD-1, CTLA-4, LAG-3, TIM-3, TIGIT, BTLA and their interactions within the TME. In addition, this review focuses on the overcoming resistance by targeting immunosuppressive cells, normalizing tumor blood vessels, blocking two or three checkpoints simultaneously, combining vaccines, oncolytic viruses and metabolic inhibitors with ICIs or other therapies. This review also focuses on the necessity of finding predictive markers for the stratification of patients and to check response of ICIs treatment. It remains to be made certain by new research and intelligent innovations how these discoveries of the TME and its interplay facilitate ICI treatment and change the face of cancer treatment.
Collapse
Affiliation(s)
- Gurpreet Singh Gill
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Simmi Kharb
- Department of Biochemistry, Pt. B.D. Sharma Postgraduate Institute of Medical Sciences, Rohtak, India
| | - Gitanjali Goyal
- Department of Biochemistry, All India Institute of Medical Sciences, Bathinda, India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kailash Chand Kurdia
- Department of GI Surgery & Liver Transplantation, All India Institute of Medical Sciences, New Delhi, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
8
|
Saito S, Kato S, Arai U, En A, Tsunezumi J, Mizushima T, Tateishi K, Adachi N. HR eye & MMR eye: one-day assessment of DNA repair-defective tumors eligible for targeted therapy. Nat Commun 2025; 16:4239. [PMID: 40355434 PMCID: PMC12069580 DOI: 10.1038/s41467-025-59462-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
Homologous recombination (HR) and mismatch repair (MMR) act as guardians of the human genome, and defects in HR or MMR are causative in at least a quarter of all malignant tumors. Although these DNA repair-deficient tumors are eligible for effective targeted therapies, fully reliable diagnostic strategies based on functional assay have yet to be established, potentially limiting safe and proper application of the molecular targeted drugs. Here we show that transient transfection of artificial DNA substrates enables ultrarapid detection of HR and MMR. This finding led us to develop a diagnostic strategy that can determine the cellular HR/MMR status within one day without the need for control cells or tissues. Notably, the accuracy of this method allowed the discovery of a pathogenic RAD51D mutation, which was missed by existing companion diagnostic tests. Our methods, termed HR eye and MMR eye, are applicable to frozen tumor tissues and roughly predict the response to therapy. Overall, the findings presented here could pave the way for accurately assessing malignant tumors with functional defects in HR or MMR, a step forward in accelerating precision medicine.
Collapse
Affiliation(s)
- Shinta Saito
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Shingo Kato
- Department of Clinical Cancer Genomics, Yokohama City University Hospital, Yokohama, 236-0004, Japan
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Usaki Arai
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Atsuki En
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Jun Tsunezumi
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Taichi Mizushima
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Kensuke Tateishi
- Department of Neurosurgery, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Noritaka Adachi
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan.
| |
Collapse
|
9
|
Capello Ingold G, Martins da Fonseca J, Kolenda Zloić S, Verdan Moreira S, Kago Marole K, Finnegan E, Yoshikawa MH, Daugėlaitė S, Souza E Silva TX, Soato Ratti MA. Preoperative radiomics models using CT and MRI for microsatellite instability in colorectal cancer: a systematic review and meta-analysis. Abdom Radiol (NY) 2025:10.1007/s00261-025-04981-1. [PMID: 40347255 DOI: 10.1007/s00261-025-04981-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/12/2025]
Abstract
OBJECTIVE Microsatellite instability (MSI) is a novel predictive biomarker for chemotherapy and immunotherapy response, as well as prognostic indicator in colorectal cancer (CRC). The current standard for MSI identification is polymerase chain reaction (PCR) testing or the immunohistochemical analysis of tumor biopsy samples. However, tumor heterogeneity and procedure complications pose challenges to these techniques. CT and MRI-based radiomics models offer a promising non-invasive approach for this purpose. MATERIALS AND METHODS A systematic search of PubMed, Embase, Cochrane Library and Scopus was conducted to identify studies evaluating the diagnostic performance of CT and MRI-based radiomics models for detecting MSI status in CRC. Pooled area under the curve (AUC), sensitivity, and specificity were calculated in RStudio using a random-effects model. Forest plots and a summary ROC curve were generated. Heterogeneity was assessed using I² statistics and explored through sensitivity analyses, threshold effect assessment, subgroup analyses and meta-regression. RESULTS 17 studies with a total of 6,045 subjects were included in the analysis. All studies extracted radiomic features from CT or MRI images of CRC patients with confirmed MSI status to train machine learning models. The pooled AUC was 0.815 (95% CI: 0.784-0.840) for CT-based studies and 0.900 (95% CI: 0.819-0.943) for MRI-based studies. Significant heterogeneity was identified and addressed through extensive analysis. CONCLUSION Radiomics models represent a novel and promising tool for predicting MSI status in CRC patients. These findings may serve as a foundation for future studies aimed at developing and validating improved models, ultimately enhancing the diagnosis, treatment, and prognosis of colorectal cancer.
Collapse
|
10
|
Wang X, Gu Y, Yi L, Wu T, Wang L, Tian J, Lu Y, Jin P, Yang X, Yang Y. Pathologic complete response after Sintilimab combined with FOLFOX therapy in MSI-H type patients with locally advanced GC: a case report. Front Oncol 2025; 15:1560450. [PMID: 40406249 PMCID: PMC12094948 DOI: 10.3389/fonc.2025.1560450] [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: 02/07/2025] [Accepted: 03/27/2025] [Indexed: 05/26/2025] Open
Abstract
As one of the most common gastrointestinal tumors, Gastric Cancer (GC) poses a serious threat to human health due to its high morbidity and mortality. The current treatment strategy is a comprehensive treatment program mainly based on surgery, especially for advanced GC patients. The emergence of immune checkpoint inhibitors has completely changed this status quo, and the synergistic effect of neoadjuvant immunotherapy combined with chemotherapy has significantly improved the resection and radical rate and overall survival of patients with advanced local GC. We present a case of locally advanced GC (cT4N0Mx) with microsatellite instability high (MSI-H) and PD-L1 Combined Positive Score (CPS)=2. The patient received neoadjuvant therapy with Sintilimab combined with FOLFOX (folinic acid (leucovorin), 5-fluorouracil (5-FU), and oxaliplatin), and significantly reduced tumor volume after 3 cycles of treatment. Then she underwent subtotal gastrectomy with gastrojejunostomy and D2 lymph node dissection. The postoperative pathological results showed that no cancerous tissue remained in the tumor tissue, and pathologic complete response (pCR) was achieved. The first cycle of adjuvant therapy with the same protocol was received after surgery. During adjuvant therapy, patients mainly experienced side effects such as dyspepsia, nausea and mild myelosuppression. Therefore, immunotherapy with Sintilimab combined with FOLFOX chemotherapy has the potential to be an effective treatment option for patients with resectable locally advanced MSI-H GC.
Collapse
Affiliation(s)
- Xiaoke Wang
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
- First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yuanhui Gu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Lin Yi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Tao Wu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Ling Wang
- Pathology Department, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Jiao Tian
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yuyuan Lu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Penghui Jin
- First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xin Yang
- First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yan Yang
- Pharmacy Department, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
11
|
Zhu J, Fu S, Zou X, Zeng H, Cui G, Peng Y, Tang D, Zhang F, Shen H, Zeng S, Han Y. PRMT5 Inhibitor Synergizes with Chemotherapy to Induce Resembling Mismatch Repair Deficiency and Enhance Anti-TIGIT Therapy in Microsatellite-Stable Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500271. [PMID: 40344511 DOI: 10.1002/advs.202500271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/06/2025] [Indexed: 05/11/2025]
Abstract
Microsatellite stable (MSS) colorectal cancer (CRC) is considered an "immune-cold" tumor, accounting for ≈85% of all CRC cases. The overall response rate to chemotherapy combined with immune checkpoint inhibitors in MSS CRC is typically less than 10%. The specific mechanism that enhances chemotherapy sensitivity and mediated immunogenicity renders MSS CRC more responsive to immunotherapy remains elusive. Experiments in this study identify a DNA damage repair-related epigenetic gene, protein arginine methyltransferase 5 (PRMT5), whose inhibition enhances Irinotecan (CPT-11) sensitivity and synergistically induces a postmeiotic segregation increased 2 (PMS2)-deficient-like state, leading to the release of cytosolic double-stranded DNA. This activates the cyclic GMP-AMP synthase (cGAS)-stimulator of the IFN gene (STING) signaling pathway, thereby enhancing anti-tumor immunotherapy through dendritic cell-T cell-dependent functions. Importantly, combining the epigenetic anti-tumor drug GSK3326595 with CPT-11 significantly upregulates the immune receptor tyrosine-based inhibitory motif (TIGIT) level on CD8+ T cells and subsequently demonstrates impressive anti-tumor efficacy in vivo when additional anti-TIGIT is included. Collectively, this study reveals the crucial role of PRMT5 blockade combined with CPT-11 in inducing a mismatch repair deficiency-like state and provides a novel triple-drug combination therapy strategy as a potential treatment for patients with MSS CRC.
Collapse
Affiliation(s)
- Jiang Zhu
- Department of Oncology, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Shenao Fu
- Department of Oncology, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Xi Zou
- Department of Oncology, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Hanjiang Zeng
- Department of Nephrology, Xingsha Campus, Hunan Provincial People's Hospital, Changsha, Hunan, 410100, P. R. China
| | - Guangzu Cui
- Department of Oncology, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Yinghui Peng
- Department of Oncology, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Diya Tang
- Department of Oncology, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Fan Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, P. R. China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Kaifu District, Central South University, Changsha, Hunan, 410008, P. R. China
| |
Collapse
|
12
|
Kaplan Z, Prezioso E, Jain A, Lavu H, Yeo CJ, Bowne WB, Nevler A. Clinical Implications of Mismatch Repair Deficiency in Pancreatic Ductal Adenocarcinoma. Cancer Med 2025; 14:e70960. [PMID: 40366030 PMCID: PMC12076359 DOI: 10.1002/cam4.70960] [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: 02/25/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive and lethal disease, characterized by a limited response to chemotherapy and overall poor prognosis. Pancreatic cancers with a distinct mismatch repair deficiency, although relatively rare, have been shown to be associated with markedly better outcomes in comparison. Furthermore, whereas pancreatic cancers are generally unresponsive to current immunotherapy, this specific group of tumors has been shown to have a notable susceptibility to immune checkpoint inhibitors. AIMS In this review, we aim to summarize the relevant literature regarding mismatch-repair associated pancreatic cancers, the impacted biological mechanisms, and the resulting vulnerabilities for potential opportunistic immunotherapeutic treatment approaches. We will also review the current clinical studies assessing survival outcomes of mismatch repair deficient pancreatic cancers and ongoing clinical trials in this emerging field. RESULTS AND CONCLUSIONS Patients with dMMR/MSI-H pancreatic cancers harbor a distinct phenotype that has increased immune activation, greater responsiveness to immune checkpoint inhibitor therapy and better overall survival when compared to other pancreatic cancers. Although this molecular subtype makes up a small minority of cases, emerging data suggest immunotherapy may offer benefit to these patients.
Collapse
Affiliation(s)
- Zachary Kaplan
- Sidney Kimmel Medical CollegePhiladelphiaPennsylvaniaUSA
| | | | - Aditi Jain
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Harish Lavu
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Charles J. Yeo
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Wilbur B. Bowne
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Avinoam Nevler
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
13
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [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: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
14
|
Jia H, Bian Y, Yuan J, Zhang Y, Zhang S. The Potential Role of C4 MYH11+ Fibroblasts and the MDK-SDC2 Ligand-Receptor Pair in Lung Adenocarcinoma: Implications for Prognosis and Therapeutic Strategies. Transl Oncol 2025; 55:102364. [PMID: 40121996 PMCID: PMC11982484 DOI: 10.1016/j.tranon.2025.102364] [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: 11/05/2024] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) posed a significant threat to global human health. This study employed single-cell RNA sequencing (scRNA-seq) to analyze transcriptomic data from nine LUAD patients at different stages of tumor infiltration, aiming to elucidate the tumor microenvironment and key biological processes of LUAD. METHODS In this study, we processed the scRNA-seq data using the Seurat package and sequentially applied principal component analysis followed by the Harmony package to effectively correct for batch effects, identifying 105,725 high-quality cells. Through cell clustering and gene expression profiling, we identified critical cell subpopulations and gene expression patterns in LUAD patients. RESULTS Our analysis revealed that the C4 MYH11+ Fibroblasts subtype was primarily involved in biological processes related to muscle function. Further investigations uncovered the MDK-SDC2 ligand-receptor pair as a critical regulator of tumor cell invasion, proliferation, and migration, driving LUAD progression. Additionally, we developed a gene-based prognostic model that effectively predicted patient survival, providing valuable clinical insights. CONCLUSION This study provided a comprehensive atlas of the LUAD tumor microenvironment, highlighted the role of the C4 MYH11+ Fibroblasts in tumor progression. It also proposed the MDK-SDC2 ligand-receptor pair as a novel mechanism, addressing a significant gap in this area of research. And presented a gene-based prognostic model as a novel perspective for research into immunotherapy and drug sensitivity in LUAD.
Collapse
Affiliation(s)
- Hongling Jia
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.; The first clinical medical college of Shandong university of Traditional Chinese Medicine, Jinan, China
| | - Yanjie Bian
- Xinxiang Medical University, Xinxiang, China
| | - Jie Yuan
- Sijing Town Community Healthcare Center, Shanghai, China
| | - Yi Zhang
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China..
| | - Shengyi Zhang
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China..
| |
Collapse
|
15
|
Thomas QD, Vendrell JA, Khellaf L, Cavaillon S, Quantin X, Solassol J, Cabello‐Aguilar S. Artificial intelligence-driven microsatellite instability profiling reveals distinctive genetic features in patients with lung cancer. Cancer 2025; 131:e35882. [PMID: 40297960 PMCID: PMC12038786 DOI: 10.1002/cncr.35882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/10/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Microsatellite instability (MSI) has emerged as a predictive biomarker for immunotherapy response in various cancers, but its role in non-small cell lung cancer (NSCLC) is not fully understood. METHODS The authors used the bioinformatics tool MIAmS to assess microsatellite status from next-generation sequencing (NGS) data using a tailored microsatellite score. Immunohistochemistry (IHC) assays were also performed to evaluate the correspondence between MSI and deficient mismatch repair (dMMR) status. A retrospective analysis of 1547 lung cancer patients was conducted, focusing on those with an MSI phenotype. Clinical characteristics, co-occurring molecular alterations, tumor mutation burden (TMB), and homologous recombination deficiency (HRD) status were evaluated in this subset. RESULTS Of the 1547 patients analyzed, eight (0.52%) were identified as having MSI through MIAmS, with six (0.39%) of these cases also being dMMR on IHC. All patients with dMMR had an MS score ≥2 and a history of smoking. Most patients showed loss of MLH1 and PMS2 staining on IHC. No correlation was found between MSI status and programmed death-ligand 1 expression, although all MSI patients exhibited high TMB, averaging 21.4 ± 5.6 mutations per megabase. DISCUSSION MSI/dMMR in lung cancer is exceedingly rare, affecting less than 1% of cases. NGS-based analysis combined with bioinformatics tools provides a robust method to identify MSI/dMMR patients, potentially guiding immunotherapy decisions. This comprehensive approach integrates molecular genotyping and MSI detection, offering personalized treatment options for lung cancer patients. NGS-based MSI testing is emerging as the preferred method for detecting microsatellite instability in various tumor types, including rare cancers.
Collapse
Affiliation(s)
- Quentin Dominique Thomas
- Department of Medical OncologyInstitute du Cancer de MontpellierMontpellier UniversityMontpellierFrance
- Oncogenic Pathways in Lung CancerMontpellier Cancer Research InstituteUniversity of MontpellierMontpellierFrance
| | - Julie Adèle Vendrell
- Solid Tumor LaboratoryDepartment of Pathology and OncobiologyMontpellier University Hospital MontpellierArnaud de Villeneuve HospitalMontpellierFrance
| | - Lakhdar Khellaf
- Department of PathologyInstitute du Cancer de MontpellierMontpellier UniversityMontpellierFrance
| | - Sarah Cavaillon
- Department of Medical OncologyInstitute du Cancer de MontpellierMontpellier UniversityMontpellierFrance
| | - Xavier Quantin
- Department of Medical OncologyInstitute du Cancer de MontpellierMontpellier UniversityMontpellierFrance
- Oncogenic Pathways in Lung CancerMontpellier Cancer Research InstituteUniversity of MontpellierMontpellierFrance
| | - Jérôme Solassol
- Solid Tumor LaboratoryDepartment of Pathology and OncobiologyMontpellier University Hospital MontpellierArnaud de Villeneuve HospitalMontpellierFrance
| | - Simon Cabello‐Aguilar
- Solid Tumor LaboratoryDepartment of Pathology and OncobiologyMontpellier University Hospital MontpellierArnaud de Villeneuve HospitalMontpellierFrance
- Montpellier BioInformatics for Clinical DiagnosisMolecular Medicine and Genomics PlatformMontpellier University Hospital MontpellierMontpellierFrance
| |
Collapse
|
16
|
Gong W, Wen S, Chen Y, Wu F, Yang M, Sun P, Guo X, Li M, Chen D, Zhao H, Wang L. Deciphering ERR family genes as prognostic and immunological biomarkers through pan-cancer analysis with validation in gallbladder cancer. Front Oncol 2025; 15:1525635. [PMID: 40356747 PMCID: PMC12066295 DOI: 10.3389/fonc.2025.1525635] [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: 11/10/2024] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
Background The estrogen-related receptor family genes (ERRs), including ESRRA, ESRRB, and ESRRG, have been implicated in a few tumors, exhibiting distinct roles through diverse mechanisms. The purpose of our research is to explore the commonalities and underlying mechanism of ERRs in malignancies from a pan-cancer perspective and to validate the role and mechanisms of ESRRG in gallbladder cancer (GBC). Methods We leveraged public databases such as TCGA and GTEx to systematically investigate the potential functions of ERRs in malignancies. ESRRG expression was analyzed through immunohistochemical staining in gallbladder cancer and cholecystitis tissues. For functional validation, ESRRG was knocked down in GBC cell lines, followed by CCK-8, colony formation, scratch wound healing, Transwell migration, and invasion assays. Western blot, qPCR, and immunofluorescence were performed to evaluate the relationship between ESRRG, PD-L1, and CD8+ T cells. Results Compared to adjacent normal tissues, ESRRA is overexpressed in most tumors, ESRRB is generally underexpressed, and ESRRG exhibits significant expression alterations across various tumors. All three ERRs demonstrate significant prognostic value across different cancers. Notably, the strong associations of ERRs with key immunological features-stromal scores, immune cell infiltration, microsatellite instability (MSI), and tumor mutational burden (TMB)-suggest their involvement in immune evasion and their potential utility in guiding immunotherapy strategies. All three ERRs display a positive correlation with advanced tumor stages in cholangiocarcinoma (CHOL). Specifically, in CHOL, ESRRG expression is closely associated with lymphatic metastasis, poorer overall survival, reduced immune infiltration, elevated PD-L1 expression, epithelial-mesenchymal transition (EMT), and DNA damage response. In GBC tissues, we subsequently confirmed that ESRRG expression positively correlates with pathological staging and PD-L1 expression, while negatively correlating with prognosis and CD8+ T cell infiltration. Knockdown of ESRRG in gallbladder cancer cells results in decreased proliferation, migration, and invasion. Moreover, the expression of PD-L1, MSH2, BRCA1, MMP2, and VIMENTIN decreased with ESRRG knockdown. Conclusion Our pan-cancer analysis reveals ERRs as critical regulators of tumor immunity and progression, with ESRRG emerging as a key oncogenic driver in GBC. The mechanistic link between ESRRG and PD-L1/EMT suggests its potential as a therapeutic target to enhance immunotherapy efficacy. These findings underscore the need for tissue-specific targeting strategies for ERR family members in precision oncology.
Collapse
Affiliation(s)
- Wanwan Gong
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Sijia Wen
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yu Chen
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fan Wu
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Mengmeng Yang
- Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Ping Sun
- Department of Pathology, Jiangnan University Medical Center, Wuxi, China
| | - Xingmei Guo
- Department of Pathology, Jiangnan University Medical Center, Wuxi, China
| | - Meiqin Li
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
| | - Daozhen Chen
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Hui Zhao
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
| | - Lei Wang
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
- Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
- Department of Hepatopancreatobiliary Surgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
17
|
Marra A, Morganti S, Pareja F, Campanella G, Bibeau F, Fuchs T, Loda M, Parwani A, Scarpa A, Reis-Filho JS, Curigliano G, Marchiò C, Kather JN. Artificial intelligence entering the pathology arena in oncology: current applications and future perspectives. Ann Oncol 2025:S0923-7534(25)00112-7. [PMID: 40307127 DOI: 10.1016/j.annonc.2025.03.006] [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: 12/18/2024] [Revised: 02/19/2025] [Accepted: 03/07/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Artificial intelligence (AI) is rapidly transforming the fields of pathology and oncology, offering novel opportunities for advancing diagnosis, prognosis, and treatment of cancer. METHODS Through a systematic review-based approach, the representatives from the European Society for Medical Oncology (ESMO) Precision Oncology Working Group (POWG) and international experts identified studies in pathology and oncology that applied AI-based algorithms for tumour diagnosis, molecular biomarker detection, and cancer prognosis assessment. These findings were synthesised to provide a comprehensive overview of current AI applications and future directions in cancer pathology. RESULTS The integration of AI tools in digital pathology is markedly improving the accuracy and efficiency of image analysis, allowing for automated tumour detection and classification, identification of prognostic molecular biomarkers, and prediction of treatment response and patient outcomes. Several barriers for the adoption of AI in clinical workflows, such as data availability, explainability, and regulatory considerations, still persist. There are currently no prognostic or predictive AI-based biomarkers supported by level IA or IB evidence. The ongoing advancements in AI algorithms, particularly foundation models, generalist models and transformer-based deep learning, offer immense promise for the future of cancer research and care. AI is also facilitating the integration of multi-omics data, leading to more precise patient stratification and personalised treatment strategies. CONCLUSIONS The application of AI in pathology is poised to not only enhance the accuracy and efficiency of cancer diagnosis and prognosis but also facilitate the development of personalised treatment strategies. Although barriers to implementation remain, ongoing research and development in this field coupled with addressing ethical and regulatory considerations will likely lead to a future where AI plays an integral role in cancer management and precision medicine. The continued evolution and adoption of AI in pathology and oncology are anticipated to reshape the landscape of cancer care, heralding a new era of precision medicine and improved patient outcomes.
Collapse
Affiliation(s)
- A Marra
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - S Morganti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA; Department of Medicine, Harvard Medical School, Boston, USA; Gerstner Center for Cancer Diagnostics, Broad Institute of MIT and Harvard, Boston, USA
| | - F Pareja
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - G Campanella
- Hasso Plattner Institute for Digital Health, Mount Sinai Medical School, New York, USA; Department of AI and Human Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - F Bibeau
- Department of Pathology, University Hospital of Besançon, Besancon, France
| | - T Fuchs
- Hasso Plattner Institute for Digital Health, Mount Sinai Medical School, New York, USA; Department of AI and Human Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - M Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA; Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, USA
| | - A Parwani
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, USA
| | - A Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy; ARC-Net Research Center, University of Verona, Verona, Italy
| | - J S Reis-Filho
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - G Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - C Marchiò
- Candiolo Cancer Institute, FPO IRCCS, Candiolo, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - J N Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany; Department of Medicine I, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
18
|
Evrard C, Rochelle T, Martel M, Al Achkar A, Ferru A, Randrian V, Karayan-Tapon L, Tougeron D. Prognostic and Predictive Value of Microsatellite Instability Analysis in Circulating Tumor DNA Using Digital Droplet PCR for Patients With Microsatellite Instability Colorectal Cancers. J Transl Med 2025; 105:104176. [PMID: 40246285 DOI: 10.1016/j.labinv.2025.104176] [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: 11/19/2024] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Deficient mismatch repair (dMMR) and/or microsatellite instability (MSI) colorectal cancer (CRC) is highly sensitive to immune checkpoint inhibitors (ICI). It is thus becoming increasingly relevant to monitor circulating tumor DNA (ctDNA) and to determine the MSI status (ctDNA-MSI) in CRC. So far, few studies have explored this, even though it could be particularly relevant in evaluating treatment efficacy in patients with dMMR and/or MSI CRC. The ctDNA DIgestive cancers MSI study (ADI-MSI) aims to assess the value of ctDNA-MSI as a predictor of ICI efficacy. Blood samples were collected prospectively in a single-center cohort to analyze circulating cell-free DNA (cfDNA) and ctDNA-MSI before the start of and during treatment. ctDNA-MSI was measured using digital droplet PCR with the 5 microsatellite markers of the Pentaplex panel (Promega Corporation). The primary endpoint was to evaluate ctDNA-MSI levels as a predictor of progression-free survival (PFS). We included 54 patients with dMMR and/or MSI CRC, most of whom had metastatic disease (77.8%) treated in the first (25.9%) or second line (42.6%) with ICI. High-baseline cfDNA and ctDNA-MSI were associated with worse PFS and overall survival. ctDNA-MSI kinetics, but not cfDNA kinetics, was associated with treatment response (P = .006), PFS (P = .03), and overall survival (P = .04). ctDNA-MSI kinetics divided into 3 groups (increase, decrease, and negative) correlated strongly with PFS (PFS at 24 months was 0%, 53.0%, and 77.0%, respectively; P < .001) and remained significant in multivariate analysis (hazard ratio = 7.93; 95% CI, 2.23-28.21; P = .005). As there is no strong predictor of ICI efficacy in patients with dMMR and/or MSI CRC, these results suggest that ctDNA-MSI could help physicians in treatment decision-making in the future.
Collapse
Affiliation(s)
- Camille Evrard
- PRoDiCeT, Université de Poitiers, Poitiers, France; Service d'Oncologie Médicale, CHU de Poitiers, Poitiers, France
| | - Tristan Rochelle
- Service de Cancérologie Biologique, CHU de Poitiers, Poitiers, France
| | - Marine Martel
- Service de Cancérologie Biologique, CHU de Poitiers, Poitiers, France
| | - Anis Al Achkar
- PRoDiCeT, Université de Poitiers, Poitiers, France; Service de Cancérologie Biologique, CHU de Poitiers, Poitiers, France
| | - Aurélie Ferru
- Service d'Oncologie Médicale, CHU de Poitiers, Poitiers, France
| | - Violaine Randrian
- PRoDiCeT, Université de Poitiers, Poitiers, France; Service d'Hépato-Gastro-Entérologie, CHU de Poitiers, Poitiers, France
| | - Lucie Karayan-Tapon
- PRoDiCeT, Université de Poitiers, Poitiers, France; Service de Cancérologie Biologique, CHU de Poitiers, Poitiers, France
| | - David Tougeron
- PRoDiCeT, Université de Poitiers, Poitiers, France; Service d'Hépato-Gastro-Entérologie, CHU de Poitiers, Poitiers, France.
| |
Collapse
|
19
|
Tan Y, Xing Y, Yuan S, Sun F, Lin X, Bao S, Jiang D, Zhang J, Sun SL. Potential Value of AURKA and CDK6 Amplification for the Response of Patients With Gastric Cancer to Neoadjuvant Chemotherapy. Mol Carcinog 2025. [PMID: 40222043 DOI: 10.1002/mc.23921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/19/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025]
Abstract
Many patients respond poorly to neoadjuvant chemotherapy (NACT), negatively affecting the surgical success rate. Identifying effective biomarkers and understanding the potential resistance mechanisms are urgently needed. Data of 18 patients with advanced stomach cancer who were treated with NACT categorized according to tumor regression grade into major histological response (MJHR) and nonhistological response (NHR) groups were retrospectively analyzed. Genomic signatures associated with the response to NACT were identified using whole-exome and RNA sequencing. Extraction of molecular signatures revealed increased deficient mismatch repair signature and tumor mutation levels in the NHR group. Compared to the MJHR group, the NHR group was also characterized by a greater number of copy number alterations (p = 0.08), which was further confirmed by RNA sequencing, and upregulation of aurora kinase A (AURKA) (p = 0.05) and cyclin-dependent kinase 6 (CDK6) (p = 0.049). Western blot analysis and immunohistochemical analyses further confirmed high CDK6 (p < 0.01/p < 0.0001) and AURKA (p < 0.01/p < 0.001) expression levels in the NHR group. Finally, palbociclib, an inhibitor of CDK4/6, effectively inhibited the proliferation (p < 0.05) and induced apoptosis of oxaliplatin-resistant gastric cancer cells (p < 0.01) in vitro. These findings support the potential value of AURKA and CDK6 amplification, as well as their effects on the tumor microenvironment, in predicting poor outcomes of NACT in patients with locally advanced gastric cancer. Thus, CDK4/6 inhibitors could be used to treat NACT-resistant patients with gastric cancer.
Collapse
Affiliation(s)
- Yuen Tan
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Yao Xing
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Shuai Yuan
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Fan Sun
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Xiaohui Lin
- School of Computer Science and Technology, Dalian University of Technology, Dalian, China
| | - Simeng Bao
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Dongyue Jiang
- Key Laboratory of Ocean Energy Utilization and Energy Conservation of Ministry of Education, Dalian University of Technology, Dalian, China
| | - Jianjun Zhang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Shu-Lan Sun
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| |
Collapse
|
20
|
Cui S, Zheng H, Xu Y, Wu Q, Liu W, Cai Y, Fan L, Tian Y, Qian H, Ding Y, Zhang X, Zhang J, Wu X, Wang R, Li X, Chen X. Plasma proteomic biomarkers predict therapeutic responses in advanced biliary tract cancer patients receiving Camrelizumab plus the GEMOX treatment. NPJ Precis Oncol 2025; 9:102. [PMID: 40195413 PMCID: PMC11977001 DOI: 10.1038/s41698-025-00879-9] [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: 10/26/2024] [Accepted: 03/13/2025] [Indexed: 04/09/2025] Open
Abstract
Biliary tract cancer (BTC) has greatly influenced patient survival for years. Nowadays, immunotherapy represents a promising breakthrough and proteomics is one of powerful technologies in biomarker research. We collected plasma and tissue samples from 37 patients with advanced BTC and 92 proteins were analyzed by proximity extension assay (PEA). Through linear mixed effect models, compared to partial response (PR) group, 8 proteins, IL7, ANGPT2, IL15, HO-1, CXCL1, CXCL5, IL33, and VEGFA, exhibited significantly higher expression in stable disease and progressive disease (SD_PD) group in response-effect analysis. It was also revealed that a subset of proteins increased over time, including PDCD1, TNFRSF4, DCN, CRTAM, VEGFR-2 and ADA in PR group and PDCD1, IL10, ADA, CD28, and PTN in SD_PD group. In interaction-effect analysis, HO-1, ANGPT2, IL15 were three significant differentially expressed proteins (DEPs). Receiver operating characteristic (ROC) analysis further demonstrated that HO-1, ANGPT2, IL15 showed high accuracy in patients with immune checkpoint blockade (ICB) treatment plus chemotherapy (AUC = 0.74). In addition, based on the obtained plasma and tissue samples, two nomogram models were constructed for predicting the prognosis of BTC by genome combined with proteomics. Collectively meaningful proteomic biomarkers are beneficial to evaluate the efficacy of immunotherapy, and these discovered biomarkers may be included in the scope of treatments' evaluation and improvement in future study.
Collapse
Affiliation(s)
- Shiyun Cui
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, P. R. China
- Department of oncology, Chongqing Hospital of Jiangsu Province Hospital (The People's Hospital of Qijiang District), Chongqing, 401420, China
| | - Hejian Zheng
- Department of oncology, Chongqing Hospital of Jiangsu Province Hospital (The People's Hospital of Qijiang District), Chongqing, 401420, China
| | - Yiyang Xu
- Nanjing Medical University, Nanjing, 210029, China
| | - Qiuyu Wu
- Nanjing Medical University, Nanjing, 210029, China
| | - Weici Liu
- Nanjing Medical University, Nanjing, 210029, China
| | - Yucheng Cai
- Nanjing Medical University, Nanjing, 210029, China
| | - Lei Fan
- Department of General Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Yitong Tian
- Nanjing Medical University, Nanjing, 210029, China
| | - Hao Qian
- Nanjing Medical University, Nanjing, 210029, China
| | - Yuting Ding
- Nanjing Medical University, Nanjing, 210029, China
| | - Xinyi Zhang
- Nanjing Medical University, Nanjing, 210029, China
| | | | - Xiaofeng Wu
- Hepatobiliar k77y Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Rong Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, P. R. China.
| | - Xiangcheng Li
- Hepatobiliar k77y Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Xiaofeng Chen
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, P. R. China.
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, P. R. China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, P. R. China.
| |
Collapse
|
21
|
Lebedeva A, Taraskina A, Grigoreva T, Belova E, Kuznetsova O, Ivanilova D, Sergeeva A, Kavun A, Veselovsky E, Nikulin V, Aliyarova S, Belyaeva L, Tryakin A, Fedyanin M, Mileyko V, Ivanov M. The Role of MSI Testing Methodology and Its Heterogeneity in Predicting Colorectal Cancer Immunotherapy Response. Int J Mol Sci 2025; 26:3420. [PMID: 40244273 PMCID: PMC11989282 DOI: 10.3390/ijms26073420] [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: 02/27/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
MSI is a crucial biomarker for selecting CRC patients for immunotherapy. Here, we analyze the first results from the observational prospective trial BLOOMSI (NCT06414304), which investigated the impact of MSI/dMMR testing methods and baseline tumor heterogeneity on treatment outcomes. Thirty MSI/dMMR+ CRC patients, who were candidates for immunotherapy, were enrolled. Depending on the local test used for MSI/dMMR, central PCR/IHC was performed. Baseline FFPE and liquid biopsy (LB) were analyzed with NGS. ORR (objective response rate) in the ITT population was 50% (95% CI, 31.3-68.7%). Concordance between local/central dMMR/MSI testing was 81%, and the concordance of IHC, PCR, NGS/FFPE, and NGS/LB was 68.4%. The ORR was similar for IHC+, PCR+, NGS/FFPE+, and NGS/LB+ patients (55.6%, 55.6%, 55%, and 57.9%, respectively). The ORR among patients with discordant IHC/PCR results was 0%, and the ORR among patients with NGS/LB-ORR was 25% (2/8 CR). Next, we performed quantitative MSI analysis, reflecting the clonality of MSI+ tumor cells. Multivariate analysis identified MSI clonality in FFPE (HR 0.63, 95% CI, 0.39-0.99, p = 0.0487) and LB (HR 3.05, 95% CI, 2.01-4.65, p < 0.00001) as independent predictors of progression. The ORR in patients with high clonality (≥7%, n = 4, NGS/LB) was 25%. We describe baseline methodological predictors of non-response to immunotherapy and propose a strategy for selecting potential non-responders. These findings warrant further investigation.
Collapse
Affiliation(s)
- Alexandra Lebedeva
- OncoAtlas LLC, Leninskiy Prospekt, 4c1A, Office #1, Moscow 119049, Russia; (A.L.); (A.T.); (T.G.); (O.K.); (A.K.); (E.V.); (S.A.); (V.M.); (M.I.)
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Trubetskaya Ulitsa, 8/2, Moscow 119048, Russia;
| | - Anastasiia Taraskina
- OncoAtlas LLC, Leninskiy Prospekt, 4c1A, Office #1, Moscow 119049, Russia; (A.L.); (A.T.); (T.G.); (O.K.); (A.K.); (E.V.); (S.A.); (V.M.); (M.I.)
| | - Tatiana Grigoreva
- OncoAtlas LLC, Leninskiy Prospekt, 4c1A, Office #1, Moscow 119049, Russia; (A.L.); (A.T.); (T.G.); (O.K.); (A.K.); (E.V.); (S.A.); (V.M.); (M.I.)
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Trubetskaya Ulitsa, 8/2, Moscow 119048, Russia;
| | - Ekaterina Belova
- OncoAtlas LLC, Leninskiy Prospekt, 4c1A, Office #1, Moscow 119049, Russia; (A.L.); (A.T.); (T.G.); (O.K.); (A.K.); (E.V.); (S.A.); (V.M.); (M.I.)
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Trubetskaya Ulitsa, 8/2, Moscow 119048, Russia;
- Faculty of Physics, Lomonosov Moscow State University, Leninskiye Gory, 1, Moscow 119991, Russia
| | - Olesya Kuznetsova
- OncoAtlas LLC, Leninskiy Prospekt, 4c1A, Office #1, Moscow 119049, Russia; (A.L.); (A.T.); (T.G.); (O.K.); (A.K.); (E.V.); (S.A.); (V.M.); (M.I.)
- Federal State Budgetary Institution N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye Highway, 23, Moscow 115478, Russia; (A.S.); (V.N.); (A.T.); (M.F.)
| | - Daria Ivanilova
- State Budgetary Institution of Health Care of the City of Moscow, Moscow Multidisciplinary Clinical Center “Kommunarka”, Department of Health of the City of Moscow, Ulitsa Sosenskiy Stan, 8c3, Moscow 142770, Russia;
| | - Anastasia Sergeeva
- Federal State Budgetary Institution N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye Highway, 23, Moscow 115478, Russia; (A.S.); (V.N.); (A.T.); (M.F.)
| | - Alexandra Kavun
- OncoAtlas LLC, Leninskiy Prospekt, 4c1A, Office #1, Moscow 119049, Russia; (A.L.); (A.T.); (T.G.); (O.K.); (A.K.); (E.V.); (S.A.); (V.M.); (M.I.)
| | - Egor Veselovsky
- OncoAtlas LLC, Leninskiy Prospekt, 4c1A, Office #1, Moscow 119049, Russia; (A.L.); (A.T.); (T.G.); (O.K.); (A.K.); (E.V.); (S.A.); (V.M.); (M.I.)
| | - Vladislav Nikulin
- Federal State Budgetary Institution N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye Highway, 23, Moscow 115478, Russia; (A.S.); (V.N.); (A.T.); (M.F.)
| | - Saida Aliyarova
- OncoAtlas LLC, Leninskiy Prospekt, 4c1A, Office #1, Moscow 119049, Russia; (A.L.); (A.T.); (T.G.); (O.K.); (A.K.); (E.V.); (S.A.); (V.M.); (M.I.)
| | - Laima Belyaeva
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Trubetskaya Ulitsa, 8/2, Moscow 119048, Russia;
| | - Alexey Tryakin
- Federal State Budgetary Institution N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye Highway, 23, Moscow 115478, Russia; (A.S.); (V.N.); (A.T.); (M.F.)
| | - Mikhail Fedyanin
- Federal State Budgetary Institution N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye Highway, 23, Moscow 115478, Russia; (A.S.); (V.N.); (A.T.); (M.F.)
- State Budgetary Institution of Health Care of the City of Moscow, Moscow Multidisciplinary Clinical Center “Kommunarka”, Department of Health of the City of Moscow, Ulitsa Sosenskiy Stan, 8c3, Moscow 142770, Russia;
- Department of Oncology, Pirogov Russian National Research Medical University, Ostrovityanova Ulitsa, 1, Moscow 117997, Russia
| | - Vladislav Mileyko
- OncoAtlas LLC, Leninskiy Prospekt, 4c1A, Office #1, Moscow 119049, Russia; (A.L.); (A.T.); (T.G.); (O.K.); (A.K.); (E.V.); (S.A.); (V.M.); (M.I.)
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Trubetskaya Ulitsa, 8/2, Moscow 119048, Russia;
| | - Maxim Ivanov
- OncoAtlas LLC, Leninskiy Prospekt, 4c1A, Office #1, Moscow 119049, Russia; (A.L.); (A.T.); (T.G.); (O.K.); (A.K.); (E.V.); (S.A.); (V.M.); (M.I.)
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Trubetskaya Ulitsa, 8/2, Moscow 119048, Russia;
| |
Collapse
|
22
|
Jiang X, Zhai W, Song J, Shao W, Zhang A, Duan S, Qu F, Cheng W, Luo C, Wu F, Liu X, Chen T. Associations between MRI radiomic phenotypes and clinical outcomes in endometrial cancer: Implications for preoperative risk stratification. Magn Reson Imaging 2025; 117:110298. [PMID: 39645007 DOI: 10.1016/j.mri.2024.110298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/20/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVES This study aimed to investigate the correlation between imaging phenotypes of endometrial cancer (EC) and clinical, pathologic, and molecular characteristics, as well as disease-free survival (DFS). METHODS The clinical, pathologic, and molecular characteristics, along with MRI radiomics features, of 356 patients with EC were collected retrospectively. The patients were divided into 2 groups based on radiomics features using unsupervised machine learning. The obtained characteristics and DFS of patients were compared between the various imaging phenotypes. RESULTS The lesions with deep myometrial invasion (DMI), lymphovascular space invasion (LVSI), cervical stromal invasion (CSI), lymph node metastasis, aggressive histologic type, advanced postoperative International Federation of Gynecology and Obstetrics (FIGO) stage, overexpression of p53, and absent expression of estrogen receptor or progesterone receptor were associated with poor DFS. Two clusters were identified and defined as imaging phenotype 1 and 2, respectively. Compared with phenotype 2, phenotype 1 exhibited a higher correlation with DMI (33.7 % vs 13.0 %), LVSI (23.8 % vs 9.2 %), CSI (16.3 % vs 3.8 %), aggressive histologic type (36.0 % vs 17.4 %), and advanced FIGO stage (IB or higher, 43.6 % vs 22.3 %) (p < 0.001). The incidence of p53 overexpression was higher in phenotype 1 than in phenotype 2 (20.2 % vs 8.5 %, p = 0.022). Survival analysis exhibited a higher risk of poor DFS in phenotype 1 than in phenotype 2 (log-rank p = 0.002). CONCLUSION EC imaging phenotypes identified through MRI radiomics features were associated with pathologic, molecular characteristics, and DFS, suggesting potential for preoperative risk stratification.
Collapse
Affiliation(s)
- Xiaoting Jiang
- Department of Radiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Weiling Zhai
- Department of Radiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Jiacheng Song
- Department of Radiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wenhui Shao
- Department of Radiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Aining Zhang
- Department of Radiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Shaofeng Duan
- Central Research Institute, UIH Group, Shanghai, China
| | - Feifei Qu
- MR Research Collaboration, Siemens Healthineers, Shanghai, China
| | - Wenjun Cheng
- Department of Gynecology and Obstetrics, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Chengyan Luo
- Department of Gynecology and Obstetrics, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Feiyun Wu
- Department of Radiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Xisheng Liu
- Department of Radiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| | - Ting Chen
- Department of Radiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
23
|
Schneider D, Brown EDL, Gluski J, Mishra A, Shah HA, Sciubba DM, Lo SFL. Subtype-Specific Patterns of Tumor Purity and Mutation Load Suggest Treatment Implications: A Cross-Sectional Analysis of 7494 Soft Tissue and Bone Sarcomas (MSK Cohort). Am J Clin Oncol 2025; 48:185-192. [PMID: 40085522 DOI: 10.1097/coc.0000000000001161] [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: 03/16/2025]
Abstract
OBJECTIVES Sarcomas are complex mesenchymal malignancies whose molecular characteristics can significantly influence treatment strategies. This study aimed to investigate the relationship between tumor purity, mutation load, and clinical characteristics across sarcoma subtypes, focusing on potential implications for therapeutic stratification. METHODS This study analyzed the molecular characteristics of 7494 sarcoma cases from the Soft Tissue and Bone Sarcoma (MSK, Nat Commun 2022) data set using available case analysis. Correlations between tumor purity, mutation load, age, and sex were analyzed using nonparametric methods, with subtype-specific analyses conducted using Kruskal-Wallis tests and Bonferroni-corrected post hoc comparisons. A comprehensive analysis of mutation patterns was performed using microsatellite instability (MSI) status. RESULTS Significant correlations between mutation load and tumor purity (ρ=0.320, P <0.001) were identified, with marked heterogeneity across subtypes. Tumor purity ranged from 20.0% in brain sarcomas to 78.5% in dermatofibrosarcoma protuberans. Age-related molecular changes were observed in brain (ρ=0.711, P =0.006) and skin sarcomas (ρ=0.450, P =0.006), suggesting distinct evolutionary patterns. A subset of hypermutated, microsatellite stable cases (0.15%) with mutation loads exceeding 100 mutations/mb were identified, suggesting alternative mechanisms of genomic instability. MSI-high status was rare (0.24%) but associated with higher mutation loads (median: 25.84 vs. 2.42, P <0.001), particularly in uterine sarcomas (0.7% prevalence). CONCLUSIONS The identification of distinct molecular patterns across sarcoma subtypes challenge existing morphology-based classification systems and may hold implications for therapeutic stratification. These findings may help inform future immunotherapeutic and molecular-guided approaches to treatment in sarcoma patients, particularly for elderly patients with brain sarcomas or females with uterine sarcomas.
Collapse
Affiliation(s)
- Daniel Schneider
- Department of Neurosurgery, Donald and Barbara Zucker Hofstra School of Medicine at Northwell, Manhasset, NY
| | | | | | | | | | | | | |
Collapse
|
24
|
Obermair A, Gebski V, Goh J, Kuchel A, Brand A, Mak B, McNally O, Baxter E, Jobling T, Mileshkin L. Phase 2b, open-label, single-arm, multicenter pilot study of the efficacy, safety, and tolerability of dostarlimab in women with early-stage mismatch repair-deficient endometrioid endometrial adenocarcinoma. Int J Gynecol Cancer 2025; 35:101644. [PMID: 39955187 DOI: 10.1016/j.ijgc.2025.101644] [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: 11/04/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND The standard treatment for endometrial cancer is hysterectomy with or without bilateral salpingo-oophorectomy; however, this may not be an optimal choice for women who have not completed childbearing or who are at a high risk of surgical complications. Conservative treatment with levonorgestrel intrauterine devices appear to be effective in patients with early-stage endometrial cancer; however, patients with mismatch repair-deficient (dMMR) tumors have a low likelihood of responding to levonorgestrel intrauterine devices. PRIMARY OBJECTIVE To assess the efficacy of dostarlimab, an active immune checkpoint inhibitor that targets the programmed cell death protein-1 receptor, in patients with early-stage dMMR endometrioid endometrial adenocarcinoma. STUDY HYPOTHESIS Administration of 4 3-weekly cycles of 500 mg dostarlimab followed by a 3-week rest period and 3 6-weekly cycles of 1000 mg dostarlimab will be safe and efficacious in early-stage dMMR endometrial cancer patients. TRIAL DESIGN Non-randomized, open-label, pilot, multicenter phase2b study designed to evaluate the efficacy and safety of dostarlimab in 10 women aged ≥18 years with a clinically confirmed diagnosis of early-stage and dMMR endometrioid endometrial adenocarcinoma. MAJOR INCLUSION/EXCLUSION CRITERIA Eligible patients must have histologically proven stage I, International Federation of Gynecology and Obstetrics grade 1 or 2 dMMR endometrioid endometrial adenocarcinoma and desire for fertility preservation. Exclusions include, but are not limited to, patients with other high-risk endometrial cancer cell types, a poor medical risk due to uncontrolled medical conditions, or those who experienced grade 3 or higher immune-related adverse events from prior immunotherapy. PRIMARY ENDPOINT(S) The primary endpoint is the number of participants achieving investigator-assessed pathological complete response within 6 months of treatment. SAMPLE SIZE Ten (10) women ≥18 years of age will be enrolled. ESTIMATED DATES FOR COMPLETING ACCRUAL AND PRESENTING RESULTS Accruals are expected to be completed by 2027, with the presentation of results by 2029. TRIAL REGISTRATION NCT06278857.
Collapse
Affiliation(s)
- Andreas Obermair
- Royal Brisbane and Women's Hospital Campus, UQ Centre for Clinical Research, The University of Queensland, Queensland Centre for Gynaecological Cancer (QCGC) Research, Herston, QLD, Australia.
| | - Val Gebski
- The University of Sydney, Faculty of Medicine and Health, NHMRC Clinical Trials Centre, Camperdown, NSW, Australia
| | - Jeffrey Goh
- Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Anna Kuchel
- Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, QLD, Australia; University of Queensland, The School of Medicine, Herston, QLD, Australia
| | - Alison Brand
- Westmead Hospital, The Crown Princess Mary Cancer Centre, Westmead, NSW, Australia; Westmead Centre for Gynaecological Oncology, The University of Sydney, Camperdown, NSW, Australia
| | - Blossom Mak
- The University of Sydney, Faculty of Medicine and Health, NHMRC Clinical Trials Centre, Camperdown, NSW, Australia; Westmead Hospital, The Crown Princess Mary Cancer Centre, Westmead, NSW, Australia
| | - Orla McNally
- Oncology and Dysplasia Service, The Royal Women's Hospital, Parkville, VIC, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Eva Baxter
- Royal Brisbane and Women's Hospital Campus, UQ Centre for Clinical Research, The University of Queensland, Queensland Centre for Gynaecological Cancer (QCGC) Research, Herston, QLD, Australia
| | - Thomas Jobling
- Monash University, Department of Obstetrics and Gynaecology, Clayton, VIC, Australia; Monash Health, Women's and Newborn Program, Clayton, VIC, Australia
| | - Linda Mileshkin
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
25
|
Wu YT, Gao M, Cheng KY, Li L, Wang BQ, He YN, Zhang Y, Liu XY, Du RL, Li GQ, Liang YX, Zhang JF, Zhang XD, Liu Y. VRK2 promotes colorectal cancer growth and impedes immunotherapy and 5-FU treatment efficacy. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167729. [PMID: 39978443 DOI: 10.1016/j.bbadis.2025.167729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/31/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
Vaccinia-Related Kinase 2 (VRK2), a member of the vaccinia virus-related protein kinase family, is crucial in regulating apoptosis and tumor cell growth signaling pathways. Despite its established roles in various cancers, investigations into its functions in colorectal cancer have been relatively limited. Utilizing The Cancer Genome Atlas and Genotype-Tissue Expression databases, this study assesses VRK2 expression across 33 cancer types, highlighting significant upregulation and diagnostic relevance, particularly in colorectal cancer, where it marks poor prognosis. VRK2's influence extends across multiple cancer-related signaling pathways, with focused experiments confirming its vital role in the E2F signaling pathway through transcriptomic sequencing and dual-luciferase reporter assays. Deletion of VRK2 markedly inhibits proliferation, cell cycle progression, migration, and tumorigenesis in colorectal cancer cells, whereas overexpression enhances these oncogenic traits. Additionally, VRK2 expression correlates with genomic instability and the tumor microenvironment, influencing antitumor immunity and response to immunotherapy. Importantly, our analysis reveals that VRK2 modulates the chemosensitivity of tumor cells, specifically enhancing resistance to the chemotherapeutic agent 5-FU. These findings underscore VRK2's multifaceted role in promoting colorectal cancer development and suggest its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yu-Tong Wu
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Meng Gao
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Kun-Yang Cheng
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Le Li
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Bai-Qi Wang
- Department of Radiation Oncology, The Second Affiliated Hospital University of South China Clinical Research Center, For Prevention and Treatment of Breast & Thyroid Disease In Hunan Province, Hengyang, Hunan, China
| | - Ya-Nan He
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Yue Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Xue-Yi Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Run-Lei Du
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Guo-Qing Li
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yue-Xiu Liang
- Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions & Department of Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Jian-Feng Zhang
- Xuancheng Institutes of Food and Drug Control, Xuancheng, China
| | - Xiao-Dong Zhang
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Yi Liu
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
26
|
Helderman NC, Strobel F, Bohaumilitzky L, Terlouw D, van der Werf-'t Lam AS, van Wezel T, Morreau H, von Knebel Doeberitz M, Nielsen M, Kloor M, Ahadova A. Lower Degree of Microsatellite Instability in Colorectal Carcinomas From MSH6-Associated Lynch Syndrome Patients. Mod Pathol 2025; 38:100757. [PMID: 40118460 DOI: 10.1016/j.modpat.2025.100757] [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: 08/12/2024] [Revised: 03/01/2025] [Accepted: 03/09/2025] [Indexed: 03/23/2025]
Abstract
Numerous observational and molecular studies focusing on Lynch syndrome (LS) have revealed significant variation in the phenotype and molecular characteristics among carriers of pathogenic variants in mismatch repair genes (path_MMR). Recently, we demonstrated that colorectal carcinomas in path_MSH6 carriers exhibit fewer insertion/deletion mutations compared with cumulative colorectal cancers (CRCs) from other MMR groups, raising the question of whether MSH6-mutated CRCs might display a relatively lower degree of microsatellite instability (MSI). Mutations at 20 coding microsatellites (cMS) were analyzed in 39 MSH6-, 18 MLH1-, 16 MSH2-, and 22 PMS2-mutated CRCs and 35 sporadic MSI CRCs, and mutation frequencies and mutant allele ratios were compared among the different MMR-deficient groups. Considering factors such as HLA-A∗02:01 type, B2M status, and the anticipated immunogenicity of frameshift peptides derived from cMS mutations, the identified cMS mutation profiles of MSH6-mutated CRCs were further investigated to assess their potential impact on immunotherapeutic strategies. MSH6-mutated CRCs exhibited lower mutation frequencies and mutant allele ratios across most cMS. Variation in cMS mutation patterns was observed both between different tumor regions and between tumor tissue and adjacent adenomatous tissue. The cMS mutations in MSH6-mutated CRCs demonstrated inverse correlations with the predicted immunogenicity of the resulting frameshift peptides, which may suggest a negative selection of cell clones bearing highly immunogenic frameshift peptides. Overall, MSH6-mutated CRCs display a relatively lower degree of MSI and represent a biologically distinct subgroup of LS-associated CRCs. This lower MSI level may implicate an altered immune response compared with other MSI CRCs, which could have theoretical implications for the success of immunotherapy in MSH6-mutated CRCs. Future studies should carefully evaluate this possibility. If confirmed, these results would reinforce the notion of classifying LS as distinct syndromes associated with specific MMR genes.
Collapse
Affiliation(s)
- Noah C Helderman
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Fabian Strobel
- Department of Applied Tumor Biology, Heidelberg University Hospital, Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Lena Bohaumilitzky
- Department of Applied Tumor Biology, Heidelberg University Hospital, Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Diantha Terlouw
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Tom van Wezel
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Magnus von Knebel Doeberitz
- Department of Applied Tumor Biology, Heidelberg University Hospital, Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Maartje Nielsen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Matthias Kloor
- Department of Applied Tumor Biology, Heidelberg University Hospital, Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Aysel Ahadova
- Department of Applied Tumor Biology, Heidelberg University Hospital, Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Centre (DKFZ), Heidelberg, Germany.
| |
Collapse
|
27
|
Cereda V, D’Andrea MR. Pancreatic cancer: failures and hopes-a review of new promising treatment approaches. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002299. [PMID: 40124650 PMCID: PMC11926728 DOI: 10.37349/etat.2025.1002299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/22/2025] [Indexed: 03/25/2025] Open
Abstract
Pancreatic cancer is a challenging disease with limited treatment options and a high mortality rate. Just few therapy advances have been made in recent years. Tumor microenvironment, immunosuppressive features and mutational status represent important obstacles in the improvement of survival outcomes. Up to now, first-line therapy did achieve a median overall survival of less than 12 months and this discouraging data lead clinicians all over the world to focus their efforts on various fields of investigation: 1) sequential cycling of different systemic therapy in order to overcome mechanisms of resistance; 2) discovery of new predictive bio-markers, in order to target specific patient population; 3) combination treatment, in order to modulate the tumor microenvironment of pancreatic cancer; 4) new modalities of the delivery of drugs in order to pass the physical barrier of desmoplasia and tumor stroma. This review shows future directions of treatment strategies in advanced pancreatic cancer through a deep analysis of these recent macro areas of research.
Collapse
Affiliation(s)
- Vittore Cereda
- Asl Roma 4, Hospital S. Paolo Civitavecchia, 00053 Civitavecchia, Italy
| | | |
Collapse
|
28
|
Zhang Z, Luo T, Yan M, Shen H, Tao K, Zeng J, Yuan J, Fang M, Zheng J, Bermejo I, Dekker A, Ruysscher DD, Wee L, Zhang W, Jiang Y, Ji Y. Voxel-level radiomics and deep learning for predicting pathologic complete response in esophageal squamous cell carcinoma after neoadjuvant immunotherapy and chemotherapy. J Immunother Cancer 2025; 13:e011149. [PMID: 40090670 PMCID: PMC11911808 DOI: 10.1136/jitc-2024-011149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/04/2025] [Indexed: 03/18/2025] Open
Abstract
BACKGROUND Accurate prediction of pathologic complete response (pCR) following neoadjuvant immunotherapy combined with chemotherapy (nICT) is crucial for tailoring patient care in esophageal squamous cell carcinoma (ESCC). This study aimed to develop and validate a deep learning model using a novel voxel-level radiomics approach to predict pCR based on preoperative CT images. METHODS In this multicenter, retrospective study, 741 patients with ESCC who underwent nICT followed by radical esophagectomy were enrolled from three institutions. Patients from one center were divided into a training set (469 patients) and an internal validation set (118 patients) while the data from the other two centers was used as external validation sets (120 and 34 patients, respectively). The deep learning model, Vision-Mamba, integrated voxel-level radiomics feature maps and CT images for pCR prediction. Additionally, other commonly used deep learning models, including 3D-ResNet and Vision Transformer, as well as traditional radiomics methods, were developed for comparison. Model performance was evaluated using accuracy, area under the curve (AUC), sensitivity, specificity, and prognostic stratification capabilities. The SHapley Additive exPlanations analysis was employed to interpret the model's predictions. RESULTS The Vision-Mamba model demonstrated robust predictive performance in the training set (accuracy: 0.89, AUC: 0.91, sensitivity: 0.82, specificity: 0.92) and validation sets (accuracy: 0.83-0.91, AUC: 0.83-0.92, sensitivity: 0.73-0.94, specificity: 0.84-1.0). The model outperformed other deep learning models and traditional radiomics methods. The model's ability to stratify patients into high and low-risk groups was validated, showing superior prognostic stratification compared with traditional methods. SHAP provided quantitative and visual model interpretation. CONCLUSIONS We present a voxel-level radiomics-based deep learning model to predict pCR to neoadjuvant immunotherapy combined with chemotherapy based on pretreatment diagnostic CT images with high accuracy and robustness. This model could provide a promising tool for individualized management of patients with ESCC.
Collapse
Affiliation(s)
- Zhen Zhang
- Zhejiang Cancer Hospital,Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Department of Radiation Oncology (Maastro),GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | - Meng Yan
- Department of Radiation Oncology (Maastro),GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Haixia Shen
- Zhejiang Cancer Hospital,Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Kaiyi Tao
- Zhejiang Cancer Hospital,Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jian Zeng
- Zhejiang Cancer Hospital,Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Min Fang
- Zhejiang Cancer Hospital,Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jian Zheng
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | | | - Andre Dekker
- Department of Radiation Oncology (Maastro),GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (Maastro),GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Leonard Wee
- Department of Radiation Oncology (Maastro),GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Wencheng Zhang
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Youhua Jiang
- Zhejiang Cancer Hospital,Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Yongling Ji
- Zhejiang Cancer Hospital,Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Prevention Diagnosis and Therapy for Gastrointestinal Cancer, Hangzhou, Zhejiang, China
| |
Collapse
|
29
|
Alsina Maqueda M, Teijo Quintáns A, Cuatrecasas M, Fernández Aceñero MJ, Fernández Montes A, Gómez Martín C, Jiménez Fonseca P, Martínez Ciarpaglini C, Rivera Herrero F, Iglesias Coma M. Biomarkers in gastroesophageal cancer 2025: an updated consensus statement by the Spanish Society of Medical Oncology (SEOM) and the Spanish Society of Pathology (SEAP). Clin Transl Oncol 2025:10.1007/s12094-025-03865-6. [PMID: 40072752 DOI: 10.1007/s12094-025-03865-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 03/14/2025]
Abstract
Gastroesophageal carcinomas, including gastroesophageal adenocarcinoma (GEA) and esophageal squamous cell carcinoma (ESCC), pose a global health challenge due to their heterogeneity. The approach to diagnosis and treatment should first differentiate between GEA and ESCC. Over the past decade, therapies for metastatic or advanced GEA/ESCC have expanded, with several new therapeutic targets alongside trastuzumab for metastatic HER2-positive GEA. Four key biomarkers are essential for targeted therapy: HER2 overexpression/amplification, deficient mismatch repair/microsatellite instability (dMMR/MSI), PD-L1, and Claudin18.2 expression. Immunohistochemistry is the recommended method for these biomarkers evaluation. In addition, the assessment of biomarkers like FGFR2b is likely to become routine in the near future. Experts from the Spanish Society of Pathology (SEAP) and the Spanish Society of Medical Oncology (SEOM) have formed a consensus to optimize biomarker detection and usage in clinical practice. Their recommendations aim to improve personalized treatment strategies for GEA and ESCC patients, integrating new diagnostic insights into routine care.
Collapse
Affiliation(s)
- Maria Alsina Maqueda
- Medical Oncology Department, Unidad de Oncología Médica Traslacional, Hospital Universitario de Navarra, Navarrabiomed - Centro de Investigación Sanitaria de Navarra, Pamplona, Spain.
| | - Ana Teijo Quintáns
- Pathology Department, Gastrointestinal and Neuroendocrine Tumors Research Group, Hospital Universitario 12 de Octubre, Research Institute (Imas12), Madrid, Spain
| | - Miriam Cuatrecasas
- Pathology Department, Hospital Clinic de Barcelona, Biomedical Research Institute IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Maria Jesús Fernández Aceñero
- Department of Legal Medicine, Psychiatry and Pathology, Surgical Pathology Department, Hospital Clínico Universitario San Carlos, nstituto de Investigación Sanitaria Clínico San Carlos (IdISSC), Universidad Complutense, Madrid, Spain
- , Madrid, Spain
| | - Ana Fernández Montes
- Medical Oncology Department, Complexo Hospitalario Universitario de Ourense, Ourense, Spain
| | - Carlos Gómez Martín
- Gastrointestinal Cancer and Early Clinical-Translational Research Units, Medical Oncology Division, 12 de Octubre University Hospital, Madrid, Spain
| | - Paula Jiménez Fonseca
- Department of Medical Oncology, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Carolina Martínez Ciarpaglini
- Pathology Department, Hospital Clínico Universitario de Valencia, Biomedical Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | - Fernando Rivera Herrero
- Medical Oncology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Mar Iglesias Coma
- Pathology Department, Hospital del Mar, Pompeu Fabra University, Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
30
|
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
|
31
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
32
|
de la Fouchardière C, Cammarota A, Svrcek M, Alsina M, Fleitas-Kanonnikoff T, Lordick Obermannová R, Wagner AD, Yap Wei Ting D, Enea D, Petrillo A, Smyth EC. How do I treat dMMR/MSI gastro-oesophageal adenocarcinoma in 2025? A position paper from the EORTC-GITCG gastro-esophageal task force. Cancer Treat Rev 2025; 134:102890. [PMID: 39933210 DOI: 10.1016/j.ctrv.2025.102890] [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: 12/16/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
In less than a decade, immune checkpoint inhibitors (ICIs) have transformed the management of mismatch repair-deficient (dMMR) and microsatellite instability-high (MSI) cancers. However, beyond colorectal cancer (CRC), much of the evidence is mostly derived from non-randomized phase II studies or post-hoc analyses of broader clinical trials. dMMR/MSI tumours represent a specific subgroup of gastro-esophageal adenocarcinomas (GEA), accounting for approximately 9 % of cases, with a higher prevalence in early-stage compared to advanced-stage disease and older female patients. These tumours are predominantly sporadic, often linked to MLH1 promoter methylation, and rarely exhibit HER2 overexpression/ERBB2 amplification or other oncogenic drivers. The treatment landscape for early stage dMMR/MSI GEA is likely to change substantially soon, as ICIs have shown high pathological complete response (pCR) rates in small phase II trials, raising questions on optimisation of neoadjuvant therapy, and paving the way for organ preservation. The standard of treatment for untreated patients with advanced dMMR/MSI GEA is chemotherapy + ICI irrespectively of PDL-1 status. However, the role of chemotherapy-free regimen consisting of CTLA-4 plus PD-1 inhibitors remains undetermined. This review addresses these and other emerging questions, offering expert opinions and insights into the future therapeutic landscape for dMMR/MSI GEA.
Collapse
Affiliation(s)
- Christelle de la Fouchardière
- Institut PAOLI-CALMETTES, 232 Boulevard Sainte-Marguerite 13009, Marseille, France; Unicancer GI (UCGI) Group, Paris, France; EORTC-GITC Group, Brussels, Belgium.
| | - Antonella Cammarota
- EORTC-GITC Group, Brussels, Belgium; Hepatobiliary Immunopathology Lab, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Magali Svrcek
- Sorbonne Université, AP-HP, Saint-Antoine Hospital, Department of Pathology, France; LIMICS, UMRS 1142, Campus des Cordeliers 75006, Paris, France
| | - Maria Alsina
- EORTC-GITC Group, Brussels, Belgium; Hospital Universitario de Navarra, Navarrabiomed - IdiSNA, c. de Irunlarrea 3 31008, Pamplona, Spain
| | - Tania Fleitas-Kanonnikoff
- EORTC-GITC Group, Brussels, Belgium; Hospital Clínico Universitario de Valencia, INCLIVA, Valencia, Spain
| | - Radka Lordick Obermannová
- EORTC-GITC Group, Brussels, Belgium; Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute and Faculty of Medicine, Masaryk University, Czech Republic
| | - Anna Dorothea Wagner
- EORTC-GITC Group, Brussels, Belgium; Anna Dorothea Wagner, Department of Oncology, Division of Medical Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), 1011, Lausanne, Switzerland
| | | | - Diana Enea
- Sorbonne Université, AP-HP, Saint-Antoine Hospital, Department of Pathology, France
| | - Angelica Petrillo
- EORTC-GITC Group, Brussels, Belgium; Medical Oncology Unit, Ospedale del Mare, Naples, Italy
| | - Elizabeth C Smyth
- EORTC-GITC Group, Brussels, Belgium; Oxford NIHRBiomedical Research Centre, Churchill Hospital, Oxford OX3 7LE, UK
| |
Collapse
|
33
|
Wang Y, He K, Zhang Y, Chen Y, Wang S, Zhao K, Liu Z, Hu M. Peptide-based immuno-PET/CT monitoring of dynamic PD-L1 expression during glioblastoma radiotherapy. J Pharm Anal 2025; 15:101082. [PMID: 40177067 PMCID: PMC11964630 DOI: 10.1016/j.jpha.2024.101082] [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: 04/08/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 04/05/2025] Open
Abstract
Real-time, noninvasive programmed death-ligand 1 (PD-L1) testing using molecular imaging has enhanced our understanding of the immune environments of neoplasms and has served as a guide for immunotherapy. However, the utilization of radiotracers in the imaging of human brain tumors using positron emission tomography/computed tomography (PET/CT) remains limited. This investigation involved the synthesis of [18F]AlF-NOTA-PCP2, which is a novel peptide-based radiolabeled tracer that targets PD-L1, and evaluated its imaging capabilities in orthotopic glioblastoma (GBM) models. Using this tracer, we could noninvasively monitor radiation-induced PD-L1 changes in GBM. [18F]AlF-NOTA-PCP2 exhibited high radiochemical purity (>95%) and stability up to 4 h after synthesis. It demonstrated specific, high-affinity binding to PD-L1 in vitro and in vivo, with a dissociation constant of 0.24 nM. PET/CT imaging, integrated with contrast-enhanced magnetic resonance imaging, revealed significant accumulation of [18F]AlF-NOTA-PCP2 in orthotopic tumors, correlating with blood-brain barrier disruption. After radiotherapy (15 Gy), [18F]AlF-NOTA-PCP2 uptake in tumors increased from 9.51% ± 0.73% to 12.04% ± 1.43%, indicating enhanced PD-L1 expression consistent with immunohistochemistry findings. Fractionated radiation (5 Gy × 3) further amplified PD-L1 upregulation (13.9% ± 1.54% ID/cc) compared with a single dose (11.48% ± 1.05% ID/cc). Taken together, [18F]AlF-NOTA-PCP2 may be a valuable tool for noninvasively monitoring PD-L1 expression in brain tumors after radiotherapy.
Collapse
Affiliation(s)
- Yong Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Kewen He
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Yang Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Yunhao Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Shijie Wang
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Kunlong Zhao
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Zhiguo Liu
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Man Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| |
Collapse
|
34
|
Abu-Freha N, Hozaeel W, Weissmann S, Lerner Z, Ben-Shoshan L, Ganayem R, Katz LH. Lynch Syndrome: Similarities and Differences of Recommendations in Published Guidelines. J Gastroenterol Hepatol 2025; 40:564-573. [PMID: 39797698 DOI: 10.1111/jgh.16881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/23/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
BACKGROUND In this review, we aimed to compare the recommendations for Lynch syndrome (LS). METHODS We compared the LS's guidelines of different medical societies, including recommendations for cancer surveillance, aspirin treatment, and universal screening. RESULTS Most guidelines for LS patients recommend intervals of 1-2 years for performing colonoscopy, though there is disagreement regarding the age to begin CRC screening (dependent on status as a MLH1/MSH2 or MSH6/PMS2 carrier). There are inconsistencies between LS guidelines for gastric cancer surveillance. Most guidelines do not recommend routine surveillance of the pancreas and small bowel. Most but not all of the guidelines support endometrial and ovarian surveillance with transvaginal ultrasound and endometrial biopsy. Only two societies recommend urological surveillance, while others recommend surveillance among high-risk carriers with family history only. There is significant disagreement between the guidelines about the recommendation for limited or extended bowel resection among patients with CRC. Aspirin use is recommended by most societies, though some with reservations, and most of them recommend universal screening. CONCLUSIONS There are significant disparities and disagreements in the guidelines and recommendations for patients with LS, causing confusion and difficulties for clinicians. Harmonization and cooperation are needed between the societies creating LS guidelines.
Collapse
Affiliation(s)
- Naim Abu-Freha
- The Institute of Gastroenterology and Hepatology, Soroka University Medical Center, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Wael Hozaeel
- The Legacy Heritage Oncology Center & Dr Larry Norton Institute, Soroka Medical Center & ben-Gurion University, Beer-Sheva, Israel
| | - Sarah Weissmann
- Clinical Research Center, Soroka University Medical Center, Beer-Sheva, Israel
| | - Zlata Lerner
- The Institute of Gastroenterology and Hepatology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Liza Ben-Shoshan
- The Institute of Gastroenterology and Hepatology, Soroka University Medical Center, Beer-Sheva, Israel
| | | | - Lior H Katz
- Department of Gastroenterology and Hepatology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
35
|
DeWitt JT, Jimenez-Tovar D, Mazumder A, Haricharan S. Advances in diagnostic and therapeutic applications of mismatch repair loss in cancer. DNA Repair (Amst) 2025; 147:103822. [PMID: 40068557 DOI: 10.1016/j.dnarep.2025.103822] [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: 01/02/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/17/2025]
Abstract
Mismatch repair (MMR) is a highly conserved, fundamental DNA damage repair pathway that maintains genomic fidelity during cell replication. MMR dysregulation contributes to tumor formation by promoting genomic instability thereby increasing the frequency of potentially oncogenic mutational events. Therefore, MMR dysregulation, in its tumor suppressor role, is largely studied in the context of genomic instability and associated response to immune checkpoint blockade therapies. However, a growing body of literature suggests that the impact of MMR dysregulation on tumor phenotypes is more nuanced than a concerted impact on genomic stability. Rather, loss of individual MMR genes promotes distinct cancer-relevant biological phenotypes, and these phenotypes are further modulated by the tissue of tumor origin. Here, we explore relevant literature and review the prognostic and predictive significance of these non-canonical discoveries.
Collapse
Affiliation(s)
- J T DeWitt
- Dept of Biology, San Diego State University, San Diego, CA, USA; Cancer Biology and Signaling Program, UCSD Moores Cancer Center, San Diego, CA, USA
| | - D Jimenez-Tovar
- Dept of Biology, San Diego State University, San Diego, CA, USA
| | - A Mazumder
- Dept of Biology, San Diego State University, San Diego, CA, USA
| | - S Haricharan
- Dept of Biology, San Diego State University, San Diego, CA, USA; Cancer Biology and Signaling Program, UCSD Moores Cancer Center, San Diego, CA, USA.
| |
Collapse
|
36
|
Jantus-Lewintre E, Rappa A, Ruano D, van Egmond D, Gallach S, Gozuyasli D, Durães C, Costa JL, Camps C, Lacroix L, Kashofer K, van Wezel T, Barberis M. Multicenter In-House Evaluation of an Amplicon-Based Next-Generation Sequencing Panel for Comprehensive Molecular Profiling. Mol Diagn Ther 2025; 29:249-261. [PMID: 39798063 PMCID: PMC11860996 DOI: 10.1007/s40291-024-00766-2] [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: 12/09/2024] [Indexed: 01/13/2025]
Abstract
BACKGROUND Predicting response to targeted cancer therapies increasingly relies on both simple and complex genetic biomarkers. Comprehensive genomic profiling using high-throughput assays must be evaluated for reproducibility and accuracy compared with existing methods. METHODS This study is a multicenter evaluation of the Oncomine™ Comprehensive Assay Plus (OCA Plus) Pan-Cancer Research Panel for comprehensive genomic profiling of solid tumors. A series of 193 research samples (125 DNA and 68 RNA samples) was analyzed to evaluate the correlation and concordance of the OCA Plus panel with orthogonal methods, as well as its reproducibility (n = 5 DNA samples) across laboratories. RESULTS The success rate for DNA and RNA sequencing was 96.6% and 89.7%, respectively. In a single workflow, the OCA Plus panel provided a detailed genomic profile with a high success rate for all biomarkers tested: single nucleotide variants/indels, copy number variants, and fusions, as well as complex biomarkers such as microsatellite instability, tumor mutational burden, and homologous recombination deficiency. The concordance for single nucleotide variants/indels was 94.8%, for copy number variants 96.5%, for fusions 94.2%, for microsatellite instability 80.8%, for tumor mutational burden 81.3%, and for homologous recombination deficiency 100%. The results showed high reproducibility across the five European research centers, each analyzing shared pre-characterized tissue biopsies (average of 1890 single nucleotide variants/indels per sample). CONCLUSIONS This multicenter evaluation of the OCA Plus panel confirms the results of previous single-center studies and demonstrates the high reproducibility and accuracy of this assay.
Collapse
Affiliation(s)
- Eloisa Jantus-Lewintre
- Fundación Investigación Hospital General Universitario de València, Universitat Politècnica de València, CIBERONC, Valencia, Spain
| | - Alessandra Rappa
- Istituto Europeo di Oncologia, IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Dina Ruano
- Leiden University Medical Center, Leiden, The Netherlands
| | | | - Sandra Gallach
- Fundación Investigación Hospital General Universitario de València, Universitat Politècnica de València, CIBERONC, Valencia, Spain
| | | | | | | | - Carlos Camps
- Consorcio Hospital General Universitario de València, Universitat de València, CIBERONC, Valencia, Spain
| | | | | | - Tom van Wezel
- Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Massimo Barberis
- Istituto Europeo di Oncologia, IRCCS, Via Adamello 16, 20139, Milan, Italy.
| |
Collapse
|
37
|
Michálková R, Šafanda A, Hájková N, Hojný J, Krkavcová E, Kendall Bártů M, Švajdler M, Shatokhina T, Laco J, Matěj R, Méhes G, Hausnerová J, Škarda J, Hácová M, Náležinská M, Zima T, Dundr P, Němejcová K. The Molecular Landscape of 227 Adult Granulosa Cell Tumors of the Ovary: Insights into the Progression from Primary to Recurrence. J Transl Med 2025; 105:102201. [PMID: 39615884 DOI: 10.1016/j.labinv.2024.102201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/04/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Adult granulosa cell tumors (AGCTs) of the ovary are characterized by their propensity for late recurrences and are primarily managed surgically due to the limited efficacy of systemic treatment. The FOXL2 p.C134W somatic mutation has been identified in ∼95% of AGCT cases, and TERT promoter alterations have been linked to worse overall survival. This study highlights the potential prognostic significance of FOXO1 mutations, suggesting that they may be associated with poorer overall survival and shorter time to recurrence. A total of 183 primary AGCTs and 44 recurrences without corresponding primary tumors were analyzed. The primary AGCTs were categorized into 3 groups: 77 nonrecurrent tumors, 18 tumors that later recurred (including 9 cases with matched primary-recurrence pairs), and 88 tumors with unknown recurrence status. Targeted next-generation sequencing was conducted on 786 cancer-related genes to investigate their genetic profile. The study aimed to identify the molecular alterations associated with AGCT pathogenesis and recurrence rate, comparing primary versus recurrent tumors, and primary recurrent versus primary nonrecurrent cases. Our findings confirmed the high prevalence (99%) of the FOXL2 p.C134W mutation in AGCTs. Secondary truncating FOXL2 mutations were observed in 5% of cases. Two cases with typical AGCT morphology were FOXL2 wild-type, harboring mutations in KRAS or KMT2D instead, suggesting alternative genetic pathways. TERT promoter mutations were found in 43% of cases, more frequently in recurrences. Other recurrent mutations detected in the cohort included KMT2D (10%), FOXO1 (7%), CHEK2 (5%), TP53 (3.5%), PIK3CA (3.5%), and AKT1 (3%). Two recurrent, FOXL2-mutated cases also carried DICER1 mutations. One tumor exhibited MSI-high status and a tumor mutation burden of 19 mut/Mb.Our results indicate the need for further investigation into the role of FOXO1 as a potential prognostic marker in AGCTs.
Collapse
Affiliation(s)
- Romana Michálková
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Adam Šafanda
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Nikola Hájková
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Jan Hojný
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Eva Krkavcová
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Michaela Kendall Bártů
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Marián Švajdler
- Šikl's Department of Pathology, The Faculty of Medicine and Faculty Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Tetiana Shatokhina
- Department of Oncological Pathology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Jan Laco
- The Fingerland Department of Pathology, Charles University Faculty of Medicine in Hradec Králové and University Hospital, Hradec Králové, Czech Republic
| | - Radoslav Matěj
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic; Department of Pathology, Charles University, 3rd Faculty of Medicine, University Hospital Královské Vinohrady, Prague, Czech Republic; Department of Pathology and Molecular Medicine, Third Faculty of Medicine, Charles University, Thomayer University Hospital, Prague, Czech Republic
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Jitka Hausnerová
- Department of Pathology, University Hospital Brno and Medical Faculty, Masaryk University, Brno, Czech Republic
| | - Jozef Škarda
- Department of Pathology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Mária Hácová
- Department of Pathology, The Regional Hospital Pardubice, Pardubice, Czech Republic
| | - Monika Náležinská
- Division of Gynecologic Oncology, Department of Surgical Oncology, Masaryk Memorial Cancer Institute and Medical Faculty of Masaryk University, Brno, Czech Republic
| | - Tomáš Zima
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Pavel Dundr
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Kristýna Němejcová
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| |
Collapse
|
38
|
Chen Y, Lubinga SJ, Williams T, Carlson JJ, Ramsey S. Real-World Molecular Testing Rates and Patterns in Patients With Primary Advanced or Recurrent Endometrial Cancer in the United States. JCO Precis Oncol 2025; 9:e2400815. [PMID: 40153685 PMCID: PMC11974621 DOI: 10.1200/po-24-00815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/21/2025] [Accepted: 02/18/2025] [Indexed: 03/30/2025] Open
Abstract
PURPOSE This retrospective cohort study estimated the real-world utilization of biomarker testing among patients with primary advanced/recurrent endometrial cancer (pA/rEC) and characterized testing according to demographic and clinical characteristics. MATERIALS AND METHODS A nationwide electronic health record-derived deidentified database was used. Records from January 1, 2013, to August 31, 2023, for women age 18 years and older with pA/rEC were searched for DNA mismatch repair (MMR)/microsatellite instability (MSI), human epidermal growth factor receptor 2 (HER2), and estrogen receptor (ER) or progesterone receptor (PR) testing; a subsample data set (advEndo Spotlight) was searched from April 1, 2013, to November 30, 2022, for additional biomolecular testing. Testing rates were reported by index year and molecular marker. Multivariate logistic regression analyses were conducted to identify characteristics associated with testing. RESULTS The full data set included 2,982 patients, of whom 53% were age 65 years and older; most were non-Hispanic White (56%) and received care in a community setting (73%). The advEndo Spotlight subsample (n = 509) had similar characteristics. From 2013 to 2021, testing for any biomarker increased from 53% to 89% (MMR/MSI, 17% to 81%; ER/PR, 45% to 62%; HER2, 15% to 43%). Patients who received care at an academic versus community facility, had commercial/other insurance versus Medicare/Medicaid, had primary advanced versus recurrent EC, had endometrioid versus nonendometrioid carcinoma, or had no previous surgery as part of primary treatment were more likely to receive testing. CONCLUSION Molecular testing rates in pA/rEC have increased over time, likely due in part to incorporation of biomarker testing into treatment guidelines. This highlights an unmet need to ensure universal access to testing in patients with pA/rEC. Understanding these factors can inform approaches to increase access to molecular testing and increase testing rates.
Collapse
Affiliation(s)
| | | | | | | | - Scott Ramsey
- Curta, Seattle, WA
- University of Washington, Seattle, WA
| |
Collapse
|
39
|
Zhou J, Liu J, Yu Y, Nie H, Hong Y, Ning Y, Yang C, Lai J, Wang H, Tang X, Wang F, Zhao Q. Melanoma Cell Adhesion Molecule Plays a Pivotal Role in Proliferation, Migration, Tumor Immune Microenvironment, and Immunotherapy in Colorectal Cancer. Cancer Med 2025; 14:e70740. [PMID: 40042109 PMCID: PMC11880918 DOI: 10.1002/cam4.70740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 05/12/2025] Open
Abstract
INTRODUCTION MCAM, alternatively referred to as CD146, is an integral membrane glycoprotein belonging to the immunoglobulin superfamily. However, its importance in the tumorigenesis of colorectal cancer is still partially understood. Therefore, this study was designed to investigate the significance of MCAM in colorectal cancer. METHODS MCAM expression was analyzed by TCGA and GEO databases. qRT-PCR and IHC analysis were conducted to validate MCAM expression in patient tissues. The tumor-inhibiting ability of MCAM was further assessed by CCK-8 assay, colony formation assay, and wound-healing assay. qRT-PCR and WB analysis were conducted to evaluate the expression of EMT markers and MMP2/9. qRT-PCR analysis was utilized to detect the polarization status of macrophages. Kaplan-Meier curve, univariate, and multivariate cox analyses were employed to verify the ability of MCAM in prognosis prediction. TIDE scores were used to assess the impact of MCAM on immunotherapy. RESULTS The expression of MCAM was significantly downregulated in CRC, and low MCAM expression revealed poor prognosis in CRC patients. Moreover, MCAM overexpression inhibited the proliferation, migration, and invasive ability of CRC cells. Additionally, MCAM overexpression suppressed N-cadherin and MMP2/9 expression. Furthermore, MCAM impacted M1 macrophage polarization. MCAM is an independent predictor of CRC patient prognosis through Cox regression analysis. Lastly, TIDE score analysis indicated that elevated expression of MCAM increased immunotherapy efficacy. CONCLUSION The findings of this research suggest that MCAM impacts M1 macrophage polarization and enhances immunotherapy efficacy, underscoring its potential as a therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Jingkai Zhou
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jing Liu
- Emergency Medicine CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yali Yu
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Haihang Nie
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yuntian Hong
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yumei Ning
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Chao Yang
- Department of RadiologyXiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Jun Lai
- The Infirmary of Hangzhou Power Supply Company of State Grid, Zhejiang Electric Power Co., LtdHangzhouChina
| | - Haizhou Wang
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xuelian Tang
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Fan Wang
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Qiu Zhao
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Provincial Clinical Research Center for Intestinal and Colorectal DiseasesWuhanChina
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
40
|
Ivanov M, Lebedeva A, Belova E, Grigoreva T, Veselovsky E, Kavun A, Taraskina A, Kuznetsova O, Nikulin V, Belyaeva L, Kravchuk D, Lisitsa T, Barinov A, Pospekhova N, Aliyarova S, Khomenko E, Tryakin A, Demidova I, Stroganova A, Fedyanin M, Mileyko V. Analytical and Clinical Validation of Solo-Test Driver: A Targeted Amplicon-Based NGS Test-System for FFPE and cfDNA Analysis in Clinical Oncology Setting. J Clin Lab Anal 2025; 39:e70008. [PMID: 40056420 PMCID: PMC11937170 DOI: 10.1002/jcla.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Next-generation sequencing (NGS) is increasingly integrated into cancer patient management, necessitating cost-effective, reliable tests for companion diagnostics. We present the validation of the Solo-test Driver panel, a custom NGS amplicon-based tool for DNA analysis of 34 oncogenes, addressing key clinical needs. METHODS The panel's performance was validated for detecting SNVs, CNVs, and MSI. Analytical validation used 182 samples, while clinical validation involved 130 samples, both encompassing diverse tumor types and specimen formats. RESULTS The Solo-test Driver panel has the potential to identify an additional 18.3%, 16.5%, and 8.7% of RAS+ colorectal, PIK3CA+ breast, and EGFR+ lung cancer patients, respectively, when compared to FDA-approved PCR tests. Analytical validation demonstrated high intra-lab robustness (coefficient of variation of coverage uniformity 6.4%) and high inter-lab robustness (PCC of per-amplicon coverage 0.82, 0.84, 0.9 for three different labs). Estimated in silico sensitivity was 100% for detecting clinically actionable SNVs at 250x, corresponding to only 60,000 read pairs per sample. In vitro mixing experiments determined LoD starting from 3.3% VAF. Estimated in silico LoD ranged from 0.5% to 5% at 1000× (1% to 5% at 650x). Clinical validation demonstrated PPA/NPA of 100%/80%, 95%/100%, and 100%/100% for detecting MSI, SNVs, and CNVs, respectively. CONCLUSIONS The Solo-test Driver panel offers a reliable, cost-effective solution for detecting somatic alterations and genomic signatures, making it suitable for routine companion diagnostics in solid tumors.
Collapse
Affiliation(s)
- Maxim Ivanov
- OncoAtlas LLCMoscowRussia
- Sechenov First Moscow State Medical UniversityMoscowRussia
| | - Alexandra Lebedeva
- OncoAtlas LLCMoscowRussia
- Sechenov First Moscow State Medical UniversityMoscowRussia
| | - Ekaterina Belova
- OncoAtlas LLCMoscowRussia
- Sechenov First Moscow State Medical UniversityMoscowRussia
- Lomonosov Moscow State UniversityMoscowRussia
| | - Tatiana Grigoreva
- OncoAtlas LLCMoscowRussia
- Sechenov First Moscow State Medical UniversityMoscowRussia
| | | | | | | | - Olesya Kuznetsova
- OncoAtlas LLCMoscowRussia
- Federal State Budgetary Institution N.N. Blokhin National Medical Research Center of OncologyMoscowRussian Federation
| | - Vladislav Nikulin
- Federal State Budgetary Institution N.N. Blokhin National Medical Research Center of OncologyMoscowRussian Federation
| | - Laima Belyaeva
- Sechenov First Moscow State Medical UniversityMoscowRussia
| | - Daria Kravchuk
- State Budgetary Institution of Health Care of the City of Moscow “Moscow Multidisciplinary Clinical Center” “Kommunarka” of the Department of Health of the City of MoscowMoscowRussia
| | - Tatyana Lisitsa
- Federal State Budgetary Institution N.N. Blokhin National Medical Research Center of OncologyMoscowRussian Federation
| | - Alexey Barinov
- Moscow City Oncology Hospital No 62 of the Moscow Health DepartmentIstraRussia
| | - Natalia Pospekhova
- Federal State Budgetary Institution N.N. Blokhin National Medical Research Center of OncologyMoscowRussian Federation
| | | | | | - Alexey Tryakin
- Federal State Budgetary Institution N.N. Blokhin National Medical Research Center of OncologyMoscowRussian Federation
| | - Irina Demidova
- Moscow City Oncology Hospital No 62 of the Moscow Health DepartmentIstraRussia
| | - Anna Stroganova
- Federal State Budgetary Institution N.N. Blokhin National Medical Research Center of OncologyMoscowRussian Federation
| | - Mikhail Fedyanin
- Federal State Budgetary Institution N.N. Blokhin National Medical Research Center of OncologyMoscowRussian Federation
- State Budgetary Institution of Health Care of the City of Moscow “Moscow Multidisciplinary Clinical Center” “Kommunarka” of the Department of Health of the City of MoscowMoscowRussia
- Federal State Budgetary Institution “National Medical and Surgical Center Named After N.I. Pirogov” of the Ministry of Health of the Russian FederationMoscowRussia
| | - Vladislav Mileyko
- OncoAtlas LLCMoscowRussia
- Sechenov First Moscow State Medical UniversityMoscowRussia
| |
Collapse
|
41
|
Landre T, Des Guetz G. Microsatellite instability-high status as a pan-cancer biomarker for immunotherapy efficacy. Cancer Immunol Immunother 2025; 74:122. [PMID: 39998698 PMCID: PMC11861458 DOI: 10.1007/s00262-025-03980-x] [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: 01/10/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Microsatellite instability-high (MSI-H) cancers are linked to exceptional benefit from immune checkpoint inhibitors (ICIs), but studies on their efficacy across various MSI-H cancer types are limited. METHODS Randomized clinical trials (RCTs) comparing ICIs to chemotherapy in advanced MSI-H/dMMR cancers were systematically reviewed. Eligible studies included 13 RCTs with 1633 MSI-H patients across colorectal, gastric, and endometrial cancers. Data were analyzed using hazard ratios for progression-free survival (PFS) and overall survival (OS), with subgroup analyses by tumor type. Statistical heterogeneity was assessed using Cochrane's Q and I2. RESULTS Immunotherapy significantly improved PFS and OS in MSI-H patients, with an HR for OS of 0.35 (95% CI 0.27-0.46; p < 0.00001) versus 0.81 for MSS patients. PFS showed a 64% reduced risk of progression (HR = 0.36, 95% CI 0.28-0.46; p < 0.0001). Subgroup analyses highlighted PFS benefits across tumor types: colorectal (HR = 0.28, 95% CI 0.11-0.73), gastric (HR = 0.43, 95% CI 0.27-0.68), and endometrial cancers (HR = 0.34, 95% CI 0.27-0.42). CONCLUSIONS This meta-analysis establishes MSI-H as a predictive biomarker for ICIs, supporting its role in therapy selection and underscoring the need for MSI-H/dMMR-focused clinical trials.
Collapse
Affiliation(s)
- Thierry Landre
- Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital René Muret, Hôpitaux Universitaires de Paris Seine-St-Denis, Avenue du Dr Schaeffner, 93270, Sevran, France.
| | - Gaëtan Des Guetz
- Service d'Oncologie Médicale, Faculté de Médecine, Centre Hospitalier de St-Denis, Université Sorbonne Paris Nord, St-Denis, France
| |
Collapse
|
42
|
Venetis K, Frascarelli C, Bielo LB, Cursano G, Adorisio R, Ivanova M, Mane E, Peruzzo V, Concardi A, Negrelli M, D'Ercole M, Porta FM, Zhan Y, Marra A, Trapani D, Criscitiello C, Curigliano G, Guerini-Rocco E, Fusco N. Mismatch repair (MMR) and microsatellite instability (MSI) phenotypes across solid tumors: A comprehensive cBioPortal study on prevalence and prognostic impact. Eur J Cancer 2025; 217:115233. [PMID: 39827722 DOI: 10.1016/j.ejca.2025.115233] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Mismatch repair deficiency (MMR-d) and microsatellite instability (MSI) are prognostic and predictive biomarkers in oncology. Current testing for MMR/MSI relies on immunohistochemistry (IHC) for MMR proteins and molecular assays for MSI detection. This combined diagnostic strategy, however, lacks tumor specificity and does not account for gene variants. This study provides an in-depth analysis of MMR mutations frequency, spectrum, and distribution in solid tumors. Data from 23,893 patients across 11 tumor types, using 66 publicly available studies, were analyzed. MMR-mutated (MMR-m) status was defined by alterations in MLH1, PMS2, MSH2, and/or MSH6; MSI was assessed by MSIsensor. Cases with indeterminate labelling were excluded. Survival was analyzed using the Kaplan-Meier method. Among 19,353 tumors, 949 MMR variants were identified, comprising 432 pathogenic and 517 variants of unknown significance (VUS), as defined by OncoKB. MSH6 mutations were the most frequent (n = 279, 29.4 %), followed by MSH2 (n = 198, 20.9 %), MLH1 (n = 187, 19.7 %), and PMS2 (n = 161, 16.9 %). MMR-m cases were more frequent in endometrial (EC, 20.5 %), colorectal (CRC, 8.2 %), bladder (BLCA, 8.7 %), and gastroesophageal cancers (GEC, 5.4 %). Pathogenic mutations were more common than non-pathogenic in EC, CRC, and GEC (p < 0.001, p = 0.01, p = 0.32, respectively). MMR-m status was not associated with MSI in 247 (48.9 %) cases, including 67 (13.2 %) with pathogenic mutations. The highest concordance between MMR-m and MSI was observed in CRC (65.7 %), EC (91.2 %), and GEC (69.6 %), while the lowest in pancreatic (0.2 %) and lung cancers (0.1 %). MMR-m GECs showed improved overall survival compared to MMR-wt (p = 0.009), a relationship not observed in other tumor types. This study demonstrates that the MMR spectrum is extremely hetoerogeneous in solid tumors, highliting the need for comprehensive and tumor-specific testing strategies.
Collapse
Affiliation(s)
| | - Chiara Frascarelli
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Luca Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Giulia Cursano
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Riccardo Adorisio
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Mariia Ivanova
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Eltjona Mane
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Virginia Peruzzo
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Alberto Concardi
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Marianna D'Ercole
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Yinxiu Zhan
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Antonio Marra
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Dario Trapani
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
43
|
André T, Elez E, Lonardi S. Microsatellite-Instability-High Metastatic Colorectal Cancer. Reply. N Engl J Med 2025; 392:726-727. [PMID: 39938109 DOI: 10.1056/nejmc2416383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Affiliation(s)
| | - Elena Elez
- Vall d'Hebron University Hospital, Barcelona
| | - Sara Lonardi
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| |
Collapse
|
44
|
Kravchuk D, Lebedeva A, Kuznetsova O, Kavun A, Taraskina A, Belova E, Grigoreva T, Veselovsky E, Mileyko V, Nikulin V, Nekrasova L, Tryakin A, Fedyanin M, Ivanov M. Dynamics of blood microsatellite instability (bMSI) burden predicts outcome of a patient treated with immune checkpoint inhibitors: a case report of hyperprogressive disease. Front Immunol 2025; 16:1492296. [PMID: 39975556 PMCID: PMC11836019 DOI: 10.3389/fimmu.2025.1492296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025] Open
Abstract
Microsatellite instability (MSI) is a widely studied molecular signature, which is associated with long-term benefit in patients treated with immune checkpoint inhibitor therapy. This approach has been proven to be effective in the treatment of patients with MSI-positive colorectal cancer (CRC). Analysis of serial liquid biopsy samples allows to detect changes in the tumor in response to therapy. Typically, somatic mutations are used for tracing the dynamics of the tumor, and the assessment of DNA signatures such as MSI is not currently used for these purposes. Here, we describe a case of a MSI-positive CRC, who received nivolumab monotherapy. Sequential sampling of the patient's plasma demonstrated an increase in MSI burden (bMSI), which was found to correlate with the increase of driver mutation burden one month after starting nivolumab, and hyperprogressive disease. Thus, analysis of bMSI in liquid biopsy via NGS may be a promising method for timely assessment of the treatment effectiveness received by patients with MSI-positive CRC.
Collapse
Affiliation(s)
- Daria Kravchuk
- Moscow Multidisciplinary Clinical Center “Kommunarka” of the Department of Health of the City of Moscow, State Budgetary Institution of Healthcare, Moscow, Russia
| | - Alexandra Lebedeva
- R&D Department, OncoAtlas LLC, Moscow, Russia
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Olesya Kuznetsova
- R&D Department, OncoAtlas LLC, Moscow, Russia
- N.N. Blokhin Russian Cancer Research Center, Moscow, Russia
| | | | | | - Ekaterina Belova
- R&D Department, OncoAtlas LLC, Moscow, Russia
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia
| | - Tatiana Grigoreva
- R&D Department, OncoAtlas LLC, Moscow, Russia
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Vladislav Mileyko
- R&D Department, OncoAtlas LLC, Moscow, Russia
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Lidia Nekrasova
- P. Hertsen Moscow Oncology Research Institute (MORI), Moscow, Russia
| | - Alexey Tryakin
- N.N. Blokhin Russian Cancer Research Center, Moscow, Russia
| | - Mikhail Fedyanin
- Moscow Multidisciplinary Clinical Center “Kommunarka” of the Department of Health of the City of Moscow, State Budgetary Institution of Healthcare, Moscow, Russia
- N.N. Blokhin Russian Cancer Research Center, Moscow, Russia
- Federal State Budgetary Institution “National Medical and Surgical Center named after N.I. Pirogov” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Maxim Ivanov
- R&D Department, OncoAtlas LLC, Moscow, Russia
- Institute for Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
45
|
Kabatnik S, Zheng X, Pappas G, Steigerwald S, Padula MP, Mann M. Deep visual proteomics reveals DNA replication stress as a hallmark of signet ring cell carcinoma. NPJ Precis Oncol 2025; 9:37. [PMID: 39910169 PMCID: PMC11799539 DOI: 10.1038/s41698-025-00819-7] [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: 07/22/2024] [Accepted: 01/17/2025] [Indexed: 02/07/2025] Open
Abstract
Signet Ring Cell Carcinoma (SRCC) is a rare and highly malignant form of adenocarcinoma with increasing incidence and poor prognosis due to late diagnosis and limited treatment options. We employed Deep Visual Proteomics (DVP), which combines AI-directed cell segmentation and classification with laser microdissection and ultra-high sensitivity mass spectrometry, for cell-type-specific proteomic analysis of SRCC across the bladder, prostate, seminal vesicle, and a lymph node of a single patient. DVP identified significant alterations in DNA damage response (DDR) proteins, particularly within the ATR and mismatch repair (MMR) pathways, indicating replication stress as a crucial factor in SRCC mutagenicity. Additionally, we observed substantial enrichment of immune-related proteins, reflecting high levels of cytotoxic T lymphocyte infiltration and elevated PD-1 expression. These findings suggest that pembrolizumab immunotherapy may be more effective than conventional chemotherapy for this patient. Our results provide novel insights into the proteomic landscape of SRCC, identify potential targets, and open up for personalized therapeutic strategies in managing SRCC.
Collapse
Affiliation(s)
- Sonja Kabatnik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Xiang Zheng
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Georgios Pappas
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Sophia Steigerwald
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Matthew P Padula
- School of Life Sciences and Proteomics Core Facility, Faculty of Science, University of Technology Sydney, Ultimo, Australia.
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark.
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
46
|
Cai Y, Chen X, Chen J, Liao J, Han M, Lin D, Hong X, Hu H, Hu J. Deep learning-assisted colonoscopy images for prediction of mismatch repair deficiency in colorectal cancer. Surg Endosc 2025; 39:859-867. [PMID: 39623175 DOI: 10.1007/s00464-024-11426-1] [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: 08/06/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025]
Abstract
BACKGROUND Deficient mismatch repair or microsatellite instability is a major predictive biomarker for the efficacy of immune checkpoint inhibitors of colorectal cancer. However, routine testing has not been uniformly implemented due to cost and resource constraints. METHODS We developed and validated a deep learning-based classifiers to detect mismatch repair-deficient status from routine colonoscopy images. We obtained the colonoscopy images from the imaging database at Endoscopic Center of the Sixth Affiliated Hospital, Sun Yat-sen University. Colonoscopy images from a prospective trial (Neoadjuvant PD-1 blockade by toripalimab with or without celecoxib in mismatch repair-deficient or microsatellite instability-high locally advanced colorectal cancer) were used to test the model. RESULTS A total of 5226 eligible images from 892 tumors from the consecutive patients were utilized to develop and validate the deep learning model. 2105 colorectal cancer images from 306 tumors were randomly selected to form model development dataset with a class-balanced approach. 3121 images of 488 proficient mismatch repair tumors and 98 deficient mismatch repair tumors were used to form the independent dataset. The model achieved an AUROC of 0.948 (95% CI 0.919-0.977) on the test dataset. On the independent validation dataset, the AUROC was 0.807 (0.760-0.854), and the NPV in was 94.2% (95% CI 0.918-0.967). On the prospective trial dataset, the model identified 29 tumors among the 33 deficient mismatch repair tumors (87.88%). CONCLUSIONS The model achieved a high NPV in detecting deficient mismatch repair colorectal cancers. This model might serve as an automatic screening tool.
Collapse
Affiliation(s)
- Yue Cai
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China
| | - Xijie Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery (Gastric Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Junguo Chen
- Department of Thoracic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - James Liao
- Guangzhou Aptiligent Technology Co. Ltd., Guangzhou, Guangdong, China
| | - Ming Han
- Guangzhou Aptiligent Technology Co. Ltd., Guangzhou, Guangdong, China
| | - Dezheng Lin
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery (Endoscopic Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoling Hong
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery (Endoscopic Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huabin Hu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China.
| | - Jiancong Hu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Department of General Surgery (Endoscopic Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
47
|
Haldar SD, Kopetz S. Dual checkpoint blockade for microsatellite instability-high colorectal cancer. Lancet 2025; 405:354-356. [PMID: 39874978 DOI: 10.1016/s0140-6736(25)00144-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Affiliation(s)
- S Daniel Haldar
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77584, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77584, USA.
| |
Collapse
|
48
|
Cai Y, Xiao H, Zhou Q, Lin J, Liang X, Xu W, Cao Y, Zhang X, Wang H. Comprehensive Analyses of PANoptosome with Potential Implications in Cancer Prognosis and Immunotherapy. Biochem Genet 2025; 63:331-353. [PMID: 38436818 PMCID: PMC11832696 DOI: 10.1007/s10528-024-10687-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 01/04/2024] [Indexed: 03/05/2024]
Abstract
Cell death resistance significantly contributes to poor therapeutic outcomes in various cancers. PANoptosis, a unique inflammatory programmed cell death (PCD) pathway activated by specific triggers and regulated by the PANoptosome, possesses key features of apoptosis, pyroptosis, and necroptosis, but these cannot be accounted for by any of the three PCD pathways alone. While existing studies on PANoptosis have predominantly centered on infectious and inflammatory diseases, its role in cancer malignancy has been understudied. In this comprehensive investigation, we conducted pan-cancer analyses of PANoptosome component genes across 33 cancer types. We characterized the genetic, epigenetic, and transcriptomic landscapes, and introduced a PANoptosome-related potential index (PANo-RPI) for evaluating the intrinsic PANoptosome assembly potential in cancers. Our findings unveil PANo-RPI as a prognostic factor in numerous cancers, including KIRC, LGG, and PAAD. Crucially, we established a significant correlation between PANo-RPI and tumor immune responses, as well as the infiltration of diverse lymphoid and myeloid cell subsets across nearly all cancer types. Moreover, a high PANo-RPI was consistently associated with improved immunotherapy response and efficacy, as evidenced by re-analysis of multiple immunotherapy cohorts. In conclusion, our study suggests that targeting PANoptosome components and modulating PANoptosis may hold tremendous therapeutic potential in the context of cancer.
Collapse
Affiliation(s)
- Yonghua Cai
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Heng Xiao
- Southern Medical School, No. 1023, South Shatai Road, Baiyun District, Guangzhou, 510515, Guangdong, China
| | - Qixiong Zhou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Jie Lin
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Xianqiu Liang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Wei Xu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Yongfu Cao
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Xian Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
| | - Hai Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
| |
Collapse
|
49
|
Zgura A, Chipuc S, Bacalbasa N, Haineala B, Rodica A, Sebastian V. Evaluating Tumour Mutational Burden as a Key Biomarker in Personalized Cancer Immunotherapy: A Pan-Cancer Systematic Review. Cancers (Basel) 2025; 17:480. [PMID: 39941847 PMCID: PMC11816366 DOI: 10.3390/cancers17030480] [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: 12/29/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Tumour mutational burden (TMB) is an emerging biomarker for predicting the efficacy of immune checkpoint inhibitors (ICIs) in cancer therapy. While its role is well established in lung cancer and melanoma, its predictive value for breast and prostate cancers remains unclear. OBJECTIVE This systematic review aimed to assess the predictive value of TMB for ICI therapy across four major cancer types-lung, melanoma, breast, and prostate-and to explore factors contributing to the variability in its effectiveness as a biomarker. METHODS A systematic search and a review of the literature were conducted in accordance with PRISMA guidelines. Studies examining the relationship between TMB levels and clinical outcomes following ICI therapy in the specified cancers were analyzed. The data were synthesized to evaluate TMB's predictive value and identify gaps in the current research. RESULTS High TMB consistently correlated with improved outcomes in lung cancer and melanoma, confirming its predictive utility in these cancers. Conversely, the findings for breast and prostate cancers were inconclusive. The variability in TMB's predictive value for these cancers suggests the need for complementary biomarkers or refined criteria to enhance its reliability. Methodological inconsistencies in TMB evaluation were also noted as a significant limitation. CONCLUSIONS TMB serves as a robust biomarker for predicting ICI response in lung cancer and melanoma, but demonstrates limited predictive utility in breast and prostate cancers. Future research should prioritize standardizing TMB assessment protocols and investigating additional biomarkers to improve treatment personalization for these cancer types.
Collapse
Affiliation(s)
- Anca Zgura
- Department of Oncology-Radiotherapy, Prof. Dr. Alexandru Trestioreanu Institute of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.Z.)
- Prof. Dr. Alexandru Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Stefania Chipuc
- Prof. Dr. Alexandru Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Nicolae Bacalbasa
- Department of Surgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (N.B.); (V.S.)
| | - Bogdan Haineala
- Department of Urology, “Fundeni” Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Anghel Rodica
- Department of Oncology-Radiotherapy, Prof. Dr. Alexandru Trestioreanu Institute of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.Z.)
- Prof. Dr. Alexandru Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Vâlcea Sebastian
- Department of Surgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (N.B.); (V.S.)
| |
Collapse
|
50
|
Palacios Diaz RD, de Unamuno Bustos B, Pozuelo Ruiz M, Llavador Ros M, Palanca Suela S, Botella Estrada R. Study of Microsatellite Instability by Immunohistochemistry in a Cohort of Patients With Melanoma. J Cutan Pathol 2025; 52:162-171. [PMID: 39581581 DOI: 10.1111/cup.14758] [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: 04/01/2024] [Revised: 09/30/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Microsatellite instability (MSI) has prognostic value and impacts therapy strategies in several malignancies. Data regarding MSI in melanoma are scarce. The aim of this study was to assess MSI through the analysis of MMR protein expression in patients with melanoma. METHODS An observational retrospective single-center study was designed based on patients with primary melanoma. We assessed MSI through immunohistochemical staining with anti-MLH1, anti-MSH2, anti-MSH6, and anti-PMS2 on full-thickness excision tissue. RESULTS Ninety-three patients were included in this study. The complete absence of nuclear staining in tumoral cells was extremely rare, with only one melanoma not expressing MSH6. Most melanomas showed an expression index for MLH1 (77.7%), MSH2 (87.2%), and PMS2 (78.6%) ≥ 75%. Most melanomas (57.8%) exhibited an MSH6 expression index in the range of 1%-74%. A low MSH6 expression index and a reduced combined MMR protein expression index (MMR-e) were significantly associated with higher melanoma-specific survival. A mild PMS2 staining intensity was significantly associated with a higher melanoma-specific survival. The patients with high MMR-e who received immunotherapy progressed and died more frequently than those with reduced MMR-e (75% vs. 33.3%). CONCLUSION More studies are needed to further define the role of MSI in melanoma prognosis and response to immunotherapy.
Collapse
Affiliation(s)
| | - Blanca de Unamuno Bustos
- Dermatology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
- Dermatology, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Mónica Pozuelo Ruiz
- Dermatology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | | | - Sarai Palanca Suela
- Molecular Biology Laboratory, Service of Clinical Analysis, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Rafael Botella Estrada
- Dermatology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
- Dermatology, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Department of Medicine, Universitat de València, Valencia, Spain
| |
Collapse
|