1
|
Movva S, Seier K, Bradic M, Charalambous K, Rosenbaum E, Kelly CM, Cohen SM, Hensley ML, Avutu V, Banks LB, Chan JE, Chi P, D'Angelo S, Dickson MA, Gounder MM, Keohan ML, Maki RG, Green A, Makker V, Rubinstein MM, Saunds S, Cho JM, Lefkowitz RA, Erinjeri J, Qin LX, Shah R, Wong P, Tap W. Phase II study of rucaparib and nivolumab in patients with leiomyosarcoma. J Immunother Cancer 2025; 13:e012020. [PMID: 40514070 PMCID: PMC12164637 DOI: 10.1136/jitc-2025-012020] [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: 03/11/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Objective responses to immune checkpoint inhibitors (ICI) in leiomyosarcoma (LMS) are rare. Response rates may be increased by combination with other drugs known to promote immune infiltration, such as poly(ADP-ribose) polymerase (PARP) inhibitors, which have led to benefit in BRCA-altered uterine LMS. We therefore evaluated the combination of a PARP inhibitor, rucaparib, and the anti-programmed death receptor-1 monoclonal antibody, nivolumab, in patients with advanced LMS and investigated its effects on the tumor immune microenvironment. METHODS This was an open-label, single-center, single-arm, phase II study in patients with advanced refractory LMS. Full protocol available Patients were treated with rucaparib 600 mg orally, two times daily, continuously and nivolumab 480 mg intravenously on day 1 of a 28-day cycle. Re-staging scans were performed every 8 weeks. Blood and tissue samples were collected at baseline and at week 8 on treatment. The primary objective was the best objective response rate by 24 weeks using Response Evaluation Criteria in Solid Tumour (RECIST V.1.1). Secondary objectives included treatment-related toxicity, progression-free survival, overall survival, and changes in immune pathways in blood and tumor. RESULTS 20 patients with LMS were enrolled. There was one partial response (PR) (5%) in a patient with uterine LMS and a somatic BRCA deep deletion. 19 (95%) patients had a treatment-related adverse event (TRAE) and 7 (35%) had a grade 3 or higher TRAE. Interferon (IFN) α and γ hallmark pathways were more highly expressed in patients who derived benefit from treatment (at least stable disease by 16 weeks) vs those who did not in both baseline (adjusted p=0.005 for IFN-α, 0.03 for IFN-γ) and on-treatment biopsies (adjusted p=0.0002 for IFN-α, 0.0001 for IFN-γ), but the abundance of tumor immune cell populations did not differ between these groups at either time point. CONCLUSION The addition of a PARP inhibitor did not improve the efficacy of ICI in LMS. Adverse events, especially due to overlapping toxicities, were frequent and often led to dose delays and modifications.
Collapse
Affiliation(s)
- Sujana Movva
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Kenneth Seier
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Martina Bradic
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Karmelina Charalambous
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Evan Rosenbaum
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Ciara M Kelly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Seth M Cohen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Martee L Hensley
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Viswatej Avutu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Lauren B Banks
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jason E Chan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Ping Chi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Sandra D'Angelo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Mark A Dickson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Mrinal M Gounder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Mary L Keohan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Robert G Maki
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Angela Green
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Vicky Makker
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Maria M Rubinstein
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Sara Saunds
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jae-Mun Cho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Robert A Lefkowitz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Joseph Erinjeri
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Li-Xuan Qin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ronak Shah
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Phillip Wong
- Immune Monitoring Facility, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - William Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
2
|
Kokkali S, Dolcan A, Boye K, Kyriazoglou A, Boukovinas I, Kalofonou F, Koumarianou A, Asimakopoulou N, Vorrias E, Tsapakidis K, Georgaki E, Boulouta A, Mavroeidis L, Harneshaug M, Theocharis S, Jones RL, Digklia A. Real-world data on immune checkpoint inhibitors in advanced sarcomas across multiple European institutions. Acta Oncol 2025; 64:761-768. [PMID: 40495323 DOI: 10.2340/1651-226x.2025.43135] [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: 02/11/2025] [Accepted: 05/01/2025] [Indexed: 06/18/2025]
Abstract
BACKGROUND Following the success of immune checkpoint inhibitors (ICI) in other cancer types, their role is being evaluated in sarcomas. They have been assessed as monotherapy, or in combination with other ICI, chemotherapeutic drugs and tyrosine kinase inhibitors (TKI) in several clinical trials. So far the results have been limited to non-selected sarcoma populations. Further work is required to select patients who will benefit from immunotherapy. PATIENTS AND METHODS We conducted a pooled retrospective analysis of the use of ICI in patients with advanced sarcomas in multiple European institutions. ICI-based treatments included ICI monotherapy (n = 43, 59.7%), double ICI (n = 5, 6.9%), ICI plus TKI (n = 21, 29.2%) and ICI plus chemotherapy (n = 3, 4.2%). RESULTS Seventy-two patients from 10 European institutions, with metastatic (87.5%) or locally advanced (12.5%) disease were included. The most common subtype was undifferentiated pleomorphic sarcoma (16.7%), followed by leiomyosarcoma (12%), liposarcoma (10%) and angiosarcoma (9.7%). The median number of prior lines of systemic therapy was 2 (0-8). The objective response rate was 34.4% and was higher in combination regimens versus ICI monotherapy. With a median follow-up of 20.7 months, median progression-free survival (PFS) was 4.6 and median overall survival (OS) 18.8 months. Line of therapy (1st/2nd vs. ≥ 3rd line) and best response to ICI was significantly associated with PFS and OS. Histological subtype was significantly associated with OS. Toxicity was in general manageable; only six (8.3%) patients discontinued therapy for AE. INTERPRETATION Our study provided additional real-world data on the outcome of ICI in patients with advanced sarcomas.
Collapse
Affiliation(s)
- Stefania Kokkali
- Oncology Unit, Second Department of Medicine, University of Athens, Hippocratio General Hospital of Athens, Athens, Greece
| | - Ana Dolcan
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Kjetil Boye
- Department of Oncology, Oslo University Hospital, Oslo, Norway.
| | - Anastasios Kyriazoglou
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | | | - Foteini Kalofonou
- The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | - Anna Koumarianou
- Fourth Department of Internal Medicine, Attikon University Hospital, Athens, Greece
| | | | - Eleftherios Vorrias
- Department of Medical Oncology, General University Hospital of Heraklion, Heraklion, Greece
| | | | - Eleni Georgaki
- Oncology Unit, Second Department of Medicine, University of Athens, Hippocratio General Hospital of Athens, Athens, Greece
| | - Anna Boulouta
- Second Department of Internal Medicine, Oncology Unit, Attikon University Hospital, Athens, Greece
| | - Leonidas Mavroeidis
- The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | | | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Robin L Jones
- The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | - Antonia Digklia
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
3
|
Hua Y, Wang C, Li F, Han Y, Zuo D, Lv Y, Sun M, Yuan P, Yuan R, Zhang F, Ma L, Wang Y, Wu H, Zhou G, Lin Q, Wang S, Li N, Lu Y, North China Petroleum Bureau General Hospital. Phase 1, open-label, multicenter, dose escalation safety and tolerability study of oncolytic virus OVV-01 in advanced solid tumors. J Immunother Cancer 2025; 13:e011517. [PMID: 40480657 PMCID: PMC12142144 DOI: 10.1136/jitc-2025-011517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/25/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND OVV-01 is a genetically engineered vesicular stomatitis virus oncolytic virus designed to selectively amplify in tumor cells and express tumor-associated antigen NY-ESO-1. This study was designed to evaluate the safety, tolerability, and efficacy of OVV-01 in patients with advanced solid tumors. METHODS This is a phase 1, first-in-human, open-label, multicenter study of OVV-01 in patients with advanced solid tumors. OVV-01 was intratumorally injected biweekly (every two weeks, Q2W), 3 weeks after the first dose for a total of six doses. Dose escalation follows a 3+3 design at four doses of 6×107 Plaue-Forming Unit (PFU), 6×108 PFU, 6×109 PFU, and 1.2×1011 PFU. The primary endpoints were safety and tolerability. The second endpoints included overall response rate (ORR) and disease control rate (DCR) of OVV-01, by investigators per Response Evaluation Criteria in Solid Tumors V.1.1. RESULTS 18 patients were enrolled into four dose groups, among whom 6 were soft tissue sarcoma (STS). No dose-limiting toxicities and treatment-related severe adverse events were observed. 11 patients were evaluated for efficacy, and the ORR was 27.3%, and the DCR was 63.6%. Among the four evaluable patients with advanced STS, the ORR was 75%. Two patients with STS achieved CR at doses above 6.0×109 PFU. CONCLUSIONS The intratumor injection of OVV-01 was safe and well-tolerated in patients with advanced solid tumors. A significant response was observed in patients with STS. TRIAL REGISTRATION NUMBER NCT04787003.
Collapse
Affiliation(s)
- Yingqi Hua
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chongren Wang
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Li
- GoBroad Medical (Hematology), Beijing Research Center / Beijing GoBroad Boren Hospital, Beijing, China
| | - Yanjie Han
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Dongqing Zuo
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Lv
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengxiong Sun
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Yuan
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruirong Yuan
- Dowlin Biomed, New Hampshire, New Hampshire, USA
| | - Fan Zhang
- Joint Biosciences (SH) Ltd, Shanghai, Shanghai, China
| | - Liang Ma
- Joint Biosciences (SH) Ltd, Shanghai, Shanghai, China
| | - Yan Wang
- Joint Biosciences (SH) Ltd, Shanghai, Shanghai, China
| | - Hui Wu
- Joint Biosciences (SH) Ltd, Shanghai, Shanghai, China
| | - Guoqing Zhou
- Joint Biosciences (SH) Ltd, Shanghai, Shanghai, China
| | - Qiang Lin
- North China Petroleum Bureau General Hospital, Cangzhou, Hebei, China
| | - Shuhang Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ning Li
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yinying Lu
- The 5th medical Center of PLA general Hospital, Beijing, China
| | | |
Collapse
Collaborators
Haitao Liu,
Collapse
|
4
|
Bai G, Zhao S, Zhao M, Chen L, Chen W. The phosphatase CTDSPL2 promotes proliferation, invasion, metastasis and regorafenib resistance in osteosarcoma. J Bone Oncol 2025; 52:100684. [PMID: 40352265 PMCID: PMC12063119 DOI: 10.1016/j.jbo.2025.100684] [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: 02/04/2025] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
Osteosarcoma is the most common bone malignancy in children and adolescents. Patients with metastatic and recurrent osteosarcoma have poor prognosis. Regorafenib is a multi-kinase inhibitor recommended as a complement to standard chemotherapy in the treatment of advanced osteosarcoma. The mechanisms associated with regorafenib resistance remains unclear. In this study we performed transcriptomics, proteomics and phosphorylated proteomics using regorafenib-treated osteosarcoma cell lines (MG-63, HOS-MNNG for transcriptomics, HOS-MNNG for proteomics and phosphorylated proteomics). After comprehensive multiomics and verification analyses of differentially expressed genes, essential genes for the malignancy of osteosarcoma cells were identified. The effects of essential genes on the proliferation, invasion, and migration of osteosarcoma were determined. The study also evaluated their role in the apoptosis of osteosarcoma cells. The up-regulation of essential genes was determined by immunohistochemistry assays. Using comprehensive multiomics and verification analyses we found that the CTDSPL2 gene might play a role in the malignancy and Regorafenib resistance in osteosarcoma. In vitro and clinical specimen assays demonstrated that CTDSPL2 promotes the proliferation, invasion and metastasis of osteosarcoma cells, while inhibiting tumor cell apoptosis. In conclusion CTDSPL2 was identified as an essential gene for survival of osteosarcoma cells. Knockdown of CTDSPL2 expression significantly inhibited the proliferation, invasion, and metastasis of osteosarcoma cells, suggesting that it is involved in the formation and development of osteosarcoma tumors. Our data showed that CTDSPL2 is a potential therapeutic target for patients with osteosarcoma.
Collapse
Affiliation(s)
- Guannan Bai
- Department of Orthopedics, Children’s Hospital, Zhejiang University School of Medicine, National Children’s Regional Medical Center, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, Zhejiang Province 310052, China
| | - Shaobo Zhao
- Department of Orthopedics, Children’s Hospital, Zhejiang University School of Medicine, National Children’s Regional Medical Center, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, Zhejiang Province 310052, China
| | - Manli Zhao
- Department of Pathology, Children’s Hospital, Zhejiang University School of Medicine, National Children’s Regional Medical Center, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, Zhejiang Province 310052, China
| | - Limiao Chen
- Children’s Hospital, Zhejiang University School of Medicine, National Children’s Regional Medical Center, National Clinical Research Center for Child Health, 3333 Bingsheng Road, Hangzhou, Zhejiang Province 310052, China
| | - Wenhao Chen
- Department of Orthopedics, Children’s Hospital, Zhejiang University School of Medicine, National Children’s Regional Medical Center, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, Zhejiang Province 310052, China
| |
Collapse
|
5
|
Chen X, Tian B, Wang Y, Zheng J, Kang X. Harnessing multi‑omics to revolutionize understanding and management of osteosarcoma: A pathway to precision medicine (Review). Int J Mol Med 2025; 55:92. [PMID: 40242955 PMCID: PMC12021390 DOI: 10.3892/ijmm.2025.5533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Osteosarcoma, the most prevalent primary bone malignancy in children and adolescents, poses significant challenges due to its aggressive nature and propensity for metastasis. Despite advances in treatment, survival rates for high‑risk patients remain unsatisfactory, underscoring the urgent need for innovative approaches. This review explores the vital role of multi‑omics‑integrating genomics, transcriptomics, proteomics and metabolomics‑in unraveling the complex biological landscapes of osteosarcoma. By providing comprehensive insights into tumor heterogeneity, signaling pathways and metabolic reprogramming, multi‑omics facilitates the identification of novel biomarkers and therapeutic targets. The objective of the present study was to highlight the transformative potential of multi‑omics in enhancing the understanding and management of osteosarcoma, ultimately paving the way for personalized treatment strategies and improved patient outcomes. Through this synthesis, the study calls for a concerted effort to integrate multi‑omics into clinical practice, fostering a more precise approach to osteosarcoma care.
Collapse
Affiliation(s)
| | | | | | - Jiang Zheng
- Sports Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Xin Kang
- Sports Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
6
|
Crane E, Gaillard S, Hensley ML. Rare Uterine Tumors: What to Do? Am Soc Clin Oncol Educ Book 2025; 45:e473106. [PMID: 40340459 DOI: 10.1200/edbk-25-473106] [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: 05/10/2025]
Abstract
Rare uterine malignancies present treatment challenges because of their clinical and biological heterogeneity. Among the rarest of the uterine cancers are leiomyosarcomas, uterine stromal tumors, and the mesonephric-like and serous carcinomas. In this article, we review recent advancements in diagnostic precision, risk stratification, and identification of biomarker-guided therapeutic options for these rare subtypes of uterine tumors. The improved understanding of the molecular profile of these tumors has led to the development of targeted treatment approaches. Further progress will depend on a coordinated, global effort to further characterize these diseases and enroll patients on biomarker-driven clinical trials.
Collapse
Affiliation(s)
- Erin Crane
- Levine Cancer, Atrium Health, Charlotte, NC
| | | | | |
Collapse
|
7
|
Ullah A, Goodbee M, Malham K, Khan Yasinzai AQ, Mirza MB, Tareen B, Khan A, Idrees K. Comparative analysis of bone and soft tissue vs. visceral synovial sarcoma: Demographic, clinical, and survival outcomes; a retrospective population-based study. Surgeon 2025; 23:167-173. [PMID: 39528367 DOI: 10.1016/j.surge.2024.11.003] [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: 09/05/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Synovial sarcoma (SS) is typically diagnoses in young adults and usually appears in the extremities and soft tissues. However, it can sometimes arise in visceral organs. This study examines the differences in patient demographics, clinical features, and survival rates between soft tissue and visceral synovial sarcoma. METHODS We analyzed data from the Surveillance, Epidemiology, and End Results (SEER) database spanning 2000-2018. ANOVA (analysis of variance) was used to identify significant factors for multivariate analysis. Cox regression modeling was used to determine independent risk factors for survival. RESULTS Of the 2,776 SS patients included, the median age was 39.0 years, predominating male (53.0 %) and white (81.3 %). Bone and soft tissue sarcomas were more common, accounting for 86.4 % of cases (p-value<0.001), while the rest were diagnosed as visceral sarcomas. Visceral SS patients were typically older (p-value<0.001), male (56.8 %), and white (84.8 %). They also presented more frequently with distant metastasis (HR 3.8, 95 % CI, 2.4-6.1), had larger tumors on average (HR for tumors >10 cm: 2.9, 95 % CI, 2.0-4.1), and were less likely to undergo surgery (HR 0.4, 95 % CI, 0.3-0.6). Despite receiving multimodal treatments, including surgery, radiation, and/or chemotherapy, visceral SS patients exhibited poorer overall survival compared to their bone and soft tissue SS counterparts (p-value<0.001). CONCLUSION Visceral SS often presents in older patients with advanced-stage and larger tumor size as compared to bone and soft tissue SS, which likely contributes to poorer survival. Advanced age, regional spread, and larger tumor size were all found to worsen outcomes, while surgery and radiation were found to be protective factors.
Collapse
Affiliation(s)
- Asad Ullah
- Department of Pathology, Texas Tech University Health Sciences, Lubbock, TX, USA.
| | - Mya Goodbee
- Medical College of Georgia, Augusta, GA, USA
| | - Kali Malham
- Medical College of Georgia, Augusta, GA, USA
| | | | | | | | - Aimal Khan
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kamran Idrees
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
8
|
Yang J, Lu X, Cai Q, Liu M, Xia T, Hong D, Le L, Zhang X, Zhang X. Loss of TACC2 impairs chemokine CCL3 and CCL4 expression and reduces response to anti-PD-1 therapy in soft tissue sarcoma. Mol Cancer 2025; 24:158. [PMID: 40442694 PMCID: PMC12123857 DOI: 10.1186/s12943-025-02354-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 05/14/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Soft tissue sarcoma (STS) is a rare, heterogeneous malignancy with limited treatment options for metastatic disease. Despite advances in immunotherapy, including PD-1 inhibitors, clinical outcomes remain suboptimal, highlighting the need for novel biomarkers and therapeutic strategies. This study investigated the role of TACC2 in STS, focusing on its impact on the immune microenvironment and immunotherapy response. METHODS Whole-exome sequencing was performed to characterize TACC2-related genomic alterations in STS cohorts, complemented by immunohistochemistry for protein-level validation. Mechanistic insights were obtained through chromatin immunoprecipitation (ChIP) and co-immunoprecipitation assays, focusing on TACC2's interaction with the NuRD/CoREST complex. The efficacy of anti-PD-1 therapy was evaluated in TACC2-overexpressing mouse models, and clinical relevance was analyzed using patient survival and treatment response data. RESULTS TACC2 acted as a tumor suppressor in STS, with low expression associated with poor overall survival. Mechanistically, TACC2 enhanced CCL3 and CCL4 transcription, promoting CD8 + T cell infiltration by inhibiting NuRD/CoREST nuclear translocation. In vivo, TACC2 overexpression synergized with PD-1 blockade therapy, leading to a significant reduction in tumor volume and prolonged survival. Clinically, high TACC2 expression was associated with improved responses to immunotherapy. CONCLUSIONS In conclusion, TACC2 is an important regulator of the immune response in STS, functioning as a tumor suppressor and a modulator of response to PD-1 blockade. Its role in modulating chemokine expression and CD8 + T cell infiltration highlights its potential as a therapeutic target and predictive biomarker for STS immunotherapy.
Collapse
Affiliation(s)
- Jing Yang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Xiuxia Lu
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P.R. China
| | - Qiyan Cai
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Mengmeng Liu
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang University, Nanchang, Jiangxi, 330000, P.R. China
| | - Tianliang Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Dongchun Hong
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Liyuan Le
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Xinke Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xing Zhang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China.
| |
Collapse
|
9
|
Kumar R, Sporn K, Khanna A, Paladugu P, Gowda C, Ngo A, Jagadeesan R, Zaman N, Tavakkoli A. Integrating Radiogenomics and Machine Learning in Musculoskeletal Oncology Care. Diagnostics (Basel) 2025; 15:1377. [PMID: 40506947 PMCID: PMC12155258 DOI: 10.3390/diagnostics15111377] [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: 05/04/2025] [Revised: 05/21/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025] Open
Abstract
Musculoskeletal tumors present a diagnostic challenge due to their rarity, histological diversity, and overlapping imaging features. Accurate characterization is essential for effective treatment planning and prognosis, yet current diagnostic workflows rely heavily on invasive biopsy and subjective radiologic interpretation. This review explores the evolving role of radiogenomics and machine learning in improving diagnostic accuracy for bone and soft tissue tumors. We examine integrating quantitative imaging features from MRI, CT, and PET with genomic and transcriptomic data to enable non-invasive tumor profiling. AI-powered platforms employing convolutional neural networks (CNNs) and radiomic texture analysis show promising results in tumor grading, subtype differentiation (e.g., Osteosarcoma vs. Ewing sarcoma), and predicting mutation signatures (e.g., TP53, RB1). Moreover, we highlight the use of liquid biopsy and circulating tumor DNA (ctDNA) as emerging diagnostic biomarkers, coupled with point-of-care molecular assays, to enable early and accurate detection in low-resource settings. The review concludes by discussing translational barriers, including data harmonization, regulatory challenges, and the need for multi-institutional datasets to validate AI-based diagnostic frameworks. This article synthesizes current advancements and provides a forward-looking view of precision diagnostics in musculoskeletal oncology.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.G.); (A.N.)
| | - Kyle Sporn
- Norton College of Medicine, Upstate Medical University, Syracuse, NY 13210, USA;
| | - Akshay Khanna
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.K.); (P.P.)
| | - Phani Paladugu
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.K.); (P.P.)
- Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Chirag Gowda
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.G.); (A.N.)
| | - Alex Ngo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.G.); (A.N.)
| | - Ram Jagadeesan
- Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA;
- Cisco AI Systems, Cisco Inc., San Jose, CA 95134, USA
| | - Nasif Zaman
- Department of Computer Science, University of Nevada Reno, Reno, NV 89557, USA; (N.Z.); (A.T.)
| | - Alireza Tavakkoli
- Department of Computer Science, University of Nevada Reno, Reno, NV 89557, USA; (N.Z.); (A.T.)
| |
Collapse
|
10
|
Jonczak E, Trent J, Roland C, Haddad EN. Liposarcoma: Novel Approaches to Systemic Therapy and Multidisciplinary Care. Hematol Oncol Clin North Am 2025:S0889-8588(25)00048-6. [PMID: 40414786 DOI: 10.1016/j.hoc.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Retroperitoneal well-differentiated liposarcoma (WDLPS) and dedifferentiated liposarcoma (DDLPS) are rare tumors presenting as bulky abdominal tumors requiring multidisciplinary management in high-volume centers. Surgery is the mainstay of treatment through complete en bloc resection with the goal of achieving macroscopically complete resection, with a single specimen encompassing the tumor and involved contiguous organs. Preoperative radiation therapy is not standard of care and the role of preoperative chemotherapy is under investigation. If the tumor is not resectable or metastatic, the preferred treatment is doxorubicin-based chemotherapy in the case of DDLPS, whereas WDLPS are generally thought of as chemo-resistant.
Collapse
Affiliation(s)
- Emily Jonczak
- Department of Sarcoma Medical Oncology, University of Miami Sylvester Comprehensive Cancer Center, 1475 NW 12th Avenue, Miami, FL 33136, USA.
| | - Jonathan Trent
- Department of Sarcoma Medical Oncology, University of Miami Sylvester Comprehensive Cancer Center, 1475 NW 12th Avenue, Miami, FL 33136, USA
| | - Christina Roland
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Elise Nassif Haddad
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| |
Collapse
|
11
|
Fuchs B, Schelling G, Glanzmann C, Studer G, on behalf of the Swiss Sarcoma Network. From Intensification to Optimization: Balancing Efficacy, Safety, and Costs in High-Risk Localized Soft Tissue Sarcomas. Cancers (Basel) 2025; 17:1724. [PMID: 40427221 PMCID: PMC12109959 DOI: 10.3390/cancers17101724] [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: 03/14/2025] [Revised: 04/24/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: The SU2C-SARC032 randomized controlled trial (RCT) tested pembrolizumab combined with preoperative normofractionated radiotherapy as an intensified treatment for high-risk stage III resectable soft tissue sarcoma (STS), demonstrating a moderate improvement in disease-free survival (DFS) compared to preoperative radiotherapy alone, but accompanied by significantly increased toxicity, prolonged treatment durations, elevated resource source, and limited real-world applicability. To address the gap between highly controlled trial outcomes and routine clinical practice, this comparative analysis evaluated a streamlined ultra-hypofractionated preoperative radiotherapy (uhpRT) protocol using real-world data (RWD) as a potentially more balanced approach. Methods: Prospectively collected observational RWD from 54 consecutive patients with Stage III (T2 N0 M0) high-risk resectable STS treated at a single institution with uhpRT (25 Gy in 5 fractions in one week, no systemic therapy, median interval of 14 days to surgery) were analyzed. Survival endpoints (overall survival [OS], DFS, local disease-free survival [LDFS], distant disease-free survival [DDFS]), toxicity, and treatment duration were compared qualitatively with published outcomes from the SU2C-SARC032 trial's intensified pembrolizumab arm and control arm. Results: At 2 years, the optimized uhpRT protocol achieved OS (90%), DFS (66%), and DDFS (70%) comparable to the intensified pembrolizumab arm (OS: 88%, DFS: 67%, DDFS (67%)) and clearly exceeded outcomes of the control arm (OS/DFS/DDFS: 85%/52%/52%). Importantly, the uhpRT protocol markedly reduced treatment-related toxicities (0% Grade 3/4 events vs. 56% in the intensified trial arm) and total treatment duration (<1 month vs. 3-11 months). Conclusions: These findings challenge the necessity of broad treatment intensification for high-risk localized STS, strongly supporting the concept of therapeutic optimization. Given substantial real-world variability in treatment practices and feasibility highlighted by recent research, our findings advocate for treatment strategies that prioritize realistic applicability, patient safety, and value-based care principles over pure intensification.
Collapse
Affiliation(s)
- Bruno Fuchs
- Faculty of Health Sciences & Medicine, University Lucerne, Frohburgstrasse 3, 6002 Luzern, Switzerland
- Sarcoma Service, Department of Orthopedics and Trauma, Sarcoma Center, LUKS University Hospital, 6000 Luzern, Switzerland
| | - Georg Schelling
- Sarcoma Service, Department of Orthopedics and Trauma, Sarcoma Center, LUKS University Hospital, 6000 Luzern, Switzerland
| | - Christoph Glanzmann
- Sarcoma Service, Department of Radiation Oncology, Sarcoma Center, LUKS University Hospital, 6000 Luzern, Switzerland
| | - Gabriela Studer
- Faculty of Health Sciences & Medicine, University Lucerne, Frohburgstrasse 3, 6002 Luzern, Switzerland
- Sarcoma Service, Department of Radiation Oncology, Sarcoma Center, LUKS University Hospital, 6000 Luzern, Switzerland
| | | |
Collapse
|
12
|
Xu T, Xiao W, Li W, Xu X, Zhang H, Zhang X. Exploring the causal relationship between immune factors and chondrosarcoma: a Mendelian randomization study. Discov Oncol 2025; 16:801. [PMID: 40382743 PMCID: PMC12086138 DOI: 10.1007/s12672-025-02654-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 05/09/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Previous studies have investigated the potential role of immune factors in chondrosarcoma (CHS). However, the causal relationship is unknown. METHODS A two-sample Mendelian randomization (MR) was used to explore potential correlations between 731 immunocyte phenotypes, 91 inflammatory proteins, and CHS. The data were derived from published summary statistics of genome-wide association studies. Inverse-variance weighted was employed as the primary method. Furthermore, a range of analytical methods, including MR-Egger, weighted mode, and weighted median was used to enhance the robustness of the results. A two-step MR was used to assess the mediating effects of inflammatory proteins. Subsequently, sensitivity and MR Steiger directionality tests were performed. RESULTS MR analyses showed that 12 immunocyte phenotypes were positively correlated with CHS (P < 0.05, OR > 1), and 11 immunocyte phenotypes were negatively correlated with CHS (P < 0.05, OR < 1). Five inflammatory proteins were positively associated with CHS (P < 0.05, OR > 1). No heterogeneous or horizontal pleiotropy was found. The MR Steiger analysis found no statistically significant evidence of reverse causation. Mediation analysis did not identify any potential mediating effects. CONCLUSION Our study underscores the pivotal role of immune factors in CHS and offers insights that can inform future research.
Collapse
Affiliation(s)
- Taichuan Xu
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Wentao Xiao
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Wenjie Li
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Xianfa Xu
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Haiwen Zhang
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Xian Zhang
- Department of Spine, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, 214072, Jiangsu, China.
| |
Collapse
|
13
|
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
|
14
|
Miwa S, Yamamoto N, Hayashi K, Taniguchi Y, Yonezawa H, Morinaga S, Demura S. Current and emerging systemic treatment options for malignant fibrous histiocytoma/undifferentiated pleomorphic sarcoma. Int J Clin Oncol 2025:10.1007/s10147-025-02712-6. [PMID: 40366546 DOI: 10.1007/s10147-025-02712-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/23/2025] [Indexed: 05/15/2025]
Abstract
Undifferentiated pleomorphic sarcoma (UPS)/malignant fibrous histiocytoma (MFH) is the second most common soft-tissue sarcoma. The standard treatment options for UPS/MFH include tumor excision with appropriate surgical margins, radiation therapy, and chemotherapy. Preferable clinical outcomes can be expected in patients with resectable disease, whereas the clinical outcomes in patients with metastatic disease are unsatisfactory despite multidisciplinary treatment. Although patients with metastatic diseases require chemotherapy, the response rate to conventional chemotherapy has been reported to be only 27-33% in previous reports. Systemic treatment is required to eliminate metastatic disease and improve clinical outcomes in patients with UPS/MFH. Recent clinical studies have investigated the optimal period of conventional chemotherapy and the efficacy of various combinations of anticancer agents. Furthermore, molecular targeted drugs and immune checkpoint inhibitors have shown superior outcomes compared to standard treatments for various types of malignancies. Therefore, these anticancer agents are considered as new treatment options for patients with UPS/MFH. Recent clinical trials have demonstrated the safety and efficacy of these agents in patients with soft-tissue sarcomas, including UPS/MFH. In particular, a high response rate to immune checkpoint inhibitors combined with doxorubicin has been reported in recent clinical trials; however, combination therapy needs to be assessed in a large number of patients with UPS/MFH. In this review article, recent clinical studies on the systemic treatment of UPS/MFH are discussed.
Collapse
Affiliation(s)
- Shinji Miwa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan.
| | - Norio Yamamoto
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan
| | - Katsuhiro Hayashi
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan
| | - Yuta Taniguchi
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan
| | - Hirotaka Yonezawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan
| | - Sei Morinaga
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan
| | - Satoru Demura
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan
| |
Collapse
|
15
|
Farooq MS, Shafique N, Vargas GM, Guo J, Miura JT, Lefler DS, Karakousis GC. Neoadjuvant Immunotherapy for Resectable Dedifferentiated Liposarcoma: A National Cohort Analysis. J Surg Oncol 2025. [PMID: 40358214 DOI: 10.1002/jso.28155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/03/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Neoadjuvant immunotherapy (NIT) with checkpoint blockade has been increasingly studied for soft tissue sarcomas, however, survival outcomes data are limited, and dedifferentiated liposarcoma (DDLPS) histology remains underrepresented in recent trial cohorts. We assessed the impact of NIT with or without radiation therapy (RT) on overall survival (OS) for resectable DDLPS. METHODS The National Cancer Database (NCDB) was used to identify patients diagnosed with nonmetastatic DDLPS who received NIT and underwent surgical resection between 2016 and 2022. Primary outcome was 5-year OS. RESULTS A total of 3414 patients with DDLPS met the inclusion criteria and NIT was administered to 31 (1%) patients. Factors associated with receipt of NIT were receipt of neoadjuvant RT (NRT, odds ratio [OR]: 5.75, p < 0.001) and male sex (OR: 3.33, p = 0.036). NIT was associated with a hazard ratio (HR) for mortality of 0.89 (p = 0.786). No difference was found in 5-year OS in the overall cohort (NIT 72% vs. 61% no NIT, p = 0.320) or in the propensity-matched cohort (68% vs. 65%, p = 0.848). Subanalysis between NIT with NRT versus NRT-only also did not find any significant difference in 5-year OS (88% vs. 59%, p = 0.331). CONCLUSION In this retrospective NCDB analysis of patients with resectable DDLPS, administration of NIT did not significantly affect OS.
Collapse
Affiliation(s)
- Mohammad S Farooq
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Neha Shafique
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gracia M Vargas
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jennifer Guo
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John T Miura
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel S Lefler
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Giorgos C Karakousis
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
16
|
Liu H, Wang X, Wang X, Qiu F, Zhou B. Challenges and hope: latest research trends in the clinical treatment and prognosis of liposarcoma. Front Pharmacol 2025; 16:1529755. [PMID: 40421219 PMCID: PMC12104207 DOI: 10.3389/fphar.2025.1529755] [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: 11/17/2024] [Accepted: 04/21/2025] [Indexed: 05/28/2025] Open
Abstract
Liposarcoma, as a complex disease, is characterized by intricate interactions between distinct histopathological subtypes and corresponding clinical outcomes, emphasizing the necessity of personalized approaches in diagnosis and treatment strategies. This malignant tumor originating from adipose tissue is classified into different subtypes with specific molecular markers, which not only distinguish them but also guide treatment directions. The main approach for treating liposarcoma is surgical resection, with the aim of complete excision and achieving clean margins (R0 resection) to minimize the risk of recurrence. This surgical principle emphasizes the critical need for precise preoperative planning, and in certain cases, the integration of neoadjuvant therapy may be needed to reduce the tumor to a surgically manageable size. In addition to surgery, systemic therapy plays a key role in the advanced stages of the disease, especially when resistance to traditional treatment arises. The emergence of novel systemic therapies, including chemotherapy, targeted therapy, and immunotherapy, has opened new avenues for treating this challenging malignancy. These systemic therapies are selected on the basis of the specific molecular features of the tumor, highlighting the importance of detailed molecular diagnostics. As our understanding of the molecular basis of liposarcoma deepens, integrating clinical and molecular features is crucial for optimizing treatment outcomes. This comprehensive approach, which combines surgical precision with systemic therapy innovations, will change the treatment landscape for patients with liposarcoma, advancing toward more personalized and effective treatment strategies.
Collapse
Affiliation(s)
- Hongliang Liu
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xi Wang
- Department of Oncology, Women and Children’s Hospital Affiliated to Qingdao University, Qingdao, China
| | - Xiaoyu Wang
- Department of Anesthesiology Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fabo Qiu
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery and Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
17
|
Wu H, Sun Z, Zeng G, Yang J, Deng C, Li C. Mapping Knowledge Structure and Themes Trends of Ewing Sarcoma: A Text-Mining Study. Ann Surg Oncol 2025; 32:3724-3740. [PMID: 39971856 DOI: 10.1245/s10434-025-16978-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Ewing sarcoma (ES) is a rare type of cancer that primarily affects children and young adults. In recent years, there have been notable advancements in the treatment of ES due to the introduction of neoadjuvant chemotherapy and targeted therapy, as well as a wealth of related studies in this field published by scholars around the world. However, there is still a lack of a comprehensive report to aid researchers in quickly understanding the knowledge structure and emerging trends of this area. MATERIALS AND METHODS We conducted a comprehensive search in the Web of Science Core Collection for relevant studies published from 2000 to 2023. To conduct a thorough scientometric analysis and create network visualizations, we utilized three different tools, namely, an online analysis platform, CiteSpace, and VOSviewer. These tools enabled us to examine various aspects such as annual publication output; active journals; contributions from countries, institutions, and authors; references; and keywords. RESULTS A total of 5163 qualified publications were included. The annual number of publications on ES showed a significant linear growth trend (R2 = 0.927). The USA stands out as the most productive country. The institution and author that contributed the largest number of papers were University of Texas System and Dirksen U., respectively. As can be seen from highly cited studies and reference analysis and application of Surveillance, Epidemiology, and End Results (SEER) database, preclinical and clinical trials of multiple combination treatment strategies, especially with different approach of immunotherapy such as CAR-T cells or anti-PD-1 antibody for ES, are the primary focus of the current study. Keyword analysis identified several research topics including immunotherapy, tumor microenvironment, microRNA, biomarkers, EWSR1, metastasis, epigenetics, epithelial mesenchymal transition, mesenchymal stem cell, and combination therapy, which have emerged as frontiers in the field and hold significant potential as ongoing research hotspots. CONCLUSIONS ES is a field that has continued to receive increasing attention in recent years. Harnessing current immunotherapy approaches to improve outcomes of patients with ES is an area of intense interest. This study has provided a comprehensive knowledge map, development landscape, and future directions for ES research, and offered a practical and valuable reference for scholars to obtain better understanding of the current state of research in this field.
Collapse
Affiliation(s)
- Haiyang Wu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zaijie Sun
- Department of Orthopaedic Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Guiping Zeng
- Department of Orthopaedic Surgery, Yangxin People's Hospital, Yangxin, Hubei, China
| | - Jia Yang
- Department of Orthopaedics, Jincheng General Hospital, Jincheng, Shanxi Province, China.
| | - Changxu Deng
- Department of Sports Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Cheng Li
- Department of Spine Surgery, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- Center for Musculoskeletal Surgery (CMSC), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt University of Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
18
|
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
|
19
|
Yu C, Jiang X, Wei S, Zhou C, Zhou C, Wei Y, Su Z. Exploration of research hotspots and evolutionary trends in osteosarcoma pulmonary metastasis: A comprehensive bibliometric analysis spanning five decades. J Orthop 2025; 63:181-195. [PMID: 40291605 PMCID: PMC12018099 DOI: 10.1016/j.jor.2025.03.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Objective Pulmonary metastasis frequently complicates osteosarcoma, making the investigation of its mechanisms and therapeutic strategies a focal point in both clinical medicine and biosciences. The explosive growth of the relevant literature has still not been systematically sorted out and summarized. Methods Bibliometric methods were used in this study, Literature was retrieved from the Web of Science Core Collection, SCOPUS, and PubMed up until July 1, 2024. Bibliometric indicators were analyzed and research trends and hotspots in the field visualized using the Bibliometrix package, VOSviewer 1.6.19 and Citespace 6.3.R1 software. Results A total of 1148 publications were reviewed, revealing that over the past five decades, the cumulative number of publications on pulmonary metastasis of osteosarcoma has gradually increased. China leads in the number of published papers, while the United States exhibits the most collaborative relationships with other countries. Italy is noted for the highest quality of research. Sun Yat-sen University is the most prolific institution, and the most productive author is Eugenie S. Kleinerman from the University of Texas MD Anderson Cancer Center. "Cancer" is the journal with the most publications, and Zhou Yan's 2020 paper received the highest local citations. Keywords such as "angiogenesis," "tumor microenvironment," and "surgery" appeared frequently, suggesting these topics are current research hotspots. Conclusion "Immunotherapy", "survival", "chemotherapy", "prognosis", "biomarkers", "metastasectomy", "microRNA", "angiogenesis", "tumor microenvironment", and "surgery" define key research areas. Current hot topics include tumor-associated macrophages and the tumor microenvironment, which may hold the key to future therapeutic breakthroughs.
Collapse
Affiliation(s)
- Chaojie Yu
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xiaohua Jiang
- Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Shutian Wei
- Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Chengxing Zhou
- Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Chengyu Zhou
- Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Yanshan Wei
- Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhiping Su
- Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
20
|
Torres MB, Leung CH, Zoghbi M, Lazcano R, Ingram D, Wani K, Keung EZ, Zarzour MA, Scally CP, Hunt KK, Conley A, Bishop AJ, Guadagnolo BA, Farooqi A, Mitra D, Yoder AK, Nakazawa MS, Araujo D, Livingston A, Ratan R, Patel S, Ravi V, Lazar AJ, Roland CL, Somaiah N, Nassif Haddad EF. Dedifferentiated liposarcomas treated with immune checkpoint blockade: the MD Anderson experience. Front Immunol 2025; 16:1567736. [PMID: 40370451 PMCID: PMC12075363 DOI: 10.3389/fimmu.2025.1567736] [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] [Accepted: 04/11/2025] [Indexed: 05/16/2025] Open
Abstract
Background Dedifferentiated liposarcoma (DDLPS) is one of the most common types of soft tissue sarcoma (STS) characterized by liposarcomatous differentiation and a predilection for the retroperitoneum. Despite the growing number of histology-specific immune checkpoint blockade (ICB) trials in STS, it is still difficult to identify the radiographic objective response rate (ORR) for DDLPS in the real world setting. This study aimed to evaluate the ORR and survival of patients with DDLPS treated with ICB at a single center. Methods We conducted a retrospective study of 31 patients with pathologically confirmed DDLPS treated with ICB at MD Anderson Cancer Center between 2018 and 2023. Patient demographics, disease characteristics, treatment history, and response to ICB were analyzed. Immunohistochemical analysis was performed on tumor samples to assess immune-related markers. Results ORR by RECIST 1.1 was 3.2% (n=1/31). Among all patients (n=31), 6% achieved partial radiographic response, while 39% had stable disease, and 55% showed progressive disease. Median progression-free survival (PFS) was 3.5 (95%CI:1.9, 4.7) months, and overall survival (OS) after ICB initiation was 19.7 (95%CI: 8.8, not reached) months. Patients without prior systemic therapy demonstrated better OS (p=0.004). Immunohistochemistry revealed no relationship between pre- or post-ICB expression of CD8, CD20, CD21 and PDL-1 and response. Conclusion While the response to ICB in DDLPS remains limited, specific immune markers may influence treatment outcomes. CD20/21 post-ICB appear more important for prognosis. Further research is warranted to identify predictive factors for ICB efficacy in DDLPS.
Collapse
Affiliation(s)
- Madeline B. Torres
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Surgery, Cooper University Hospital, Cooper Medical School of Rowan University, Camden, NJ, United States
| | - Cheuk Hong Leung
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marianne Zoghbi
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rossana Lazcano
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Davis Ingram
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Khalida Wani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Emily Z. Keung
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - M. Alejandra Zarzour
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Christopher P. Scally
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kelly K. Hunt
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anthony Conley
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andrew J. Bishop
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - B. Ashleigh Guadagnolo
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ahsan Farooqi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Devarati Mitra
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alison K. Yoder
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael S. Nakazawa
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dejka Araujo
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andrew Livingston
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ravin Ratan
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shreyaskumar Patel
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vinod Ravi
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alexander J. Lazar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Christina L. Roland
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elise F. Nassif Haddad
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
21
|
Ginn MP, Denu RA, Ingram DR, Wani KM, Lazar AJ, Harrison DJ, Nakazawa MS, Conley AP, Patel S, Livingston JA. TFCP2 Fusion-Positive Rhabdomyosarcomas: A Report of 10 Cases and a Review of the Literature. Cancers (Basel) 2025; 17:1441. [PMID: 40361368 PMCID: PMC12070825 DOI: 10.3390/cancers17091441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: The fusion of the TFCP2 gene with either EWSR1 or FUS typically results in a spindle cell and/or epithelioid variant of rhabdomyosarcoma. This is an ultra-rare type of sarcoma, with most of our knowledge about these coming from case reports and small case series. Herein, we describe the clinical characteristics and treatment course of 10 patients with TFCP2 fusion sarcomas. Methods: We identified 10 patients in our hospital system with TFCP2 fusion sarcomas and 43 previously reported cases in the literature. We assessed primary tumor characteristics, treatment regimens, and survival rates among all cases. Results: We find that TFCP2 fusion sarcomas most commonly occur in young adults (median age: 33 years) and arise in craniofacial bones (7/10, 70%). Concomitant ALK alterations and ALK overexpression is nearly universal, and two of our patients were treated with ALK inhibitors; one patient had a near complete response before eventual progression, while the other patient had progressive disease after 2 months. For most, the prognosis was poor. The median overall survival in this cohort was 24.7 months (range: 5.9-29.7 months). Four patients were treated with upfront surgery, and all four developed recurrent disease. The median time to recurrence following upfront surgery was 2.1 months (range: 0.73-6.9 months). Five patients received systemic therapy, and the median progression-free survival from the start of treatment to progression was 1.6 months (range: 0.97-2.7). We also review the 53 total cases of TFCP2 fusion sarcomas in the literature, again highlighting the dismal outcomes in this disease. Conclusions:TFCP2 fusion sarcomas are proven to be aggressive and have poor prognosis. Additional work is needed to define the optimal treatment course for TFCP2 fusion sarcomas.
Collapse
Affiliation(s)
- Madison P. Ginn
- Departments of Internal Medicine & Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Ryan A. Denu
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Davis R. Ingram
- Department of Pathology, Division of Pathology & Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Khalida M. Wani
- Department of Pathology, Division of Pathology & Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alexander J. Lazar
- Department of Pathology, Division of Pathology & Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Douglas J. Harrison
- Department of Pediatrics Patient Care, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael S. Nakazawa
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anthony P. Conley
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shreyaskumar Patel
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - John Andrew Livingston
- Department of Pediatrics Patient Care, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
22
|
Aobo Z, Xiao Z, Chengfei X, Zhe X, Yingxue C, Chenhe Z, Fuan X, Fan Y, Mengmeng X, Feng Y, Wengang L. Combination of immune checkpoint inhibitors and anthracyclines as a potential first-line regimen for dedifferentiated liposarcoma: systematic review and meta-analysis. Cancer Immunol Immunother 2025; 74:179. [PMID: 40257618 PMCID: PMC12011665 DOI: 10.1007/s00262-025-04007-1] [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: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 04/22/2025]
Abstract
INTRODUCTION Dedifferentiated liposarcoma (DDLPS) is a rare and aggressive subtype of soft tissue sarcoma, characterized by limited treatment options and poor prognosis. Despite surgical resection being the only potentially curative treatment for localized DDLPS, the recurrence rate remains high, and systemic chemotherapy, typically anthracycline-based, shows limited efficacy in advanced stages. While immune checkpoint inhibitors (ICIs) have shown promise in various sarcoma subtypes, including DDLPS, their role as a first-line treatment remains unclear. METHODS We conducted a systematic meta-analysis to evaluate the efficacy of ICIs in treating patients with DDLPS. A total of 25 studies encompassing 245 patients were included. Data on overall response rate (ORR), progression-free survival, and grade III-V treatment-related adverse events were analyzed. We assessed treatment efficacy based on the line of therapy and treatment regimens, including ICI monotherapy, dual ICI therapy, and ICI combinations with other modalities. RESULTS The pooled ORR for all ICI-based treatments was 7%. First-line ICI therapy yielded a significantly higher ORR of 22%, compared to 4% in later-line treatment. The combination of ICI with anthracyclines demonstrated the highest ORR of 52%. In contrast, ICI regimens combined with trabectedin or other agents showed limited efficacy. Sensitivity analysis confirmed the stability of results, and publication bias was not detected. CONCLUSION This meta-analysis supports the potential role of ICIs, particularly in combination with anthracyclines, as a first-line therapeutic strategy for DDLPS. These results provide a foundation for future prospective studies aimed at optimizing immunotherapy approaches for this rare and challenging malignancy.
Collapse
Affiliation(s)
- Zhuang Aobo
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhou Xiao
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xu Chengfei
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China
| | - Xi Zhe
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Chen Yingxue
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhang Chenhe
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xie Fuan
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yang Fan
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiao Mengmeng
- Department of Retroperitoneal Tumor Surgery, Peking University People's Hospital, Beijing, China.
| | - Ye Feng
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Li Wengang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
23
|
De Lauretis F, Sanchez AM, Accetta C, Carnassale B, D’Archi S, Di Leone A, Franco A, Gagliardi F, Magno S, Mason EJ, Moschella F, Scardina L, Silenzi M, Bucaro A, Pirrottina CV, D’Alessandris N, Mulè A, Santoro A, Marazzi F, Masiello V, Fabi A, Orlandi A, Palazzo A, Paris I, Foschini MP, Masetti R, Franceschini G. Malignant Mesenchymal Tumors of the Breast: Current Challenges and New Perspectives on Primary Sarcomas and Malignant Phyllodes Tumors. Life (Basel) 2025; 15:673. [PMID: 40283227 PMCID: PMC12028549 DOI: 10.3390/life15040673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Mesenchymal tumors of the breast constitute a rare and heterogeneous group of neoplasms, representing only 0.5% to 1% of all breast tumors. Originating from mesenchymal tissues, these tumors include various histological subtypes. They are particularly aggressive, characterized by a high propensity for local recurrence and an overall poor prognosis. The rarity of these cases has impeded the development of comprehensive clinical studies, leading to a lack of standardized diagnostic protocols and treatment guidelines. This review provides a thorough synthesis of current knowledge on breast mesenchymal tumors with a specific focus on malignant variants such as phyllodes tumors and breast sarcomas. It also addresses the diagnostic challenges faced by clinicians, evaluates current therapeutic strategies, and emphasizes the crucial role of surgical treatment. Additionally, it examines the evolving roles of chemotherapy and radiotherapy in enhancing patient outcomes.
Collapse
Affiliation(s)
- Flavia De Lauretis
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Alejandro Martin Sanchez
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Cristina Accetta
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Beatrice Carnassale
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Sabatino D’Archi
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Alba Di Leone
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Franco
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Federica Gagliardi
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Stefano Magno
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Elena Jane Mason
- Breast Surgery, Center for Women’s and Newborn Health, Isola Tiberina Hospital, Gemelli Isola, 00153 Rome, Italy
| | - Francesca Moschella
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Lorenzo Scardina
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Marta Silenzi
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Angela Bucaro
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Chiara V. Pirrottina
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Nicoletta D’Alessandris
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Antonino Mulè
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Angela Santoro
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Fabio Marazzi
- Division of Radiotherapy, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Valeria Masiello
- Division of Radiotherapy, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Alessandra Fabi
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Armando Orlandi
- Division of Medical Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Antonella Palazzo
- Division of Medical Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Ida Paris
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Maria Pia Foschini
- Breast Unit, Bellaria Hospital, AUSL Bologna, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy
| | - Riccardo Masetti
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Gianluca Franceschini
- Multidisciplinary Breast Center, Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
24
|
FENG LIWEN, CHEN YUTING, JIN WENYI. Research progress on cancer-associated fibroblasts in osteosarcoma. Oncol Res 2025; 33:1091-1103. [PMID: 40296919 PMCID: PMC12033999 DOI: 10.32604/or.2024.054207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/26/2024] [Indexed: 04/30/2025] Open
Abstract
Osteosarcoma (OS) is a prevalent primary bone malignancy with limited treatment options. Therefore, it is imperative to investigate and understand the mechanisms underlying OS pathogenesis. Cancer-associated fibroblasts (CAFs) are markedly abundant in tumor stromal cells and are essentially involved in the modulation of tumor occurrence and development. In recent years, CAFs have become a hotspot as researchers aim to elucidate CAF mechanisms that regulate tumor progression. However, most studies on CAFs are limited to a few common cancers, and their association with OS remains elusive. This review describes the role and current knowledge of CAFs in OS, focusing on their potential cellular origin, classification, and diverse functionality. It was found that CAFs influenced OS tumor cell signaling, proliferation, invasion, metastasis, epithelial-mesenchymal transition, stemness maintenance, angiogenesis, and the ability to modify immune system components. Furthermore, findings on other common cancers indicated that effective therapeutic strategies included the manipulation of CAF activation, targeting CAF-derived components, and depletion of CAFs by biomarkers. This review provides new insights and a theoretical basis for OS research.
Collapse
Affiliation(s)
- LIWEN FENG
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - YUTING CHEN
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - WENYI JIN
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430022, China
| |
Collapse
|
25
|
Fradin JJ, Charlson JA. Review of Adoptive Cellular Therapies for the Treatment of Sarcoma. Cancers (Basel) 2025; 17:1302. [PMID: 40282478 PMCID: PMC12026197 DOI: 10.3390/cancers17081302] [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: 02/21/2025] [Revised: 04/02/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Sarcomas are a heterogeneous group of malignancies with limited therapeutic options, particularly in the metastatic setting. Adoptive cellular therapies (ACTs), including tumor-infiltrating lymphocyte (TIL) therapy, chimeric antigen receptor (CAR) T-cell therapy, and T-cell receptor (TCR) gene-modified T-cell therapy, offer promising novel approaches for these refractory tumors. TIL-based therapy has demonstrated early efficacy in melanoma and myeloma, with ongoing trials exploring its role in sarcoma. CAR T-cell strategies targeting HER2, GD2, and B7-H3 antigens are in development, though challenges such as tumor microenvironment-mediated resistance and antigen escape remain significant. Engineered TCRs, particularly those targeting MAGE-A4 and NY-ESO-1, have shown promising clinical results in synovial sarcoma (SS) and myxoid/round cell liposarcoma (MRCLS), leading to the recent FDA approval of afamitresgene autoleucel (afami-cel) and letetresgene autoleucel (lete-cel). Despite encouraging preliminary data, ACT implementation faces barriers including limited antigen specificity, off-tumor toxicity, immune evasion, and manufacturing scalability. Future research will focus on optimizing lymphodepleting regimens, mitigating toxicity, enhancing in vivo persistence, and combining ACT with other therapeutic agents. As clinical trials expand, ACT holds the potential to revolutionize sarcoma treatment by offering durable, targeted therapies for previously refractory disease.
Collapse
Affiliation(s)
- James J. Fradin
- Division of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John A. Charlson
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| |
Collapse
|
26
|
Saoud C, Gundem G, Vanderbilt CM, Wexler LH, Reed DR, Tap W, Singer S, Villafania LB, Papaemmanouil E, Benhamida J, Bale TA, Antonescu CR. Undifferentiated Pleomorphic Sarcoma in Children and Young Adults: A Comprehensive Clinicopathologic, Genomic, and Epigenetic Comparison With Adult Counterparts. Mod Pathol 2025; 38:100769. [PMID: 40222653 DOI: 10.1016/j.modpat.2025.100769] [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: 02/25/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
Undifferentiated pleomorphic sarcoma (UPS) occurs primarily in older adults and remains a diagnosis of exclusion due to its lack of differentiation and specific molecular alterations. Its occurrence in children is rare and controversial, with an unclear relationship to its adult counterpart. In this study, we aimed to investigate a cohort of 6 pediatric undifferentiated pleomorphic sarcoma (P-UPS, mean 10 years old) and 19 young-adult undifferentiated pleomorphic sarcoma (YA-UPS, mean 30 years old) cases by conducting a comprehensive comparative analysis of their clinicopathologic, genomic, and epigenetic features relative to their adult undifferentiated pleomorphic sarcoma counterparts (A-UPS, n = 100). Histologically, P-UPS and YA-UPS exhibited broad morphologic spectrum. The most frequent alterations across all groups were TP53, CDKN2A/B, and ATRX, with no significant differences among subsets. Notably, RB1 alterations were absent in P-UPS, although representing the second most common alteration in YA-UPS (32%) and A-UPS (41%). PTEN alterations were significantly more prevalent in YA-UPS (26%) compared with that in P-UPS (0%) and A-UPS (6%). Deletions in chromosomes 10, 16q, and 13q, along with amplification of 20q, were the most common across all groups. Except for a higher frequency of 17q amplification in P-UPS (33%) and YA-UPS (26%) compared with that in A-UPS (6%), no other arm-level differences were observed. P-UPS showed a lower mean fraction genome altered compared with YA-UPS and A-UPS, whereas all UPS age groups showed a low tumor mutational burden (mean <10 mut/MB). Pathogenic germline variants of high clinical significance (TP53, NF1, MLH1, CHEK2, and BARD1) were observed only in YA-UPS (31%) and A-UPS (12%) cases. By T-distributed stochastic neighborhood embedding and hierarchical clustering of DNA methylation, the majority of P-UPS and a small subset of YA-UPS grouped in a distinct cluster, characterized by a lower genomic index compared to A-UPS. In contrast, most UPS occurring in young adults genomically parallel their older adults' counterparts. P-UPS and YA-UPS cases exhibited a better disease-specific and progression-free survival, compared with A-UPS cases.
Collapse
Affiliation(s)
- Carla Saoud
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gunes Gundem
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York; Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chad M Vanderbilt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Leonard H Wexler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Damon R Reed
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William Tap
- Department of Medicine, Sarcoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Samuel Singer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Liliana B Villafania
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elli Papaemmanouil
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York; Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jamal Benhamida
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tejus A Bale
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cristina R Antonescu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
27
|
Wilky BA, Schwartz GK, Gordon MS, El-Khoueiry AB, Bullock AJ, Henick B, Agulnik M, Singh A, Mahadevan D, Stebbing J, Delepine C, Chand D, Avagyan M, Wu W, Johnson B, Grossman JE, O'Day S, Trent JC, Jones RL, Tsimberidou AM. Botensilimab (Fc-enhanced anti-cytotoxic lymphocyte-association protein-4 antibody) Plus Balstilimab (anti-PD-1 antibody) in Patients With Relapsed/Refractory Metastatic Sarcomas. J Clin Oncol 2025; 43:1358-1368. [PMID: 39869830 PMCID: PMC11974637 DOI: 10.1200/jco-24-02524] [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: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/19/2024] [Indexed: 01/29/2025] Open
Abstract
PURPOSE Outcomes for patients with advanced sarcomas are poor and there is a high unmet need to develop novel therapies. The purpose of this phase I study was to define the safety and efficacy of botensilimab (BOT), an Fc-enhanced anti-cytotoxic lymphocyte-association protein-4 antibody, plus balstilimab (BAL), an anti-PD-1 antibody, in advanced sarcomas. METHODS BOT was administered intravenously (IV) at 1 mg/kg or 2 mg/kg once every 6 weeks in combination with BAL IV at 3 mg/kg once every 2 weeks for up to 2 years. The primary end point was to determine dose-limiting toxicities during the dose-escalation period. Secondary end points include objective response rate (ORR), duration of response (DOR), disease control rate, and progression-free survival (PFS) by RECIST 1.1. Exploratory end points include assessing patient biomarkers including tumor mutational burden, cytokines, and PD-L1 expression. RESULTS Overall, 64 patients with sarcoma were treated; all were evaluable for safety and 52 for efficacy. The most common treatment-related adverse event (TRAE) was diarrhea/colitis occurring in 35.9% of patients, with grade 3 in 6.3% of patients. No grade 4 or 5 TRAEs were reported. For all evaluable patients, ORR was 19.2% (95% CI, 9.6 to 32.5), and 27.8% (95% CI, 9.7 to 53.5) for evaluable patients with angiosarcoma (n = 18); 33.3% in visceral and 22.2% in cutaneous subtypes. Median PFS for evaluable patients was 4.4 months (95% CI, 2.8 to 6.1), with a 6-month PFS rate of 36% (95% CI, 22 to 50) and a median DOR of 21.7 months (95% CI, 1.9 to not reached). CONCLUSION The combination of BOT/BAL demonstrated promising efficacy and safety in a large cohort of heavily pretreated sarcoma patients. This encouraging activity warrants further investigation (ClinicalTrials.gov identifier: NCT03860272).
Collapse
Affiliation(s)
| | | | | | | | | | - Brian Henick
- Herbert Irving Comprehensive Cancer Center at Columbia University School of Medicine, New York, NY
| | - Mark Agulnik
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA
| | - Arun Singh
- University of California Los Angeles, Los Angeles, CA
| | - Daruka Mahadevan
- The University of Texas Health Sciences Center at San Antonio, San Antonio, TX
| | - Justin Stebbing
- Anglia Ruskin University, School of Life Sciences, Cambridge, United Kingdom
| | | | | | | | - Wei Wu
- Agenus Inc, Lexington, MA
| | | | | | | | - Jonathan C. Trent
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Robin L. Jones
- The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | | |
Collapse
|
28
|
Kloos J, Ehrlich M, Taback B, Matushansky I. UV-induced multifocal cutaneous high-grade leiomyosarcoma. BMJ Case Rep 2025; 18:e264326. [PMID: 40216414 DOI: 10.1136/bcr-2024-264326] [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: 04/16/2025] Open
Abstract
Cutaneous sarcomas almost never present as multifocal disease. We describe a case of a man in his mid-90s with history of prior squamous cell carcinomas and an atypical fibroxanthoma involving the scalp who presented with multiple focal cutaneous nodules localised to the scalp, with some >3 cm. Biopsy with histopathology confirmed high-grade desmin-positive mesenchymal malignancy, with 99% of cells expressing PD-1, consistent with leiomyosarcoma (LMS). Gene expression profiling indicated a UV gene signature. The patient was treated with radiation therapy to the affected area with significant response but then developed additional cutaneous nodules inferior to the radiation field. He was subsequently treated with pembrolizumab with almost immediate resolution of the remaining nodules. This case is unique as it highlights several unusual features of cutaneous high-grade LMSs, notably presentation of multifocal disease, a UV signature and high responsiveness to anti-PD-1 therapy.
Collapse
Affiliation(s)
- Jacqueline Kloos
- New York-Presbyterian/Columbia University Irving Medical Center, New York, NY, USA
| | - Matthew Ehrlich
- New York-Presbyterian/Columbia University Irving Medical Center, New York, NY, USA
| | - Bret Taback
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Igor Matushansky
- Medical Oncology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
29
|
Li D, Li Y, Cang J, Yan X, Wu F, Sun X, Zhang W. Synergistic chemo-immunotherapy for osteosarcoma via a pH-responsive multi-component nanoparticle system. Front Pharmacol 2025; 16:1584245. [PMID: 40264674 PMCID: PMC12011790 DOI: 10.3389/fphar.2025.1584245] [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: 02/27/2025] [Accepted: 03/26/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction Osteosarcoma (OS) is the most common primary malignant bone tumor in pediatric populations. Its treatment is complicated by chemotherapy-induced toxicity and limited induction of immunogenic cell death (ICD). Methods To address these challenges, we developed a pH-responsive, multi-component nanoparticle system designed to co-deliver doxorubicin (DOX), monophosphoryl lipid A (MPLA), and a PD-1/PD-L1-targeting peptide, integrated with the immune-modulating polymer PEG-PC7A. The system was optimized using both one-factor-at-a-time (OFAT) and Box-Behnken design (BBD). Results The optimized nanoparticles had a hydrodynamic size of 110 nm, high encapsulation efficiency (97.15%), and pH-sensitive drug release (91% at pH 6.5). In vitro studies showed enhanced ICD markers, including calreticulin exposure and ATP/HMGB1 release, aswell as synergistic dendritic cell maturation via dual STING/TLR4 pathway activation. In an orthotopic LM8 osteosarcoma model, the nanoparticles significantly suppressed tumor growth, promoted cytotoxic T lymphocyte infiltration, reduced regulatory T cells, and established long-term immune memory. Discussion The combination of ICD induction, innate immune activation, and checkpoint blockade reprogrammed the tumor microenvironment, amplifying anti-tumor immune responses. These results demonstrate the potential of this multifunctional nanoparticle platform as an effective immunochemotherapeutic strategy for osteosarcoma, offering enhanced therapeutic efficacy and reduced systemic toxicity.
Collapse
Affiliation(s)
- Dapeng Li
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yuanfan Li
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jie Cang
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xianwen Yan
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Feipeng Wu
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xuan Sun
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wenchao Zhang
- Department of Orthopedics, Affiliated Jintan Hospital of Jiangsu University, Jintan, Jiangsu, China
| |
Collapse
|
30
|
Zhang Q, Wang Z, Zeng X, Ding Y, Wang C. Evaluation of tumorous LCP1 and ADPGK as predictive biomarker for immune-related adverse events in bone and soft tissue sarcomas treated with anti-PD-1 and anti-PD-L1 antibodies. BMC Cancer 2025; 25:619. [PMID: 40189514 PMCID: PMC11974013 DOI: 10.1186/s12885-025-14007-0] [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: 09/25/2024] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Immune checkpoint inhibitor (ICI) have been utilized in bone and soft tissues sarcoma patients under multiple circumstances in combination with surgeries and chemotherapy. Regretfully, immune-related adverse events (irAE) increases as the usage of ICI increases. Since a large portion of bone and soft tissues sarcoma patients gain long survival times after successful removal of the tumors which makes clinicians to avoid regimens that causes adverse events, especially lifetime irAE. Hence, predicting the development of irAE are of special significance for utilizing ICI in bone and soft tissues sarcoma patients. We have retrospectively stained tumorous LCP1 and ADPGK, two biomarkers previously reported to predict ICI induced irAE, with surgical removed, formalin-fixed and parrffin-embedded samples in a cohort of 50 bone and soft tissues sarcoma patients. We observed that the most common irAE in bone and soft tissues sarcoma patients received ICI is hyperglycemia and high grade irAE happens predominately in patients over 30 years old. Immunochemistry revealed that both LCP1 and ADPGK were elevated in tumorous tissues of patients developed irAE and bivariate-model of LCP1 and ADPGK severs as a better biomarker in comparison to LCP1 or ADPGK alone in the entire cohort. In osteosarcoma, LCP1 alone exhibited an outstanding predication value with an AUC of 0.9244 (P value of 0.0013 and a 95% CI of 0.8178 to 1.000). LCP1 and ADPGK bivariate-model serves as a promising biomarker for predicting ICI induced irAE in bone and soft tissues sarcoma patients while LCP1 alone works better in bone malignancy especially in osteosarcoma.
Collapse
Affiliation(s)
- Qing Zhang
- Beijing Jishuitan Hospital, Capital Medical University, Beijing, China.
| | - Zhe Wang
- Beijing Chaoyang Integrative Medicine Rescue and First AID Hospital, Beijing, China
| | - Xueqin Zeng
- Pathology Department, Renji Hospital of Shanghai Jiaotong University, Shanghai, 201210, China
| | - Yi Ding
- Beijing Jishuitan Hospital, Capital Medical University, Beijing, China.
| | - Chen Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
31
|
Kastner L, Kandalaft W, Mahant Mahant A, Crimella J, Hakim S, Peng X, Isakoff MS, Hayashi M, Loeb DM. Cytokine Profiling of Children, Adolescents, and Young Adults Newly Diagnosed with Sarcomas Demonstrates a Role for IL-1β in Osteosarcoma Metastasis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.05.25325205. [PMID: 40297413 PMCID: PMC12036375 DOI: 10.1101/2025.04.05.25325205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Background Sarcomas are a heterogeneous group of mesenchymal tumors frequently diagnosed in pediatric and young adult patients. These tumors respond poorly to conventional immunotherapy, though the precise reason for this is not known. We sought to characterize the systemic immune response to sarcomas by measuring the levels of circulating cytokines in the plasma of sarcoma patients, testing the hypothesis that the natures of a patient's immune response to their tumor directly affects outcome. Methods Plasma was collected from newly diagnosed, treatment-naive pediatric sarcoma patients participating in an ongoing clinical trial, MCC20320. A panel of 18 cytokines was selected and cytokine levels were measured using the Luminex platform. Cytokine levels were analyzed based on clinicopathological parameters such as gender, age, stage, and survival. Results We found that the cytokine profile in patients newly diagnosed with sarcoma is distinct from healthy controls, but different sarcomas were not distinguishable. Patients with osteosarcoma who had elevated levels of multiple cytokines had inferior overall survival compared to those with fewer or no elevated levels. Similarly, elevated levels of individual cytokines and chemokines, including IL-24, CXCL5, and CXCL10, were associated with inferior event-free or overall survival in patients with osteosarcoma. Perhaps most significantly, elevated IL-1β at diagnosis was associated with metastatic presentation and inferior event-free survival in patients with osteosarcoma. Conclusion These findings suggest that pediatric sarcoma patients mount a systemic immune response that may affect event-free or overall survival. IL-1β in particular may be a valuable target for immunotherapy for osteosarcoma patients.
Collapse
Affiliation(s)
- Laurel Kastner
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - William Kandalaft
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | | | - Sydney Hakim
- Department of Pediatrics-Hematology/Oncology and Bone Marrow Transplantation, University of Colorado, Aurora, Colorado 80045
| | - Xiao Peng
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | - Masanori Hayashi
- Department of Pediatrics-Hematology/Oncology and Bone Marrow Transplantation, University of Colorado, Aurora, Colorado 80045
| | - David M Loeb
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461
- Cancer Dormancy Institute, Albert Einstein College of Medicine, Bronx, NY 10461
- Marilyn and Stanley M. Katz Institute for Immunotherapy for Cancer and Inflammatory Disorders, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
32
|
Ferraro DA, Bisig B, Rotzinger DC, Pareja F, Missiaglia E, Voutsadakis I, Homicsko K, Digklia A. Case Report: Lasting complete response to pembrolizumab in mismatch repair-deficient cardiac sarcoma: a genomic characterization. Front Oncol 2025; 15:1485386. [PMID: 40248199 PMCID: PMC12003144 DOI: 10.3389/fonc.2025.1485386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Sarcomas are traditionally considered "cold" tumors with poor response to immunotherapy. However, evidence accumulating over the last years shows that immune checkpoint inhibitors (ICIs) may have a role in selected sarcoma patients according to predictive markers. Here, we report the case of a woman diagnosed with a primary cardiac undifferentiated sarcoma. Following failure of standard first line chemotherapy, high-throughput sequencing (HTS) revealed a high tumor mutational burden (TMB), pathogenic mutations in FAT1 and NOTCH2 and a microsatellite instability (MSI)-associated signature. Immunohistochemistry confirmed mismatch repair-deficiency (MMRd) and abundant CD8+ tumor-infiltrating lymphocytes (TILs), in the absence of tertiary lymphoid structures. The patient was, therefore, treated with the ICI pembrolizumab, reaching a complete response that continues to persist at last follow-up, more than seven years from initial diagnosis and nearly six years from initiation of ICI treatment. This case illustrates the importance of performing HTS in rare sarcomas given the availability of efficient therapies, such as those for tumors displaying high TMB or MMRd/MSI. In agreement with other reports, it supports the contention that MMRd/MSI status and high numbers of TILs are valuable predictive markers of response to immunotherapy in sarcomas.
Collapse
Affiliation(s)
- Daniela A. Ferraro
- Department of Medical Oncology, CHUV University Hospital, Lausanne, Switzerland
| | - Bettina Bisig
- Institute of Pathology, Department of Laboratory Medicine and Pathology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - David C. Rotzinger
- Department of Radiology, CHUV University Hospital, Lausanne, Switzerland
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Edoardo Missiaglia
- Institute of Pathology, Department of Laboratory Medicine and Pathology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Ioannis Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, ON, Canada
- Division of Clinical Sciences, Section of Internal Medicine, Northern Ontario School of Medicine, Sudbury, ON, Canada
| | - Krisztian Homicsko
- Department of Medical Oncology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Antonia Digklia
- Department of Medical Oncology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
33
|
Hashimoto K, Nishimura S, Goto K. PD‑1/PD‑L1 immune checkpoint in bone and soft tissue tumors (Review). Mol Clin Oncol 2025; 22:31. [PMID: 39989606 PMCID: PMC11843085 DOI: 10.3892/mco.2025.2826] [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: 12/08/2024] [Accepted: 01/22/2025] [Indexed: 02/25/2025] Open
Abstract
Anti-programmed cell death 1 (PD-1)/PD-1 ligand-1 (PD-L1) drugs have been used clinically, including those for skin cancer, with reasonable efficacy. Despite extensive ongoing research on bone and soft tissue sarcomas, there is a paucity of reviews that present a coherent picture. The present article is a comprehensive narrative review on the role of the PD-1/PD-L1 immune checkpoint in bone and soft tissue tumors. The review outlines the biological functions and mechanisms of action of PD-1/PD-L1 and its expression and clinical significance in various tumor types, including osteosarcoma and soft tissue sarcoma. Clinical trial results of immune checkpoint inhibitors, their association with prognosis, mechanisms of resistance to therapy, immune-related adverse events, and their potential in combination therapies, were also discussed.
Collapse
Affiliation(s)
- Kazuhiko Hashimoto
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama, Osaka 589-8511, Japan
| | - Shunji Nishimura
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama, Osaka 589-8511, Japan
| | - Koji Goto
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama, Osaka 589-8511, Japan
| |
Collapse
|
34
|
Assi T, Moussa T, Le Cesne A. Exploring the role of immune checkpoint inhibitors in solitary fibrous tumors. Immunotherapy 2025; 17:305-307. [PMID: 40162892 PMCID: PMC12045572 DOI: 10.1080/1750743x.2025.2485670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Affiliation(s)
- Tarek Assi
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France
| | - Tania Moussa
- Radiology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Axel Le Cesne
- Division of International Patients Care, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
35
|
Rieth J, Monga V, Milhem M. The Decline and Fall of the Current Chemotherapy Paradigm in Soft Tissue Sarcoma. Cancers (Basel) 2025; 17:1203. [PMID: 40227810 PMCID: PMC11987756 DOI: 10.3390/cancers17071203] [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: 02/20/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Despite conventional cytotoxic chemotherapy treatments, soft tissue sarcoma continues to remain a terminal diagnosis for most patients. Numerous chemotherapeutic agents have been trialed in soft tissue sarcoma, with marginal improvement in overall survival. Novel therapeutic approaches are needed to improve outcomes for this entity. METHODS the literature was reviewed, including a summary of pertinent adjuvant/neoadjuvant clinical trials and trials for metastatic disease. RESULTS Chemotherapeutic agent use in adjuvant/neoadjuvant trials provided limited if any evidence of the benefit of chemotherapy in this space. Despite multiple trials in the metastatic space, novel chemotherapeutic agents appear to have limited long-term benefits for the management of soft tissue sarcoma. Suggestions for further research, particularly with neoadjuvant clinical trials, were made. CONCLUSIONS Chemotherapy remains an inadequate treatment option for soft tissue sarcoma, and novel therapies are needed. The neoadjuvant space provides an excellent opportunity to study the effects of innovative treatments in soft tissue sarcoma.
Collapse
Affiliation(s)
- John Rieth
- Holden Comprehensive Cancer Center, University of Iowa Health Care, Iowa City, IA 52242, USA;
| | - Varun Monga
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA;
| | - Mohammed Milhem
- Holden Comprehensive Cancer Center, University of Iowa Health Care, Iowa City, IA 52242, USA;
| |
Collapse
|
36
|
Callan AK, Alexander JH, Montgomery NI, Lindberg AW, Scharschmidt TJ, Binitie O. Contemporary surgical management of osteosarcoma and Ewing sarcoma. Pediatr Blood Cancer 2025; 72 Suppl 2:e31374. [PMID: 39410791 DOI: 10.1002/pbc.31374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 04/08/2025]
Abstract
The incidence of malignant bone tumors has remained relatively stable over the past two decades between 8% and 9% per 1,000,000 in North America. Multidisciplinary treatment is paramount for optimal care combining surgical resection, chemotherapy, and rehabilitation. Surgical treatment aims for a negative margin resection of the sarcoma with a personalized reconstruction plan. Limb salvage surgery (LSS) is possible in the majority of cases; however, amputation (including rotationplasty) may be required or preferred. Reconstruction can be achieved utilizing endoprostheses, allograft, autograft, or a combination of these techniques. Emerging technologies such as 3D printing of implants and cutting guides, and intraoperative navigation have helped to improve options for LSS.
Collapse
Affiliation(s)
- Alexandra K Callan
- Department of Orthopaedics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John H Alexander
- Department of Orthopaedics, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | - Odion Binitie
- Department of Sarcoma, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
37
|
Takagi S, Nakajima M, Koike S, Takami M, Sugiura Y, Sakata S, Baba S, Takemoto A, Huang T, Seto Y, Saito M, Funauchi Y, Ae K, Takeuchi K, Fujita N, Katayama R. Frequent copy number gain of MCL1 is a therapeutic target for osteosarcoma. Oncogene 2025; 44:794-804. [PMID: 39663392 PMCID: PMC11913727 DOI: 10.1038/s41388-024-03251-6] [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: 11/29/2023] [Revised: 11/26/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Osteosarcoma (OS) is a primary malignant bone tumor primarily affecting children and adolescents. The lack of progress in drug development for OS is partly due to unidentified actionable oncogenic drivers common to OS. In this study, we demonstrate that copy number gains of MCL1 frequently occur in OS, leading to vulnerability to therapies based on Mcl-1 inhibitors. Fluorescence in situ hybridization analysis of 41 specimens revealed MCL1 amplification in 46.3% of patients with OS. Genetic inhibition of MCL1 induced significant apoptosis in MCL1-amplified OS cells, and the pharmacological efficacy of Mcl-1 inhibitors was correlated with MCL1 copy numbers. Chromosome 1q21.2-3 region, where MCL1 is located, contains multiple genes related to the IGF-1R/PI3K pathway, including PIP5K1A, TARS2, OUTD7B, and ENSA, which also showed increased copy numbers in MCL1-amplified OS cells. Furthermore, combining Mcl-1 inhibitors with IGF-1R inhibitors resulted in synergistic cell death by overcoming drug tolerance conferred by the activation of IGF signaling and suppressed tumor growth in MCL1-amplified OS xenograft models. These results suggest that genomic amplification of MCL1 in the 1q21.2-3 region, which occurred in approximately half of OS patients, may serve as a predictive biomarker for the combination therapy with an Mcl-1 inhibitor and an IGF1R inhibitor.
Collapse
Affiliation(s)
- Satoshi Takagi
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
| | - Mikako Nakajima
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, The University of Tokyo, Tokyo, Japan
| | - Sumie Koike
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
| | - Miho Takami
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
| | - Yoshiya Sugiura
- Division of Pathology, Cancer Institute, JFCR, Tokyo, Japan
- Department of Pathology, Toho University Medical Center, Sakura Hospital, Sakura, Japan
| | - Seiji Sakata
- Division of Pathology, Cancer Institute, JFCR, Tokyo, Japan
- Department of Pathology, Cancer Institute Hospital, JFCR, Tokyo, Japan
- Pathology Project for Molecular Targets, Cancer Institute, JFCR, Tokyo, Japan
| | - Satoko Baba
- Division of Pathology, Cancer Institute, JFCR, Tokyo, Japan
- Department of Pathology, Cancer Institute Hospital, JFCR, Tokyo, Japan
- Pathology Project for Molecular Targets, Cancer Institute, JFCR, Tokyo, Japan
| | - Ai Takemoto
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
| | - Tianyi Huang
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, The University of Tokyo, Tokyo, Japan
| | - Yosuke Seto
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
| | - Masanori Saito
- Department of Orthopedic Oncology, Cancer Institute Hospital, JFCR, Tokyo, Japan
| | - Yuki Funauchi
- Department of Orthopedic Oncology, Cancer Institute Hospital, JFCR, Tokyo, Japan
- Department of Orthopedic Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Keisuke Ae
- Department of Orthopedic Oncology, Cancer Institute Hospital, JFCR, Tokyo, Japan
| | - Kengo Takeuchi
- Division of Pathology, Cancer Institute, JFCR, Tokyo, Japan
- Department of Pathology, Cancer Institute Hospital, JFCR, Tokyo, Japan
- Pathology Project for Molecular Targets, Cancer Institute, JFCR, Tokyo, Japan
| | | | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan.
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
38
|
Chen TWW. Angiosarcoma: Role of Immunotherapy. Curr Treat Options Oncol 2025; 26:242-250. [PMID: 40056281 DOI: 10.1007/s11864-025-01307-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
OPINION STATEMENT Recent clinical trials have investigated immune checkpoint inhibitors (ICIs) alone, in combination with tyrosine kinase inhibitors (TKIs), or with chemotherapy in angiosarcoma, yielding mixed results across subtypes. Single-agent ICI therapy, such as cemiplimab (ORR 27.8%), has shown responses primarily in UV- and radiation-associated angiosarcomas, likely due to their higher tumor mutation burden (TMB), while dual ICI therapy (SWOG S1609, ORR 25%) suggests potential benefit but remains limited in cutaneous disease. ICI-TKI combinations, such as cabozantinib plus nivolumab (Alliance A091902, second-line, ORR 59%), demonstrated significant efficacy in both cutaneous and non-cutaneous angiosarcomas, suggesting anti-angiogenic therapy may enhance ICI responses in tumors historically resistant to checkpoint blockade. ICI-chemotherapy combinations have been less consistent. The Alliance A091902 first-line trial (paclitaxel ± nivolumab) found no overall PFS benefit, though scalp/face angiosarcoma patients appeared to fare better, raising the question of whether ICI alone might be equally effective in this subset. The South Korean paclitaxel + avelumab trial (ORR 50%) showed promising response rates, but the lack of detailed subgroup analysis limits interpretation. Other chemotherapy-ICI combinations, such as doxorubicin plus pembrolizumab, have shown isolated responses but require further study in larger cohorts. Moving forward, better biomarkers are critical for identifying which patients benefit most from ICIs, and while TKI-ICI combinations appear to hold the most promise, chemotherapy-ICI strategies need further refinement to optimize sequencing and patient selection in angiosarcoma treatment.
Collapse
Affiliation(s)
- Tom Wei-Wu Chen
- Department of Oncology and Center of Excellence for Sarcoma, National Taiwan University Hospital, 7 Chung Shan South Rd, Taipei, Taiwan, 100.
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan.
| |
Collapse
|
39
|
Griffin KH, Sagheb IS, Coonan TP, Fierro FA, Randall RL, Leach JK. Macrophage and osteosarcoma cell crosstalk is dependent on oxygen tension and 3D culture. BIOMATERIALS ADVANCES 2025; 169:214154. [PMID: 39708660 DOI: 10.1016/j.bioadv.2024.214154] [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: 06/26/2024] [Revised: 11/29/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Osteosarcoma (OS), the most common form of primary bone cancer in young adults, has had no improvements in clinical outcomes in 50 years. This highlights a critical need to advance mechanistic understanding of OS to further therapeutic discovery, which will only be possible with accurate models of the disease. Compared to traditional monolayer studies and preclinical models, in vitro models that better replicate the three-dimensional (3D) bone marrow microenvironment will facilitate methodical investigations of the events and factors that drive OS progression. Herein, we use fibrin-alginate interpenetrating network (FA IPN) hydrogels to model the hematological bone marrow environment. We interrogated the effects of oxygen tension, 3D culture, and macrophage phenotype on OS behavior and specifically examine the immunomodulatory crosstalk between OS and macrophages. We observe that OS is more sensitive to oxygen tension when cultured in 3D. Specifically, both highly and less metastatic OS exhibit decreased changes in DNA content over time in 3D, but then demonstrate diverging behaviors in heterotypic culture with macrophages. OS response to macrophages differs as a function of metastatic potential, where highly metastatic OS shows increased immunosuppression that varies with oxygen tension but relies on direct coculture conditions. To our knowledge, this is among the first work to report the effects of 3D culture on the interplay between OS and macrophages in a coculture microenvironment. Together, these data introduce FA IPNs as a promising platform for cancer research and emphasize the importance of novel models for the mechanistic study of OS.
Collapse
Affiliation(s)
- Katherine H Griffin
- School of Veterinary Medicine, University of California, Davis, CA, USA; Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, USA
| | - Isabel S Sagheb
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Thomas P Coonan
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Fernando A Fierro
- Department of Cell Biology and Human Anatomy, UC Davis Health, Sacramento, CA, USA
| | - R Lor Randall
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, USA; Department of Biomedical Engineering, University of California, Davis, CA, USA.
| |
Collapse
|
40
|
Sunbul FS, Almuqbil RM, Zhang H, Alhudaithi SS, Fernandez ME, Aldaqqa RR, Garcia VA, Robila V, Halquist MS, Gordon SW, Bos PD, da Rocha SRP. An improved experimental model of osteosarcoma lung metastases to investigate innovative therapeutic interventions and sex as a biological variable. Int J Pharm 2025; 673:125372. [PMID: 39971171 DOI: 10.1016/j.ijpharm.2025.125372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy, with OS lung metastasis (OSLM) being the leading cause of death in OS patients. No curative pharmacotherapies for OSLM are available, highlighting the clinical need for new therapies. Improved and rigorous preclinical models of OSLM are key in supporting advancements in this field. We aimed to develop an immunocompetent mouse model of OSLM that allows monitoring pharmacotherapies' effect on the lung metastatic burden over time and assessing the impact of sex as a biological variable in tumor growth and response to therapy. We transformed K7M2 cells to express bioluminescence and fluorescence, enabling real-time tracking of OSLM in BALB/c mice following tail vein injection. Metastasis was confined to the lungs and exhibited exponential growth with typical downregulated Fas receptor expression. In vivo bioluminescence correlated strongly with ex vivo, suggesting its reliability for evaluating metastatic progression and therapy response. Fluorescence from tdT was stable upon tissue processing, providing unique opportunities to probe the tumor characteristics ex vivo. We also assessed the effect of local lung-delivered gemcitabine, which was well-tolerated and significantly reduced OSLM burden without causing pulmonary toxicity. However, treatment did not resolve metastatic disease. We also explored the effect of sex on tumor growth and response to therapy; while no difference was observed in tumor growth between male and female mice, females showed a better response to local gemcitabine administration. In sum, we established a robust and rigorous immunocompetent mouse model of OSLM that will facilitate exploring new pharmacotherapies for OSLM.
Collapse
Affiliation(s)
- Fatemah S Sunbul
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Rashed M Almuqbil
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Hanming Zhang
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Sulaiman S Alhudaithi
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Matthew E Fernandez
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Raneem R Aldaqqa
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Victoria A Garcia
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Valentina Robila
- Department of Pathology - School of Medicine, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Matthew S Halquist
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Sarah W Gordon
- Department of Medical Oncology, Sidney Kimmel Comprehensive Cancer Center, Thomas JeffersonUniversity, Philadelphia, PA, the United States of America
| | - Paula D Bos
- Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Department of Pathology - School of Medicine, Virginia Commonwealth University, Richmond, VA, the United States of America; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Sandro R P da Rocha
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, the United States of America.
| |
Collapse
|
41
|
Liu Z, Weitao Y, Cui K, Gao S, Wang X, Zhang P, Wang J. The outcomes and treatment strategies in metastatic soft tissue sarcoma treated with immunotherapy-based therapy: a three-center study. Front Immunol 2025; 16:1504117. [PMID: 40226622 PMCID: PMC11985850 DOI: 10.3389/fimmu.2025.1504117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Background Preclinical studies showed that cytotoxic agents and antiangiogenic agents had regulatory effects in the tumor immune microenvironment of soft tissue sarcoma (STS), and then enhance the antitumor effect of immunotherapy. This study was to investigate the efficacy and safety of immunotherapy-based therapy in metastatic STS. Methods We conducted a retrospective analysis in three centers where some patients received immunotherapy-based therapy consisting of immunotherapy alone or in combination with systemic agents (cytotoxic agents and/or antiangiogenic agents). The primary endpoints were median progression-free survival (mPFS) and median overall survival (mOS), and Kaplan-Meier method was used to compare survival. Results A total of 79 patients were included in this study. With the median follow-up of 14.2 months, the mPFS and mOS was 7.5 months and 19.5 months, respectively. The PFS (P < 0.01) and OS (P < 0.01) were significantly better in the alveolar soft part sarcoma (ASPS) group compared to the non-ASPS group. Patients who treated in ≤2 lines had longer PFS (P < 0.01) and OS (P < 0.01) compared to those in subsequent lines. Further analysis was performed according to histopathological types, in patients with ASPS, the combination of immunotherapy-based therapy resulted in a longer PFS (P < 0.01) compared to immunotherapy in monotherapy. Similarly, the patients treated in ≤2 lines had longer PFS (P=0.03) and OS (P < 0.01) compared to in subsequent lines. In patients with non-ASPS, patients with potentially sensitive sarcomas (undifferentiated pleomorphic sarcoma, dedifferentiated liposarcoma, myxofibrosarcoma, and angiosarcoma) had a longer PFS (P = 0.02) and OS (P = 0.03) compared to other subtypes. The OS (P = 0.03) for patients with potentially sensitive sarcomas treated in ≤2 lines showed a long trend compared to subsequent lines. Most adverse events reported were mild and tolerable. Conclusions The immunotherapy-based therapy showed promising activity in survival, especially in certain histological subtypes (undifferentiated pleomorphic sarcoma, dedifferentiated liposarcoma, myxofibrosarcoma, and angiosarcoma), as well as in combination treatment and in early lines. Prospective researches are needed to confirm the potential benefits.
Collapse
Affiliation(s)
- Zhiyong Liu
- Department of Bone and Soft Tissue Tumor, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Yao Weitao
- Department of Bone and Soft Tissue Tumor, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Kang Cui
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Songtao Gao
- Department of Orthopedic, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xin Wang
- Department of Bone and Soft Tissue Tumor, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Peng Zhang
- Department of Bone and Soft Tissue Tumor, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jiaqiang Wang
- Department of Bone and Soft Tissue Tumor, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
42
|
Babatunde OO, Coca Membribes S, Anthonescu C, Bradic M, O'Malley B, Linkov I, Bartlett E, Momtaz P, Alektiar K, Gounder MM, Rosenbaum E, Tap WD, D'Angelo SP, Kelly CM. Immunologic correlates in a CIC::DUX4 fusion-positive sarcoma responsive to dual immune checkpoint blockade. NPJ Precis Oncol 2025; 9:85. [PMID: 40128305 PMCID: PMC11933392 DOI: 10.1038/s41698-025-00878-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/13/2025] [Indexed: 03/26/2025] Open
Abstract
CIC::DUX4 sarcoma (CDS) is a rare and aggressive subtype of soft tissue sarcoma with poor prognosis and limited treatment options. Immunotherapy has not been studied in this disease. To our knowledge, response to immune checkpoint blockade (ICB) has not been previously reported. Here, we present the first case of a patient with CDS responding to dual ICB with nivolumab and relatlimab. Immunohistochemical (IHC) analysis of pre-treatment samples revealed minimal immune cell infiltration, with scarce CD3+, CD8+, and FOXP3+ T-cells and negligible expression of PD-L1 and PD-1 markers. Post-treatment tumor samples revealed a significant shift in the immune microenvironment, with increased CD8 + T-cell infiltration and co-expression of exhaustion markers PD-1 and LAG-3 following treatment. These findings suggest that doublet ICB can activate an antitumor immune response in CDS, overcoming the immune cold phenotype typically associated with this sarcoma. This case provides the first evidence of dual PD-1/LAG-3 blockade inducing an immune response in CDS. The favorable response and tolerability observed in this patient highlight the potential of dual ICB as a therapeutic option in CDS that merits further investigation.
Collapse
Affiliation(s)
- Olayode O Babatunde
- Medical Oncology/Hematology Fellowship Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | | | - Cristina Anthonescu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martina Bradic
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bernard O'Malley
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irina Linkov
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Edmund Bartlett
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Parisa Momtaz
- Melanoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kaled Alektiar
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mrinal M Gounder
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Evan Rosenbaum
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - William D Tap
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Sandra P D'Angelo
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Ciara M Kelly
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
43
|
Zhang H, Zhang J, Zhu K, Li S, Liu J, Guan B, Zhang H, Chen C, Liu Y. Identification and characterization of mitochondrial autophagy-related genes in osteosarcoma and predicting clinical prognosis. Sci Rep 2025; 15:10158. [PMID: 40128298 PMCID: PMC11933398 DOI: 10.1038/s41598-025-95173-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/19/2025] [Indexed: 03/26/2025] Open
Abstract
Osteosarcoma (OS), the most prevalent primary malignant bone tumor, is characterized by a poor prognosis and high metastatic potential. Mitochondrial autophagy has been implicated in cancer suppression. This study aimed to identify prognostic genes associated with mitochondrial autophagy in OS. Public datasets, including TARGET-OS, GSE99671, and GSE21257, were retrieved for analysis. Differentially expressed genes (DEGs1) between OS and normal samples were identified from GSE99671. Single-sample Gene Set Enrichment Analysis (ssGSEA) was applied to quantify the enrichment scores of 29 mitochondrial autophagy-related genes (MARGs) in OS samples from TARGET-OS, categorizing them into high- and low-score groups to extract DEGs2. The intersection of DEGs1 and DEGs2 yielded mitochondrial autophagy-associated differentially expressed genes (MDGs). Prognostic genes were subsequently screened through a multi-step regression analysis, and a risk score was computed. TARGET-OS samples were stratified into high- and low-risk groups based on the optimal cutoff value of the risk score. GSEA was conducted between the two risk groups. Additionally, associations between prognostic genes and the immune microenvironment were explored. A total of 31 MDGs were identified from the overlap of 3,207 DEGs1 and 622 DEGs2. Five prognostic genes-KLK2, NRXN1, HES5, OR2W3, and HS3ST4-were further selected. Kaplan-Meier survival analysis indicated significantly reduced survival in the high-risk group. GSEA revealed enrichment in ABC transporter activity and glycolysis/gluconeogenesis pathways. Immunoanalysis demonstrated significant differences in 11 immune cell populations and three immune functions between risk groups, notably myeloid-derived suppressor cells (MDSCs) and Type 1 T helper cells. HS3ST4 exhibited the strongest positive correlation with macrophages, whereas NRXN1 showed the most pronounced negative correlation with memory B cells. Expressions of HAVCR2 and PDCD1LG2 were elevated in the low-risk group. Functional analysis indicated significant differences in dysfunction patterns between risk groups. This study identified five mitochondrial autophagy-related prognostic genes and constructed a risk model, offering novel insights into OS diagnosis and therapeutic strategies.
Collapse
Affiliation(s)
- Hongliang Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Jingyu Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Kai Zhu
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Shuang Li
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Jinwei Liu
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Boya Guan
- Department of Pharmacy, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Hong Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Changbao Chen
- Department of Spinal Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Yancheng Liu
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China.
| |
Collapse
|
44
|
Mu H, Zhang Q, Zuo D, Wang J, Tao Y, Li Z, He X, Meng H, Wang H, Shen J, Sun M, Jiang Y, Zhao W, Han J, Yang M, Wang Z, Lv Y, Yang Y, Xu J, Zhang T, Yang L, Lin J, Tang F, Tang R, Hu H, Cai Z, Sun W, Hua Y. Methionine intervention induces PD-L1 expression to enhance the immune checkpoint therapy response in MTAP-deleted osteosarcoma. Cell Rep Med 2025; 6:101977. [PMID: 39983717 PMCID: PMC11970323 DOI: 10.1016/j.xcrm.2025.101977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/10/2024] [Accepted: 01/28/2025] [Indexed: 02/23/2025]
Abstract
Osteosarcoma (OS), a malignant bone tumor with limited treatment options, exhibits low sensitivity to immune checkpoint therapy (ICT). Through genomics and transcriptomics analyses, we identify a subgroup of OS with methylthioadenosine phosphorylase (MTAP) deletion, which contributes to ICT resistance, leading to a "cold" tumor microenvironment. MTAP-deleted OS relies on methionine metabolism and is sensitive to methionine intervention, achieved through either dietary restriction or inhibition of methionine adenosyltransferase 2a (MAT2A), a key enzyme in methionine metabolism. We further demonstrate that methionine intervention triggers programmed death-ligand 1 (PD-L1) transcription factor IKAROS family zinc finger 1 (IKZF1) and enhances PD-L1 expression in MTAP-deleted OS cells. Methionine intervention also activates the immune-related signaling pathways in MTAP-deleted OS cells and attracts CD8+ T cells, thereby enhancing the efficacy of ICT. Combining methionine intervention with ICT provides a significant survival benefit in MTAP-deleted OS murine models, suggesting a rationale for combination regimens in OS ICT.
Collapse
Affiliation(s)
- Haoran Mu
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Qi Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongqing Zuo
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Jinzeng Wang
- National Research Center for Translational Medicine at Shanghai, State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yining Tao
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Zhen Li
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China; Simcere Zaiming Pharmaceutical Co., Ltd., Shanghai, China
| | - Xin He
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Huanliang Meng
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Hongsheng Wang
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Jiakang Shen
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Mengxiong Sun
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Yafei Jiang
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Weisong Zhao
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Jing Han
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Mengkai Yang
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Zhuoying Wang
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Yu Lv
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Yuqin Yang
- Department of Laboratory Animal Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Xu
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Tao Zhang
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Liu Yang
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Jun Lin
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Tang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China; Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing, China
| | - Renhong Tang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China; Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing, China; Simcere Zaiming Pharmaceutical Co., Ltd., Shanghai, China
| | - Haiyan Hu
- The Drug and Device Phase I Clinical Research Ward/Demonstration Research Ward of Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengdong Cai
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China
| | - Wei Sun
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China.
| | - Yingqi Hua
- Department of Orthopedic Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Bone Tumor Institution, Shanghai, China.
| |
Collapse
|
45
|
Han Z, Chen G, Wang D. Emerging immunotherapies in osteosarcoma: from checkpoint blockade to cellular therapies. Front Immunol 2025; 16:1579822. [PMID: 40170852 PMCID: PMC11958959 DOI: 10.3389/fimmu.2025.1579822] [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: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 04/03/2025] Open
Abstract
Osteosarcoma remains a highly aggressive bone malignancy with limited therapeutic options, necessitating novel treatment strategies. Immunotherapy has emerged as a promising approach, yet its efficacy in osteosarcoma is hindered by an immunosuppressive tumor microenvironment and resistance mechanisms. This review explores recent advancements in checkpoint blockade, cellular therapies, and combination strategies aimed at enhancing immune responses. We highlight key challenges, including tumor heterogeneity, poor immune infiltration, and the need for predictive biomarkers. By integrating immunotherapy with chemotherapy, radiotherapy, and targeted therapy, emerging approaches seek to improve treatment outcomes. This review provides a comprehensive analysis of the evolving landscape of osteosarcoma immunotherapy, offering insights into future directions and potential breakthroughs. Researchers and clinicians will benefit from understanding these developments, as they pave the way for more effective and personalized therapeutic strategies in osteosarcoma.
Collapse
Affiliation(s)
- Zhiwei Han
- Department of Orthopedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guomin Chen
- Laboratory Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Dongchen Wang
- Department of Orthopedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
46
|
Levy A, Morel D, Texier M, Rodriguez-Ruiz ME, Bouarroudj L, Bouquet F, Bustillos A, Quevrin C, Clémenson C, Mondini M, Meziani L, Sun R, Zaghdoud N, Tselikas L, Assi T, Faron M, Honoré C, Ngo C, Verret B, Le Péchoux C, Le Cesne A, Ginhoux F, Massard C, Bahleda R, Deutsch E. Monocyte-lineage tumor infiltration predicts immunoradiotherapy response in advanced pretreated soft-tissue sarcoma: phase 2 trial results. Signal Transduct Target Ther 2025; 10:103. [PMID: 40097400 PMCID: PMC11914280 DOI: 10.1038/s41392-025-02173-3] [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: 09/24/2024] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Immunoradiotherapy holds promise for improving outcomes in patients with advanced solid tumors, including in soft-tissue sarcoma (STS). However, the ideal combination of treatment modalities remains to be determined, and reliable biomarkers to predict which patients will benefit are lacking. Here, we report the results of the STS cohort of the SABR-PDL1 phase II trial that evaluated the anti-PDL1 atezolizumab combined with stereotactic body radiation therapy (SBRT) delivered concurrently with the 2nd cycle to at least one tumor site. Eligible patients received atezolizumab until progression or unmanageable toxicity, with SBRT at 45 Gy in 3 fractions). The primary endpoint was one-year progression-free survival (PFS) rate with success defined as 13 patients achieving 1-year PFS. Sixty-one heavily pretreated patients with STS (median 5 prior lines; 52% men; median age 54 years; 28% leiomyosarcoma) were enrolled across two centers (France, Spain). SBRT was delivered to 55 patients (90%), with the lung being the most commonly irradiated site (50%). After a median follow-up of 45 months, the one-year PFS rate was 8.3% [95% CI: 3.6-18.1]. Median PFS and overall survival were 2.5 and 8.6 months, respectively. Best responses included partial responses (5%) and stable disease (60%). Immune profiling revealed increased immunosuppressive tumor-associated macrophages (e.g., IL4I1, HES1) and monocyte-recruiting chemokines in non-responders. Higher monocyte/lymphocyte ratios (MonoLR) in tumor and blood correlated with progression. PD-L1 status, lymphoid infiltration, and tertiary-lymphoid structures were not predictive. Although the primary endpoint was not met, this study highlights MonoLR imbalance as a potential biomarker to identify STS patients likely to benefit from immunoradiotherapy. EudraCT No. 2015-005464-42; Clinicaltrial.gov number: NCT02992912.
Collapse
Affiliation(s)
- Antonin Levy
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France.
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France.
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France.
- Sarcoma unit, Gustave Roussy, Villejuif, France.
| | - Daphné Morel
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
| | - Matthieu Texier
- Biostatistics and Epidemiology Office, Gustave Roussy, Villejuif, France
- Oncostat 1018 Inserm, University Paris-Saclay, Villejuif, France
| | | | - Lisa Bouarroudj
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
- Bioinformatic platform, Gustave Roussy, Villejuif, France
| | - Fanny Bouquet
- Product Development Medical Affairs, F Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Alberto Bustillos
- Product Development Medical Affairs, F Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Clément Quevrin
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
| | - Céline Clémenson
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
| | - Michele Mondini
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
| | - Lydia Meziani
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
| | - Roger Sun
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Nadia Zaghdoud
- Biostatistics and Epidemiology Office, Gustave Roussy, Villejuif, France
| | - Lambros Tselikas
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Department of Interventional Radiology, Gustave Roussy, Villejuif, France
| | - Tarek Assi
- Sarcoma unit, Gustave Roussy, Villejuif, France
| | - Matthieu Faron
- Sarcoma unit, Gustave Roussy, Villejuif, France
- Oncostat 1018 Inserm, University Paris-Saclay, Villejuif, France
| | | | - Carine Ngo
- Sarcoma unit, Gustave Roussy, Villejuif, France
| | | | - Cécile Le Péchoux
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- Sarcoma unit, Gustave Roussy, Villejuif, France
| | | | - Florent Ginhoux
- Gustave Roussy, Inserm U1015, Université Paris-Saclay, Villejuif, France
| | - Christophe Massard
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Drug Development Department (DITEP) Gustave Roussy-Cancer Campus, Villejuif, France
| | - Rastilav Bahleda
- Sarcoma unit, Gustave Roussy, Villejuif, France
- Drug Development Department (DITEP) Gustave Roussy-Cancer Campus, Villejuif, France
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France.
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France.
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France.
| |
Collapse
|
47
|
Jiauddin M, Reddy K, Ravi HP, Ramachandran B. Druggable upregulated proteins in EWS-FLI-driven Ewing sarcoma as emerging new therapeutic targets. Am J Transl Res 2025; 17:1580-1603. [PMID: 40225989 PMCID: PMC11982847 DOI: 10.62347/ymeu1808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/11/2025] [Indexed: 04/15/2025]
Abstract
Ewing sarcoma (ES) is a highly aggressive soft tissue tumor that primarily affects the long bones of children and young adults. It is distinguished by a characteristic chromosomal translocation between the Ewing sarcoma breakpoint region 1 (EWS) gene and the erythroblast transformation-specific (ETS) family of genes, most commonly resulting in the EWS-friend leukemia integration 1 (EWS-FLI1) fusion gene. This translocation is observed in approximately 80%-85% of ES cases. This fusion gene encodes a non-physiological chimeric fusion protein that plays a central role in tumorigenesis by interacting with numerous partner proteins. Several studies have demonstrated the tumorigenic potential of the EWS-FLI1 protein when transfected into non-cancer cell lines. However, targeting EWS-FLI1 directly remains a significant challenge, as no drug to date has been reported to bind to and inhibit its activity effectively. An alternative therapeutic strategy involves targeting key overexpressed protein complexes implicated in ES tumorigenesis, many of which may be downstream interacting partners of EWS-FLI1. This review explores emerging protein targets as potential therapeutic avenues in ES treatment.
Collapse
Affiliation(s)
- Moinuddin Jiauddin
- Department of Molecular Oncology, Cancer Institute (W.I.A) No. 38, Sardar Patel Road, Adyar, Chennai 600036, India
| | - Kirtana Reddy
- Department of Molecular Oncology, Cancer Institute (W.I.A) No. 38, Sardar Patel Road, Adyar, Chennai 600036, India
| | - Hashiya Preeya Ravi
- Department of Molecular Oncology, Cancer Institute (W.I.A) No. 38, Sardar Patel Road, Adyar, Chennai 600036, India
| | - Balaji Ramachandran
- Department of Molecular Oncology, Cancer Institute (W.I.A) No. 38, Sardar Patel Road, Adyar, Chennai 600036, India
| |
Collapse
|
48
|
Blomain ES, Soudi S, Wang Z, Somani A, Subramanian A, Nouth SCL, Oladipo E, New C, Kenney DE, Nemat-Gorgani N, Kindler T, Avedian RS, Steffner RJ, Mohler DG, Hiniker SM, Chin AL, Kalbasi A, Binkley MS, Fried M, Gaida MM, van de Rijn M, Moding EJ. Evolutionary Pressures Shape Undifferentiated Pleomorphic Sarcoma Development and Radiotherapy Response. Cancer Res 2025; 85:1162-1174. [PMID: 39808162 DOI: 10.1158/0008-5472.can-24-3281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/19/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
Radiotherapy is an integral component in the treatment of many types of cancer, with approximately half of patients with cancer receiving radiotherapy. Systemic therapy applies pressure that can select for resistant tumor subpopulations, underscoring the importance of understanding how radiation impacts tumor evolution to improve treatment outcomes. We integrated temporal genomic profiling of 120 spatially distinct tumor regions from 20 patients with undifferentiated pleomorphic sarcomas (UPS), longitudinal circulating tumor DNA analysis, and evolutionary biology computational pipelines to study UPS evolution during tumorigenesis and in response to radiotherapy. Most unirradiated UPSs displayed initial linear evolution, followed by subsequent branching evolution with distinct mutational processes during early and late development. Metrics of genetic divergence between regions provided evidence of strong selection pressures during UPS development that further increased during radiotherapy. Subclone abundance changed after radiotherapy with subclone contraction tied to alterations in calcium signaling, and inhibiting calcium transporters radiosensitized sarcoma cells. Finally, circulating tumor DNA analysis accurately measured subclone abundance and enabled noninvasive monitoring of subclonal changes. These results demonstrate that radiation exerts selective pressures on UPSs and suggest that targeting radioresistant subclonal populations could improve outcomes after radiotherapy. Significance: Radiotherapy mediates tumor evolution by leading to the expansion of resistant subclonal cancer cell populations, indicating that developing approaches to target resistant subclones will be crucial to improve radiotherapy response.
Collapse
Affiliation(s)
- Erik S Blomain
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Shaghayegh Soudi
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Ziwei Wang
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Anish Somani
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Ajay Subramanian
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Serey C L Nouth
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Eniola Oladipo
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Christin New
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Deborah E Kenney
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Neda Nemat-Gorgani
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Thomas Kindler
- Department of Internal Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- TRON, Translational Oncology at the University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Raffi S Avedian
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Robert J Steffner
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - David G Mohler
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Susan M Hiniker
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Alexander L Chin
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Anusha Kalbasi
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Michael S Binkley
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Marius Fried
- Department of Internal Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias M Gaida
- TRON, Translational Oncology at the University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Pathology, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matt van de Rijn
- Department of Pathology, Stanford University, Stanford, California
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, California
- Stanford Cancer Institute, Stanford University, Stanford, California
| |
Collapse
|
49
|
Jin Y, Jia Z, Xia X, Gordon NB, Ludwig JA, Somaiah N, Li S. Anti-CD137 agonist antibody-independent and clinically feasible preparation of tumor-infiltrating lymphocytes from soft tissue sarcoma and osteosarcoma. Front Immunol 2025; 16:1557006. [PMID: 40145091 PMCID: PMC11936977 DOI: 10.3389/fimmu.2025.1557006] [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: 01/07/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Background Tumor infiltrating lymphocytes (TILs) therapy has been proved for treatment of metastatic melanoma and is under investigation for other types of solid tumors. However, these successes are threatened by discontinued supply of GMP-grade anti-CD137 agonist, a key TIL preparation reagent. Therefore, exploring a GMP-adherent method for expanding endogenous TILs without anti-CD137 agonist is urgent. Toward this end, we aimed to establish an anti-CD137-independent and clinically feasible TIL expansion protocol to prepare TILs from under investigated sarcoma tumors. Methods We collected resected tumors from patients and cut tissues into fragments. We used IL-2 and T-cell activator CD3/CD28 without anti-CD137 agonist to expand nonselected TILs in 2-3 weeks, then rapidly expanded them over 2 weeks. Their phenotypes were characterized using flow cytometry. Their antitumor activity was validated in vitro using cytotoxic T lymphocyte assays measuring CD107a on the TILs and the viability of tumor cells and in vivo using an autologous patient-derived xenograft (PDX) tumor model. Results We successfully expanded TILs in > 90% of collected samples. TILs generated preferentially increased CD8+ T cells but suppressed CD4+ T cells. A small portion of TILs were resident memory T cells. The expanded TILs reduced autologous tumor cells by 37.5% within 24 hours. Infusion of TILs in mice bearing autologous PDX tumors strongly inhibited liposarcoma growth. FDA has approved use of this GMP-feasible protocol in our clinical trial (IND 30562). Conclusion It is feasible to generate antitumor TILs using CD3/CD28 activator to replace the unavailable anti-CD137 agonist. Our study supports the further development of TIL-based therapy.
Collapse
Affiliation(s)
- Yining Jin
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Zhiliang Jia
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xueqing Xia
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nancy B. Gordon
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joseph A. Ludwig
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shulin Li
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
50
|
Paudel A, Chattopadhyay P, Rose B, Watson A, D’Amato G, Trent J, Bialick S, Jonczak E. Systemic Treatment in Soft Tissue Sarcomas: Are We Making a Difference? Cancers (Basel) 2025; 17:889. [PMID: 40075735 PMCID: PMC11898467 DOI: 10.3390/cancers17050889] [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: 02/05/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Soft tissue sarcomas [STSs] are rare tumors of mesodermal origin that arise in diverse tissues such as muscles, fat, and nerves. There are over 100 subtypes of STS, each with distinct clinical behaviors and responses to treatment. Recent advances in treatment have moved towards histology-specific approaches, emphasizing the integration of pathological, immunohistochemical, and molecular features to guide treatment. Localized STS is primarily treated with surgery, often supplemented by neoadjuvant or adjuvant radiation and/or chemotherapy. However, about half of patients with localized disease will progress to an advanced stage, which is typically managed with systemic therapies including anthracycline-based chemotherapy such as doxorubicin or epirubicin. Despite these treatments, the survival rates for most subtypes of advanced metastatic STS remain relatively low. While anthracycline-based chemotherapy remains the mainstay of treatment, ongoing research into the biology of STSs is enhancing our understanding and approach to these complex tumors with an expansion beyond chemotherapy to include targeted therapy and immunotherapy to improve response rates and survival outcomes. This review focuses on STS other than gastrointestinal stromal tumors [GISTs], examines the current systemic treatment strategies, highlights recent advances, and explores future directions in the systemic therapy of sarcoma patients.
Collapse
Affiliation(s)
- Amrit Paudel
- Department of Medicine, Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; (A.W.); (G.D.); (J.T.); (S.B.)
| | - Priya Chattopadhyay
- Department of Internal Medicine, Jackson Health System, University of Miami, Miami, FL 33136, USA; (P.C.); (B.R.)
| | - Brandon Rose
- Department of Internal Medicine, Jackson Health System, University of Miami, Miami, FL 33136, USA; (P.C.); (B.R.)
| | - Aleksandra Watson
- Department of Medicine, Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; (A.W.); (G.D.); (J.T.); (S.B.)
| | - Gina D’Amato
- Department of Medicine, Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; (A.W.); (G.D.); (J.T.); (S.B.)
| | - Jonathan Trent
- Department of Medicine, Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; (A.W.); (G.D.); (J.T.); (S.B.)
| | - Steven Bialick
- Department of Medicine, Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; (A.W.); (G.D.); (J.T.); (S.B.)
| | - Emily Jonczak
- Department of Medicine, Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; (A.W.); (G.D.); (J.T.); (S.B.)
| |
Collapse
|