1
|
Zhang T, Chen Y, Xiang Z. Machine learning-based integration develops a disulfidptosis-related lncRNA signature for improving outcomes in gastric cancer. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2025; 53:1-13. [PMID: 39701937 DOI: 10.1080/21691401.2024.2440415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 11/05/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024]
Abstract
Gastric cancer remains one of the deadliest cancers globally due to delayed detection and limited treatment options, underscoring the critical need for innovative prognostic methods. Disulfidptosis, a recently discovered programmed cell death triggered by disulphide stress, presents a fresh avenue for therapeutic exploration. This research examines disulfidptosis-related long noncoding RNAs (DRLs) in gastric cancer, with the goal of leveraging these lncRNAs as potential markers to enhance patient outcomes and treatment approaches. Comprehensive genomic and clinical data from stomach adenocarcinoma (STAD) were obtained from The Cancer Genome Atlas (TCGA). Employing least absolute shrinkage and selection operator (LASSO) regression analysis, a prognostic model was devised incorporating five key DRLs to forecast survival rates. The effectiveness of this model was validated using Kaplan-Meier survival plots, receiver operating characteristic (ROC) curves, and extensive functional enrichment studies. The importance of select lncRNAs and the expression variability of genes tied to disulfidptosis were validated via quantitative real-time PCR (qRT-PCR) and Western blot tests, establishing a solid foundation for their prognostic utility. Analyses of functional enrichment and tumour mutation burden highlighted the biological importance of these DRLs, connecting them to critical cancer pathways and immune responses. These discoveries broaden our comprehension of the molecular framework of gastric cancer and bolster the development of tailored treatment plans, highlighting the substantial role of DRLs in clinical prognosis and therapeutic intervention.
Collapse
Affiliation(s)
- Tianze Zhang
- Department of Gastrointestinal Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Yuqing Chen
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Zhiping Xiang
- Head and Neck Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
2
|
Rocha P, Bach R, Masfarré L, Hernandez S, Navarro-Gorro N, Rossell A, Villanueva X, Giner M, Sanchéz I, Galindo M, Del Rey-Vergara R, Iñañez A, Sanchéz-Espiridion B, Lu W, Acedo-Terrades A, Berenguer-Molins P, Sánchez-Font A, Chalela R, Curull V, Taus Á, Hardy-Werbin M, Sausen M, Georgiadis A, White J, Jackson JB, Moliner L, Clavé S, Bellosillo B, Rovira A, Wistuba I, Soto LMS, Perera-Bel J, Arriola E. Molecular and immunological features associated with long-term benefits in metastatic NSCLC patients undergoing immune checkpoint blockade. Oncoimmunology 2025; 14:2469377. [PMID: 39991958 PMCID: PMC11853546 DOI: 10.1080/2162402x.2025.2469377] [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: 07/03/2024] [Revised: 01/22/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025] Open
Abstract
INTRODUCTION Immunotherapy is firmly established as a treatment regimen in various solid tumors, driven by its exceptional benefits in a selected group of patients. Despite widespread adoption of immune checkpoint blockade (ICB) across diverse solid tumors, the quest for a clinically informative biomarker for long-term benefit remains unmet. METHODS A total of 49 patients with metastatic NSCLC treated with ICB were included. Long-term (LTR) and short-term responders (STR) were defined as those with a response to ICB lasting more than 24 months or less than 6 months, respectively. Longitudinal blood specimens were collected before ICB treatment initiation and early-on treatment. Plasma ctDNA next-generation sequencing panel (NGS) and serum proteomics were performed. GeoMx DSP on baseline tumor tissue was performed in a subset of patients. RESULTS Our analysis revealed specific characteristics of LTR compared with STR, namely higher PD-L1 in tumor cells (p = 0.005) and higher incidence of irAEs (p = 0.001). Genomic features associated with lack of benefit from ICB included co-occurring mutations in KRAS/STK11 and TP53/KMT2D (p < 0.05). At a baseline, LTR patients exhibited higher serum levels of proteins related with apoptosis (CASP8, PRKRA), chemotaxis, immune proteasome, processing of MHC class I (S100A4, PSMD9, RNF41) and immune homeostasis (HAVCR1, ARG1) (p < 0.05). Protein spatial profiling of tumor samples showed higher levels of proteins linked with the presence of immune cells (CD45), T cells (CD8), antigen presentation (HLA-DR) and immune regulation proteins (PD-L1, IDO1) within the tumor and tumor stroma component (p < 0.05) in LTR patients. Serum longitudinal analysis identified a set of proteins that presented distinct dynamics in LTR compared to STR, making them interesting candidates to evaluate as early predictors of treatment efficacy. CONCLUSIONS Our multimodal analysis of patients with metastatic NSCLC treated with ICB identified clinicopathological and immunological features associated with long-term benefits. The presence of preexisting antitumor immunity emerged as a strong predictor of long-term benefits, providing insights for potential biomarkers and therapeutic strategies for enhancing ICB outcomes in metastatic NSCLC.
Collapse
Affiliation(s)
- Pedro Rocha
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Rafael Bach
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Laura Masfarré
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Sharia Hernandez
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Adrià Rossell
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | | | - Mario Giner
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | | | - Miguel Galindo
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | | | - Albert Iñañez
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Beatriz Sanchéz-Espiridion
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Lu
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | - Victor Curull
- Pulmonology Department, Hospital del Mar, Barcelona, Spain
| | - Álvaro Taus
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | | | | | | | | | | | | | - Sergi Clavé
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | - Beatriz Bellosillo
- Pathology Department, Hospital del Mar, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ana Rovira
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Luisa M Solis Soto
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Edurne Arriola
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
3
|
Tacelli M, Gentiluomo M, Biamonte P, Castano JP, Berković MC, Cives M, Kapitanović S, Marinoni I, Marinovic S, Nikas I, Nosáková L, Pedraza-Arevalo S, Pellè E, Perren A, Strosberg J, Campa D, Capurso G. Pancreatic neuroendocrine neoplasms (pNENs): Genetic and environmental biomarkers for risk of occurrence and prognosis. Semin Cancer Biol 2025; 112:112-125. [PMID: 40158764 DOI: 10.1016/j.semcancer.2025.03.005] [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: 01/17/2025] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Pancreatic neuroendocrine neoplasms (pNENs) are rare and heterogeneous tumors arising from neuroendocrine cells, representing approximately 10 % of all Gastro-Entero-Pancreatic neuroendocrine neoplasms. While most pNENs are sporadic, a subset is associated with genetic syndromes such as multiple endocrine neoplasia type 1 (MEN1) or von Hippel-Lindau disease (VHL). pNENs are further classified into functioning and non-functioning tumors, with distinct clinical behaviors, prognoses, and treatment approaches. This review explores genetic and environmental biomarkers that influence the risk, prognosis, and therapeutic responses in pNENs. The epidemiology of pNENs reveals an increasing incidence, primarily due to advancements in imaging techniques. Genetic factors play a pivotal role, with germline mutations in MEN1, VHL, and other genes contributing to familial pNENs. Somatic mutations, including alterations in the mTOR pathway and DNA maintenance genes such as DAXX and ATRX, are critical in sporadic pNENs. These mutations, along with epigenetic dysregulation and transcriptomic alterations, underpin the diverse clinical and molecular phenotypes of pNENs. Emerging evidence suggests that epigenetic changes, including DNA methylation profiles, can stratify pNEN subtypes and predict disease progression. Environmental and lifestyle factors, such as diabetes, smoking, and chronic pancreatitis, have been linked to an increased risk of sporadic pNENs. While the association between these factors and tumor progression is still under investigation, their potential role in influencing therapeutic outcomes warrants further study. Advances in systemic therapies, including somatostatin analogs, mTOR inhibitors, and tyrosine kinase inhibitors, have improved disease management. Biomarkers such as Ki-67, somatostatin receptor expression, and O6-methylguanine-DNA methyltransferase (MGMT) status are being evaluated for their predictive value. Novel approaches, including the use of circulating biomarkers (NETest, circulating tumor cells, and ctDNA) and polygenic risk scores, offer promising avenues for non-invasive diagnosis and monitoring. Despite these advancements, challenges remain, including the need for large, well-annotated datasets and validated biomarkers. Future research should integrate multi-omics approaches and leverage liquid biopsy technologies to refine diagnostic, prognostic, and therapeutic strategies. Interdisciplinary collaborations and global consortia are crucial for overcoming current limitations and translating research findings into clinical practice. These insights hold promise for improving prevention, early detection, and tailored treatments, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Matteo Tacelli
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Paolo Biamonte
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Justo P Castano
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Reina Sofia University Hospital, Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Maja Cigrovski Berković
- Department for Sport and Exercise Medicine, Faculty of Kinesiology University of Zagreb, Zagreb 10000, Croatia
| | - Mauro Cives
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy; Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Sanja Kapitanović
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Ilaria Marinoni
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Sonja Marinovic
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Ilias Nikas
- Medical School, University of Cyprus, Nicosia, Cyprus
| | - Lenka Nosáková
- Clinic of Internal Medicine - Gastroenterology, JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Bratislava, Slovakia
| | - Sergio Pedraza-Arevalo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Reina Sofia University Hospital, Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Eleonora Pellè
- Department of GI Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Aurel Perren
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Jonathan Strosberg
- Department of GI Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
4
|
Subbiah V, Othus M, Palma J, Cuglievan B, Kurzrock R. Designing Clinical Trials for Patients With Rare Cancers: Connecting the Zebras. Am Soc Clin Oncol Educ Book 2025; 45:e100051. [PMID: 40228175 DOI: 10.1200/edbk-25-100051] [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: 04/16/2025]
Abstract
The field of rare cancer research is rapidly transforming, marked by significant progress in clinical trials and treatment strategies. Rare cancers, as defined by the National Cancer Institute, occur in fewer than 150 cases per million people each year, yet they collectively represent a significant portion of all cancer diagnoses. Because of their infrequency, these cancers pose distinct challenges for clinical trials, including limited patient populations, geographical dispersion, and a general lack of awareness of treatment options. Economic limitations further complicate drug development, making initiatives such as the Orphan Drug Act essential for incentivizing research. The advent of next-generation sequencing (NGS) and precision medicine has been instrumental in identifying actionable genetic alterations in parallel with an explosion in the development of genomically targeted therapies, immunotherapies, and antibody drug conjugates. Advances in clinical NGS, precision medicine, and tumor-agnostic therapies have become central to the progress in rare cancer research. The development and approval of tumor-agnostic drugs, such as BRAF, NTRK, and RET inhibitors, and immunotherapy for mismatch repair deficient/microsatellite instability-high status cancers highlight the potential of personalized treatments across diverse cancer types and across the age spectrum. Collaborative trials from cooperative groups including SWOG DART, ASCO TAPUR, NCI-MATCH, pediatric COG-match, DRUP, IMPRESS, and innovative registrational basket and platform trials (eg, VE-Basket, ROAR, LIBRETTO-001, ARROW), along with patient advocacy group-run trials like TRACK, are enhancing access to clinical trials. In addition, artificial intelligence has the potential to improve the trial matching process. An integrated approach, combining these innovations in collaboration with multiple stakeholders, is crucial for advancing rare cancer research, offering hope for better patient outcomes and quality of life.
Collapse
Affiliation(s)
| | - Megan Othus
- SWOG Cancer Research Network Statistical Center, Seattle, WA
- Division of Public Health, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jim Palma
- TargetCancer Foundation, Rare Cancer Patient Advocacy Group, Cambridge, MA
| | - Branko Cuglievan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Razelle Kurzrock
- Genomic Sciences and Precision Medicine Center, and Medical College of Wisconsin Cancer Center, Milwaukee, WI
- WIN Consortium, Paris, France
- University of Nebraska, Lincoln, NE
| |
Collapse
|
5
|
Nakazato Y, Hirano K, Mitsuma T, Arimasu Y, Hirokawa T, Chiba T, Fujiwara M, Tanaka R, Kondo H, Kamma H. Regulatory SNP of TERT promoter accompanied by C228T and BRAFV 600E is an exacerbating factor of papillary thyroid carcinoma. Oncol Lett 2025; 29:267. [PMID: 40235685 PMCID: PMC11997643 DOI: 10.3892/ol.2025.15013] [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: 05/15/2024] [Accepted: 10/09/2024] [Indexed: 04/17/2025] Open
Abstract
Despite the increased incidence of thyroid cancer due to enhanced precision of ultrasound technology and extensive utilization of puncture aspiration cytology, the mortality rate remains low, raising concerns about overdiagnosis. Papillary thyroid carcinoma (PTC) is the most common type, primarily diagnosed through cell nuclei examination. Recent advancements in identifying genetic mutations and tumor classification have refined diagnostic methods. Point mutations in the telomerase reverse transcriptase promoter (TERTp), specifically -124 C >T (C228T) and -146 C >T (C250T), and the regulatory single nucleotide polymorphism -245 T >C, C allele of rs2853669 (TrSNP) are potential thyroid cancer biomarkers. The present study tested the hypothesis that the coexistence of BRAF mutations in driver genes upstream of the MAPK pathway and late mutations unrelated to signaling, such as point mutations in TERTp, increases tumor virulence. A total of 133 patients with PTC who underwent surgery between January 2014 and November 2021 were included in the study. Blood and tumor tissue samples were collected, and DNA was extracted for genetic mutation analysis using PCR and Sanger sequencing. The TrSNP analysis of blood and surgical tissue samples showed a 97.7% agreement rate. TrSNP was detected in 70 of 133 patients (52.6%) and was significantly associated with tumor size, particularly in tumors >2.0 cm. TERTp point mutations were identified in 29 of 133 patients (21.8%), with C228T strongly associated with tumor size, particularly in tumors >4.0 cm, and extraglandular invasion. BRAF V600E was detected in 82 patients (61.7%) but showed no significant association with clinicopathological parameters. However, the coexistence of BRAF V600E with C228T and TrSNP affected tumor size and progression. The findings indicated that TrSNPs, along with C228T and BRAF V600E, may serve as potential molecular markers to predict PTC growth or exacerbation. Notably, coexistence of C228T and TrSNP is a preoperative indicator of disease progression.
Collapse
Affiliation(s)
- Yoko Nakazato
- Department of General Thoracic and Thyroid Surgery, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Koichi Hirano
- Department of General Thoracic and Thyroid Surgery, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Tomoya Mitsuma
- Department of General Thoracic and Thyroid Surgery, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Yu Arimasu
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine Research Institute, Tokyo 162-8655, Japan
| | | | - Tomohiro Chiba
- Department of Pathology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 112-0012, Japan
| | - Masachika Fujiwara
- Department of Pathology, Kyorin University School of Medicine, Tokyo 181-8611, Japan
| | - Ryota Tanaka
- Department of General Thoracic and Thyroid Surgery, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Haruhiko Kondo
- Department of General Thoracic and Thyroid Surgery, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Hiroshi Kamma
- Nasu Institute of Medical Sciences, Kamma Memorial Hospital, Nasushiobara, Tochigi 325-0046, Japan
| |
Collapse
|
6
|
Guo Y, Zhai J, Yang Y, Wei Q, Li S, Huo R, Tong G, Xu E, Chen Y, Han S, Chen D. Long-term survival of an ALK fusion lung adenocarcinoma patient with high mutation burden and microsatellite instability high: a case report. Anticancer Drugs 2025; 36:427-431. [PMID: 39908227 DOI: 10.1097/cad.0000000000001693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Immune checkpoint blockage (ICB) therapy has shown minimal effectiveness in anaplastic lymphoma kinase (ALK)-positive nonsmall cell lung cancer (NSCLC) regardless of Programmed death-ligand 1 expression. ALK fusion accompanied by mismatch repair deficiency or microsatellite instability-high (MMRd/MSI-H) and high tumor mutation burden (TMB-H) are extremely rare in NSCLC, and the efficacy of ALK inhibitors or ICB-based therapies is unclear. Here, we report the case of a 60-year-old female patient with metastatic lung adenocarcinoma accompanied by EML4-ALK fusion, TMB-H, MMRd/MSI-H, and pathogenic mutations in TP53, MLH1, and STK11. The patient experienced progression on initial iruplinalkib and subsequent alectinib therapy within 5 months. After the failure of third-line therapy with cisplatin-pemetrexed combined with bevacizumab, she received sintilimab plus anlotinib which led to a progression-free survival of 6.5 months. She received sintilimab combined with albumin-paclitaxel plus carboplatin and achieved partial response after 6 months. She developed adverse events after one cycle of sintilimab plus albumin-paclitaxel treatment. Then she was continued with sintilimab plus anlotinib as a maintenance therapy due to intolerance to chemotherapy. After progression on ICB-based therapy, the patient was treated with lorlatinib and still under follow-up with overall survival of more than 3 years. Our findings highlight the therapeutic potential of ICB-based regimens in patients with MSI-H and ALK-rearranged NSCLC.
Collapse
Affiliation(s)
- Yanrong Guo
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Jinfang Zhai
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Yanli Yang
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Qin Wei
- Department of Medicine, Xiamen Spacegen Co., Ltd, Xiamen
| | - Shengshu Li
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Rujie Huo
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Guoping Tong
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Enwei Xu
- Department of Pathology, Shanxi Province Cancer Hospital, Taiyuan, China
| | - Yan Chen
- Department of Medicine, Xiamen Spacegen Co., Ltd, Xiamen
| | - Songyan Han
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Deyi Chen
- Department of Medicine, Xiamen Spacegen Co., Ltd, Xiamen
| |
Collapse
|
7
|
Shao YF, Baca Y, Hinton A, Xiu J, VanderWalde A, Hadfield M, Park SJ, Darabi S, Sato T, Moser JC. Immune Profiling of Uveal Melanoma Liver Metastases and Response to Checkpoint Inhibitors. J Immunother 2025; 48:189-195. [PMID: 40231356 PMCID: PMC12052074 DOI: 10.1097/cji.0000000000000558] [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: 12/02/2024] [Accepted: 03/13/2025] [Indexed: 04/16/2025]
Abstract
Responses to immune checkpoint inhibitors (ICIs) differ significantly between uveal melanoma (UM) and cutaneous melanoma (CM) patients. We investigated the immune profile of metastatic UM(mUM) patients and identified markers that are predictive of improved survival. Metastatic liver samples from 189 UM patients and 48 CM patients were analyzed at genomic and transcriptional levels, and survival analysis was performed on a subgroup of 76 ICI-treated mUM patients. UM liver metastases seem to preserve the genomic and immune characteristics of primary UM (pUM), with a low prevalence of ICI markers and high mutation rates of GNA11, GNAQ, BAP1, and SF3B. Compared with mCM, UM liver metastasis showed lower infiltration of several immune cells, but a greater proportion of M2 macrophages. Compared with UM liver metastases, CM liver metastases showed higher expression of G2M checkpoint and EF2 target genes. Among the mUM and mCM samples, expression of G2M and E2F pathway genes was highest in tumors with high TMB values and T-cell inflamed scores. A longer median overall survival (OS) was observed in mUM patients with higher expression of LAG3, HLA class I, or HLA class II; which may represent a small proportion of immune hot tumors. Expression patterns of G2M checkpoint and E2F targets suggest a possible contribution to immunotherapy response.
Collapse
Affiliation(s)
- Yusra F. Shao
- Karmanos Cancer Institute and Wayne State University, Detroit, MI
| | - Yasmine Baca
- Medical Affairs, Caris Life Sciences, Phoenix, AZ
| | | | - Joanne Xiu
- Medical Affairs, Caris Life Sciences, Phoenix, AZ
| | | | | | - Soo J. Park
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | | | - Takami Sato
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | |
Collapse
|
8
|
Reiss KA, Soares KC, Torphy RJ, Gyawali B. Treatment Innovations in Pancreatic Cancer: Putting Patient Priorities First. Am Soc Clin Oncol Educ Book 2025; 45:e473204. [PMID: 40173379 DOI: 10.1200/edbk-25-473204] [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: 04/04/2025]
Abstract
Pancreatic adenocarcinoma remains one of the most aggressive and difficult-to-treat solid tumor malignancies, with a high mortality-to-incidence ratio. Globally, pancreatic cancer ranks 12th in terms of incidence but sixth for mortality signifying its aggressive behavior and limited treatment options. While the mortality rates for many other solid tumors have substantially improved over the past few decades, temporal trends in pancreatic cancer mortality rates are quite sobering. In the United States, from 2000 to 2020, the mortality rates from pancreatic cancer have increased, whereas at the same time, mortality rates from other cancers, such as lung, colorectal, or kidney, have fallen appreciably. Is this for lack of treatment innovation? How do we improve survival for patients with pancreatic cancer? In this chapter, we discuss the recent advances and future directions with targeted therapies and immunotherapies in the treatment of pancreatic cancer, and provide the reasons for both optimism and caution for the future of systemic treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Kim A Reiss
- Division of Hematology/Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Kevin C Soares
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert J Torphy
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bishal Gyawali
- Department of Oncology, Queen's University, Kingston, Canada
- Division of Cancer Care and Epidemiology, Queen's University, Kingston, Canada
- Department of Public Health Sciences, Queen's University, Kingston, Canada
| |
Collapse
|
9
|
Rajak P. Immune checkpoint inhibitors: From friend to foe. Toxicol Rep 2025; 14:102033. [PMID: 40353246 PMCID: PMC12063143 DOI: 10.1016/j.toxrep.2025.102033] [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: 11/30/2024] [Revised: 04/18/2025] [Accepted: 04/20/2025] [Indexed: 05/14/2025] Open
Abstract
Immune checkpoints are crucial in regulating the activation of cell-mediated and humoral immune responses. However, cancer cells hijack this mechanism to evade the immune surveillance and anti-cancer response. Typically, receptors like PD-1 and CTLA4, expressed on immune cells, prevent the activation and differentiation of T cells. They also inhibit the development of autoimmune reactions. However, ligands such as PD-L1 for the receptor PD-1 are also expressed on the surface of cancer cells that help prevent the activation of anti-cancer immune responses by blocking the signalling pathways mediated by PD-1 and CTLA4. Immune checkpoint inhibitors (ICIs) have promising therapeutic efficacy for treating several cancers by activating T cells and their differentiation into effector cells against tumours. Nonetheless, hyperactivated immune cells usually contribute to detrimental issues, also known as immune-related adverse effects (IrAE). IrAEs have been observed in multiple organs, leading to neurological issues, colitis, endocrine dysfunction, renal issues, hepatitis, pneumonitis, and dermatitis. The interplay between hyperactivated T cells and Treg cells helps in orchestrating the development of autoimmunity. Moreover, the crosstalk between proinflammatory interleukins and the development of autoantibodies also mediates the multiorgan effects of ICIs in cancer patients. IrAEs are generally managed by terminating the ICI therapy, reducing the ICI dose, and by using corticosteroids to subvert inflammation. Therefore, the present review aims to delineate the impacts of ICIs on the development of autoimmune diseases and inflammatory outcomes in cancer patients. In addition, mechanistic insight involving immune cells, cytokines, and autoantibodies for ICI-mediated IrAEs will also be discussed with updated findings in this field.
Collapse
Affiliation(s)
- Prem Rajak
- Toxicology Research Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| |
Collapse
|
10
|
Stenzinger A, Westphalen CB, Budczies J, Kazdal D, Ploeger C, Altbürger C, Evert M, Malek N, Schirmacher P, Klauschen F. Comprehensive genomic profiling requires a blended ecosystem of learning healthcare and clinical trials. Br J Cancer 2025; 132:866-868. [PMID: 40251279 DOI: 10.1038/s41416-025-03009-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/03/2025] [Accepted: 03/28/2025] [Indexed: 04/20/2025] Open
Affiliation(s)
- Albrecht Stenzinger
- Center for Personalized Medicine (ZPM), Heidelberg, Germany.
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.
| | - C Benedikt Westphalen
- Comprehensive Cancer Center and Department of Medicine III, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jan Budczies
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Daniel Kazdal
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Carolin Ploeger
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Altbürger
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Nisar Malek
- Center for Personalized Medicine (ZPM), Tübingen, Germany
- Department for Internal Medicine 1, Tübingen University Hospital, Tübingen, Germany
| | - Peter Schirmacher
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Frederick Klauschen
- Institute of Pathology, Ludwig Maximilian University of Munich, Munich, Germany
- BIFOLD-Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| |
Collapse
|
11
|
Jiang Y, Immadi MS, Wang D, Zeng S, On Chan Y, Zhou J, Xu D, Joshi T. IRnet: Immunotherapy response prediction using pathway knowledge-informed graph neural network. J Adv Res 2025; 72:319-331. [PMID: 39097091 DOI: 10.1016/j.jare.2024.07.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/10/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024] Open
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) are potent and precise therapies for various cancer types, significantly improving survival rates in patients who respond positively to them. However, only a minority of patients benefit from ICI treatments. OBJECTIVES Identifying ICI responders before treatment could greatly conserve medical resources, minimize potential drug side effects, and expedite the search for alternative therapies. Our goal is to introduce a novel deep-learning method to predict ICI treatment responses in cancer patients. METHODS The proposed deep-learning framework leverages graph neural network and biological pathway knowledge. We trained and tested our method using ICI-treated patients' data from several clinical trials covering melanoma, gastric cancer, and bladder cancer. RESULTS Our results demonstrate that this predictive model outperforms current state-of-the-art methods and tumor microenvironment-based predictors. Additionally, the model quantifies the importance of pathways, pathway interactions, and genes in its predictions. A web server for IRnet has been developed and deployed, providing broad accessibility to users at https://irnet.missouri.edu. CONCLUSION IRnet is a competitive tool for predicting patient responses to immunotherapy, specifically ICIs. Its interpretability also offers valuable insights into the mechanisms underlying ICI treatments.
Collapse
Affiliation(s)
- Yuexu Jiang
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Manish Sridhar Immadi
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA
| | - Duolin Wang
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Shuai Zeng
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Yen On Chan
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA; MU Institute for Data Science and Informatics, University of Missouri-Columbia, Columbia, MO, USA
| | - Jing Zhou
- Department of Surgery, University of Missouri-Columbia, Columbia, MO, USA
| | - Dong Xu
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri-Columbia, Columbia, MO, USA; MU Institute for Data Science and Informatics, University of Missouri-Columbia, Columbia, MO, USA
| | - Trupti Joshi
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri-Columbia, Columbia, MO, USA; MU Institute for Data Science and Informatics, University of Missouri-Columbia, Columbia, MO, USA; Department of Biomedical Informatics, Biostatistics and Medical Epidemiology, University of Missouri-Columbia, Columbia, MO, USA.
| |
Collapse
|
12
|
Wu H, Li C, Yuan H, Zhao J, Li S. Brain Delivery Strategies for Biomacromolecular Drugs: Intranasal Administration. Int J Nanomedicine 2025; 20:6463-6487. [PMID: 40420915 PMCID: PMC12105674 DOI: 10.2147/ijn.s520768] [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: 02/05/2025] [Accepted: 05/03/2025] [Indexed: 05/28/2025] Open
Abstract
Macromolecular Drugs (including monoclonal antibodies, recombinant proteins, and nucleic acid therapies) have become a cornerstone strategy for intervening in complex pathological mechanisms such as cancer, autoimmune diseases, and genetic disorders due to their high specificity for disease targets and low off-target toxicity. However, compared to traditional small-molecule drugs, the high molecular weight (>10 kDa) and structural complexity of macromolecular drugs result in extremely low transmembrane permeability. This is particularly challenging in the treatment of central nervous system (CNS) diseases, where the blood-brain barrier (BBB) imposes stringent selectivity, further limiting drug delivery efficiency. This review focuses on the breakthrough strategy of nose-to-brain (NtB) drug delivery. On one hand, the NtB pathway bypasses the BBB, enabling direct CNS drug delivery. On the other hand, nanocarrier technology can synergistically achieve systemic delivery and brain-targeted transport. Based on the latest research advances, this article systematically examines the feasibility of delivering macromolecular drugs via NtB administration. We comprehensively summarize relevant delivery carriers and discuss the potential advantages of intranasal-brain delivery for CNS disease treatment. Notably, while significant progress has been made in this field, further exploration is still needed regarding the mechanisms of NtB delivery and challenges in clinical translation.
Collapse
Affiliation(s)
- Huanhuan Wu
- The First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Dalian Medical University, Dalian, People’s Republic of China
| | - Chenyu Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Dalian Medical University, Dalian, People’s Republic of China
| | - Hong Yuan
- Central Hospital of Dalian University of Technology, Dalian, People’s Republic of China
| | - Jingyuan Zhao
- Central Hospital of Dalian University of Technology, Dalian, People’s Republic of China
| | - Shuai Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| |
Collapse
|
13
|
Harel M, Dahan N, Lahav C, Jacob E, Elon Y, Puzanov I, Kelly RJ, Shaked Y, Leibowitz R, Carbone DP, Gandara DR, Dicker AP. Decoding resistance to immune checkpoint inhibitors in non-small cell lung cancer: a comprehensive analysis of plasma proteomics and therapeutic implications. J Immunother Cancer 2025; 13:e011427. [PMID: 40404205 PMCID: PMC12097049 DOI: 10.1136/jitc-2024-011427] [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: 01/13/2025] [Accepted: 05/05/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have shown substantial benefit for patients with advanced non-small cell lung cancer (NSCLC). However, resistance to ICIs remains a major clinical challenge. Here, we perform a comprehensive bioinformatic analysis of plasma proteomic profiles to explore the underlying biology of treatment resistance in NSCLC. METHODS The analysis was performed on 388 "resistance-associated proteins" (RAPs) that were previously described as pretreatment plasma proteomic predictors within the PROphet computational model designed to predict ICI clinical benefit in NSCLC. Putative tissue origins of the RAPs were explored using publicly available datasets. Enrichment analyses were performed to investigate RAP-related biological processes. Plasma proteomic data from 50 healthy subjects and 272 patients with NSCLC were compared, where patients were classified as displaying clinical benefit (CB; n=76) or no CB (NCB; n=196). Therapeutic agents targeting RAPs were identified in drug and clinical trial databases. RESULTS The RAP set was significantly enriched with proteins associated with lung cancer, liver tissue, cell proliferation, extracellular matrix, invasion, and metastasis. Comparison of RAP expression in healthy subjects and patients with NSCLC revealed five distinct RAP subsets that provide mechanistic insights. The RAP subset displaying a pattern of high expression in the healthy population relative to the NSCLC population included multiple proteins associated with antitumor activities, while the subset displaying a pattern of highest expression in the NCB population included proteins associated with various hallmarks of treatment resistance. Analysis of patient-specific RAP profiles revealed inter-patient diversity of potential resistance mechanisms, suggesting that RAPs may aid in developing personalized therapeutic strategies. Furthermore, examination of drug and clinical trial databases revealed that 17.5% of the RAPs are drug targets, highlighting the RAP set as a valuable resource for drug development. CONCLUSIONS The study provides insight into the underlying biology of ICI resistance in NSCLC and highlights the potential clinical value of RAP profiles for developing personalized therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- The Roswell Park Comprehensive Cancer Center Data Bank and BioRepository, Buffalo, New York, USA
| | - Ronan J Kelly
- Department of Hematology and Oncology, Baylor University Medical Center at Dallas, Dallas, Texas, USA
| | - Yuval Shaked
- Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | | | | | - David R Gandara
- Division of Hematology/Oncology, UC Davis Comprehensive Cancer Center, Sacramento, California, USA
| | - Adam P Dicker
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Lee KW, Zang DY, Kim HD, Kim JW, Kim BJ, Kang YK, Ryu MH, Kim HK. Multicenter phase Ib/II study of second-line durvalumab and tremelimumab in combination with paclitaxel in patients with biomarker-selected metastatic gastric cancer. Br J Cancer 2025:10.1038/s41416-025-03052-y. [PMID: 40399487 DOI: 10.1038/s41416-025-03052-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 04/24/2025] [Accepted: 05/01/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND This multicenter phase Ib/II trial aimed to evaluate the safety and efficacy of combining durvalumab, tremelimumab, and paclitaxel as second-line treatment for biomarker-selected patients with metastatic gastric cancer. METHODS In phase Ib, the standard 3 + 3 dose escalation method was used. Durvalumab and tremelimumab were administered every 4 weeks for 13 and 4 cycles, respectively, combining paclitaxel 80 mg/m2 (dose level 2) or 60 mg/m2 (dose level 1) on days 1, 8, and 15. The primary outcome for phase II was the objective response rate (ORR). RESULTS In phase Ib (n = 7), dose level-1 was selected as the recommended phase II dose. In phase II, 48 patients were enrolled: microsatellite instability-high or deficient mismatch repair protein tumors (n = 16); EBV-positive tumors (n = 15); high tumor mutation burden ( ≥ 5/Mb) (n = 11); CD274 amplification (n = 5); and POLD1 mutation (n = 1). The ORR was 52.1%, meeting the primary endpoint. The median progression-free survival and overall survival were 5.3 and 13.1 months, respectively. The most common any-grade and grade 3-4 adverse events were anemia (41.7%) and neutropenia (10.4%), respectively. CONCLUSIONS Durvalumab-tremelimumab with paclitaxel was tolerable and efficacious in biomarker-selected gastric cancer patients as a second-line treatment, highlighting the importance of biomarker-based approaches for immunotherapy in gastric cancer. CLINICAL TRIAL REGISTRATION NCT03751761.
Collapse
Affiliation(s)
- Keun Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Da Young Zang
- Division of Hematology-Oncology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Hyung-Don Kim
- Departmentof Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin-Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Bum Jun Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Yoon-Koo Kang
- Departmentof Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min-Hee Ryu
- Departmentof Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Hark Kyun Kim
- Center for Gastric Cancer, National Cancer Center, Goyang, Republic of Korea.
| |
Collapse
|
15
|
Thorn GJ, Gadaleta E, Dayem Ullah AZM, James LGE, Abdollahyan M, Barrow-McGee R, Jones LJ, Chelala C. The clinical and molecular landscape of breast cancer in women of African and South Asian ancestry. Nat Commun 2025; 16:4237. [PMID: 40394000 DOI: 10.1038/s41467-025-59144-z] [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: 05/15/2024] [Accepted: 04/07/2025] [Indexed: 05/22/2025] Open
Abstract
Addressing existing racial disparity in breast cancer is crucial to ensure equitable benefit across diverse communities. We evaluate the molecular and clinical effects of genetic ancestry in African and South Asian women compared to European using a combined cohort of 7136 breast cancer patients. We find that non-European patients present significantly earlier and die at a younger age. The African group has an increased prevalence of higher grade and hormone receptor negative disease. The South Asian group shows tendency towards lower stage at diagnosis and tumour mutational burden. We observe differences and similarities in the somatic mutational landscape, and differences in germline mutation rates relevant to genetic testing and breast cancer predisposition. Potential therapeutic candidates are identified, with a higher propensity for homologous recombination deficiency serving as a therapy response indicator. We harness breast cancer multimodal data to improve understanding of ancestry-associated differences and highlight opportunities to advance health equity.
Collapse
Affiliation(s)
- Graeme J Thorn
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Emanuela Gadaleta
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Abu Z M Dayem Ullah
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Lewis G E James
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Maryam Abdollahyan
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Rachel Barrow-McGee
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Louise J Jones
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Claude Chelala
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK.
| |
Collapse
|
16
|
Cui Y, Qiao Y, An R, Pan X, Tu J. The application of compressed sensing on tumor mutation burden calculation from overlapped pooling sequencing data. BMC Bioinformatics 2025; 26:129. [PMID: 40394464 PMCID: PMC12090583 DOI: 10.1186/s12859-025-06148-7] [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: 11/10/2023] [Accepted: 04/24/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Tumor Mutation Burden (TMB) is commonly characterized as the number of non-synonymous somatic SNVs per megabase within the gene region identified through whole exon sequencing or targeted sequencing in a tumor sample. It has been statistically demonstrated that TMB was related to the ability of neoantigen production and used to predict the efficacy of immunotherapy for various types of cancers. However, screening for TMB in patients poses challenges due to the extensive labor and financial resources required for the preparation of large quantities of parallel sequencing libraries. RESULTS In this study, we employed compressed sensing (CS) to calculate TMB from overlapped pooling sequencing data, aiming to reduce the sequencing cost by minimizing the number of library builds. Over 90% SNPs could still be detected without a significant loss of mutation information even when the data is pooled from ten different samples. Based on this, the orthogonal matching pursuit (OMP) algorithm and the basic pursuit (BP) algorithm were used to reconstruct TMB from pooling sequencing data. The performance of these two algorithms was evaluated. The BP algorithm consistently performed well across all cases, albeit necessitating extended computational time. The OMP algorithm has been proved to be suitable for scenarios where the original matrix was sparse but it showed low overall performance. Based on an accurate calculation of TMB, we determined that the number of sequencing runs could be reduced to 0.6 times the total number of samples, resulting in a 40% reduction in sequencing cost. CONCLUSIONS In conclusion, we calculated TMB from overlapped pooling sequencing data utilizing compressed sensing strategy to reduce sequencing cost. Our findings confirm that the SNP calling from ten samples' pooling sequencing data is feasible. Additionally, we performed an assessment of the reconstruction efficiency of both the BP model and the OMP model.
Collapse
Affiliation(s)
- Yue Cui
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yi Qiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Rongming An
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Monash University-Southeast University Joint Research Institute, Suzhou, 215123, China
| | - Xuan Pan
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China.
| | - Jing Tu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
17
|
Shang Y, Pang Y, Liu T, Wang W. Application of mass cytometry in the immune microenvironment of breast cancer. Med Oncol 2025; 42:215. [PMID: 40388018 DOI: 10.1007/s12032-025-02770-w] [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: 02/08/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
The rapid development of immunotherapy has shown preliminary clinical efficacy and significant anti-tumor effects in some cancer patients. Although immunotherapy has been approved for breast cancer, some breast cancer patients still do not benefit from it due to issues such as immunotherapy insensitivity and resistance. Mass cytometry, as a mature single-cell proteomic analysis method, with its high-throughput capabilities, has been widely used in the analysis of tumor immune microenvironments and immune cell subpopulations. Using mass cytometry to analyze the immune microenvironment of breast cancer and explore new immunotherapy targets can help improve the current status of breast cancer immunotherapy and develop personalized treatment plans for more patients. This review surveys the recent advancements in analyzing the single-cell components of breast cancer using mass cytometry technology and reviews the immune microenvironment of breast cancer as well as potential targets for immunotherapy. These results provide new insights for the subsequent research of the immune microenvironment of breast cancer and targeted immunotherapy.
Collapse
Affiliation(s)
- Yuefeng Shang
- Department of Radiation Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Yuheng Pang
- Department of Radiation Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Tong Liu
- Department of Radiation Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen Wai, Fengtai District, Beijing, 100069, People's Republic of China.
| |
Collapse
|
18
|
Yamakawa K, Ogata D, Namikawa K, Nakano E, Yamaguchi Y, Yamazaki N. A review of cutaneous angiosarcoma: epidemiology, diagnosis, prognosis, and treatment options. Jpn J Clin Oncol 2025:hyaf071. [PMID: 40381218 DOI: 10.1093/jjco/hyaf071] [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: 02/03/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025] Open
Abstract
Cutaneous angiosarcoma (cAS) is a rare and aggressive malignant vascular tumor that arises from endothelial cells lining the blood vessels. It can occur in any part of the body, but most commonly, it affects the skin and soft tissues. cAS has a poor prognosis with a 5-year survival rate of only 9%. This review summarizes the current understanding of angiosarcoma pathogenesis, clinical presentation, diagnosis, and treatment approaches. Recent advances in molecular characterization have identified recurrent genetic alterations that may lead to the development of novel targeted therapies. Multidisciplinary management combining surgery, radiation, and systemic therapy remains the mainstay of treatment; however, outcomes remain poor for metastatic disease. Ongoing research into the molecular drivers of cAS and immunotherapeutic approaches offers hope for improving the outcomes of this challenging malignancy.
Collapse
Affiliation(s)
- Kohei Yamakawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Dai Ogata
- Department of Dermatologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Eiji Nakano
- Department of Dermatologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Yukie Yamaguchi
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
19
|
Barroso-Sousa R, Zanudo JGT, Li T, Reddy SM, Emens LA, Kuntz TM, Silva CAC, AlDubayan SH, Chu H, Overmoyer B, Lange P, DiLullo MK, Montesion M, Kasparian J, Hughes ME, Attaya V, Basta A, Lin NU, Tayob N, Jeselsohn R, Mittendorf EA, Tolaney SM. Nivolumab plus low-dose ipilimumab in hypermutated HER2-negative metastatic breast cancer: a phase II trial (NIMBUS). Nat Commun 2025; 16:4430. [PMID: 40360544 PMCID: PMC12075640 DOI: 10.1038/s41467-025-59695-1] [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] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
In the phase II NIMBUS trial, patients with human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer (MBC) and high tumor mutational burden (TMB ≥ 9 mut/Mb) received nivolumab (3 mg/kg biweekly) and low-dose ipilimumab (1 mg/kg every 6 weeks) for 2 years or until progression. The primary endpoint was objective response rate (ORR) per RECIST 1.1 criteria. Among 30 patients enrolled, the median TMB was 10.9 mut/Mb (range: 9-110) and the confirmed objective response rate was 20%. Secondary endpoints included progression-free survival, overall survival, clinical benefit rate, and safety and tolerability, including immune-related adverse events (irAEs). A prespecified correlative outcome was to evaluate the ORR in patients with a TMB ≥ 14 mut/Mb. Patients with TMB ≥ 14 mut/Mb (n = 6) experienced higher response rates (60% vs 12%; p = 0.041) and showed a trend towards improved progression-free survival and overall survival compared to patients with TMB < 14 mut/Mb. Exploratory genomic analyses suggested that ESR1 and PTEN mutations may be associated with poor response, while clinical benefit was associated with a decrease or no change in tumor fraction by serial circulating tumor DNA during treatment. Stool microbiome analysis revealed that baseline blood TMB, PD-L1 positivity, and immune-related diarrhea are associated with distinct taxonomic profiles. In summary, some patients with hypermutated HER2-negative MBC experience extended clinical benefit with a dual immunotherapy regimen; a higher TMB, and additional genomic and microbiome biomarkers may optimize patient selection for therapy with nivolumab plus low-dose ipilimumab. (Funded by Bristol Myers Squibb; ClinicalTrials.gov identifier, NCT03789110).
Collapse
Affiliation(s)
| | - Jorge Gomez Tejeda Zanudo
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tianyu Li
- Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Leisha A Emens
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Thomas M Kuntz
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | | | | | - Hoyin Chu
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Beth Overmoyer
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Paulina Lange
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Molly K DiLullo
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | | | - Julie Kasparian
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Melissa E Hughes
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Victoria Attaya
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Ameer Basta
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Nancy U Lin
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nabihah Tayob
- Harvard Medical School, Boston, MA, USA
- Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rinath Jeselsohn
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Mittendorf
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Sara M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Camus V, Molina T, Desmots F, Blanc-Durand P, Kanoun S, Moslemi A, Ruminy P, Le Gouill S, Ghesquières H, Oberic L, Morschhauser F, Tilly H, Ribrag V, Houot R, Thieblemont C, Maisonneuve H, Claves F, Bouabdallah K, Haioun C, Damaj GL, Fornecker LM, Noel R, Feugier P, Sibon D, Cartron G, Bonnet C, Bernard W, Kraeber-Bodéré F, Bodet-Milin C, Jais JP, Brière J, Rossi C, Elsensohn MH, Chartier L, Itti E, Jardin F, Fest T. Interim PET after 4 cycles predicts outcome in histomolecularly confirmed primary mediastinal B-cell lymphoma. Blood Adv 2025; 9:2232-2246. [PMID: 40030008 PMCID: PMC12088757 DOI: 10.1182/bloodadvances.2024015577] [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: 12/10/2024] [Accepted: 02/05/2025] [Indexed: 05/01/2025] Open
Abstract
ABSTRACT The GAINED study was a randomized phase 3 trial comparing obinutuzumab (G) with rituximab (R) plus ACVBP (doxorubicin, cyclophosphamide, and prednisone, combined with either vindesine or bleomycin) or CHOP14 (cyclophosphamide, doxorubicin, vincristine, and prednisone, administered on a 14-day schedule) induction, followed by positron emission tomography (PET)-guided consolidation. This post hoc analysis aimed to detail the outcomes of patients with primary mediastinal B-cell lymphoma (PMBL), verified through expert pathological review and the use of gene expression profiling (GEP) and next-generation sequencing. Of 620 centrally reviewed patients, 138 (22.3%) confirmed PMBL cases were analyzed. Baseline characteristics included a median age of 33.5 years, 63.8% female, 55.1% stage III to IV, 90.6% elevated lactate dehydrogenase, 87.6% Eastern Cooperative Oncology Group performance status score of 0 to 1, 62.3% extranodal involvement, 52.6% age-adjusted International Prognostic Index (aaIPI) of 2% to 3%, and 53.6% bulk (>10 cm). Induction regimens were R/G-CHOP14 (56.9%) and R/G-ACVBP (43.1%). Postinduction treatments, based on interim PET results, included: standard consolidation chemotherapy (59.8%) if change in maximum standardized uptake value (ΔSUVmax) of >66% after cycle 2 and >70% after cycle 4 (PET2-/4-), intensive treatment and autologous transplantation (26.8%) if PET2+/4-, and salvage therapy (13.4%) if PET4+ (ΔSUVmax of ≤70%). Among patients with GEP data (n = 107), 38 (35.5%) were PDL1high/PDL2high. Key somatic mutations data (n = 87) included SOCS1 (70.1%), B2M (56.3%), STAT6 (49.4%), TNFAIP3 (47.1%), GNA13 (39.1%), CIITA (37.9%), CD58 (36.8%), and TP53 (29.9%). After a median follow-up of 39.5 months, 2-year progression-free survival (PFS) and overall survival (OS) rates were 86.2% and 93.2%, respectively. In a multivariate model including bulk, aaIPI, and ΔSUVmax PET2/PET4, only bulk and ΔSUVmax PET4 of ≤70% were associated with shorter PFS (hazard ratio, 4.39 [95% confidence interval (CI), 1.28-15.11] and 4.95 [95% CI, 1.71-14.3], respectively), whereas none were associated with OS. The ΔSUVmax-based interim PET4 response emerged as the strongest predictor of patient outcomes in this selected clinical trial population. This trial was registered at www.ClinicalTrials.gov as #NCT01659099.
Collapse
Affiliation(s)
- Vincent Camus
- Department of Hematology, Centre Henri Becquerel, Rouen, France
- INSERM U1245, Centre Henri Becquerel, University of Rouen, Rouen, France
| | - Thierry Molina
- Department of Pathology, Assistance Publique–Hôpitaux de Paris, Necker and Robert Debré Hospital, Université Paris Cité, Paris, France
| | - Fabienne Desmots
- Laboratoire d'Hématologie, Team B_DEVIL, UMR_S1236, Centre Hospitalier Universitaire de Rennes, Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, Rennes, France
| | - Paul Blanc-Durand
- Department of Nuclear Medicine, CHU Henri Mondor, Nuclear Medicine, Paris-Est University, Créteil, France
| | - Salim Kanoun
- INSERM, UMR 1037, Cancer Research Center of Toulouse, Toulouse, France
| | - Amine Moslemi
- Department of Pathology, Centre Hospitalier Universitaire Amiens, Amiens, France
| | - Philippe Ruminy
- INSERM U1245, Centre Henri Becquerel, University of Rouen, Rouen, France
| | | | - Hervé Ghesquières
- Department of Hematology, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Lucie Oberic
- Department of Hematology, Institut Universitaire du Cancer, Toulouse-Oncopole, Toulouse, France
| | - Franck Morschhauser
- Department of Hematology, Claude Huriez Hospital, Lille University Hospital, Lille, France
| | - Hervé Tilly
- Department of Hematology, Centre Henri Becquerel, Rouen, France
- INSERM U1245, Centre Henri Becquerel, University of Rouen, Rouen, France
| | - Vincent Ribrag
- Department of Hematology, Gustave Roussy, Villejuif, France
| | - Roch Houot
- Department of Hematology, Centre Hospitalier Universitaire Rennes, University of Rennes, INSERM U1236, Etablissement Français du Sang, Rennes, France
| | - Catherine Thieblemont
- Department of Hemato-oncology, Assistance Publique–Hôpitaux de Paris, Hôpital Saint-Louis, Université de Paris, Paris, France
| | - Hervé Maisonneuve
- Department of Hematology, Centre Hospitalier Departemental de Vendée, La Roche sur Yon, France
| | - Fabien Claves
- Department of Hematology, Grenoble University Hospital, Grenoble, France
| | - Krimo Bouabdallah
- Department of Hematology, Bordeaux University Hospital, Bordeaux, France
| | - Corinne Haioun
- Department of Hematology, Henri Mondor University Hospital, Créteil, France
| | | | | | - Robin Noel
- Department of Hematology, Institut Paoli-Calmettes, Marseille, France
| | - Pierre Feugier
- Department of Hematology, Hôpital de Brabois, Nancy University Hospital, Nancy, France
| | - David Sibon
- Department of Hematology, Assistance Publique–Hôpitaux de Paris, Hôpital Necker, Université de Paris, Paris, France
| | - Guillaume Cartron
- Department of Hematology, Montpellier University Hospital, Montpellier, France
| | - Christophe Bonnet
- Department of Hematology, Centre Hospitalier Universitaire Liege, Liege, Belgium
| | - Wivine Bernard
- Department of Hematology, CHU Université Catholique de Louvain Namur, Site Godinne, Namur, Belgium
| | | | | | - Jean-Philippe Jais
- Department of Biostatistics, Hôpital Necker, Institut Imagine, Unité INSERM 1163, University of Paris, Paris, France
| | - Josette Brière
- Department of Hematology, Hôpital Saint-Louis, Assistance Publique–Hôpitaux de Paris, Université Paris Diderot, Paris, France
| | - Cedric Rossi
- Department of Hematology, Dijon University Hospital, Dijon, France
| | - Mad-Hélénie Elsensohn
- Biostatistics Department, The Lymphoma Academic Research Organisation, Lyon-Sud Hospital, Pierre-Bénite, France
| | - Loïc Chartier
- Biostatistics Department, The Lymphoma Academic Research Organisation, Lyon-Sud Hospital, Pierre-Bénite, France
| | - Emmanuel Itti
- Department of Nuclear Medicine, CHU Henri Mondor, Nuclear Medicine, Paris-Est University, Créteil, France
| | - Fabrice Jardin
- Department of Hematology, Centre Henri Becquerel, Rouen, France
- INSERM U1245, Centre Henri Becquerel, University of Rouen, Rouen, France
| | - Thierry Fest
- Laboratoire d'Hématologie, Team B_DEVIL, UMR_S1236, Centre Hospitalier Universitaire de Rennes, Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, Rennes, France
| |
Collapse
|
21
|
Zhu Y, Gao Y, Huang X, Chen B, Wang X, Wu Y, Sun J, Huang X. Pan-cancer analysis reveals TRA16 as a master regulator of human carcinogenesis. Front Oncol 2025; 15:1543419. [PMID: 40432923 PMCID: PMC12106028 DOI: 10.3389/fonc.2025.1543419] [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: 12/11/2024] [Accepted: 04/16/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction The nuclear receptor TR4 binding protein, TRA16, has been implicated in lung carcinogenesis; however, its broader role across diverse human cancers remains poorly understood. Understanding TRA16's involvement in cancer biology could uncover novel regulatory mechanisms and potential therapeutic targets. Methods We conducted a comprehensive pan-cancer analysis of TRA16 expression and function across multiple human malignancies. Gene co-expression networks, pathway enrichment, transcription factor analysis, organoid modeling, and intercellular communication profiling were employed. Tumor mutation burden (TMB) and microenvironmental features were also assessed in relation to TRA16 expression, stratified by TP53 mutation status. Results Correlation analysis identified the cell cycle as the top enriched pathway among TRA16-associated genes, with key transcription factors, including RB-E2F, MYC, and TP53, regulating genes co-expressed with TRA16. In liver cancer organoid models, TRA16 and its co-expressed genes were significantly upregulated. Intercellular communication analysis showed that TRA16-positive cells exhibited increased autocrine signaling and overall signaling activity. Importantly, patients with high TRA16 expression demonstrated elevated TMB and decreased stromal and immune features. Discussion These findings highlight TRA16 as a potential master regulator of oncogenic processes, contributing to tumor progression through coordinated regulation of cell cycle genes, intercellular signaling, and genomic instability. Our results provide new insights into TRA16's role across cancers and support its potential as a novel oncogene.
Collapse
Affiliation(s)
- Yanyan Zhu
- Department of Oncology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Yike Gao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaoqing Huang
- Intelliphecy Center for Systems Medicine, Intelliphecy, Shenzhen, China
| | - Bowang Chen
- Intelliphecy Center for Systems Medicine, Intelliphecy, Shenzhen, China
- Department of Data Science, Intelliphecy, Nanjing, China
| | - Xinyi Wang
- Department of Dermatology, Southern University of Science and Technology Yantian Hospital, Shenzhen, China
| | - Ying Wu
- Department of Obstetrics and Gynecology, Southern University of Science and Technology Yantian Hospital, Shenzhen, China
| | - Jian Sun
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaoyun Huang
- Intelliphecy Center for Systems Medicine, Intelliphecy, Shenzhen, China
- College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
22
|
Lu J, Luo F. MYB Proto-Oncogene Like 2 identified as a biomarker for uterine corpus endometrial carcinoma: evidence from bioinformatics and clinical validation. Front Oncol 2025; 15:1595485. [PMID: 40432915 PMCID: PMC12106007 DOI: 10.3389/fonc.2025.1595485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Background Endometrial carcinoma (EC) is the sixth most prevalent malignancy among women globally, posing a significant clinical challenge due to limited therapeutic options for advanced or recurrent cases. The identification of novel prognostic biomarkers and therapeutic targets is crucial for improving patient outcomes. This study aimed to investigate the multifaceted roles of MYB Proto-Oncogene Like 2 (MYBL2) in uterine corpus endometrial carcinoma (UCEC). Methods We employed multiple bioinformatics algorithms (GEPIA, TCGA, TIMER2.0) to analyze MYBL2 expression across different cancer types and in UCEC specifically. Expression patterns were validated using quantitative real-time PCR (qPCR) on clinical samples. Epigenetic analyses focused on promoter methylation status, and immune infiltration patterns were assessed using MethSurv, CIBERSORT and TIMER2.0. Drug sensitivity profiling was performed using the CPADS web platform. Results MYBL2 was found to be significantly upregulated in UCEC tumors compared to normal tissues. Elevated MYBL2 expression correlated with advanced histologic grade and clinical stage, indicating its potential as a biomarker for disease progression. Epigenetic analysis revealed promoter hypomethylation in tumors, suggesting a regulatory mechanism driving MYBL2 overexpression. MYBL2 demonstrated dynamic interactions with the tumor immune microenvironment, including associations with immune cell infiltration patterns and co-expression with immune checkpoint molecules and chemokines. Drug sensitivity profiling highlighted differential therapeutic responses linked to MYBL2 expression levels. Conclusion This study establishes MYBL2 as a critical regulator of UCEC progression, bridging epigenetic dysregulation, immune modulation, and clinical outcomes. The findings provide a foundation for exploring MYBL2-targeted strategies in precision immunotherapy and personalized therapeutic interventions.
Collapse
Affiliation(s)
- Jiaoyun Lu
- Department of Oncology, Xi’an NO.3 Hospital, The Affiliated Hospital of Northwest University, Xi’an, Shaanxi, China
| | - Furong Luo
- Department of Traditional Chinese Medicine, Xi’an NO.3 Hospital, The Affiliated Hospital of Northwest University, Xi’an, Shaanxi, China
| |
Collapse
|
23
|
Kouhen F, El Ghanmi A, Inghaoun H, Miftah H, Ghazi B, Badou A. The promise of PD1/PDL1 targeted immunotherapy in locally advanced cervical cancer: a game-changer for patients outcome? Front Immunol 2025; 16:1573576. [PMID: 40433369 PMCID: PMC12106400 DOI: 10.3389/fimmu.2025.1573576] [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: 02/09/2025] [Accepted: 04/14/2025] [Indexed: 05/29/2025] Open
Abstract
Locally advanced cervical cancer remains a significant therapeutic challenge, with high rates of recurrence and metastasis despite advances in chemoradiation. Immunotherapy, particularly immune checkpoint inhibitors targeting the PD-1/PD-L1 axis, has emerged as a promising strategy to enhance treatment efficacy. This review explores the integration of immunotherapy with standard chemoradiation, highlighting the potential of PD-1 inhibitors, such as pembrolizumab, in improving progression-free survival (PFS) among high-risk patients. Furthermore, the role of predictive biomarkers, including microsatellite instability (MSI) and tumor mutational burden (TMB), is examined to refine patient selection and personalize therapeutic approaches. Emerging strategies, including the use of nivolumab, ipilimumab, and maintenance immunotherapy, are also discussed. While preliminary clinical data are encouraging, further research is required to optimize treatment combinations, establish robust patient selection criteria, and enhance long-term outcomes in cervical cancer management.
Collapse
Affiliation(s)
- Fadila Kouhen
- Mohammed VI Faculty of Medicine, Mohammed VI University of Sciences and Health (UM6SS), Rabat, Morocco
- Laboratory of Neurooncology, Oncogenetic and Personalized Medicine, Faculty of Medicine, Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
- Department of Radiotherapy, International University Hospital Cheikh Khalifa, Casablanca, Morocco
| | - Adil El Ghanmi
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
- Department of Gynecology and Obstetrics, Mohammed VI International University Hospital, Bouskoura, Morocco
| | - Hanane Inghaoun
- Laboratory of Neurooncology, Oncogenetic and Personalized Medicine, Faculty of Medicine, Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
| | - Hayat Miftah
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy, Casablanca, Morocco
| | - Bouchra Ghazi
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
- Department of Gynecology and Obstetrics, Mohammed VI International University Hospital, Bouskoura, Morocco
| | - Abdallah Badou
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy, Casablanca, Morocco
| |
Collapse
|
24
|
Rödel F, Fleischmann M, Diefenhardt M, Dapper H, Hoffmann A, Rödel C, Martin D, Fokas E. Emerging advances and future opportunities in the molecular and therapeutic landscape of anal cancer. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01025-x. [PMID: 40360682 DOI: 10.1038/s41571-025-01025-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2025] [Indexed: 05/15/2025]
Abstract
Anal squamous cell carcinoma (ASCC) is a rare malignancy with an increasing incidence. Primary chemoradiotherapy (CRT) is the standard-of-care treatment for patients with localized ASCC. In the metastatic setting, trials testing immune-checkpoint inhibitor monotherapy have demonstrated outcomes similar to those of patients receiving chemotherapy. Conversely, adding the anti-PD-1 antibody retifanlimab to chemotherapy in patients with recurrent or metastatic ASCC has been shown to significantly improve outcomes. Despite considerable efforts to develop personalized therapy, treatment guidance and prognosis remain reliant on baseline clinical characteristics. An improved understanding of the molecular characteristics of ASCC has provided insights into the mechanisms that mediate tumour progression and response to CRT. For example, human papillomavirus (HPV) infection is known to have an aetiological role in most ASCCs and can modulate cellular responses to CRT via several distinct mechanisms. In this Review, we summarize emerging advances in the molecular and therapeutic landscape of ASCC, including the implementation of biomarkers for treatment guidance and translation into new therapeutic approaches, with HPV infection constituting a global determinant of both tumour biology and clinical outcome. We also discuss the rationale for combining immune-checkpoint inhibitors with CRT in patients with HPV+ tumours.
Collapse
Affiliation(s)
- Franz Rödel
- Department of Radiotherapy and Oncology, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
| | - Maximilian Fleischmann
- Department of Radiotherapy and Oncology, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
| | - Markus Diefenhardt
- Department of Radiotherapy and Oncology, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
| | - Hendrik Dapper
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Annett Hoffmann
- Department of Radiotherapy and Oncology, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| | - Claus Rödel
- Department of Radiotherapy and Oncology, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
| | - Daniel Martin
- Department of Radiotherapy and Oncology, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
| | - Emmanouil Fokas
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany.
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
25
|
Arango-Argoty G, Bikiel DE, Sun GJ, Kipkogei E, Smith KM, Carrasco Pro S, Choe EY, Jacob E. AI-driven predictive biomarker discovery with contrastive learning to improve clinical trial outcomes. Cancer Cell 2025; 43:875-890.e8. [PMID: 40250446 DOI: 10.1016/j.ccell.2025.03.029] [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: 08/06/2024] [Revised: 12/20/2024] [Accepted: 03/26/2025] [Indexed: 04/20/2025]
Abstract
Modern clinical trials can capture tens of thousands of clinicogenomic measurements per individual. Discovering predictive biomarkers, as opposed to prognostic markers, remains challenging. To address this, we present a neural network framework based on contrastive learning-the Predictive Biomarker Modeling Framework (PBMF)-that explores potential predictive biomarkers in an automated, systematic, and unbiased manner. Applied retrospectively to real clinicogenomic datasets, particularly for immuno-oncology (IO) trials, our algorithm identifies biomarkers of IO-treated individuals who survive longer than those treated with other therapies. We demonstrate how our framework retrospectively contributes to a phase 3 clinical trial by uncovering a predictive, interpretable biomarker based solely on early study data. Patients identified with this predictive biomarker show a 15% improvement in survival risk compared to those in the original trial. The PBMF offers a general-purpose, rapid, and robust approach to inform biomarker strategy, providing actionable outcomes for clinical decision-making.
Collapse
Affiliation(s)
| | - Damian E Bikiel
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | - Gerald J Sun
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | - Elly Kipkogei
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | - Kaitlin M Smith
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | | | - Elizabeth Y Choe
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | - Etai Jacob
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA.
| |
Collapse
|
26
|
Wiertsema P, Tan YH, Haanen JBAG, Seijkens TTP, Jedema I. Advances in TIL therapy: Expanding the horizons beyond melanoma. MED 2025:100702. [PMID: 40381620 DOI: 10.1016/j.medj.2025.100702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/08/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
Tumor-infiltrating lymphocyte (TIL) therapy represents a breakthrough in solid tumor treatment, addressing unmet needs for patients with limited options. While its efficacy is established in advanced melanoma, TIL therapy shows early promise in non-small cell lung cancer, breast cancer, gynecological cancers, and head and neck cancers. However, challenges such as reduced T cell infiltration, lower tumor mutational burden (TMB), immunosuppressive tumor microenvironments (TME), and toxicity associated with the TIL therapy regimen hinder its broader application in these patient groups, compared with melanoma. To address these challenges, new approaches focus on the selection of tumor-reactive TIL, optimization of TIL expansion, combination of immune checkpoint inhibitors with TIL therapy to counteract immunosuppressive microenvironments, and genetic modification of TIL to enhance persistence and functionality. Larger clinical trials are essential to validate these innovations and standardize protocols. With continued advancements, TIL therapy has the potential to redefine the treatment landscape for advanced solid cancers.
Collapse
Affiliation(s)
- Pauline Wiertsema
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ya Hwee Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore; Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - John B A G Haanen
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands; Melanoma Clinic, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Tom T P Seijkens
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Inge Jedema
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
27
|
Teleanu MV, Schneider A, Ball CR, Leber MF, Stange C, Krieghoff-Henning E, Beck K, Heilig CE, Kreutzfeldt S, Kuster B, Lipka DB, Fröhling S. Celebrating Ulrik Ringborg: Multi-Omics-Based Patient Stratification for Precision Cancer Treatment. Biomolecules 2025; 15:693. [PMID: 40427586 DOI: 10.3390/biom15050693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/03/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Precision oncology is becoming a mainstay in the standard of care for cancer patients. Recent technological advancements have significantly lowered the cost of various tumor profiling approaches, broadening the reach of molecular diagnostics and improving patient access to precision oncology. In parallel, drug development and discovery pipelines continue to evolve, driving targeted therapeutic options forward. Yet, not all patients harboring actionable molecular alterations respond to these interventions, and existing therapies do not cover the entire spectrum of potential molecular targets. In this review, we examine the current suite of omics technologies employed in clinical settings and underscore their roles in deepening our understanding of tumor biology and optimizing patient stratification for targeted treatments. We also highlight relevant precision oncology trials and share our own experiences using multi-omics data within a molecular tumor board framework. Finally, we discuss areas for future exploration aimed at propelling precision oncology to new heights.
Collapse
Affiliation(s)
- Maria-Veronica Teleanu
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Annika Schneider
- School of Life Sciences, Technical University Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Claudia R Ball
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT)/University Cancer Center Dresden, a Partnership Between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, Dresden University of Technology (TUD), and Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Dresden University of Technology (TUD), 01069 Dresden, Germany
- Faculty of Biology, Dresden University of Technology (TUD), 01217 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany
| | - Mathias Felix Leber
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- Department of Medical Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Christoph Stange
- School of Life Sciences, Technical University Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Eva Krieghoff-Henning
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- Division of Personalized Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Personalized Oncology, DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Department of Personalized Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Katja Beck
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Christoph E Heilig
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Simon Kreutzfeldt
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Bernhard Kuster
- School of Life Sciences, Technical University Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Daniel B Lipka
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Stefan Fröhling
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
- Institute of Human Genetics, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
28
|
Sugitani I, Kiyota N, Ito Y, Onoda N, Hiromasa T, Horiuchi K, Kinuya S, Kondo T, Moritani S, Sugino K, Hara H. The 2024 revised clinical guidelines on the management of thyroid tumors by the Japan Association of Endocrine Surgery. Endocr J 2025; 72:545-635. [PMID: 40058844 PMCID: PMC12086281 DOI: 10.1507/endocrj.ej24-0644] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 12/31/2024] [Indexed: 05/09/2025] Open
Abstract
The Japan Association of Endocrine Surgery published the first edition of the "Clinical guidelines on the management of thyroid tumors" in 2010 and the revised edition in 2018. The guideline presented herein is the English translation of the revised third edition, issued in 2024. The aim is to enhance health outcomes for patients suffering from thyroid tumors by facilitating evidence-based shared decision-making between healthcare providers and patients, as well as standardizing the management of thyroid tumors. The focus is on adult patients with thyroid tumors, addressing clinically significant issues categorized into areas such as an overview of the diagnosis and treatment of thyroid nodules, treatment strategies by histological type, radioactive iodine therapy, treatment of advanced differentiated carcinoma, pharmacotherapy, and complications and safety management associated with thyroid surgery. Thirty-two clinical questions were established in these areas. Following a comprehensive search of the literature and systematic review to evaluate the overall evidence, we aimed to present optimal recommendations by considering the balance of benefits and harms from the patient's perspective. We integrated evidence and clinical experience to determine the "Certainty of evidence" and "Strength of recommendations". Based on these, we illustrated overall flows of care as "Clinical algorithms". Necessary background knowledge of diseases and established clinical procedures for understanding the recommendations are presented in "Notes", while information that may be clinically useful but for which evidence remains insufficient is included in "Columns", based on the current state of evidence. Finally, future challenges for the next revision are presented as "Future research questions".
Collapse
Affiliation(s)
- Iwao Sugitani
- Department of Endocrine Surgery, Nippon Medical School, Tokyo 113-8603, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Naomi Kiyota
- Department of Medical Oncology and Hematology, Cancer Center, Kobe University Hospital, Kobe 650-0017, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Yasuhiro Ito
- Department of Surgery, Kuma Hospital, Kobe 650-0011, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Naoyoshi Onoda
- Department of Surgery, Kuma Hospital, Kobe 650-0011, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Tomo Hiromasa
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa 920-8641, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Kiyomi Horiuchi
- Department of Endocrine Surgery, Tokyo Women’s Medical University, Tokyo 162-8666, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University, Kanazawa 920-8641, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Tetsuo Kondo
- Department of Pathology, University of Yamanashi, Yamanashi 409-3898, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Sueyoshi Moritani
- Center for Head and Neck Thyroid Surgery, Oumi Medical Center, Shiga 525-8585, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Kiminori Sugino
- Surgical Branch, Ito Hospital, Tokyo 150-8308, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| | - Hisato Hara
- Department of Breast and Endocrine Surgery, University of Tsukuba, Tsukuba 305-8576, Japan
- the Task Force of the Japan Association of Endocrine Surgery on the Guidelines for Thyroid Tumors
| |
Collapse
|
29
|
Soupir A, Ospina OE, Hampton O, Churchman M, Radmacher M, Hedges D, McKean D, Agius P, Zeeshan S, Seligson ND, Pollock R, Liebner D, Chen JL, Tinoco G, Salhia B, McCarter M, Wilky BA, Miller BJ, Cavnar MJ, Groundland JS, Schneider BP, Riedlinger G, Edge SB, Moskaluk CA, Cardona K, Naqash AR, Gonzalez RJ, Mullinax JE, Joyce DM, Binitie O, Douglas Letson G, Naghavi AO, Druta M, Reed DR, Siegel EM, Teer JK, Fridley BL, Brohl AS. Genomic, transcriptomic, and immunogenomic landscape of over 1300 sarcomas of diverse histology subtypes. Nat Commun 2025; 16:4206. [PMID: 40328759 PMCID: PMC12055966 DOI: 10.1038/s41467-025-58678-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Given their rarity and diversity, a fundamental understanding of the genomic underpinnings for many sarcoma subtypes is still lacking. To better define the molecular landscape of this group of diseases, we perform matched whole exome sequencing and RNA sequencing on a cohort of 1340 sarcoma tumor specimens. We identify recurrent somatic mutations and observe an increased mutational burden in metastatic vs. primary samples (p < 0.001). We observe frequent copy number alterations including whole genome doubling, with this feature being more common in metastatic tumors (p = 0.026). Estimation of immune cell abundances followed by hierarchical clustering identifies five immune subtypes ranging from low to high and we observe inferior overall survival in immune deplete clusters compared to immune enriched (p < 0.01). Interestingly, GIST predominantly form a distinct "immune intermediate" cluster that is marked by a specific enrichment for NK cells (FDR < 0.01).
Collapse
Affiliation(s)
- Alex Soupir
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Oscar E Ospina
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | | | | | | | | | | | | | - Saman Zeeshan
- Department of Biomedical and Health Informatics, School of Medicine, University of Missouri, Kansas City, MO, USA
| | - Nathan D Seligson
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL, USA
| | - Raphael Pollock
- Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - David Liebner
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - James L Chen
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Gabriel Tinoco
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bodour Salhia
- Department of Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | | | - Benjamin J Miller
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IO, USA
| | - Michael J Cavnar
- Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - John S Groundland
- University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Bryan P Schneider
- Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA
| | | | - Stephen B Edge
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Kenneth Cardona
- Division of Surgical Oncology, Emory University, Atlanta, GA, USA
| | - Abdul Rafeh Naqash
- Medical Oncology/Phase 1 program, Stephenson Cancer Center, University of Oklahoma Health Sciences, Oklahoma City, OK, USA
| | | | | | - David M Joyce
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL, USA
| | - Odion Binitie
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Arash O Naghavi
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Mihaela Druta
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL, USA
| | - Damon R Reed
- Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, FL, USA
| | - Erin M Siegel
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Brooke L Fridley
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
- Division of Health Services and Outcomes Research, Children's Mercy, Kansas City, MO, USA
| | - Andrew S Brohl
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
30
|
Peters S, Oliner KS, L'Hernault A, Ratcliffe M, Madison H, Lai Z, Stewart R, Mann H, Lowery C, Garon EB, Mok T, Johnson ML. Durvalumab with or without tremelimumab in combination with chemotherapy in first-line metastatic non-small-cell lung cancer: outcomes by tumor mutational burden in POSEIDON. ESMO Open 2025; 10:105058. [PMID: 40334315 DOI: 10.1016/j.esmoop.2025.105058] [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: 12/03/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND In updated analyses from the phase III POSEIDON study, after a median follow-up of >5 years, tremelimumab plus durvalumab and chemotherapy (T + D + CT) showed durable long-term overall survival (OS) benefit versus CT alone in first-line metastatic non-small-cell lung cancer (mNSCLC). In this article, we report the associations of tumor mutational burden (TMB) with outcomes of D with or without T in combination with CT versus CT alone. PATIENTS AND METHODS A total of 1013 patients with EGFR/ALK wild-type mNSCLC were randomized (1 : 1 : 1) to T + D + CT, D + CT, or CT, stratified by programmed cell death-ligand 1 (PD-L1) tumor cell (TC) expression ≥50% versus <50%, disease stage (IVA versus IVB) and histology (squamous versus nonsquamous). Patient subgroups were defined by a range of blood TMB (bTMB) values, including at a prespecified cut-off of 20 mutations (mut)/megabase (Mb) and across further subdivisions by PD-L1 TC expression ≥1% or <1% and by tissue TMB (tTMB) values. RESULTS At the primary OS data cut-off (12 March 2021), at each bTMB or tTMB cut-off, the magnitude of OS benefit appeared greater among patients in the bTMB- or tTMB-high subgroups for the T + D + CT arm versus the CT arm but was similar between subgroups for the D + CT arm versus the CT arm. Updated OS analyses in the bTMB ≥20 and <20 mut/Mb subgroups, after median follow-up of >5 years (data cut-off 24 August 2023), were similar to those obtained at the primary OS data cut-off. CONCLUSIONS First-line treatment with T (limited course) plus D (until progression) and four cycles of CT consistently improved clinical outcomes versus CT alone in both bTMB-high and -low subgroups, and also in both high and low tTMB subgroups, in patients with mNSCLC. Benefit appeared greater in the TMB-high versus TMB-low subgroups; the addition of anti-cytotoxic T lymphocyte-associated antigen-4 to anti-PD-L1 and CT seemed to increase the magnitude of this difference.
Collapse
Affiliation(s)
- S Peters
- Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland.
| | | | | | | | | | - Z Lai
- AstraZeneca, Waltham, USA
| | | | - H Mann
- AstraZeneca, Cambridge, UK
| | | | - E B Garon
- David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - T Mok
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Chinese University of Hong Kong, Hong Kong, China
| | - M L Johnson
- Sarah Cannon Research Institute, SCRI Oncology Partners, Nashville, USA
| |
Collapse
|
31
|
Kitadai R, Okuma Y, Shibata T, Kohno T, Koyama T. Tissue-agnostic target profiles and treatment efficacy in cancer patients: Insights from the C-CAT clinicogenomic repository. Eur J Cancer 2025; 220:115380. [PMID: 40163907 DOI: 10.1016/j.ejca.2025.115380] [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: 01/12/2025] [Revised: 03/02/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
PURPOSE Utilizing real-world data from Japan's C-CAT clinicogenomic repository, our study demonstrates a significant shift in cancer management through cancer genomic profiling (CGP) tests. The aim of this study is assessing the prevalence of cross-organ targeted genetic alterations and exploring the differences in treatment responses among cancer patients who underwent CGP tests. METHODS Analyzing data from 60,256 patients in the C-CAT repository, we documented the prevalence of FDA-approved biomarkers, cross-organ genetic alterations, and treatment outcomes for tissue-agnostic therapies from June 2019 to December 2023. RESULTS Biomarkers including RET rearrangement, BRAFV600E mutation, and NTRK rearrangement showed varied therapeutic responses, underscoring the need for universal CGP testing to optimize patient outcomes. Notably, our findings highlight variations in response across different age groups, suggesting the potential for age-specific treatment strategies. Comparisons with the AACR Project GENIE database revealed broader implications for the global genomic landscape. CONCLUSION This study emphasizes the crucial role of clinicogenomic repositories in advancing precision oncology across diverse populations, underscoring the utility of integrating clinical and genomic data in national repositories.
Collapse
Affiliation(s)
- Rui Kitadai
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan; Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Taro Shibata
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center, Tokyo, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
32
|
Takano Y, Mizuno K, Iwase M, Morita S, Torii N, Kikumori T, Ando Y. Tumor mutational burden status and clinical characteristics of invasive lobular carcinoma of the breast. Breast Cancer 2025:10.1007/s12282-025-01706-6. [PMID: 40314919 DOI: 10.1007/s12282-025-01706-6] [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: 12/04/2024] [Accepted: 04/18/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND High tumor mutational burden (TMB-H) is an established biomarker for a favorable response to immune checkpoint inhibitors. However, tumor mutational burden (TMB) in invasive ductal carcinoma (IDC) and invasive lobular carcinoma (ILC) has not been sufficiently investigated. METHODS We collected data of patients with ILC or IDC from the Center for Cancer Genomics and Advanced Therapeutics database between June 2019 and August 2023. Furthermore, we examined the clinicopathological factors and TMB status. RESULTS Patients with ILC (n = 170) had a median TMB score of 4.00 mut/Mb (interquartile range, 2.00-7.14 mut/Mb), whereas those with IDC (n = 2598) had a score of 3.90 mut/Mb (2.00-6.00 mut/Mb). TMB-H was more common in patients with ILC than in those with IDC (18.2% vs. 10.1%, P < 0.001), particularly in the ER+ /HER2- subtype. Multivariate analysis revealed that the pathological diagnosis of ILC (P = 0.006), tissue samples collected from metastatic sites (P < 0.001), and older age (50 years, P < 0.001) were independent factors for TMB-H. CONCLUSIONS Patients with ILC were more likely to have TMB-H than those with IDC. The findings of this study would be invaluable in selecting treatment strategies for patients with ILC.
Collapse
Affiliation(s)
- Yuko Takano
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan.
- Department of Breast and Endocrine Surgery, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan.
| | - Kazuyuki Mizuno
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Madoka Iwase
- Department of Breast and Endocrine Surgery, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Sachi Morita
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Nao Torii
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
- Department of Breast and Endocrine Surgery, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Toyone Kikumori
- Department of Breast and Endocrine Surgery, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Yuichi Ando
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| |
Collapse
|
33
|
Seino M, Sano S, Gonai Y, Horikawa S, Nakamura F, Okui Y, Matsukawa J, Sakaki H, Watanabe N, Yamauchi K, Ohta T, Hoshi Y, Suzuki S, Kawai M, Nagase S. Investigation of tumor mutation burden using the comprehensive genomic profiling data of vulvar and vaginal malignant tumors: an observational study using C-CAT database. Int J Clin Oncol 2025; 30:1033-1039. [PMID: 40192944 DOI: 10.1007/s10147-025-02730-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 02/23/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND This study aimed to reveal the gene alteration and tumor mutation burden (TMB) statuses of vulvar and vaginal malignant tumors in Japan. METHODS We investigated the cancer genomic profiling (CGP) data of 79 patients with vulvar and vaginal cancers. These data were obtained from the Center for Cancer Genomics and Advanced Therapeutics (C-CAT). RESULTS None of the patients had high microsatellite instability. Although 21.9% of the patients with vulvar and vaginal squamous cell carcinoma (SCC) had high TMB, those with other histological types did not. The top single-nucleotide variants (SNVs) in SCC were TERT, TP53, CDKN2A, KMT2D, and NOTCH1. The frequencies of ATRX and PBRM1 were significantly higher in TMB-high SCC than in non-TMB-high SCC. CONCLUSION SCC of the vulva and vagina is expected to have high TMB, and gene alteration status differed between TMB-high and non-TMB-high groups.
Collapse
Affiliation(s)
- Manabu Seino
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan.
| | - Shiori Sano
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan
| | - Yuta Gonai
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan
| | - Shota Horikawa
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan
| | - Fumihiro Nakamura
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan
| | - Yosuke Okui
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan
| | - Jun Matsukawa
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan
| | - Hirotsugu Sakaki
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan
| | - Norikazu Watanabe
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan
| | - Keiko Yamauchi
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan
| | - Tsuyoshi Ohta
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan
| | - Yuki Hoshi
- Genetic Counseling Unit, Yamagata University Hospital, Yamagata, Japan
| | - Shuhei Suzuki
- Department of Clinical Oncology, Yamagata Prefecture Shinjo Hospital, Yamagata, Japan
| | - Masaaki Kawai
- Department of Surgery I, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Satoru Nagase
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan
| |
Collapse
|
34
|
Tsilimigras DI, Kurzrock R, Pawlik TM. Molecular Testing and Targeted Therapies in Hepatobiliary Cancers: A Review. JAMA Surg 2025; 160:576-585. [PMID: 40105823 DOI: 10.1001/jamasurg.2025.0242] [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: 03/20/2025]
Abstract
Importance Hepatobiliary cancers are heterogeneous and molecularly complex. Recent advances in next-generation sequencing (NGS) have enhanced the understanding of their molecular landscape and enabled deployment of biomarker-based gene- and immune-targeted therapies. This review examines the role of molecular testing and targeted therapies in these malignant neoplasms. Observations Patients with hepatobiliary cancers have poor outcomes. Precision oncology studies have shown that while many common molecular alterations are not currently targetable in hepatocellular carcinoma (HCC), a large number of actionable alterations characterize biliary tract cancers (BTCs), with several therapies now approved by the US Food and Drug Administration. Immunotherapy is increasingly adopted in clinical practice, either as monotherapy or combined with cytotoxic chemotherapy, for both HCC and BTCs. Moreover, multiple solid cancer tumor-agnostic therapies are approved (larotrectinib, entrectinib, and repotrectinib for NTRK fusions; selpercatinib for RET fusions; dabrafenib and trametinib combination for BRAF V600E mutations; dostarlimab or pembrolizumab for tumors with high microsatellite instability and pembrolizumab for tumor mutation burden ≥10 mutations/megabase), highlighting the need for NGS as well as ERBB2 (formerly HER2) immunohistochemistry (IHC) (with the recent approval of solid tissue-agnostic deruxtecan trastuzumab for ERBB2-positive [IHC 3+] cancer) across cancers. N-of-1 clinical trials using customized drug combinations matched to the tumor's molecular profile have yielded encouraging results and provide a promising framework for future clinical trial design. Conclusions and Relevance Molecular testing and gene- and immune-targeted therapies are transforming hepatobiliary cancer treatment. Tumor-agnostic and N-of-1 clinical trials have challenged traditional clinical trial paradigms and provide the foundation for truly personalized oncology for patients with these aggressive cancers. Further work is needed to determine how to leverage these novel approaches into the management of operable disease.
Collapse
Affiliation(s)
- Diamantis I Tsilimigras
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus
| | - Razelle Kurzrock
- Medical College of Wisconsin Cancer Center and Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Milwaukee
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus
- Deputy Editor, JAMA Surgery
| |
Collapse
|
35
|
Carlson DK, Painter C, Gradecki SE, Ring KL, Williams ES, Mills AM. High-grade Endometrial Carcinomas With Solid Basaloid Morphology and Geographic Necrosis Lacking Definitive Pilomatrix-like Features: Clinicopathologic Characteristics Including Aggressive Behavior and Novel Molecular Events. Int J Gynecol Pathol 2025; 44:199-209. [PMID: 39475375 DOI: 10.1097/pgp.0000000000001081] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
High-grade endometrioid carcinomas occasionally demonstrate solid basaloid morphology with geographic necrosis (SB-GN). This pattern is among the defining features of pilomatrix-like high-grade endometrioid carcinoma (PiMHEC), a recently proposed tumor type which is additionally characterized by the presence of shadow cells, abnormal beta-catenin/ CTNNB1 mutations, strong CDX2 expression, and poor outcomes. Clinicopathologic overlap between PiMHEC and other high-grade endometrial cancers with SB-GN has not been established. We screened 300 endometrial carcinomas on tissue microarray for SB-GN histology and performed a detailed whole-section morphologic review, immunohistochemical analysis, and next-generation sequencing on all cases bearing this pattern. Four (1.3%) demonstrated SB-GN. All 3 with clinical follow-up had extremely aggressive behavior despite being MMR-deficient; in contrast, only 27% of other MMR-deficient high-grade carcinomas recurred. One SB-GN case met most of the previously outlined diagnostic criteria for PiMHEC including abnormal beta-catenin/ CTNNB1 (p.S37P variant) and strong CDX2 expression; notably, however, shadow cells were absent. This case also demonstrated a KRAS p.A59T pathogenic variant. The other 3 cases also lacked shadow cells; the 2 with sequencing data bore no CTNNB1 abnormalities but showed likely oncogenic variants involving the pilomatrixoma-associated gene FGFR2. All 3 cases with molecular results also bore somatic Notch pathway ( NOTCH1/NOTCH2/NOTCH3 ) variants. The single case treated with immunotherapy showed complete and sustained response with regression of bone metastases despite abnormal beta-catenin/ CTNNB1 , which has been associated with immunotherapeutic resistance. These data suggest that the SB-GN pattern may connote a poor prognosis even in the absence of overt pilomatrix-like differentiation, and that novel molecular events may have implications for the treatment of these tumors.
Collapse
Affiliation(s)
| | | | | | - Kari L Ring
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Virginia
| | | | | |
Collapse
|
36
|
Kret ZS, Sweder RJ, Pollock R, Tinoco G. Potential Mechanisms for Immunotherapy Resistance in Adult Soft-Tissue Sarcoma. Target Oncol 2025; 20:485-502. [PMID: 40289241 DOI: 10.1007/s11523-025-01145-5] [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: 04/13/2025] [Indexed: 04/30/2025]
Abstract
Soft-tissue sarcomas represent a diverse group of rare malignancies originating from mesenchymal tissue, accounting for less than 1% of adult cancers in the USA. With over 13,000 new cases and around 5350 deaths annually, patients with metastatic soft-tissue sarcomas face limited therapeutic options and an estimated median overall survival of 18 months. While immunotherapy has demonstrated effectiveness in several cancers, its application in soft-tissue sarcomas remains challenging owing to the tumors' largely "cold" immunological environment, characterized by low levels of tumor-infiltrating lymphocytes and a lack of soft-tissue sarcoma-specific biomarkers. This review examines potential mechanisms underlying immunotherapy resistance in soft-tissue sarcomas, including the complex interplay between innate and adaptive immunity, the tumor microenvironment, and the role of immune-related genes. Despite preliminary findings suggesting correlations between immune profiles and histological subtypes, consistent biomarkers for predicting immunotherapeutic responses across soft-tissue sarcoma types are absent. Emerging strategies focus on converting "cold" tumors to "hot" tumors, enhancing their susceptibility to immunologic activation. While research is ongoing, personalized treatment approaches may offer hope for overcoming the inherent heterogeneity and resistance seen in soft-tissue sarcomas, ultimately aiming to improve outcomes for affected patients.
Collapse
Affiliation(s)
- Zaina S Kret
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ryan J Sweder
- The Ohio State University College of Arts and Sciences and College of Medicine, Columbus, OH, USA
| | - Raphael Pollock
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Gabriel Tinoco
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 1800 Cannon Drive, 1240 Lincoln Tower, Columbus, OH, 43210, USA.
| |
Collapse
|
37
|
Fu J, Franzen N, Aas E, Koen van der Mijn JC, van Leeuwen PJ, Retel VP. Early Cost-Effectiveness Analysis of Using Whole-Genome Sequencing for Patients With Castration-Resistant Prostate Cancer. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2025; 28:720-729. [PMID: 40049327 DOI: 10.1016/j.jval.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/16/2025] [Accepted: 02/16/2025] [Indexed: 03/30/2025]
Abstract
OBJECTIVES This study aims to assess the potential cost-effectiveness of using whole-genome sequencing (WGS)-guided systemic therapy in metastatic castrate-resistant prostate cancer compared with the European Association of Urology guideline recommended diagnostics from a Dutch societal perspective. METHODS A decision analytic model combining a decision tree and partitioned survival models was developed to link diagnostic results with subsequent biomarker-guided treatments. Two diagnostic strategies, WGS and guideline-recommended practice-the genomic testing for breast cancer gene 1/2 (BRCA1/2) and deficient mismatch repair, were simulated to compare the health outcome and cost. Treatment effectiveness was estimated through survival analysis using published trial data. Sensitivity and scenario analyses were conducted to examine result robustness and to identify conditions under which WGS may be cost-effective. RESULTS WGS identified an additional 21% of patients eligible for personalized therapy (PD-1/PDL-1 inhibitors and olaparib), resulting in an incremental increase in cost (€14 260) and quality-adjusted life years (QALY = 0.05). These results yielded an incremental cost-effectiveness ratio of €289 625 per QALY gained. WGS would become cost-effective if the cost of biomarker-guided therapies decreases by 62% and when identifying a proportion of 23% more patients with actional targets. CONCLUSIONS Our findings suggest that future treatments with improved efficacy and reduced cost could potentially make the WGS strategy cost-effective. Its unaccounted potential value to identify prognostic biomarkers, diagnostic alternatives, and patient heterogeneity should be addressed in future research and considered for optimal implementation. New reimbursement options are needed considering the high prices of biomarker-guided therapies that drive the incremental cost-effectiveness ratio.
Collapse
Affiliation(s)
- Jinjing Fu
- Department of Epidemiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Nora Franzen
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Eline Aas
- Department of Health Management and Health Economics, University of Oslo, Oslo, Norway; Division of Health Services, Norwegian Institute of Public Health, Oslo, Norway
| | - J C Koen van der Mijn
- Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Pim J van Leeuwen
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Valesca P Retel
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands; Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
| |
Collapse
|
38
|
Wang X, Wang L, Liu Y. Current Status of Immune Checkpoint Inhibitors and Treatment Responsive Biomarkers for Triple-Negative Breast Cancer. Thorac Cancer 2025; 16:e70072. [PMID: 40324951 PMCID: PMC12052518 DOI: 10.1111/1759-7714.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/24/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025] Open
Abstract
Triple-negative breast cancer (TNBC), accounting for about 10%-20% of all breast cancer cases, is characterized by its aggressive nature, high recurrence rates, and poor prognosis. Unlike other breast cancer subtypes, TNBC lacks hormone receptors and specific molecular targets, limiting therapeutic options. In recent years, immune checkpoint inhibitors (ICIs) have shown promise in treating TNBC by targeting immune evasion mechanisms. Despite these advancements, several issues remain unresolved, including low response rates in programmed cell death ligand 1 (PD-L1) negative TNBC subtypes and the challenge of predicting which patients will benefit from ICIs. Consequently, there is growing interest in identifying reliable biomarkers beyond PD-L1 expression. This review synthesizes recent studies to provide a comprehensive perspective on ICI therapy in TNBC, clarifying the status of single-agent ICI therapies and combination strategies, emphasizing the need for further research into biomarkers. These insights provide clues for more personalized and effective treatment approaches, ultimately aiming to improve clinical outcomes for patients with TNBC.
Collapse
Affiliation(s)
- Xinran Wang
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Lingxia Wang
- Value & Implementation, Global Medical & Scientific AffairsMSD ChinaShanghaiChina
| | - Yueping Liu
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| |
Collapse
|
39
|
Chiaruttini MV, Proto C, Lo Russo G, Prelaj A, Segale M, Zanghì A, Galli F, Greco FG, Signorelli D, Brambilla M, Occhipinti M, De Braud F, Garassino MC, Sozzi G, Rulli E, Boeri M. Tracking the Response to Immunotherapy: Blood microRNA Dynamics in Patients With Advanced Non-Small Cell Lung Cancer. JCO Precis Oncol 2025; 9:e2400790. [PMID: 40403209 DOI: 10.1200/po-24-00790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/19/2025] [Accepted: 03/27/2025] [Indexed: 05/24/2025] Open
Abstract
PURPOSE Despite the significant improvement in outcomes for patients with advanced non-small cell lung cancer (NSCLC) treated with immune checkpoint inhibitors (ICIs), resistance, whether primary or secondary, remains a substantial challenge. Currently, reliable biomarkers to monitor ICI response are lacking, highlighting the need for minimally invasive tools like liquid biopsy to track treatment efficacy. This study aimed to identify circulating microRNAs (miRNAs) as potential biomarkers to track ICI response in patients with NSCLC. MATERIALS AND METHODS The Apollo longitudinal study enrolled patients with advanced NSCLC receiving ICI in first or subsequent lines. Plasma samples were collected at baseline and follow-up to prospectively assess miRNA profiles until progressive disease (PD). Using a custom reverse transcription-quantitative polymerase chain reaction platform, 276 ratios among 24 lung cancer-related miRNAs were analyzed. The generalized estimating equation and joint models were applied to select the miRNA ratios most associated with PD over time. To control for multiple testing, the Benjamini-Yekutieli method was applied setting a 10% false discovery rate threshold. RESULTS From the 211 patients, a total of 454 plasma samples were analyzed. Clinical and biochemical variables had little effect on miRNAs' profile. The analysis identified nine miRNA ratios, all involving miR-145-5p, as significant biomarkers for monitoring treatment response, even after adjustment for the line of therapy. These ratios exhibited a longitudinal modulation pattern consistent with radiologic response, particularly in patients who initially benefited from ICI treatment. In addition, in an independent set of 32 plasma samples from 10 patients receiving ICI as maintenance therapy, the same trends were observed. CONCLUSION A focused panel of miRNA ratios, driven by miR-145-5p, effectively reflects response to ICI therapy in patients with advanced NSCLC, highlighting their potential as biomarkers for treatment monitoring.
Collapse
Affiliation(s)
- Maria Vittoria Chiaruttini
- Laboratory of Methodology for Clinical Research, Clinical Oncology Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Unit of Biostatistics, Epidemiology and Public Health, University of Padua, Padua, Italy
| | - Claudia Proto
- Thoracic Oncology Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppe Lo Russo
- Thoracic Oncology Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Arsela Prelaj
- Thoracic Oncology Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Electronics, Information, and Bioengineering, Polytechnic University of Milan, Milan, Italy
| | - Miriam Segale
- Epigenomics and Biomarkers of Solid Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Anna Zanghì
- Epigenomics and Biomarkers of Solid Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Galli
- Laboratory of Methodology for Clinical Research, Clinical Oncology Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Francesca G Greco
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Diego Signorelli
- Thoracic Oncology Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Marta Brambilla
- Thoracic Oncology Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Mario Occhipinti
- Thoracic Oncology Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo De Braud
- Thoracic Oncology Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marina C Garassino
- Section of Hematology-Oncology, Department of Medicine, Knapp Center for Biomedical Discovery, The University of Chicago, Chicago, IL
| | - Gabriella Sozzi
- Epigenomics and Biomarkers of Solid Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Eliana Rulli
- Laboratory of Methodology for Clinical Research, Clinical Oncology Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Mattia Boeri
- Epigenomics and Biomarkers of Solid Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
40
|
Ma J, Wang Y, Zhang Z, Cai X, Xiang X, Chen Y, Sun F, Dong J. Peripheral Blood T-Cell Receptor Repertoire Diversity as a Potential Biomarker in the Diagnosis and Treatment Evaluation of Colorectal and Lung Cancers: A Prospective Observational Study. Cancer Med 2025; 14:e70937. [PMID: 40387418 PMCID: PMC12086972 DOI: 10.1002/cam4.70937] [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: 08/28/2024] [Revised: 04/24/2025] [Accepted: 04/27/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND T-cell receptor (TCR) diversity 50 (D50) values could assess peripheral blood (PB) TCR diversity and immunity. This study aimed to evaluate the potential D50 value in the diagnosis and treatment evaluation of colorectal cancer (CRC) and nonsmall-cell lung cancer (NSCLC). METHODS This prospective observational study enrolled patients with CRC, benign colorectal disease (BCD), NSCLC, or benign nodule controls (BNC) and healthy donors (HD) at Yunnan Cancer Hospital between January 2021 and June 2022. PB specimens were used for TCRβ sequencing, and D50 was calculated and compared within different groups. The area under the curve (AUC) was used to evaluate the diagnostic performance of D50 in CRC and NSCLC. RESULTS A total of 114 HD and 115 CRC, 31 BCD, 67 NSCLC, and 25 BNC patients were enrolled. Both CRC and NSCLC patients exhibited significantly lower D50 compared with HDs (p < 0.001), whereas BCD and BNC patients showed a modest decrease in TCR diversity (p < 0.05). NSCLC patients with lymph node metastases had markedly lower D50 than those without lymph node metastasis (0.05 vs. 0.11, p < 0.01). Higher D50 was found in CRC and NSCLC patients with normal carcinoembryonic antigen (CEA) levels (p < 0.05). The potential of D50 value for early detection of CRC and NSCLC was demonstrated, with an area under the receiver operating characteristic curve (AUC) of 0.736 for CRC (sensitivity: 71.30%, specificity: 68.42%) and 0.768 for NSCLC (sensitivity: 83.58%, specificity: 60.53%). Significant differences in D50 values were observed between patients with tumor regression grade (TRG) 0-1 and those with TRG 2-3 (p = 0.027), with an AUC of 0.731 (sensitivity: 68.75%, specificity: 76.92%). CONCLUSION These findings suggest that the PB TCR D50 values may have significant clinical value in cancer diagnosis and in evaluating the efficacy of neoadjuvant therapies.
Collapse
MESH Headings
- Humans
- Male
- Female
- Middle Aged
- Lung Neoplasms/diagnosis
- Lung Neoplasms/blood
- Lung Neoplasms/therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/genetics
- Prospective Studies
- Colorectal Neoplasms/diagnosis
- Colorectal Neoplasms/blood
- Colorectal Neoplasms/therapy
- Colorectal Neoplasms/immunology
- Colorectal Neoplasms/genetics
- Aged
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Carcinoma, Non-Small-Cell Lung/diagnosis
- Carcinoma, Non-Small-Cell Lung/blood
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/genetics
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/blood
- Adult
Collapse
Affiliation(s)
- Jilong Ma
- Key Laboratory of Cell Therapy Technology Transformation Medicine of Yunnan Province, the Han Weidong Expert Workstation of Yunnan Province, Yunnan Provincial Engineering Research Centre of Cell Therapy and Quality Control System, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingYunnanChina
| | - Yuanbiao Wang
- Key Laboratory of Cell Therapy Technology Transformation Medicine of Yunnan Province, the Han Weidong Expert Workstation of Yunnan Province, Yunnan Provincial Engineering Research Centre of Cell Therapy and Quality Control System, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingYunnanChina
| | - Zhixin Zhang
- Department of TechnologyChengdu ExAb Biotechnology, LTDChengduSichuanChina
| | - Xinyi Cai
- Department of Colorectal SurgeryThe Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingYunnanChina
| | - Xudong Xiang
- Department of Thoracic Surgery IIThe Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingYunnanChina
| | - Yan Chen
- Key Laboratory of Cell Therapy Technology Transformation Medicine of Yunnan Province, the Han Weidong Expert Workstation of Yunnan Province, Yunnan Provincial Engineering Research Centre of Cell Therapy and Quality Control System, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingYunnanChina
| | - Fengqiong Sun
- Department of Colorectal SurgeryThe Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingYunnanChina
| | - Jian Dong
- Key Laboratory of Cell Therapy Technology Transformation Medicine of Yunnan Province, the Han Weidong Expert Workstation of Yunnan Province, Yunnan Provincial Engineering Research Centre of Cell Therapy and Quality Control System, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer HospitalKunmingYunnanChina
| |
Collapse
|
41
|
Bitting RL, McNair C, Wyatt AW, Vandekerkhove G, Choi T, Leader AE, Blanding-Godbolt J, Gross L, Hamade K, Halabi S, Giri VN. Factors Affecting Genomic Testing in Prostate Cancer: Results From the Decision-Making, Experience, and Confidence In Determining Genomic Evaluation (DECIDE) Survey. JCO Precis Oncol 2025; 9:e2400821. [PMID: 40373262 DOI: 10.1200/po-24-00821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/28/2025] [Indexed: 05/17/2025] Open
Abstract
PURPOSE Genomic testing for prostate cancer (PCa) clinical management and hereditary cancer assessment has grown in clinical impact; however, challenges remain regarding optimal implementation and end-user confidence. The Decision-making, Experience, and Confidence In Determining Genomic Evaluation (DECIDE) survey was designed to collect information regarding utility and understanding of genomic testing from PCa health care providers, researchers, and stakeholders. METHODS The DECIDE survey was administered online from October 2022 to January 2023 with 18 multiple-response questions. Survey domains included self-confidence with ordering and interpreting germline and somatic genomic tests, process of testing and use of results, decision-making factors, and barriers to testing. Data were summarized by evaluating counts and percentages of responses, and the results were presented by descriptive statistics. RESULTS One hundred twenty-two participants completed the survey. The majority were medical oncologists (70%) and at academic medical centers (89%). Self-confidence was high in knowing indications for genomic testing (82% respondents) but lower in interpretation of results, especially from circulating tumor DNA (52%). Confidence varied in interpreting pathogenic variants (65% high confidence), variants of unknown significance (47%), and incidental findings from genomic tests (35%). Common barriers to testing were difficulty obtaining tissue (71%) and cost (35%). Testing utility was sometimes limited by inability to obtain the recommended treatment (33%). Most of the respondents (55%) agreed that lack of education and training of health care professionals regarding genomic testing is impeding clinical translation. CONCLUSION The DECIDE survey provided critical insights into challenges with genomic testing, from provider confidence in interpretating results to testing and practice barriers. The results inform next steps to further educate PCa providers and to collectively improve testing and result reporting for enhanced implementation of PCa genomic testing.
Collapse
Affiliation(s)
- Rhonda L Bitting
- Duke Cancer Institute, Duke University, Durham, NC
- Duke University School of Medicine, Durham, NC
- Durham VA Healthcare System, Durham, NC
| | - Christopher McNair
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Alexander W Wyatt
- Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
- Michael Smith Genome Sciences Centre and Clinical Cancer Genomics Program, BC Cancer, Vancouver, Canada
| | | | - Taehwa Choi
- School of Mathematics, Statistics and Data Science, Sungshin Women's University, Seoul, South Korea
| | - Amy E Leader
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | | - Laura Gross
- Yale University School of Medicine, Yale Cancer Center, New Haven, CT
| | - Khaldoun Hamade
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Susan Halabi
- Duke Cancer Institute, Duke University, Durham, NC
- Duke University School of Medicine, Durham, NC
| | - Veda N Giri
- Yale University School of Medicine, Yale Cancer Center, New Haven, CT
| |
Collapse
|
42
|
Koh GCC, Nanda AS, Rinaldi G, Boushaki S, Degasperi A, Badja C, Pregnall AM, Zhao SJ, Chmelova L, Black D, Heskin L, Dias J, Young J, Memari Y, Shooter S, Czarnecki J, Brown MA, Davies HR, Zou X, Nik-Zainal S. A redefined InDel taxonomy provides insights into mutational signatures. Nat Genet 2025; 57:1132-1141. [PMID: 40210680 DOI: 10.1038/s41588-025-02152-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
Despite their deleterious effects, small insertions and deletions (InDels) have received far less attention than substitutions. Here we generated isogenic CRISPR-edited human cellular models of postreplicative repair dysfunction (PRRd), including individual and combined gene edits of DNA mismatch repair (MMR) and replicative polymerases (Pol ε and Pol δ). Unique, diverse InDel mutational footprints were revealed. However, the prevailing InDel classification framework was unable to discriminate these InDel signatures from background mutagenesis and from each other. To address this, we developed an alternative InDel classification system that considers flanking sequences and informative motifs (for example, longer homopolymers), enabling unambiguous InDel classification into 89 subtypes. Through focused characterization of seven tumor types from the 100,000 Genomes Project, we uncovered 37 InDel signatures; 27 were new. In addition to unveiling previously hidden biological insights, we also developed PRRDetect-a highly specific classifier of PRRd status in tumors, with potential implications for immunotherapies.
Collapse
Affiliation(s)
- Gene Ching Chiek Koh
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia
| | - Arjun Scott Nanda
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Giuseppe Rinaldi
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Soraya Boushaki
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Andrea Degasperi
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Cherif Badja
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Andrew Marcel Pregnall
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Salome Jingchen Zhao
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Lucia Chmelova
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Daniella Black
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Laura Heskin
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - João Dias
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Jamie Young
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Yasin Memari
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Scott Shooter
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Jan Czarnecki
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Matthew Arthur Brown
- Genomics England, Queen Mary University of London, Dawson Hall, Charterhouse Square, London, UK
| | - Helen Ruth Davies
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Xueqing Zou
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Serena Nik-Zainal
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
43
|
Papke DJ, Chrisinger JSA, French CA, Crymes A, Krivak TC, Estape RE, Seetharam M, Patel RA, O'Connor WN, Chi AW, Gutman P, Singer S, Kim C, Bryant DA, Oberley MJ, Adeyelu T, Bridge JA, Evans MG. MAD::NUT Fusion Sarcoma: A Sarcoma Class With NUTM1, NUTM2A, and NUTM2G Fusions and Possibly Distinctive Subtypes. Mod Pathol 2025; 38:100729. [PMID: 39921028 DOI: 10.1016/j.modpat.2025.100729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/31/2024] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
NUT fusion-associated cancers are heterogeneous and include NUT carcinoma and an emerging group with non-BRD4/BRD3/NSD3 fusion partners. In this study, we characterized 11 tumors harboring MAD::NUT fusions (10/11 in female patients; median age: 48 years; range: 1-67 years), all histologically different from NUT carcinoma. Eight cases were identified via sequencing database review and 3 were diagnosed prospectively. Eight (73%) patients presented with multifocal disease, including 6 with disseminated peritoneal tumors; 3 (27%) presented with solitary colonic, pulmonary, or orbital masses. Nine (82%) tumors harbored NUTM1 fusions, with MXI1 (5/9; 56%), MXD4 (2/9; 22%), and MGA (2/9; 22%). One tumor each harbored MXD4::NUTM2G and MXI1::NUTM2A fusions. The 9 MXD4/MXI1-rearranged sarcomas were high-grade, with epithelioid-to-spindle cell cytomorphology, amphophilic cytoplasm, vesicular nuclei, and prominent nucleoli. Histologic features included infiltrative growth (7/7 assessable tumors), rhabdoid morphology (7/9; 78%), prominent collagen (3/9; 33%), multinucleated tumor cells (2/9; 22%), and myxoid stroma (1/9; 11%). MXD4/MXI1-rearranged sarcomas expressed desmin (3/7; 43%) and keratin(s) (3/7; 43%), and not p63 (6 tumors), CD34 (5 tumors), or S-100 (5 tumors). The adult MGA::NUTM1 fusion sarcoma exhibited some cytologic overlap with MXD4/MXI1-rearranged sarcomas but showed lower grade myxoid spindle cell regions, microcystic spaces, and S-100 expression. The pediatric MGA::NUTM1 fusion sarcoma was low-grade with CD34/S-100 coexpression. Immunohistochemistry demonstrated NUTM1 expression in NUTM1-rearranged sarcomas (5/5), and weak and no expression in NUTM2A- and NUTM2G-rearranged sarcomas, respectively. Gene expression profiling demonstrated sarcomas with MXD4/MXI1::NUTM1/NUTM2A/NUTM2G fusions clustered separately from NUT carcinoma. Follow-up was available for 9 patients (82%; median length: 1.8 years; range: 2 months to 8.2 years). Four of 7 patients with MXD4/MXI1-rearranged sarcomas died of disease (median survival: 1.3 years; range: 5 months to 4.8 years), 1 entered hospice at 2 months, 1 was alive with pericardial masses at 2.8 years, and 1 was alive with no evidence of disease at 8.2 years. The adult with the MGA::NUTM1 fusion sarcoma died of other causes at 4.5 years; the child was alive without disease at 11 months. We conclude that MAD::NUT fusions define a sarcoma class distinct from NUT carcinoma. Among this group, MGA::NUTM1 fusion sarcomas might represent a distinctive subset. NUTM1 immunohistochemistry does not reliably detect NUTM2A/NUTM2G-rearranged sarcomas.
Collapse
Affiliation(s)
- David J Papke
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| | - John S A Chrisinger
- Division of Anatomic and Molecular Pathology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Christopher A French
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Anthony Crymes
- Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Thomas C Krivak
- Division of Gynecologic Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, Pennsylvania
| | - Ricardo E Estape
- Hospital Corporation of America (HCA) Florida Institute for Gynecologic Oncology, HCA Florida Healthcare, Miami, Florida
| | - Mahesh Seetharam
- Division of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, Arizona
| | - Reema A Patel
- Division of Medical Oncology, Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, Kentucky
| | - William N O'Connor
- Department of Pathology and Laboratory Medicine, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Anthony W Chi
- Rockville Regional Lab, Kaiser Permanente Mid-Atlantic Medical Group, Rockville, Maryland
| | - Pablo Gutman
- Department of Pathology, Holy Cross Hospital, Silver Spring, Maryland
| | - Stephan Singer
- Institute of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Chul Kim
- Department of Medicine, Division of Hematology and Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | | | | | | | - Julia A Bridge
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, Nebraska; ProPath, Dallas, Texas
| | | |
Collapse
|
44
|
Morris VK, Liu S, Lin K, Zhu H, Prasad S, Mahvash A, Bhosale P, Sun B, Parra ER, Wistuba I, Peddireddy A, Yao J, Mendoza-Perez J, Knafl M, Woodman SE, Eng C, Halperin D. Phase II Trial of Atezolizumab and Bevacizumab for Treatment of HPV-Positive Unresectable or Metastatic Squamous Cell Carcinoma of the Anal Canal. Clin Cancer Res 2025; 31:1657-1666. [PMID: 40019482 PMCID: PMC12010964 DOI: 10.1158/1078-0432.ccr-24-1512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/31/2024] [Accepted: 02/26/2025] [Indexed: 03/01/2025]
Abstract
PURPOSE Anti-PD-L1 antibodies are associated with responses in <25% of patients with metastatic human papillomavirus-associated malignancies. VEGF signaling causes immune evasion and immune suppression within the tumor. We evaluated the anti-PD-L1 antibody atezolizumab and anti-VEGF antibody bevacizumab for patients with unresectable, advanced anal cancer. PATIENTS AND METHODS For this phase II study, participants with previously treated, immunotherapy-naïve anal cancer received atezolizumab (1,200 mg) and bevacizumab (15 mg/kg) intravenously every 21 days. Responses were evaluated every 9 weeks (RECIST version 1.1). The primary endpoint was the best radiographic response. Median survival was estimated by Kaplan-Meier and compared for selected biomarkers (including paired pre- and on-treatment biopsies) using a log-rank test. RESULTS Among 20 participants, the overall response rate was 11% [95% confidence interval (CI): 1.2-32]. Median progression-free survival and overall survival were 4.1 months (95% CI, 2.6-not assessable) and 11.6 months (95% CI, 9.5-20), respectively. One grade 5 bevacizumab-related bowel perforation occurred. Analyses of 16 paired biopsies linked increases in IFN-γ (P = 0.03) and inflammatory response (P = 0.02) gene expression signatures with prolonged progression-free survival, as did increases in CD3+CD8+PD1+ (P = 0.02) cells and decreases in CD3+FoxP3+ cells (P = 0.04) from 10 paired biopsies with multiplex immunofluorescence. A subgroup of anal cancers characterized by the SBS31 "prior-platinum" signature demonstrated shorter median overall survival (HR, 6.3; 95% CI, 1.2-32; P = 0.01). CONCLUSIONS Atezolizumab and bevacizumab demonstrate activity similar to anti-PD-1 antibodies alone for unresectable anal cancer. Our translational data identify undescribed chromosomal and transcriptomic biomarkers associated with survival for metastatic anal cancer. These correlative findings warrant confirmation and further validation in larger, prospective immunotherapy trials for advanced anal cancer.
Collapse
MESH Headings
- Humans
- Anus Neoplasms/drug therapy
- Anus Neoplasms/pathology
- Anus Neoplasms/virology
- Anus Neoplasms/mortality
- Male
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Middle Aged
- Female
- Bevacizumab/administration & dosage
- Bevacizumab/adverse effects
- Aged
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/virology
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/mortality
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Adult
- Papillomavirus Infections/virology
- Papillomavirus Infections/complications
- Papillomavirus Infections/drug therapy
- Aged, 80 and over
- Biomarkers, Tumor
- Papillomaviridae/isolation & purification
Collapse
Affiliation(s)
- Van K Morris
- Department of Gastrointestinal Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Suyu Liu
- Department of Biostatistics, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Kangyu Lin
- Department of Gastrointestinal Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Haifeng Zhu
- Department of Genomic Medicine, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Seema Prasad
- Department of Gastrointestinal Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Armeen Mahvash
- Department of Radiology, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Priya Bhosale
- Department of Radiology, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Baohua Sun
- Department of Translational Molecular Pathology, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Edwin R Parra
- Department of Translational Molecular Pathology, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | | | - James Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Julia Mendoza-Perez
- Department of Translational Molecular Pathology, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Mark Knafl
- Department of Genomic Medicine, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Scott E Woodman
- Department of Genomic Medicine, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| | - Cathy Eng
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Daniel Halperin
- Department of Gastrointestinal Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
45
|
Shitara K, Janjigian YY, Ajani J, Moehler M, Yao J, Wang X, Chhibber A, Pandya D, Shen L, Garrido M, Gallardo C, Wyrwicz L, Yamaguchi K, Skoczylas T, Bragagnoli A, Liu T, Schenker M, Yañez P, Kowalyszyn R, Karamouzis M, Zander T, Feeney K, Elimova E, Doshi P, Li M, Lei M. Nivolumab plus chemotherapy or ipilimumab in gastroesophageal cancer: exploratory biomarker analyses of a randomized phase 3 trial. Nat Med 2025; 31:1519-1530. [PMID: 40055521 PMCID: PMC12092258 DOI: 10.1038/s41591-025-03575-0] [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: 05/08/2024] [Accepted: 02/07/2025] [Indexed: 05/22/2025]
Abstract
First-line nivolumab-plus-chemotherapy demonstrated superior overall survival (OS) and progression-free survival versus chemotherapy for advanced gastroesophageal adenocarcinoma with programmed death ligand 1 combined positive score ≥ 5, meeting both primary end points of the randomized phase 3 CheckMate 649 trial. Nivolumab-plus-ipilimumab provided durable responses and higher survival rates versus chemotherapy; however, the prespecified OS significance boundary was not met. To identify biomarkers predictive of differential efficacy outcomes, post hoc exploratory analyses were performed using whole-exome sequencing and RNA sequencing. Nivolumab-based therapies demonstrated improved efficacy versus chemotherapy in hypermutated and, to a lesser degree, Epstein-Barr virus-positive tumors compared with chromosomally unstable and genomically stable tumors. Within the KRAS-altered subgroup, only patients treated with nivolumab-plus-chemotherapy demonstrated improved OS benefit versus chemotherapy. Low stroma gene expression signature scores were associated with OS benefit with nivolumab-based regimens; high regulatory T cell signatures were associated with OS benefit only with nivolumab-plus-ipilimumab. Our analyses suggest that distinct and overlapping pathways contribute to the efficacy of nivolumab-based regimens in gastroesophageal adenocarcinoma.
Collapse
Affiliation(s)
- Kohei Shitara
- National Cancer Center Hospital East, Kashiwa, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Yelena Y Janjigian
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| | - Jaffer Ajani
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Jin Yao
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Xuya Wang
- Bristol Myers Squibb, Princeton, NJ, USA
- Daiichi Sankyo Inc, Basking Ridge, NJ, USA
| | | | - Dimple Pandya
- Bristol Myers Squibb, Princeton, NJ, USA
- Eli Lilly, Indianapolis, IN, USA
| | - Lin Shen
- Peking University Cancer Hospital and Institute, Beijing, China
| | - Marcelo Garrido
- Pontificia Universidad Católica-Universidad Mayor, Santiago, Chile
| | | | | | - Kensei Yamaguchi
- Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | | | - Tianshu Liu
- Zhongshan Hospital Fudan University, Shanghai, China
| | | | | | | | | | | | - Kynan Feeney
- Notre Dame University and Edith Cowan University, Murdoch, Western Australia, Australia
| | - Elena Elimova
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Parul Doshi
- Bristol Myers Squibb, Princeton, NJ, USA
- Gilead Sciences, Foster City, CA, USA
| | | | - Ming Lei
- Bristol Myers Squibb, Princeton, NJ, USA.
| |
Collapse
|
46
|
Berg T, Ahlborn L, Jensen MB, Knoop AS, Ejlertsen B, Rossing M. Comprehensive mapping elucidates high risk genotypes in primary metastatic breast cancer. Neoplasia 2025; 63:101162. [PMID: 40121945 PMCID: PMC11981775 DOI: 10.1016/j.neo.2025.101162] [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: 01/28/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Among women with primary metastatic breast cancer (pMBC), around 5 % of women with primary invasive breast cancer, high-risk mutations associated with disease progression and poor prognosis is shown. The heterogeneity and clinical implications of these genomic alterations remains to be fully elucidated. We performed comprehensive gene mapping on 211 tumors of women diagnosed with pMBC at Rigshospitalet 2014-2021. After DNA purification 203 tumor samples were eligible for analysis. Median age in our cohort was 69 years, 68 % were ER-positive/HER2-negative, 23 % HER2-positive and 9 % triple-negative. A high tumor mutational burden (TMB), shown in 10 %, was in univariable analysis associated with a poor prognosis and a median overall survival of 5.3 months (95 % CI, 2.5-51.3) but no significant association after adjusting for subtype and age. 65 % of tumors had an actionable biomarker, including a PIK3CA mutation in 39 %. TP53 mutations were found in 33 % of tumors and were associated with an increased risk of death (adjusted HR: 1.60, 95 % CI; 1.07-2.40). We have found that for women with pMBC, the disease is driven by several targetable genetic mutations across subtypes, however our results suggest a reduced prognostic value of TMB for this complex patient group. Taken together, our findings substantiate the value of early genomic profiling to actively identify women that may be eligible for a more individualized treatment scheme.
Collapse
Affiliation(s)
- Tobias Berg
- Danish Breast Cancer Group, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
| | - Lise Ahlborn
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Maj-Britt Jensen
- Danish Breast Cancer Group, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ann Søegaard Knoop
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Bent Ejlertsen
- Danish Breast Cancer Group, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Maria Rossing
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Ozair MZ, Halmos B, D’Aiello A, Yun J, Filippi AR, Rimner A, Lin SH, Simone CB, Ohri N. Chemotherapy-Free Treatment with Radiotherapy and Immunotherapy for Locally Advanced Non-Small Cell Lung Cancer. Cancers (Basel) 2025; 17:1524. [PMID: 40361451 PMCID: PMC12071140 DOI: 10.3390/cancers17091524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/23/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Concurrent chemoradiotherapy (CRT) followed by immunotherapy is a standard treatment for locally advanced non-small cell lung cancer (LA-NSCLC), yet many patients are ineligible due to treatment-related toxicity or poor functional status. Chemotherapy-free approaches using radiotherapy (RT) and immunotherapy may offer a safer and equally effective alternative in select patient populations. Methods: A comprehensive literature review was conducted using PubMed, Google Scholar, and relevant conference proceedings focusing on trials between 2000 and 2024. Studies investigating chemotherapy-free regimens combining RT and immunotherapy in LA-NSCLC were analyzed, with emphasis on clinical outcomes, biomarker use, treatment sequencing, radiation dose/fractionation, and safety. Results: Multiple Phase I/II trials reported promising efficacy with one-year progression-free survival (PFS) ranging from 39% to 76%. Toxicity was generally acceptable, though higher-grade adverse events were more frequent in older, frail populations. Trials integrating PD-L1 expression, tumor mutational burden (TMB), and circulating tumor DNA (ctDNA) showed potential for improved patient stratification. Variation in immunotherapy timing (induction, concurrent, or consolidation) and radiation schedules highlight the need for optimization. Conclusions: Chemotherapy-free regimens represent a promising treatment strategy for patients with LA-NSCLC, especially those that are ineligible for standard CRT. Biomarker-driven patient selection and the rational integration of RT and immunotherapy are critical to improving outcomes. Randomized trials are warranted to establish the efficacy and safety of these emerging approaches.
Collapse
Affiliation(s)
- M. Zeeshan Ozair
- Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY 10461, USA; (B.H.); (A.D.); (J.Y.); (N.O.)
| | - Balazs Halmos
- Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY 10461, USA; (B.H.); (A.D.); (J.Y.); (N.O.)
| | - Angelica D’Aiello
- Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY 10461, USA; (B.H.); (A.D.); (J.Y.); (N.O.)
| | - Jaewon Yun
- Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY 10461, USA; (B.H.); (A.D.); (J.Y.); (N.O.)
| | - Andrea R. Filippi
- Department of Oncology and Hematology-Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, University of Milan, 20133 Milano, Italy;
| | - Andreas Rimner
- Department of Radiation Oncology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), Partner Site DKTK-Freiburg, 79106 Freiburg, Germany;
| | - Steven H. Lin
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Charles B. Simone
- New York Proton Center, New York, NY 10461, USA;
- Memorial Sloan Kettering Cancer Center, New York, NY 10461, USA
| | - Nitin Ohri
- Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY 10461, USA; (B.H.); (A.D.); (J.Y.); (N.O.)
| |
Collapse
|
48
|
Doi T, Ishikawa T, Moriguchi M, Itoh Y. Current status of cancer genome medicine for pancreatic ductal adenocarcinoma. Jpn J Clin Oncol 2025; 55:443-452. [PMID: 39893577 DOI: 10.1093/jjco/hyaf012] [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: 09/21/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis; however, advancements in cancer genome profiling using next-generation sequencing have provided new perspectives. KRAS mutations are the most frequently observed genomic alterations in patients with PDAC. However, until recently, it was not considered a viable therapeutic target. Although KRAS G12C mutations for which targeted therapies are already available are infrequent in PDAC, treatments targeting KRAS G12D and pan-KRAS are still under development. Similarly, new treatment methods for KRAS, such as chimeric antigen receptor T-cell therapy, have been developed. Several other potential therapeutic targets have been identified for KRAS wild-type PDAC. For instance, immune checkpoint inhibitors have demonstrated efficacy in PDAC treatment with microsatellite instability-high/deficient mismatch repair and tumor mutation burden-high profiles. However, for other PDAC cases with low immunogenicity, combination therapies that enhance the effectiveness of immune checkpoint inhibitors are being considered. Additionally, homologous recombination repair deficiencies, including BRCA1/2 mutations, are prevalent in PDAC and serve as important biomarkers for therapies involving poly (adenosine diphosphate-ribose) polymerase inhibitors and platinum-based therapies. Currently, olaparib is available for maintenance therapy of BRCA1/2 mutation-positive PDAC. Further therapeutic developments are ongoing for genetic abnormalities involving BRAF V600E and the fusion genes RET, NTRK, NRG, ALK, FGFR2, and ROS1. Overcoming advanced PDAC remains a formidable challenge; however, this review outlines the latest therapeutic strategies that are expected to lead to significant advancements.
Collapse
Affiliation(s)
- Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Medical Oncology Unit, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Michihisa Moriguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
49
|
Yamada A, Kondo T. Hereditary Colorectal Cancer: Clinical Implications of Genomic Medicine and Precision Oncology. J Anus Rectum Colon 2025; 9:167-178. [PMID: 40302859 PMCID: PMC12035340 DOI: 10.23922/jarc.2025-001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 01/17/2025] [Indexed: 05/02/2025] Open
Abstract
Approximately 10% of colorectal cancer (CRC) cases occur in the context of hereditary cancer-predisposing conditions caused by germline pathogenic variants (PVs) in cancer predisposition genes, with Lynch syndrome and familial adenomatous polyposis at the top of the list. Although the identification of hereditary CRC has traditionally relied on clinical characteristics, including familial accumulation, multiple and early onset of CRC and other related cancers, and the presence of gastrointestinal polyposis, more comprehensive approaches, such as universal tumor screening and universal germline testing, have recently been employed. From a technical standpoint, next-generation sequencing has enabled genome-wide analysis of genetic alterations in germline and somatic settings. Taking advantage of this technology, germline multigene panel testing has been utilized in genetic testing, which leads to the identification of PVs, not only in well-known hereditary CRC genes but also in rare causal genes, moderate-risk genes, and high-risk genes previously not linked to CRC predisposition. In addition, comprehensive genomic profiling and companion diagnostics for solid tumors occasionally yield unexpected hereditary CRC diagnoses. Thus, more hereditary CRCs have been identified not based on clinical phenotypes but rather by comprehensive approaches or as secondary findings of treatment drug testing. In this review, we discuss the impact of recent advances in genomic medicine on the clinical aspects of hereditary CRC, which has promoted an understanding of the entire landscape of genetic predisposition to CRC.
Collapse
Affiliation(s)
- Atsushi Yamada
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Tomohiro Kondo
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
- Department of Real-World Data Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
50
|
Inokawa Y, Mizuno H, Yamada M, Kawakatsu S, Watanabe N, Onoe S, Mizuno T, Okayama K, Okumura F, Kajikawa M, Ebata T. Pathological Complete Response after Pembrolizumab Treatment for Unresectable Perihilar Cholangiocarcinoma with High Microsatellite Instability: A Case Report. Surg Case Rep 2025; 11:25-0025. [PMID: 40308703 PMCID: PMC12041437 DOI: 10.70352/scrj.cr.25-0025] [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: 01/20/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
INTRODUCTION Pembrolizumab has been introduced to solid cancers with microsatellite instability (MSI)-high cases; however, its clinical experience for cholangiocarcinoma remains very limited. Here, we present a case who successfully underwent conversion surgery following pembrolizumab treatment for MSI-high perihilar cholangiocarcinoma, which pathologically exhibited complete response. CASE PRESENTATION A 69-year-old male with Bismuth IV perihilar cholangiocarcinoma with bulky lymphadenopathy was referred, who initially required left hepatic trisectionectomy, caudate lobectomy, bile duct resection, and portal vein resection and reconstruction (H123458-B-PV). During the waiting period after preoperative portal vein embolization, the right hepatic artery was involved by rapid tumor progression, needing a modification of the initially scheduled surgical procedure to additional hepatic artery resection and reconstruction (H123458-B-PV-HA). We revised the surgical decision of resectable to locally unresectable disease. He received systemic chemotherapy with gemcitabine and cisplatin as first-line, showing the best effect of stable disease followed by slight tumor progression and re-elevation of tumor marker after 5 courses of treatment. Cancer multi-gene panel analysis using percutaneous biopsy specimen showed the nature of MSI-high. Therefore, he received pembrolizumab treatment as second-line therapy, leading to a drastic downsize >30% in tumor diameter and normalization of the tumor marker as well after only 2 cycles of administration. After confirmation of keeping tumor shrinkage during 22 courses of pembrolizumab treatment without any severe adverse events, we decided to perform conversion surgery and performed left trisectionectomy, caudate lobectomy, and bile duct resection with portal vein resection (H123458-B-PV). Although the right hepatic artery was extensively fibrotic, there was no evidence of malignancy by frozen section histologic diagnosis. The pathological findings showed pathological complete response with no residual tumor cells. The patient is under periodical checkup without adjuvant chemotherapy, and no tumor recurrence was observed at 4 months postoperatively. CONCLUSIONS We experienced clinical partial response but pathological complete response after second-line pembrolizumab treatment for unresectable locally advanced perihilar cholangiocarcinoma with a biologic nature of MSI-high. Conversion surgery may be considered as a promising option for such effective case, whereas there is a possibility to avoid resection in the MSI-high setting.
Collapse
Affiliation(s)
- Yoshikuni Inokawa
- Department of Surgery, Gifu Prefectural Tajimi Hospital, Tajimi, Gifu, Japan
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hironori Mizuno
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Mihoko Yamada
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shoji Kawakatsu
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Nobuyuki Watanabe
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shunsuke Onoe
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Takashi Mizuno
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kohei Okayama
- Department of Gastroenterology, Gifu Prefectural Tajimi Hospital, Tajimi, Gifu, Japan
| | - Fumihiro Okumura
- Department of Gastroenterology, Gifu Prefectural Tajimi Hospital, Tajimi, Gifu, Japan
| | - Masaki Kajikawa
- Department of Surgery, Gifu Prefectural Tajimi Hospital, Tajimi, Gifu, Japan
| | - Tomoki Ebata
- Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|