1
|
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
|
2
|
Nobel TB, Stiles BM, Chudgar NP. Pulmonary Metastasectomy in Sarcoma. Thorac Surg Clin 2025; 35:201-208. [PMID: 40246409 DOI: 10.1016/j.thorsurg.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Sarcoma has a predilection to metastasize to the lungs, with pulmonary metastases occurring in up to half of patients with this disease. Resection of metastatic disease is a commonly utilized tool in the management of these patients due to limitations in systemic therapies. Prospective data are absent and recommendations for metastasectomy in the setting of sarcoma are derived from retrospective series. We describe factors associated with survival that should be evaluated in considering surgical candidacy. Approaches to resection along with management of recurrent pulmonary disease are also discussed.
Collapse
Affiliation(s)
- Tamar B Nobel
- Division of Thoracic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Brendon M Stiles
- Division of Thoracic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Neel P Chudgar
- Division of Thoracic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
3
|
Nakamura T, Hasegawa M. Treatment strategies for advanced synovial sarcoma: from chemotherapy to TCR-engineered T-cell therapy. Int J Clin Oncol 2025; 30:878-885. [PMID: 40122967 DOI: 10.1007/s10147-025-02744-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
Synovial sarcoma (SS) is the most common soft tissue sarcoma in children and adolescents. Despite the availability of new agents such as pazopanib and trabectedin, the prognosis after recurrence remains poor. Adoptive cell therapy is an emerging therapeutic strategy based on the modulation, manipulation, and selection of autologous T-cells in vitro to overcome immune system tolerance to tumor cells. Cancer-testis antigens are particularly attractive targets for immune therapy because male germ cells lack human leukocyte antigen class I molecules, limiting T-cell responses triggered by antigen presentation. T-cell receptor (TCR) engineered T-cell therapy targeting NY-ESO-1 and MAGE-A4 holds significant promise because of the high positive expression of these antigens in tumors. This approach facilitates the reprogramming of T lymphocytes by a transgenic TCR through gene transfer of TCR α and β chains specific to tumor antigens, offering potential therapeutic advances for patients with advanced SS. Clinical trials of TCR-engineered T-cell therapy targeting NY-ESO-1 and MAGE-A4 have been conducted, with an objective response rate reported to be 40-60% across several trials. This promising efficacy suggests that TCR-engineered T-cell therapy could become an attractive novel therapeutic option for advanced SS, which has limited treatment options in later stages. However, if TCR-engineered T-cell therapy is to be used in clinical practice, the standard approach following the failure of doxorubicin-based chemotherapy in patients with advanced SS must be defined. Future studies will be critical for establishing treatment strategies in this field.
Collapse
Affiliation(s)
- Tomoki Nakamura
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| | - Masahiro Hasegawa
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| |
Collapse
|
4
|
Frazzette N, Jour G. Novel Molecular Methods in Soft Tissue Sarcomas: From Diagnostics to Theragnostics. Cancers (Basel) 2025; 17:1215. [PMID: 40227789 PMCID: PMC11987812 DOI: 10.3390/cancers17071215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/27/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
Soft tissue sarcomas (STSs) are a diverse group of malignant tumors derived from mesenchymal tissues [...].
Collapse
Affiliation(s)
| | - George Jour
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA;
| |
Collapse
|
5
|
Kong P, Jiao Y, Sun M, Zhou Z, Zhang Y, Yang X, Ren J, Yang M, Dong Y, Song B. SECTM1 acts as an immune-related biomarker of poor prognosis and promotes cancer progression by modulating M2 macrophage polarization in esophageal squamous cell carcinoma. Front Immunol 2025; 16:1507227. [PMID: 39944684 PMCID: PMC11814170 DOI: 10.3389/fimmu.2025.1507227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/07/2025] [Indexed: 05/09/2025] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most prevalent primary malignant esophageal tumor in China and has a poor prognosis, but lacks effective diagnostic and prognostic biomarkers. Through single-sample gene set enrichment analysis (ssGSEA), we conducted immune genomic analysis based on 28 immune features using transcriptomic data from 155 ESCC cases. We established of two ESCC subtypes characterized by high and low immune profiles, and 352 differentially expressed immune genes were identified between the two subtypes. Performed with univariate and multivariate Cox regression, a novel prognostic prediction model was developed based on three immune-related genes (MAP3K8, SECTM1, IGLV7-43), which has been identified as a relatively accurate, independent, and specific prognostic risk model for ESCC patients in different ESCC cohorts. Furthermore, SECTM1 was upregulated in ESCC tissues and associated with adverse clinical outcomes. In cell experiments, overexpression of SECTM1 effectively promoted the proliferation, migration, and invasion of ESCC cells, while SECTM1 knockdown significantly inhibited these cellular processes. Furthermore, its overexpression promoted macrophage polarization towards the M2-like phenotype and promoted the migration of M2-like macrophage cells and C-C Motif Chemokine Ligand 5 (CCL5) was the key mediator in the pro-cancer effect of SECTM1. In a Conclusion, our study established a prognostic prediction model based on immune-related gene signature, which provided a reliable prognostic tool for ESCC and identified SECTM1 as a potential biomarker in ESCC.
Collapse
Affiliation(s)
- Pengzhou Kong
- Translational Medicine Research Center and Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
- State Key Laboratory for Pneumoconiosis of National Health Commission, Key Laboratory of Prevention, Treatment and Fundamental Studies for Respiratory Diseases of Shanxi, Department of Respiratory and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ye Jiao
- Translational Medicine Research Center and Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Meng Sun
- Translational Medicine Research Center and Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhinan Zhou
- Translational Medicine Research Center and Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yingying Zhang
- Translational Medicine Research Center and Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xin Yang
- Translational Medicine Research Center and Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Ren
- Translational Medicine Research Center and Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Mengyuan Yang
- Translational Medicine Research Center and Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanyan Dong
- Translational Medicine Research Center and Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Pathology, Shanxi Provincial People’s Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Bin Song
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
6
|
Galoian K, Bilbao D, Denny C, Campos Gallego N, Roberts E, Martinez D, Temple H. Targeting cancer stem cells by TPA leads to inhibition of refractory sarcoma and extended overall survival. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200905. [PMID: 39640862 PMCID: PMC11617462 DOI: 10.1016/j.omton.2024.200905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/23/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Refractory cancer recurrence in patients is a serious challenge in modern medicine. Tumor regrowth in a more aggressive and invasive drug-resistant form is caused by a specific sub-population of tumor cells defined as cancer stem cells (CSCs). While the role of CSCs in cancer relapse is recognized, the signaling pathways of CSCs-driven chemoresistance are less well understood. Moreover, there are no effective therapeutic strategies that involve specific inhibition of CSCs responsible for cancer recurrence and drug resistance. There is a clinical need to develop new therapies for patients with refractory sarcomas, particularly fibrosarcoma. These aggressive tumors, with poor overall survival, do not respond to conventional therapies. Standard systemic chemotherapy for these tumors includes doxorubicin (DOX). A Tyr peptide analog (TPA), developed in our laboratory, specifically targets CSCs by drastically reducing expression of the polycomb group protein enhancer of zester (EZH2) and its downstream targets, specifically ALDH1A1 and Nanog. In vivo experiments demonstrated that TPA inhibited tumor growth in nu/nu mice with relapsed DOX-treated fibrosarcoma 7-fold and led to improved overall (2-fold) survival. In an experimental metastatic model, the combination of TPA with DOX treatment extended overall survival 3-fold, suggesting that targeting CSC can become an effective strategy in the treatment of refractory/relapse fibrosarcoma.
Collapse
Affiliation(s)
- Karina Galoian
- Department of Orthopedic Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Daniel Bilbao
- Department of Pathology and Laboratory Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Carina Denny
- Department of Orthopedic Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | | | - Evan Roberts
- Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Daniel Martinez
- Department of Orthopedic Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - H.T. Temple
- Department of Orthopedic Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
7
|
Zoghbi M, Patel BA, Roulleaux Dugage M, Mezquita L, Bahleda R, Dufresne A, Brahmi M, Ray-Coquard I, Pautier P, Blay JY, Le Cesne A, Massard C, Besse B, Auclin E, Nassif Haddad EF. Association of Lung Immune Prognostic Index (LIPI) with Disease Control Rate and Progression-Free Survival in Patients with Soft-Tissue Sarcoma Treated with Immunotherapy in Early-Phase Trials. Cancers (Basel) 2024; 16:4053. [PMID: 39682239 DOI: 10.3390/cancers16234053] [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: 10/22/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND The efficacy of immunotherapies in soft-tissue sarcomas (STSs) is limited, and biomarkers of response are lacking. The lung immune prognostic index (LIPI) is a prognostic biomarker used with immunotherapy across cancer types. This study investigates the association of LIPI with the disease control rate (DCR) and progression-free survival (PFS) in patients with STS treated with immunotherapy versus other therapies in early-phase trials. METHODS This post hoc analysis was conducted with patients with STS from Gustave Roussy and Centre Léon Bérard between January 2012 and June 2021. The LIPI was calculated based on a derived neutrophil-to-lymphocyte ratio > 3 and elevated lactate dehydrogenase. Patients were categorized based on treatment (immunotherapy or other) and LIPI (good, intermediate, or poor). DCR was defined as the sum of stable disease and complete and partial response. RESULTS A total of 82 patients were enrolled in immunotherapy trials and 126 in the other therapy trials. In the immunotherapy group, DCR was higher in patients with good LIPI (76%; n = 23/30) compared with the intermediate (50%; n = 13/26) and poor LIPI groups (8%; n = 1/12; p < 0.001). The other-therapy group did not show significant differences in DCR by LIPI: DCR was 70% (n = 48/69), 70% (n = 21/30), and 60% (n = 6/10) in patients with good, intermediate, and poor LIPI, respectively (p = 0.86). In multivariate analyses, LIPI was independently associated with PFS in the immunotherapy group (hazard ratio = 5.97, p = 0.0001) and not in the control group (p = 0.71). CONCLUSIONS LIPI is a significant independent prognostic marker for DCR in patients with STS treated with immunotherapy. In early-phase trials, LIPI could be used as a screening tool for stratification at inclusion. High neutrophil levels, which correlate with a poorer LIPI score, are likely associated with immunotherapy resistance. This relationship could explain the statistical impact of poor LIPI in the immunotherapy group.
Collapse
Affiliation(s)
- Marianne Zoghbi
- Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Brina A Patel
- Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matthieu Roulleaux Dugage
- Département d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy, 94805 Villejuif, France
| | - Laura Mezquita
- Department of Medical Oncology, IDIBAPS, Hospital Clínic, 08036 Barcelona, Spain
- Department of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Rastilav Bahleda
- Département d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy, 94805 Villejuif, France
| | - Armelle Dufresne
- Département d'Oncologie Médicale, Centre Léon Bérard, 69008 Lyon, France
| | - Mehdi Brahmi
- Département d'Oncologie Médicale, Centre Léon Bérard, 69008 Lyon, France
| | | | - Patricia Pautier
- Département de Médecine Oncologique, Gustave Roussy, 94805 Villejuif, France
| | - Jean-Yves Blay
- Département d'Oncologie Médicale, Centre Léon Bérard, 69008 Lyon, France
| | - Axel Le Cesne
- International Department, Gustave Roussy, 94805 Villejuif, France
| | - Christophe Massard
- Département d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy, 94805 Villejuif, France
| | - Benjamin Besse
- Département de Médecine Oncologique, Gustave Roussy, 94805 Villejuif, France
| | - Edouard Auclin
- Department of Medical Oncology, Institut Bergonié, 33076 Bordeaux, France
| | - Elise F Nassif Haddad
- Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
8
|
Iwai Y, Baldwin XL, Feeney T, Agala CB, Yanagihara TK, Stein JN, Kim HJ, Spanheimer PM. Trends in the use of immunotherapy to treat soft tissue sarcoma. Am J Surg 2024; 236:115794. [PMID: 38879356 PMCID: PMC11392640 DOI: 10.1016/j.amjsurg.2024.115794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/29/2024] [Accepted: 06/04/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND The role of immune-oncology (IO) therapy in soft tissue sarcoma (STS) is underexplored. This study characterized IO use in STS. METHODS This is a retrospective analysis of patients with a soft tissue mass in the National Cancer Database, 2011-2021. Patients were categorized by IO receipt status. Groupwise testing and proportional trend tests were performed with Chi-squared tests. Multivariate logistic regression was performed to assess factors associated with IO receipt. RESULTS Of the 103,092 patients with STS, 1935 (1.9 %) received or were recommended IO therapy. IO use increased 10-fold (0.24 %-2.5 % from 2011 to 2021; p < 0.0001). Patients had higher odds of receiving IO when having higher grade tumors and metastatic disease, and when treated at an academic research center (all p < 0.001). CONCLUSIONS IO use in STS is low but increasing and primarily used in the metastatic setting. Future studies should identify biomarkers of IO response and facilitators for treatment receipt.
Collapse
Affiliation(s)
- Yoshiko Iwai
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xavier L Baldwin
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Timothy Feeney
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chris B Agala
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ted K Yanagihara
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jacob N Stein
- Department of Medical Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hong Jin Kim
- Division of Surgical Oncology, Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Philip M Spanheimer
- Division of Surgical Oncology, Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
9
|
Qiu Y, Zhang Q, Liu X. CCDC88A could serve as a prognostic biomarker for SARC patients. Asian J Surg 2024:S1015-9584(24)01888-8. [PMID: 39266353 DOI: 10.1016/j.asjsur.2024.08.148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/11/2024] [Accepted: 08/20/2024] [Indexed: 09/14/2024] Open
Affiliation(s)
- Yunpeng Qiu
- Department of Orthopaedics, The Second People's Hospital of Futian, Shenzhen, 27 Zhongkang Road, Shangmeilin, Futian District, 518049, Shenzhen, Guangdong, China
| | - Qiao Zhang
- Department of Orthopaedics, The Second People's Hospital of Futian, Shenzhen, 27 Zhongkang Road, Shangmeilin, Futian District, 518049, Shenzhen, Guangdong, China
| | - Xing Liu
- Department of Emergency, The Second People's Hospital of Futian, Shenzhen, 27 Zhongkang Road, Shangmeilin, Futian District, 518049, Shenzhen, Guangdong, China.
| |
Collapse
|
10
|
Feng Q, Yu W, Feng JH, Huang Q, Xiao GX. Jejunal sarcomatoid carcinoma: A case report and review of literature. World J Gastrointest Oncol 2024; 16:3723-3731. [PMID: 39171179 PMCID: PMC11334045 DOI: 10.4251/wjgo.v16.i8.3723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Sarcomatoid carcinoma (SCA) of the jejunum is a rare and aggressive neoplasm affecting the smooth muscle cells of the jejunum. This study presents a recent case of jejunal SCA, detailing its diagnosis and treatment, thereby providing a reference for clinical practice. CASE SUMMARY A 65-year-old male presented to Yichang Central People's Hospital with a chief complaint of hemorrhoids. A computed tomography (CT) scan incidentally revealed multiple abnormal signals in the liver. Subsequent positron emission tomography/CT at Wuhan Union Hospital indicated malignant tumor progression, with a primary duodenal tumor and multiple metastases in the upper left abdomen. Intraoperatively, a large tumor was identified on the omentum. Histopathological and immunohistochemical analyses of the resected specimen confirmed the diagnosis of jejunal SCA. The patient received a combination therapy of sintilimab, nanoparticle albumin-bound paclitaxel, and anlotinib. Follow-up imaging demonstrated significant reduction of hepatic and peritoneal lesions. The patient has remained stable for over one year postoperatively. CONCLUSION This case suggests that chemotherapy, immunotherapy, plus targeted therapy may represent an optimal treatment for intestinal SCA, meriting further investigation.
Collapse
Affiliation(s)
- Qian Feng
- The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, Hubei Province, China
- Department of Oncology, Yichang Central People's Hospital, Yichang 443000, Hubei Province, China
| | - Wei Yu
- The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, Hubei Province, China
- Department of Oncology, Yichang Central People's Hospital, Yichang 443000, Hubei Province, China
| | - Jing-Hui Feng
- The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, Hubei Province, China
- Department of Oncology, Yichang Central People's Hospital, Yichang 443000, Hubei Province, China
| | - Qiao Huang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, Hubei Province, China
- Department of Oncology, Yichang Central People's Hospital, Yichang 443000, Hubei Province, China
| | - Gui-Xiang Xiao
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
11
|
Lv X, Zhu L, Lan G, Huang Z, Guo Q. A clinical tool to predict overall survival of elderly patients with soft tissue sarcoma after surgical resection. Sci Rep 2024; 14:15098. [PMID: 38956230 PMCID: PMC11220034 DOI: 10.1038/s41598-024-65657-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
With the aging world population, the incidence of soft tissue sarcoma (STS) in the elderly gradually increases and the prognosis is poor. The primary goal of this research was to analyze the relevant risk factors affecting the postoperative overall survival in elderly STS patients and to provide some guidance and assistance in clinical treatment. The study included 2,353 elderly STS patients from the Surveillance, Epidemiology, and End Results database. To find independent predictive variables, we employed the Cox proportional risk regression model. R software was used to develop and validate the nomogram model to predict postoperative overall survival. The performance and practical value of the nomogram were evaluated using calibration curves, the area under the curve, and decision curve analysis. Age, tumor primary site, disease stage, tumor size, tumor grade, N stage, and marital status, are the risk variables of postoperative overall survival, and the prognostic model was constructed on this basis. In the two sets, both calibration curves and receiver operating characteristic curves showed that the nomogram had high predictive accuracy and discriminative power, while decision curve analysis demonstrated that the model had good clinical usefulness. A predictive nomogram was designed and tested to evaluate postoperative overall survival in elderly STS patients. The nomogram allows clinical practitioners to more accurately evaluate the prognosis of individual patients, facilitates the progress of individualized treatment, and provides clinical guidance.
Collapse
Affiliation(s)
- Xianmei Lv
- Department of Radiotherapy, Jinhua People's Hospital, Jinhua, Zhejiang, China
| | - Lujian Zhu
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Gaochen Lan
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Zhangheng Huang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiusheng Guo
- Department of Medical Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, No. 365 Renmin East Road, Jinhua, Zhejiang, China.
| |
Collapse
|
12
|
Su C, Kent CL, Pierpoint M, Floyd W, Luo L, Williams NT, Ma Y, Peng B, Lazarides AL, Subramanian A, Himes JE, Perez VM, Hernansaiz-Ballesteros RD, Roche KE, Modliszewski JL, Selitsky SR, Shinohara ML, Wisdom AJ, Moding EJ, Mowery YM, Kirsch DG. Enhancing radiotherapy response via intratumoral injection of a TLR9 agonist in autochthonous murine sarcomas. JCI Insight 2024; 9:e178767. [PMID: 39133651 PMCID: PMC11383182 DOI: 10.1172/jci.insight.178767] [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: 01/03/2024] [Accepted: 06/11/2024] [Indexed: 08/21/2024] Open
Abstract
Radiation therapy (RT) is frequently used to treat cancers, including soft-tissue sarcomas. Prior studies established that the toll-like receptor 9 (TLR9) agonist cytosine-phosphate-guanine oligodeoxynucleotide (CpG) enhances the response to RT in transplanted tumors, but the mechanisms of this enhancement remain unclear. Here, we used CRISPR/Cas9 and the chemical carcinogen 3-methylcholanthrene (MCA) to generate autochthonous soft-tissue sarcomas with high tumor mutation burden. Treatment with a single fraction of 20 Gy RT and 2 doses of CpG significantly enhanced tumor response, which was abrogated by genetic or immunodepletion of CD8+ T cells. To characterize the immune response to CpG+RT, we performed bulk RNA-Seq, single-cell RNA-Seq, and mass cytometry. Sarcomas treated with 20 Gy and CpG demonstrated increased CD8 T cells expressing markers associated with activation and proliferation, such as Granzyme B, Ki-67, and IFN-γ. CpG+RT also upregulated antigen presentation pathways on myeloid cells. Furthermore, in sarcomas treated with CpG+RT, TCR clonality analysis suggests an increase in clonal T cell dominance. Collectively, these findings demonstrate that CpG+RT significantly delays tumor growth in a CD8 T cell-dependent manner. These results provide a strong rationale for clinical trials evaluating CpG or other TLR9 agonists with RT in patients with soft-tissue sarcoma.
Collapse
Affiliation(s)
- Chang Su
- Department of Pharmacology and Cancer Biology and
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Collin L Kent
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Matthew Pierpoint
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Warren Floyd
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Nerissa T Williams
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Brian Peng
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Alexander L Lazarides
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Ajay Subramanian
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Jonathon E Himes
- Department of Pharmacology and Cancer Biology and
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | - Kimberly E Roche
- Tempus AI Inc., Durham, North Carolina, USA
- QuantBio LLC, Durham, North Carolina, USA
| | - Jennifer L Modliszewski
- QuantBio LLC, Durham, North Carolina, USA
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Sara R Selitsky
- Tempus AI Inc., Durham, North Carolina, USA
- QuantBio LLC, Durham, North Carolina, USA
| | - Mari L Shinohara
- Department of Integrative Immunology
- Department of Molecular Genetics and Microbiology, and
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Amy J Wisdom
- Harvard Radiation Oncology Program, Boston, Massachusetts, USA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
- Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Yvonne M Mowery
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David G Kirsch
- Department of Pharmacology and Cancer Biology and
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Radiation Oncology and
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Riley GM, Steffner R, Kwong S, Chin A, Boutin RD. MRI of Soft-Tissue Tumors: What to Include in the Report. Radiographics 2024; 44:e230086. [PMID: 38696323 DOI: 10.1148/rg.230086] [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: 05/04/2024]
Abstract
MRI serves as a critical step in the workup, local staging, and treatment planning of extremity soft-tissue masses. For the radiologist to meaningfully contribute to the management of soft-tissue masses, they need to provide a detailed list of descriptors of the lesion outlined in an organized report. While it is occasionally possible to use MRI to provide a diagnosis for patients with a mass, it is more often used to help with determining the differential diagnosis and planning of biopsies, surgery, radiation treatment, and chemotherapy (when provided). Each descriptor on the list outlined in this article is specifically aimed to assist in one or more facets of the overall approach to soft-tissue masses. This applies to all masses, but in particular sarcomas. Those descriptors are useful to help narrow the differential diagnosis and ensure concordance with a pathologic diagnosis and its accompanying grade assignment of soft-tissue sarcomas. These include a lesion's borders and shape, signal characteristics, and contrast enhancement pattern; the presence of peritumoral edema and peritumoral enhancement; and the presence of lymph nodes. The items most helpful in assisting surgical planning include a lesion's anatomic location, site of origin, size, location relative to a landmark, relationship to adjacent structures, and vascularity including feeding and draining vessels. The authors provide some background information on soft-tissue sarcomas, including their diagnosis and treatment, for the general radiologist and as a refresher for radiologists who are more experienced in tumor imaging. ©RSNA, 2024 See the invited commentary by Murphey in this issue.
Collapse
Affiliation(s)
- Geoffrey M Riley
- From the Departments of Radiology (G.M.R., R.D.B.) and Orthopedic Surgery (R.S.), Stanford University Medical Center, 300 Pasteur Dr, Stanford, CA 94305-5105; Department of Radiology, The Permanente Medical Group, Oakland, Calif (S.K.); and Department of Radiation Oncology, Stanford Cancer Institute, Stanford, Calif (A.C.)
| | - Robert Steffner
- From the Departments of Radiology (G.M.R., R.D.B.) and Orthopedic Surgery (R.S.), Stanford University Medical Center, 300 Pasteur Dr, Stanford, CA 94305-5105; Department of Radiology, The Permanente Medical Group, Oakland, Calif (S.K.); and Department of Radiation Oncology, Stanford Cancer Institute, Stanford, Calif (A.C.)
| | - Steven Kwong
- From the Departments of Radiology (G.M.R., R.D.B.) and Orthopedic Surgery (R.S.), Stanford University Medical Center, 300 Pasteur Dr, Stanford, CA 94305-5105; Department of Radiology, The Permanente Medical Group, Oakland, Calif (S.K.); and Department of Radiation Oncology, Stanford Cancer Institute, Stanford, Calif (A.C.)
| | - Alexander Chin
- From the Departments of Radiology (G.M.R., R.D.B.) and Orthopedic Surgery (R.S.), Stanford University Medical Center, 300 Pasteur Dr, Stanford, CA 94305-5105; Department of Radiology, The Permanente Medical Group, Oakland, Calif (S.K.); and Department of Radiation Oncology, Stanford Cancer Institute, Stanford, Calif (A.C.)
| | - Robert D Boutin
- From the Departments of Radiology (G.M.R., R.D.B.) and Orthopedic Surgery (R.S.), Stanford University Medical Center, 300 Pasteur Dr, Stanford, CA 94305-5105; Department of Radiology, The Permanente Medical Group, Oakland, Calif (S.K.); and Department of Radiation Oncology, Stanford Cancer Institute, Stanford, Calif (A.C.)
| |
Collapse
|
14
|
Qiu Y, Qin A, Zhao R, Ding J, Jia WWG, Singh M, Murad Y, Tan Q, Kichenadasse G. Oncolytic virotherapy stimulates anti‑tumor immune response and demonstrates activity in advanced sarcoma: Report of two cases. Oncol Lett 2024; 27:244. [PMID: 38638849 PMCID: PMC11024735 DOI: 10.3892/ol.2024.14377] [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: 10/18/2023] [Accepted: 02/27/2024] [Indexed: 04/20/2024] Open
Abstract
Sarcoma is derived from mesenchymal neoplasms and has numerous subtypes, accounting for 1% of all adult malignancies and 15% of childhood malignancies. The prognosis of metastatic or recurrent sarcoma remains poor. The current study presents two cases of sarcoma enrolled in a phase I dose escalation trial for solid tumor, who had previously failed all standard therapies. These patients were treated with VG161, an immune-stimulating herpes simplex virus type 1 oncolytic virus with payloads of IL-12, IL-15 and IL-15 receptor α unit, and a programmed cell death 1 (PD-1)/PD-1 ligand 1 blocking peptide. Both cases demonstrated stable disease as the best response, accompanied by a noteworthy prolongation of progression-free survival (11.8 months for chondrosarcoma and 11.9 months for soft tissue sarcoma, respectively) at a dose of 2.5×108 PFU/cycle. In addition, the treatment led to the activation of anti-cancer immunity, as evident from cytokine, lymphocyte subset and related pathway analyses of peripheral blood and/or tumor biopsy samples. These promising results suggest that VG161 monotherapy holds promise as an effective treatment for sarcoma and warrants further investigation through clinical trials. The two reported patients were part of a phase I clinical trial conducted and registered on the Australian New Zealand Clinical Trials Registry in Australia (registration no. ACTRN12620000244909; registration date, 26 February, 2020).
Collapse
Affiliation(s)
- Yeting Qiu
- Shanghai Virogin Biotech Ltd., Shanghai 200240, P.R. China
| | - Aijun Qin
- Shanghai Virogin Biotech Ltd., Shanghai 200240, P.R. China
| | - Ronghua Zhao
- Shanghai Virogin Biotech Ltd., Shanghai 200240, P.R. China
- Virogin Biotech Canada Ltd., Richmond, BC V6V 3A4, Canada
- China National Biotec Group (CNBG) - Virogin Biotech (Shanghai) Co., Ltd., Shanghai 200240, P.R. China
| | - Jun Ding
- Shanghai Virogin Biotech Ltd., Shanghai 200240, P.R. China
- Virogin Biotech Canada Ltd., Richmond, BC V6V 3A4, Canada
| | - William Wei-Guo Jia
- Shanghai Virogin Biotech Ltd., Shanghai 200240, P.R. China
- Virogin Biotech Canada Ltd., Richmond, BC V6V 3A4, Canada
- China National Biotec Group (CNBG) - Virogin Biotech (Shanghai) Co., Ltd., Shanghai 200240, P.R. China
| | - Manu Singh
- Virogin Biotech Canada Ltd., Richmond, BC V6V 3A4, Canada
| | - Yanal Murad
- Virogin Biotech Canada Ltd., Richmond, BC V6V 3A4, Canada
| | - Qian Tan
- Shanghai Virogin Biotech Ltd., Shanghai 200240, P.R. China
| | - Ganessan Kichenadasse
- Department of Medical Oncology, Southern Oncology Clinical Research Unit, Flinders Private Hospital, Adelaide, South Australia 5042, Australia
| |
Collapse
|
15
|
Cao Y, Li Y, Ren C, Yang C, Hao R, Mu T. Manganese-based nanomaterials promote synergistic photo-immunotherapy: green synthesis, underlying mechanisms, and multiple applications. J Mater Chem B 2024; 12:4097-4117. [PMID: 38587869 DOI: 10.1039/d3tb02844e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Single phototherapy and immunotherapy have individually made great achievements in tumor treatment. However, monotherapy has difficulty in balancing accuracy and efficiency. Combining phototherapy with immunotherapy can realize the growth inhibition of distal metastatic tumors and enable the remote monitoring of tumor treatment. The development of nanomaterials with photo-responsiveness and anti-tumor immunity activation ability is crucial for achieving photo-immunotherapy. As immune adjuvants, photosensitizers and photothermal agents, manganese-based nanoparticles (Mn-based NPs) have become a research hotspot owing to their multiple ways of anti-tumor immunity regulation, photothermal conversion and multimodal imaging. However, systematic studies on the synergistic photo-immunotherapy applications of Mn-based NPs are still limited; especially, the green synthesis and mechanism of Mn-based NPs applied in immunotherapy are rarely comprehensively discussed. In this review, the synthesis strategies and function of Mn-based NPs in immunotherapy are first introduced. Next, the different mechanisms and leading applications of Mn-based NPs in immunotherapy are reviewed. In addition, the advantages of Mn-based NPs in synergistic photo-immunotherapy are highlighted. Finally, the challenges and research focus of Mn-based NPs in combination therapy are discussed, which might provide guidance for future personalized cancer therapy.
Collapse
Affiliation(s)
- Yuanyuan Cao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Yilin Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Caixia Ren
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Chengkai Yang
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, P. R. China
| | - Rongzhang Hao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Tiancheng Mu
- Department of Chemistry, Renmin University of China, Beijing 100872, P. R. China.
| |
Collapse
|
16
|
Syed RU, Afsar S, Aboshouk NAM, Salem Alanzi S, Abdalla RAH, Khalifa AAS, Enrera JA, Elafandy NM, Abdalla RAH, Ali OHH, Satheesh Kumar G, Alshammari MD. LncRNAs in necroptosis: Deciphering their role in cancer pathogenesis and therapy. Pathol Res Pract 2024; 256:155252. [PMID: 38479121 DOI: 10.1016/j.prp.2024.155252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 04/14/2024]
Abstract
Necroptosis, a controlled type of cell death that is different from apoptosis, has become a key figure in the aetiology of cancer and offers a possible target for treatment. A growing number of biological activities, including necroptosis, have been linked to long noncoding RNAs (lncRNAs), a varied family of RNA molecules with limited capacity to code for proteins. The complex interactions between LncRNAs and important molecular effectors of necroptosis, including mixed lineage kinase domain-like pseudokinase (MLKL) and receptor-interacting protein kinase 3 (RIPK3), will be investigated. We will explore the many methods that LncRNAs use to affect necroptosis, including protein-protein interactions, transcriptional control, and post-transcriptional modification. Additionally, the deregulation of certain LncRNAs in different forms of cancer will be discussed, highlighting their dual function in influencing necroptotic processes as tumour suppressors and oncogenes. The goal of this study is to thoroughly examine the complex role that LncRNAs play in controlling necroptotic pathways and how that regulation affects the onset and spread of cancer. In the necroptosis for cancer treatment, this review will also provide insight into the possible therapeutic uses of targeting LncRNAs. Techniques utilising LncRNA-based medicines show promise in controlling necroptotic pathways to prevent cancer from spreading and improve the effectiveness of treatment.
Collapse
Affiliation(s)
- Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Hail 81442, Saudi Arabia.
| | - S Afsar
- Department of Virology, Sri Venkateswara University, Tirupathi, Andhra Pradesh 517502, India.
| | - Nayla Ahmed Mohammed Aboshouk
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | | | | | - Amna Abakar Suleiman Khalifa
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Jerlyn Apatan Enrera
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Nancy Mohammad Elafandy
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Randa Abdeen Husien Abdalla
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Omar Hafiz Haj Ali
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - G Satheesh Kumar
- Department of Pharmaceutical Chemistry, College of Pharmacy, Seven Hills College of Pharmacy, Venkataramapuram, Tirupati, India
| | - Maali D Alshammari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| |
Collapse
|
17
|
Brown JM, Patel R, Smith-Fry K, Ward M, Oliver T, Jones KB. Genetically engineered mouse model of pleomorphic liposarcoma: Immunophenotyping and histologic characterization. Neoplasia 2024; 48:100956. [PMID: 38199172 PMCID: PMC10788790 DOI: 10.1016/j.neo.2023.100956] [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: 08/21/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024]
Abstract
INTRODUCTION Pleomorphic liposarcoma is a rare and aggressive subset of soft-tissue sarcomas with a high mortality burden. Local treatment largely consists of radiation therapy and wide surgical resection, but options for systemic therapy in the setting of metastatic disease are limited and largely ineffective, prompting exploration of novel therapeutic strategies and experimental models. As with other cancers, sarcoma cell lines and patient-derived xenograft models have been developed and used to characterize these tumors and identify therapeutic targets, but these models have inherent limitations. The establishment of genetically engineered mouse models represents a more realistic framework for reproducing clinically relevant conditions for studying pleomorphic liposarcoma. METHODS Trp53fl/fl/Rb1fl/fl/Ptenfl/fl (RPP) mice were used to reliably generate an immunocompetent model of mouse pleomorphic liposarcoma through Cre-mediated conditional silencing of the Trp53, Rb1, and Pten tumor suppressor genes. Evaluation of tumor-infiltrating lymphocytes was assessed with immunostaining for CD4, CD8, and PD-L1, and flow cytometry with analysis of CD45, CD3, CD4, CD8, CD19, F4/80, CD11b, and NKp46 sub-populations. RESULTS Mice reliably produced noticeable soft-tissue tumors in approximately 6 weeks with rapid tumor growth between 100 and 150 days of life, after which mice reached euthanasia criteria. Histologic features were consistent with pleomorphic liposarcoma, including widespread pleomorphic lipoblasts. Immunoprofiling and assessment of tumor-infiltrating lymphocytes was consistent with other soft-tissue sarcomas. CONCLUSION Genetically engineered RPP mice reliably produced soft-tissue tumors consistent with pleomorphic liposarcoma, which immunological findings similar to other soft-tissue sarcomas. This model may demonstrate utility in testing treatments for this rare disease, including immunomodulatory therapies.
Collapse
Affiliation(s)
| | - Rahi Patel
- University of Utah Health Huntsman Cancer Institute, USA
| | | | - Michael Ward
- University of Utah Health Huntsman Cancer Institute, USA
| | | | - Kevin B Jones
- University of Utah Health Huntsman Cancer Institute, USA
| |
Collapse
|
18
|
Liu J, Lu J, Wang G, Gu L, Li W. Prognostic characteristics of a six-gene signature based on ssGSEA in sarcoma. Aging (Albany NY) 2024; 16:1536-1554. [PMID: 38240704 PMCID: PMC10866427 DOI: 10.18632/aging.205443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/07/2023] [Indexed: 02/06/2024]
Abstract
BACKGROUND Sarcoma is a rare malignant tumor originating of the interstitial or connective tissue with a poor prognosis. Next-generation sequencing technology offers new opportunities for accurate diagnosis and treatment of sarcomas. There is an urgent need for new gene signature to predict prognosis and evaluate treatment outcomes. METHODS We used transcriptome data from the Cancer Genome Atlas (TCGA) database and single sample gene set enrichment analysis (ssGSEA) to explore the cancer hallmarks most associated with prognosis in sarcoma patients. Then, weighted gene coexpression network analysis, univariate COX regression analysis and random forest algorithm were used to construct prognostic gene characteristics. Finally, the prognostic value of gene markers was validated in the TCGA and Integrated Gene Expression (GEO) (GSE17118) datasets, respectively. RESULTS MYC targets V1 and V2 are the main cancer hallmarks affecting the overall survival (OS) of sarcoma patients. A six-gene signature including VEGFA, HMGB3, FASN, RCC1, NETO2 and BIRC5 were constructed. Kaplan-Meier analysis suggested that higher risk scores based on the six-gene signature associated with poorer OS (P < 0.001). The receiver Operating characteristic curve showed that the risk score based on the six-gene signature was a good predictor of sarcoma, with an area under the curve (AUC) greater than 0.73. In addition, the prognostic value of the six-gene signature was validated in GSE17118 with an AUC greater than 0.72. CONCLUSION This six-gene signature is an independent prognostic factor in patients with sarcoma and is expected to be a potential therapeutic target for sarcoma.
Collapse
Affiliation(s)
- Jun Liu
- Department of Clinical Laboratory, Dongguan Hospital of Guangzhou University of Chinese Medicine, Dongguan 523005, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515000, China
| | - Jianjun Lu
- Department of Quality Control and Evaluation, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Gefei Wang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515000, China
| | - Liming Gu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515000, China
| | - Wenli Li
- Department of Clinical Laboratory, Dongguan Hospital of Guangzhou University of Chinese Medicine, Dongguan 523005, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515000, China
| |
Collapse
|
19
|
Wani AK, Singh R, Akhtar N, Prakash A, Nepovimova E, Oleksak P, Chrienova Z, Alomar S, Chopra C, Kuca K. Targeted Inhibition of the PI3K/Akt/mTOR Signaling Axis: Potential for Sarcoma Therapy. Mini Rev Med Chem 2024; 24:1496-1520. [PMID: 38265369 DOI: 10.2174/0113895575270904231129062137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 01/25/2024]
Abstract
Sarcoma is a heterogeneous group of malignancies often resistant to conventional chemotherapy and radiation therapy. The phosphatidylinositol-3-kinase/ protein kinase B /mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway has emerged as a critical cancer target due to its central role in regulating key cellular processes such as cell growth, proliferation, survival, and metabolism. Dysregulation of this pathway has been implicated in the development and progression of bone sarcomas (BS) and soft tissue sarcomas (STS). PI3K/Akt/mTOR inhibitors have shown promising preclinical and clinical activity in various cancers. These agents can inhibit the activation of PI3K, Akt, and mTOR, thereby reducing the downstream signaling events that promote tumor growth and survival. In addition, PI3K/Akt/mTOR inhibitors have been shown to enhance the efficacy of other anticancer therapies, such as chemotherapy and radiation therapy. The different types of PI3K/Akt/mTOR inhibitors vary in their specificity, potency, and side effect profiles and may be effective depending on the specific sarcoma type and stage. The molecular targeting of PI3K/Akt/mToR pathway using drugs, phytochemicals, nanomaterials (NMs), and microbe-derived molecules as Pan-PI3K inhibitors, selective PI3K inhibitors, and dual PI3K/mTOR inhibitors have been delineated. While there are still challenges to be addressed, the preclinical and clinical evidence suggests that these inhibitors may significantly improve patient outcomes. Further research is needed to understand the potential of these inhibitors as sarcoma therapeutics and to continue developing more selective and effective agents to meet the clinical needs of sarcoma patients.
Collapse
Affiliation(s)
- Atif Khurshid Wani
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Reena Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Nahid Akhtar
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Ajit Prakash
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Patrik Oleksak
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Zofia Chrienova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Suliman Alomar
- King Saud University, Zoology Department, College of Science, Riyadh, 11451, Saudi Arabia
| | - Chirag Chopra
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Králové, Czechia
| |
Collapse
|
20
|
Nielsen M, Monberg T, Sundvold V, Albieri B, Hovgaard D, Petersen MM, Krarup-Hansen A, Met Ö, Camilio K, Clancy T, Stratford R, Sveinbjornsson B, Rekdal Ø, Junker N, Svane IM. LTX-315 and adoptive cell therapy using tumor-infiltrating lymphocytes generate tumor specific T cells in patients with metastatic soft tissue sarcoma. Oncoimmunology 2023; 13:2290900. [PMID: 38125722 PMCID: PMC10732595 DOI: 10.1080/2162402x.2023.2290900] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
LTX-315 is an oncolytic peptide that elicits both local and systemic immune responses upon intratumoral injection. In the present pilot trial, we treated patients with metastatic soft tissue sarcoma with the combination of LTX-315 and adoptive T-cell therapy using in vitro expanded tumor-infiltrating lymphocytes. Six heavily pretreated patients were included in the trial and treated with LTX-315 of which four patients proceeded to adoptive T-cell therapy. Overall, the treatment was considered safe with only expected and manageable toxicity. The best overall clinical response was stable disease for 208 days, and in this patient, we detected tumor-reactive T cells in the blood that lasted until disease progression. In three patients T-cell reactivity against in silico predicted neoantigens was demonstrated. Additionally, de novo T-cell clones were generated and expanded in the blood following LTX-315 injections. In conclusion, this pilot study provides proof that it is feasible to combine LTX-315 and adoptive T-cell therapy, and that this treatment can induce systemic immune responses that resulted in stabilization of the disease in sarcoma patients with otherwise progressive disease. Further optimization of the treatment protocol is warranted to increase clinical activity. ClinicalTrials.gov Identifier: NCT03725605.
Collapse
Affiliation(s)
- Morten Nielsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Tine Monberg
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | | | - Benedetta Albieri
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Dorrit Hovgaard
- Department of Orthopedic Surgery, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Michael Mørk Petersen
- Department of Orthopedic Surgery, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | - Özcan Met
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | | | | | | | | | | | - Niels Junker
- Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
21
|
Jeong S, Afroz S, Kang D, Noh J, Suh J, Kim JH, You HJ, Kang HG, Kim YJ, Kim JH. Sarcoma Immunotherapy: Confronting Present Hurdles and Unveiling Upcoming Opportunities. Mol Cells 2023; 46:579-588. [PMID: 37853684 PMCID: PMC10590708 DOI: 10.14348/molcells.2023.0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 10/20/2023] Open
Abstract
Sarcomas are rare and heterogeneous mesenchymal neoplasms originating from the bone or soft tissues, which pose significant treatment challenges. The current standard treatment for sarcomas consists of surgical resection, often combined with chemo- and radiotherapy; however, local recurrence and metastasis remain significant concerns. Although immunotherapy has demonstrated promise in improving long-term survival rates for certain cancers, sarcomas are generally considered to be relatively less immunogenic than other tumors, presenting substantial challenges for effective immunotherapy. In this review, we examine the possible opportunities for sarcoma immunotherapy, noting cancer testis antigens expressed in sarcomas. We then cover the current status of immunotherapies in sarcomas, including progress in cancer vaccines, immune checkpoint inhibitors, and adoptive cellular therapy and their potential in combating these tumors. Furthermore, we discuss the limitations of immunotherapies in sarcomas, including a low tumor mutation burden and immunosuppressive tumor microenvironment, and explore potential strategies to tackle the immunosuppressive barriers in therapeutic interventions, shedding light on the development of effective and personalized treatments for sarcomas. Overall, this review provides a comprehensive overview of the current status and potential of immunotherapies in sarcoma treatment, highlighting the challenges and opportunities for developing effective therapies to improve the outcomes of patients with these rare malignancies.
Collapse
Affiliation(s)
- Sehan Jeong
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Sharmin Afroz
- Department of Occupational and Environmental Medicine, Ewha Womans University College of Medicine, Seoul 07985, Korea
| | - Donghyun Kang
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Jeonghwan Noh
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Jooyeon Suh
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - June Hyuk Kim
- Orthopaedic Oncology Clinic, Center for Rare Cancer, Research Institute and Hospital, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Korea
| | - Hye Jin You
- Cancer Microenvironment Branch, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Hyun Guy Kang
- Orthopaedic Oncology Clinic, Center for Rare Cancer, Research Institute and Hospital, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Korea
| | - Yi-Jun Kim
- Department of Occupational and Environmental Medicine, Ewha Womans University College of Medicine, Seoul 07985, Korea
- Department of Radiation Oncology, Ewha Womans University College of Medicine, Seoul 07985, Korea
| | - Jin-Hong Kim
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
- Bio-MAX Institute, Seoul National University, Seoul 08826, Korea
- Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang 25354, Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
22
|
Leng D, Yang Z, Sun H, Song C, Huang C, Ip KU, Chen G, Deng CX, Zhang XD, Zhao Q. Comprehensive Analysis of Tumor Microenvironment Reveals Prognostic ceRNA Network Related to Immune Infiltration in Sarcoma. Clin Cancer Res 2023; 29:3986-4001. [PMID: 37527025 PMCID: PMC10543973 DOI: 10.1158/1078-0432.ccr-22-3396] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/23/2023] [Accepted: 07/28/2023] [Indexed: 08/03/2023]
Abstract
PURPOSE Sarcoma is the second most common solid tumor type in children and adolescents. The high level of tumor heterogeneity as well as aggressive behavior of sarcomas brings serious difficulties to developing effective therapeutic strategies for clinical application. Therefore, it is of great importance to identify accurate biomarkers for early detection and prognostic prediction of sarcomas. EXPERIMENTAL DESIGN In this study, we characterized three subtypes of sarcomas based on tumor immune infiltration levels (TIIL), and constructed a prognosis-related competing endogenous RNA (ceRNA) network to investigate molecular regulations in the sarcoma tumor microenvironment (TME). We further built a subnetwork consisting of mRNAs and lncRNAs that are targets of key miRNAs and strongly correlated with each other in the ceRNA network. After validation using public data and experiments in vivo and in vitro, we deeply dug the biological role of the miRNAs and lncRNAs in a subnetwork and their impact on TME. RESULTS Altogether, 5 miRNAs (hsa-mir-125b-2, hsa-mir-135a-1, hsa-mir92a-2, hsa-mir-181a-2, and hsa-mir-214), 3 lncRNAs (LINC00641, LINC01146, and LINC00892), and 10 mRNAs (AGO2, CXCL10, CD86, CASP1, IKZF1, CD27, CD247, CD69, CCR2, and CSF2RB) in the subnetwork were identified as vital regulators to shape the TME. On the basis of the systematic network, we identified that trichostatin A, a pan-HDAC inhibitor, could potentially regulate the TME of sarcoma, thereby inhibiting the tumor growth. CONCLUSIONS Our study identifies a ceRNA network as a promising biomarker for sarcoma. This system provides a more comprehensive understanding and a novel perspective of how ceRNAs are involved in shaping sarcoma TME.
Collapse
Affiliation(s)
- Dongliang Leng
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Ziyi Yang
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Heng Sun
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Chengcheng Song
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chen Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau, SAR, China
- Stat Key laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China
| | - Ka U. Ip
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Guokai Chen
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, SAR, China
| | - Chu-Xia Deng
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, SAR, China
| | - Xiaohua Douglas Zhang
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, Kentucky
| | - Qi Zhao
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, SAR, China
| |
Collapse
|
23
|
Xie L, Liang X, Xu J, Sun X, Liu K, Sun K, Li Y, Tang X, Li X, Zhan X, Niu X, Guo W. Exploratory study of an anti-PD-L1/TGF-β antibody, TQB2858, in patients with refractory or recurrent osteosarcoma and alveolar soft part sarcoma: a report from Chinese sarcoma study group (TQB2858-Ib-02). BMC Cancer 2023; 23:868. [PMID: 37715133 PMCID: PMC10503089 DOI: 10.1186/s12885-023-11390-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Novel and effective immunotherapies are required for refractory or recurrent sarcomas. Transforming growth factor-beta (TGF-β) is a diverse regulatory and fibrogenic protein expressed in multiple sarcoma tumors that promotes epithelial-mesenchymal transition and excessive deposition of extracellular matrix. This study evaluated the efficacy and safety of the anti-PD-L1/TGF-β antibody TQB2858 in patients with refractory osteosarcoma and alveolar soft part sarcoma (ASPS). METHODS This single-arm phase 1b exploratory study included patients with refractory osteosarcoma or ASPS who had previously undergone at least two lines of systemic therapy. Patients were administered 1200 mg of TQB2858 once every 3 weeks. The primary endpoint was objective response rate (ORR), with null and alternative hypotheses of ORR ≤5% and ≥20%, respectively. Exploratory biomarker analyses using immunohistochemistry (IHC) staining (for PD-L1 and TGF-β) were performed on pre-treatment tumor samples. RESULTS Eleven eligible patients were included in this study. TQB2858 did not demonstrate evidence of efficacy as 0/5 osteosarcomas had any objective response, while 2/6 ASPS showed a partial response. The median progression-free survivals were 1.51 (1.38, Not Evaluable) and 2.86 (1.38, Not Evaluable) months for the osteosarcoma and ASPS groups, respectively. None of the administered cycles met the criteria for unacceptable toxicity. Other Grade 3 toxicities included abnormal liver function and elevation of γ-glutamyl transferase. IHC analysis revealed that functional enrichment in the TGF-β pathway or PD-L1 was not associated with treatment outcomes. CONCLUSIONS The combination of PD-L1 and TQB2858 did not significantly improve the ORR in patients with recurrent osteosarcoma. However, it improved immunogenic responses in ASPS, even after progression upon anti-PD-1/PD-L1 therapy, with an acceptable safety profile. IHC profiling with pathway enrichment analysis may not have any predictive value for survival outcomes. TRIAL REGISTRATION Prospectively registered in the Ethical Review Committee of Peking University People's Hospital. The trial registration number is 2021PHA105-001 and 2021PHA140-001 and the registration date was March 2, 2022. CLINICALTRIALS gov Identifier CTR20213001 and CTR20220390.
Collapse
Affiliation(s)
- Lu Xie
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China
| | - Xin Liang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China
| | - Jie Xu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China
| | - Xin Sun
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China
| | - Kuisheng Liu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China
| | - Kunkun Sun
- Pathology Department, Peking University People's Hospital, Beijing, 100044, China
| | - Yuan Li
- Radiology Department and Nuclear Medicine Department, Peking University People's Hospital, Beijing, 100044, China
| | - Xiaodong Tang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China.
| | - Xianan Li
- Orthopedic Oncology Department, Hunan Cancer Hospital, Changsha, 410013, China
| | - Xing Zhan
- Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiaohui Niu
- Orthopedic Oncology Department, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
24
|
Kohlmeyer JL, Lingo JJ, Kaemmer CA, Scherer A, Warrier A, Voigt E, Garay JAR, McGivney GR, Brockman QR, Tang A, Calizo A, Pollard K, Zhang X, Hirbe AC, Pratilas CA, Leidinger M, Breheny P, Chimenti MS, Sieren JC, Monga V, Tanas MR, Meyerholz DK, Darbro BW, Dodd RD, Quelle DE. CDK4/6-MEK Inhibition in MPNSTs Causes Plasma Cell Infiltration, Sensitization to PD-L1 Blockade, and Tumor Regression. Clin Cancer Res 2023; 29:3484-3497. [PMID: 37410426 PMCID: PMC10528807 DOI: 10.1158/1078-0432.ccr-23-0749] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/22/2023] [Accepted: 07/03/2023] [Indexed: 07/07/2023]
Abstract
PURPOSE Malignant peripheral nerve sheath tumors (MPNST) are lethal, Ras-driven sarcomas that lack effective therapies. We investigated effects of targeting cyclin-dependent kinases 4 and 6 (CDK4/6), MEK, and/or programmed death-ligand 1 (PD-L1) in preclinical MPNST models. EXPERIMENTAL DESIGN Patient-matched MPNSTs and precursor lesions were examined by FISH, RNA sequencing, IHC, and Connectivity-Map analyses. Antitumor activity of CDK4/6 and MEK inhibitors was measured in MPNST cell lines, patient-derived xenografts (PDX), and de novo mouse MPNSTs, with the latter used to determine anti-PD-L1 response. RESULTS Patient tumor analyses identified CDK4/6 and MEK as actionable targets for MPNST therapy. Low-dose combinations of CDK4/6 and MEK inhibitors synergistically reactivated the retinoblastoma (RB1) tumor suppressor, induced cell death, and decreased clonogenic survival of MPNST cells. In immune-deficient mice, dual CDK4/6-MEK inhibition slowed tumor growth in 4 of 5 MPNST PDXs. In immunocompetent mice, combination therapy of de novo MPNSTs caused tumor regression, delayed resistant tumor outgrowth, and improved survival relative to monotherapies. Drug-sensitive tumors that regressed contained plasma cells and increased cytotoxic T cells, whereas drug-resistant tumors adopted an immunosuppressive microenvironment with elevated MHC II-low macrophages and increased tumor cell PD-L1 expression. Excitingly, CDK4/6-MEK inhibition sensitized MPNSTs to anti-PD-L1 immune checkpoint blockade (ICB) with some mice showing complete tumor regression. CONCLUSIONS CDK4/6-MEK inhibition induces a novel plasma cell-associated immune response and extended antitumor activity in MPNSTs, which dramatically enhances anti-PD-L1 therapy. These preclinical findings provide strong rationale for clinical translation of CDK4/6-MEK-ICB targeted therapies in MPNST as they may yield sustained antitumor responses and improved patient outcomes.
Collapse
Affiliation(s)
- Jordan L Kohlmeyer
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Joshua J Lingo
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Courtney A Kaemmer
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Amanda Scherer
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Akshaya Warrier
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Ellen Voigt
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | | | - Gavin R McGivney
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
| | - Qierra R Brockman
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Amy Tang
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Center, Eastern Virginia Medical School, Norfolk, Virginia
| | - Ana Calizo
- Department of Oncology, Johns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Kai Pollard
- Department of Oncology, Johns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Xiaochun Zhang
- Division of Medical Oncology, Washington University, St. Louis, Missouri
| | - Angela C Hirbe
- Division of Medical Oncology, Washington University, St. Louis, Missouri
| | - Christine A Pratilas
- Department of Oncology, Johns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Mariah Leidinger
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Patrick Breheny
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa
| | - Michael S Chimenti
- Iowa Institute of Human Genetics, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Jessica C. Sieren
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Radiation, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Varun Monga
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Munir R Tanas
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - David K Meyerholz
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Benjamin W Darbro
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Rebecca D Dodd
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Dawn E Quelle
- Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
25
|
Wang X, Jin Z, Tang S, Huang X, Wang S, Ren X, Xu M. ATRX loss suppresses the type I interferon response in sarcoma cells through chromatin remodeling. Am J Cancer Res 2023; 13:3547-3558. [PMID: 37693131 PMCID: PMC10492102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/30/2023] [Indexed: 09/12/2023] Open
Abstract
Sarcomas constitute a heterogeneous group of mesenchymal cancers and are particularly common in children and adolescents, leading to significant lethality. Therefore, it is necessary to understand the underlying mechanisms by which genetic alterations promote sarcoma progression. Here, we demonstrate that loss-of-function of ATRX, a member of the SWI/SNF DNA-remodeling family, represses the interferon (IFN)-β response by inducing chromatin remodeling in sarcoma cells. We show that ATRX mutations are associated with worse prognosis and attenuate IFN-α/β response in patients with specific types of sarcomas. Using poly(I:C) as a stimulation model, we show that natural ATRX mutation or ATRX depletion via CRISPR/Cas9 or siRNA significantly suppresses the expression of IFNB1 and other cytokines in sarcoma cells. Moreover, RNA-seq data reveal that ATRX ablation globally influences the expression pattern of poly(I:C)-stimulated genes (PSGs). Through ATAC-seq, we show that ATRX loss enhance chromatin accessibility generally, which consistent with the heterochromatin modulating function of ATRX. However, a set of PSGs display a decrease of chromatin accessibility after ATRX depletion, indicating that ATRX promote the transcription of these genes through chromatin remodeling. Thus, we highlight that ATRX mutation plays critical roles in blocking Type I IFN signaling in sarcoma cells and point out the clinical importance of this effect on sarcoma treatment.
Collapse
Affiliation(s)
- Xinrui Wang
- Department of Cell Biology, School of Life Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Zige Jin
- Department of Cell Biology, School of Life Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Shanshan Tang
- Department of Cell Biology, School of Life Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Xiyu Huang
- Department of Cell Biology, School of Life Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Shanshan Wang
- Department of Cell Biology, School of Life Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Xiaohe Ren
- Department of Cell Biology, School of Life Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Mafei Xu
- Department of Cell Biology, School of Life Sciences, Anhui Medical University Hefei 230032, Anhui, China
| |
Collapse
|
26
|
Nery B, de Alencar Neto JF, Melo LRDS, Costa RAF, Quaggio E, de Medeiros LS, de Sousa Segundo JA, de Lima NF, Rivero RL. Olfactory groove monophasic sinovial sarcoma and von Recklinghausen's disease: A case report and literature review. Surg Neurol Int 2023; 14:231. [PMID: 37560581 PMCID: PMC10408634 DOI: 10.25259/sni_338_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/13/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Soft-tissue sarcomas are a rare and diverse group of neoplastic lesions. They represent only 1% of malignant tumors in adults and 15% in children. Synovial sarcoma (SS) is a type of soft-tissue sarcoma, accounting for 5-10% of cases, and commonly affecting extremities. Diagnosis, treatment, and prognosis remain challenging especially when localized in uncommon areas, such as intracranial lesions. CASE DESCRIPTION A 13-year-old male patient with a clinical history of neurofibromatosis Type I (NF1) presenting holocranial headache with jet vomiting and apathy 2 days before admission, without neurological deficits and/or focal findings. On magnetic resonance imaging: an extra-axial infiltrative lesion with contrast uptake at the base of the skull in the olfactory groove topography. After total tumor resection, the anatomopathological examination showed monophasic SS. The patient returned after 6 months with similar symptoms, and the lesion recurred and was reoperated. Unfortunately, 7 months after the second surgery, the patient died. CONCLUSION SS can occur extraarticulously and with a variable clinical presentation and poor prognosis despite adjuvant therapies with radiotherapy and chemotherapy. In individuals with clinical history of NF1, there is still no direct correlation between the two manifestations, although current descriptions are suggestive of a possible interaction.
Collapse
|
27
|
Chi L, Wang H, Yu F, Gao C, Dai H, Si X, Dong Y, Liu H, Zhang Q. Design, synthesis and biological evaluation of nitric oxide-releasing 5-cyano-6-phenyl-2, 4-disubstituted pyrimidine derivatives. Bioorg Med Chem Lett 2023:129389. [PMID: 37379957 DOI: 10.1016/j.bmcl.2023.129389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/09/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023]
Abstract
In this study, a series of nitric oxide (NO) -releasing 5-cyano-6-phenyl-2, 4-disubstituted pyrimidine derivatives were designed and synthesized. In the in vitro biological evaluation, compound 24l exhibited optimal antiproliferative activity against MGC-803 cells with the IC50 value of 0.95 µM, significantly better than that of the positive control 5-FU. In addition, preliminary mechanistic studies indicated that 24l inhibited colony formation and blocked MGC-803 cells in the G0/G1 phase. DAPI staining, reactive oxygen species and apoptosis assays demonstrated that 24l induced apoptosis of MGC-803 cells. Particularly, the most potent compound 24l produced the highest level of NO, and the antiproliferative activity was significantly reduced after preincubation with NO scavengers. In conclusion, compound 24l may be considered as a potential candidate antitumor agent.
Collapse
Affiliation(s)
- Lingling Chi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Institute of Drug Discovery and Development, Zhengzhou 450001, China
| | - Hao Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Institute of Drug Discovery and Development, Zhengzhou 450001, China
| | - Fuqiang Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Institute of Drug Discovery and Development, Zhengzhou 450001, China
| | - Chao Gao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Institute of Drug Discovery and Development, Zhengzhou 450001, China
| | - Honglin Dai
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Institute of Drug Discovery and Development, Zhengzhou 450001, China
| | - Xiaojie Si
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Institute of Drug Discovery and Development, Zhengzhou 450001, China
| | - Yuze Dong
- Institute of Drug Discovery and Development, Zhengzhou 450001, China; Center for Drug Safety Evaluation and Research, Zhengzhou 450001, China
| | - Hongmin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Institute of Drug Discovery and Development, Zhengzhou 450001, China; Center for Drug Safety Evaluation and Research, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450052, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou 450001, China
| | - Qiurong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Institute of Drug Discovery and Development, Zhengzhou 450001, China; Center for Drug Safety Evaluation and Research, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450052, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou 450001, China
| |
Collapse
|
28
|
Fei S, Lu Y, Chen J, Qi J, Wu W, Wang B, Han Y, Wang K, Han X, Zhou H, Wang J, Chen J. Efficacy of PD-1 Inhibitors in First-Line Treatment for Advanced Gastroesophageal Junction and Gastric Cancer by Subgroups: A Systematic Review and Meta-Analysis. Chemotherapy 2023; 68:197-209. [PMID: 37331333 DOI: 10.1159/000531457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/04/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND PD-1 inhibitors have been approved for the first-line treatment of patients with advanced gastric cancer, gastroesophageal junction cancer, or esophageal adenocarcinoma. However, the results of several clinical trials are not entirely consistent, and the dominant population of first-line immunotherapy for advanced gastric/gastroesophageal junction cancer still needs to be precisely determined. OBJECTIVE This objective of this study is to evaluate the efficacy of anti-PD-1/PD-L1 therapy in advanced gastric/gastroesophageal junction adenocarcinoma patients through a systematic review and meta-analysis of relevant clinical trials. METHOD The PubMed, Embase, and Cochrane Library electronic databases were searched up to August 1, 2022, for clinical trials of anti-PD-1/PD-L1 immunotherapy for the first-line treatment of advanced gastroesophageal cancer. Hazard ratios and 95% confidence intervals for overall survival, progression-free survival, and objective response rates were extracted and pooled for meta-analysis. Prespecified subgroups included the following: agent type, PD-L1 expression, and high microsatellite instability. RESULTS This study analyzed 5 RCTs involving 3,355 patients. Compared with the chemotherapy group, the combined immunotherapy group had a significantly higher objective response rate (OR = 0.63, 95% CI: 0.55-0.72, p < 0.00001) and prolonged overall survival (HR = 0.82, 95% CI: 0.76-0.88, p < 0.00001) and progression-free survival (HR = 0.75, 95% CI: 0.69-0.82, p < 0.00001). The combination of immunotherapy and chemotherapy prolonged OS in both MSI-H (HR = 0.38, p = 0.002) and MSS (HR = 0.78, p < 0.00001) populations, but there was a significant difference between groups (p = 0.02). However, in improving ORR, the benefit of ICI combined with chemotherapy in the MSS group and MSI-H group was not significantly different between groups (p = 0.52). Combination therapy with ICIs was more effective than chemotherapy alone in prolonging OS in the subgroup with a high CPS, regardless of the CPS cutoff for PD-L1. However, when the cutoff of CPS was 1, the difference between subgroups did not reach statistical significance (p = 0.12), while the benefit ratio of the MSI-H group was higher when the cutoff was 10 (p = 0.004) than when the cutoff value was 5 (p = 0.002). CONCLUSIONS For first-line treatment of advanced gastroesophageal cancer, an ICI combination strategy is more effective than chemotherapy. The subgroup of patients with a CPS ≥10 has a more significant benefit, and CPS ≥10 has the potential to be used as an accurate marker of the dominant population of immuno-combined therapy.
Collapse
Affiliation(s)
- Shengqi Fei
- Department of Gastroenterology Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Gastrointestinal Surgery, Changxing People's Hospital, Huzhou, China
| | - Yu Lu
- Nursing Department, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Chen
- Department of Gastroenterology Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Qi
- Department of Gastroenterology Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenxuan Wu
- Department of Gastroenterology Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Beidi Wang
- Department of Gastroenterology Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaxuan Han
- Department of Gastroenterology Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kefan Wang
- Department of Gastroenterology Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaying Han
- Department of Gastroenterology Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiyan Zhou
- Nursing Department, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Wang
- Department of Gastroenterology Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Chen
- Department of Gastroenterology Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
29
|
DePalo DK, Zager JS. Isolated Limb Infusion for Limb-Threatening, Unresectable Sarcoma: Past Progress, Current Applications, and Future Directions. J Clin Med 2023; 12:4036. [PMID: 37373729 DOI: 10.3390/jcm12124036] [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: 04/20/2023] [Revised: 05/24/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Treatment of soft tissue sarcomas (STSs) is complicated by disease heterogeneity. Further, it has not benefitted much from the recent therapeutic advances in other soft tissue malignancies. Surgical resection remains the gold standard in resectable disease, but unresectable, locally advanced STS requires alternative and multimodal approaches. Isolated limb infusion (ILI) provides regional chemotherapy to extremity STS and offers the potential for limb salvage. Despite being in use for nearly 3 decades, there is limited literature on ILI in STS. This review provides an overview of patient eligibility, the procedure, significant publications in this field, and opportunities for further progress.
Collapse
Affiliation(s)
- Danielle K DePalo
- Department of Cutaneous Oncology, Moffitt Canter, Tampa, FL 33612, USA
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Canter, Tampa, FL 33612, USA
| |
Collapse
|
30
|
Zhou MY, Bui NQ, Charville GW, Ganjoo KN, Pan M. Treatment of De-Differentiated Liposarcoma in the Era of Immunotherapy. Int J Mol Sci 2023; 24:ijms24119571. [PMID: 37298520 DOI: 10.3390/ijms24119571] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Well-differentiated/de-differentiated liposarcoma (WDLPS/DDLPS) is one of the most common histologic subtypes of soft tissue sarcoma (STS); however, treatment options remain limited. WDLPS and DDLPS both exhibit the characteristic amplification of chromosome region 12q13-15, which contains the genes CDK4 and MDM2. DDLPS exhibits higher amplification ratios of these two and carries additional genomic lesions, including the amplification of chromosome region 1p32 and chromosome region 6q23, which may explain the more aggressive biology of DDLPS. WDLPS does not respond to systemic chemotherapy and is primarily managed with local therapy, including multiple resections and debulking procedures whenever clinically feasible. In contrast, DDLPS can respond to chemotherapy drugs and drug combinations, including doxorubicin (or doxorubicin in combination with ifosfamide), gemcitabine (or gemcitabine in combination with docetaxel), trabectedin, eribulin, and pazopanib. However, the response rate is generally low, and the response duration is usually short. This review highlights the clinical trials with developmental therapeutics that have been completed or are ongoing, including CDK4/6 inhibitors, MDM2 inhibitors, and immune checkpoint inhibitors. This review will also discuss the current landscape in assessing biomarkers for identifying tumors sensitive to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Maggie Y Zhou
- Sarcoma Program, Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Nam Q Bui
- Sarcoma Program, Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Gregory W Charville
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Kristen N Ganjoo
- Sarcoma Program, Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Minggui Pan
- Sarcoma Program, Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| |
Collapse
|
31
|
Li J, Gao F, Su J, Pan T. Bioinformatics identification and validation of aging‑related molecular subtype and prognostic signature in breast cancer. Medicine (Baltimore) 2023; 102:e33605. [PMID: 37171324 PMCID: PMC10174399 DOI: 10.1097/md.0000000000033605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/01/2023] [Accepted: 04/03/2023] [Indexed: 05/13/2023] Open
Abstract
Patients with metastatic breast cancer have a poor clinical outcome, accounting for more than 90 percent of breast cancer-related deaths. Aging could regulate many biological processes in malignancies by regulating cell senescence. The role of aging has not been fully clarified. Consensus cluster analysis was performed to differentiate The Cancer Genome Atlas (TCGA) breast cancer cases. Least absolute shrinkage and selection operator (LASSO) cox regression analysis was performed to construct an aging-related prognostic signature. A total of 118 differentially expressed aging-related genes (ARGs) was obtained in breast cancer. Consensus clustering analysis identified 3 categories of TCGA-breast cancer with significant difference in prognosis and immune infiltration. We also constructed an aging-related prognostic signature for breast cancer, which had a good performance in predicting the 1-year, 3-year and 5-year OS and disease specific survival (DSS) of breast cancer patients. Further single gene analysis revealed that the expression of PIK3R1 was significantly different in different pT and pN stages of breast cancer. Moreover, low expression of PIK3R1 showed resistance to many drugs based on the data of Genomics of Drug Sensitivity in Cancer (GDSC) and Genomics of Therapeutics Response Portal (CTRP). PIK3R1 played a vital role in many well-known cancer-related pathways. The current study identified 3 clusters of TCGA-breast cancer cases with significant differences in prognosis and immune infiltration. We also constructed an aging-related prognostic signature for breast cancer. However, further in vivo and in vitro studies should be conducted to verify these results.
Collapse
Affiliation(s)
- Jingtai Li
- Department of Breast surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Fangfang Gao
- Department of Breast surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jiezhi Su
- Department of Breast surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Tao Pan
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
32
|
Hong X, Liu H, Chen C, Lai T, Lin J. Bioinformatics identification and validation of aging-related molecular subtype and prognostic signature in sarcoma. Cancer Invest 2023:1-12. [PMID: 37130077 DOI: 10.1080/07357907.2023.2209638] [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: 05/03/2023]
Abstract
Aging could regulate many biological processes in malignancies by regulating cell senescence. Consensus cluster analysis was conducted to differentiate TCGA sarcoma cases. LASSO cox regression analysis was performed to construct an aging-related prognostic signature. We identified two categories of TCGA-sarcoma with significant difference in prognosis, immune infiltration and chemotherapy and targeted therapy. Moreover, an aging-related prognostic signature was constructed for sarcoma, which had a good performance in predicting the 3-year and 5-year overall survival of sarcoma patients. We also identified a lncRNA MALAT1/miR-508-3p/CCNA2 regulatory axis for sarcoma. This stratification could provide more evidence for estimating prognosis and immunotherapy of sarcoma.
Collapse
Affiliation(s)
- Xu Hong
- Department of orthopedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou 350007, Fujian, China
| | - Hui Liu
- Department of orthopedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou 350007, Fujian, China
| | - Chu Chen
- Department of orthopedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou 350007, Fujian, China
| | - Tian Lai
- Department of orthopedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou 350007, Fujian, China
| | - Jingui Lin
- Department of orthopedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou 350007, Fujian, China
| |
Collapse
|
33
|
Gambichler T, Horny K, Mentzel T, Stricker I, Tannapfel A, Scheel CH, Behle B, Quast DR, Lee YP, Stücker M, Susok L, Becker JC. Undifferentiated pleomorphic sarcoma of the breast with neoplastic fever: case report and genomic characterization. J Cancer Res Clin Oncol 2023; 149:1465-1471. [PMID: 35501497 PMCID: PMC10020307 DOI: 10.1007/s00432-022-04000-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 03/26/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Primary breast sarcomas are extraordinary rare, in particular undifferentiated pleomorphic sarcoma (UPS). UPS with neoplastic fever (UPS-NF) of the breast has not been reported yet. Here, we present an extended UPS-NF of the breast including its comprehensive molecular workup. METHODS A 58-year-old female presented with general malaise, fever spikes, weight loss, and a massively swollen left breast. C-reactive protein and blood leucocytes were significantly increased. However, repeated blood cultures and smears were all sterile. Histopathology of the abscess-forming tumor revealed an undifferentiated malignancy with numerous of tumor giant cells as well as spindle-shaped cells with nuclear pleomorphism and hyperchromasia. Immunohistochemistry demonstrated partial, patchy desmin staining and weak heterogonous neuron-specific enolase immunoreactivity of tumor cells, but a focal staining for Melan-A. RESULTS Neither common melanoma driver mutations nor an ultraviolet mutational signature was detected by whole genome sequencing. Using FISH and RT-PCR we also excluded translocations characteristic for clear cell sarcoma. Thus, the diagnosis of inflammatory UPS-NF of the breast was considered highly probable. Despite a complete mastectomy, the tumor recurred after only three months. This recurrence was treated with a combination of ipilimumab and nivolumab based on the primary tumor's TPS score for PD-L1 of 30%. After an initial response, however, the tumor was progressive again. CONCLUSION We describe here the first case of UPS-NF of the breast, which shows great clinical and histopathologic resemblances to previously reported UPS-NF of other anatomic localizations.
Collapse
Affiliation(s)
- Thilo Gambichler
- Skin Cancer Center, Department of Dermatology, Ruhr-University Bochum, Bochum, Germany.
- Department of Dermatology, Christian Hospital Unna, Unna, Germany.
| | - Kai Horny
- Translational Skin Cancer Research, DKTK Partner Site Essen/Düsseldorf, West German Cancer Center, Dermatology, University Duisburg-Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Ingo Stricker
- Institute of Pathology, Ruhr-University Bochum, Bochum, Germany
| | | | - Christina H Scheel
- Skin Cancer Center, Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - Bertold Behle
- Skin Cancer Center, Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - Daniel R Quast
- Diabetes Division, Department of Internal Medicine, Ruhr-University Bochum, Bochum, Germany
| | - Yi-Pei Lee
- Skin Cancer Center, Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - Markus Stücker
- Skin Cancer Center, Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - Laura Susok
- Skin Cancer Center, Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - Jürgen C Becker
- Translational Skin Cancer Research, DKTK Partner Site Essen/Düsseldorf, West German Cancer Center, Dermatology, University Duisburg-Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
34
|
Fuchs JW, Schulte BC, Fuchs JR, Agulnik M. Targeted therapies for the treatment of soft tissue sarcoma. Front Oncol 2023; 13:1122508. [PMID: 36969064 PMCID: PMC10034045 DOI: 10.3389/fonc.2023.1122508] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
Soft tissue sarcomas are rare malignant tumors derived from mesenchymal cells that have a high morbidity and mortality related to frequent occurrence of advanced and metastatic disease. Over the past two decades there have been significant advances in the use of targeted therapies for the treatment of soft tissue sarcoma. The ability to study various cellular markers and pathways related to sarcomagenesis has led to the creation and approval of multiple novel therapies. Herein, we describe the current landscape of targeted medications used in the management of advanced or metastatic soft tissue sarcomas, excluding GIST. We distinguish three categories: targeted therapies that have current US Food and Drug Administration (FDA) approval for treatment of soft tissue sarcoma, non-FDA approved targeted therapies, and medications in development for treatment of patients with soft tissue sarcoma.
Collapse
Affiliation(s)
- Jeffrey W. Fuchs
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, United States
| | - Brian C. Schulte
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Joseph R. Fuchs
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, United States
| | - Mark Agulnik
- Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
- *Correspondence: Mark Agulnik,
| |
Collapse
|
35
|
Pehlivan M, İribaş A, Bilgiç B, Başaran M, Ekenel M. Clinical course and features of soft tissue sarcomas in geriatric patients: a single-center experience. J Int Med Res 2023; 51:3000605231159319. [PMID: 36879483 PMCID: PMC9996734 DOI: 10.1177/03000605231159319] [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: 03/08/2023] Open
Abstract
OBJECTIVE This study was performed to examine the disease course in geriatric patients with soft tissue sarcoma and determine the risk factors for mortality. METHODS We retrospectively analyzed patients who were treated at Istanbul University Oncology Institute from January 2000 to August 2021. RESULTS Eighty patients were included in the study. The patients' median age was 69 years (range, 65-88 years). The median overall survival of patients diagnosed between the ages of 65 and 74 years was 70 months, and that of patients diagnosed at the age of ≤75 years was significantly lower at 46 months. The median survival of patients who did and did not undergo surgical resection was 66 and 11 months, respectively, with a significant difference. The median overall survival of patients with positive and negative surgical margins was 58 and 96 months, respectively, also with a significant difference. Age at diagnosis and recurrence/metastasis significantly affected mortality. A 1-year increase in the age at diagnosis increased mortality by 1.147 times. CONCLUSION Age of >75 years, inability to undergo surgery, positive surgical margins, and head and neck location may be associated with a poor prognosis in geriatric patients with soft tissue sarcoma.
Collapse
Affiliation(s)
- Metin Pehlivan
- Department of Medical Oncology, Zonguldak Ataturk State Hospital
| | - Ayça İribaş
- Department of Radiation Oncology, Istanbul University Oncology Institute
| | - Bilge Bilgiç
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University
| | - Mert Başaran
- Department of Medical Oncology, Istanbul University Oncology Institute
| | - Meltem Ekenel
- Department of Medical Oncology, Istanbul University Oncology Institute
| |
Collapse
|
36
|
Association of SMC4 with prognosis and immune infiltration of sarcoma. Aging (Albany NY) 2023; 15:567-582. [PMID: 36719264 PMCID: PMC9925680 DOI: 10.18632/aging.204503] [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: 08/13/2022] [Accepted: 01/23/2023] [Indexed: 02/01/2023]
Abstract
OBJECTIVE This study was performed to explore the prognostic relevance of structural maintenance of chromosomes 4 (SMC4) in pan-cancer and explore the association between SMC4 and immune infiltration of sarcoma. RESULTS Elevated expression of SMC4 was detected in cancer tissues compared to normal tissue, which was confirmed in synovial sarcoma tissues with immunohistochemistry (IHC). Additionally, higher expression of SMC4 was connected to worse outcomes of sarcoma, gastric cancer, breast cancer, liver cancer or ovarian cancer. Moreover, SMC4 was positively connected to immune cell infiltrates in sarcoma. In addition, infiltrating immune cell markers including monocyte, TAM, M1 and M2 presented different SMC4-associated immune infiltration patterns. CONCLUSION The results from our study showed that SMC4 was positively related to the prognosis and immunological status of sarcoma. SMC4 could be a potential biomarker for prognosis and immune cell infiltrates in sarcoma. METHODS Several databases including ONCOMINE, GEPIA, and Kaplan-Meier Plotter were adopted to explore the expression pattern of SMC4 in sarcoma, which was confirmed by IHC. The GEPIA and TIMER datasets were adopted to investigate the associations between SMC4 and prognosis in various cancers, especially in sarcoma.
Collapse
|
37
|
A Combined Risk Score Model to Assess Prognostic Value in Patients with Soft Tissue Sarcomas. Cells 2022; 11:cells11244077. [PMID: 36552841 PMCID: PMC9776565 DOI: 10.3390/cells11244077] [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: 10/15/2022] [Revised: 11/13/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
A study by Tsvetkov et al. recently published a proposed novel form of copper-induced cell death in Science; however, few studies have looked into the possible mechanism in soft tissue sarcoma (STS). Herein, this study sought to investigate the function of cuproptosis-related genes (CRGs) in the development of tumor-associated immune cells and the prognosis of sarcoma. Herein, this study aimed to explore the role of cuproptosis-related genes (CRGs) in the development, tumor-associated immune cells, and the prognosis of sarcoma. METHODS The prognostic model was established via the least absolute shrinkage and selection operator (LASSO) algorithm as well as multivariate Cox regression analysis. The stromal scores, immune scores, ESTIMA scores, and tumor purity of sarcoma patients were evaluated by the ESTIMATE algorithm. Functional analyses were performed to investigate the underlying mechanisms of immune cell infiltration and the prognosis of CRGs in sarcoma. RESULTS Two molecular subgroups with different CRG expression patterns were recognized, which showed that patients with a higher immune score and more active immune status were prone to have better prognostic survival. Moreover, GO and KEGG analyses showed that these differentially expressed CRGs were mainly enriched in metabolic/ions-related signaling pathways, indicating that CRGs may have impacts on the immune cell infiltration and prognosis of sarcoma via regulating the bioprocess of mitochondria and consequently affecting the immune microenvironment. The expression levels of CRGs were closely correlated to the immunity condition and prognostic survival of sarcoma patients. CONCLUSIONS The interaction between cuproptosis and immunity in sarcoma may provide a novel insight into the study of molecular mechanisms and candidate biomarkers for the prognosis, resulting in effective treatments for sarcoma patients.
Collapse
|
38
|
Lynch MM, Alexiev BA, Schroeder BA, Pollack SM. Combinations of Chemotherapy and PD-1/PD-L1 Inhibitors in Sarcoma. Curr Treat Options Oncol 2022; 23:1861-1876. [PMID: 36380108 DOI: 10.1007/s11864-022-01036-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2022] [Indexed: 11/17/2022]
Abstract
OPINION STATEMENT While no PD-1 inhibitor has been FDA approved for use in sarcoma or proven efficacious in a randomized trial, the use of single agent PD-1 inhibitors is standard-of-care and recommended by the NCCN guidelines in certain specific subtypes and situations. Even while the role of immunotherapy is still being defined in sarcoma, there is rising interest in combinations of PD-1 inhibitors with standard-of-care treatments, especially chemotherapy. Recently, several early phase trials have suggested potential benefits for chemotherapy in combination with PD-1 inhibitors. Although some physicians are already combining PD-1 inhibitors and chemotherapy for sarcoma off-label in the community, we believe more data is necessary. We support further evaluation of these combinations in well-designed clinical trials.
Collapse
Affiliation(s)
- Meghan M Lynch
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Borislav A Alexiev
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Brett A Schroeder
- Department of Hematology and Medical Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Seth M Pollack
- Department of Medicine (Hematology and Oncology), Northwestern University Feinberg School of Medicine, 303 E. Superior St. #3-115, Chicago, IL, 60611, USA.
| |
Collapse
|
39
|
Li Z, Zheng H, Liu L, Fen Z, Cao H, Yang J, Wei J. A novel inflammatory signature for evaluating immune microenvironment status in soft tissue sarcoma. Front Oncol 2022; 12:990670. [PMID: 36313634 PMCID: PMC9609423 DOI: 10.3389/fonc.2022.990670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/26/2022] [Indexed: 12/03/2022] Open
Abstract
Background Tumorigenesis and progression are intimately associated with inflammation. However, the inflammatory landscape in soft tissue sarcoma (STS) and its clinical consequences are yet unknown, and more investigation is needed. Methods RNA-seq expression data for STS and corresponding normal tissues were downloaded from The Cancer Genome Atlas database and the Genotype-Tissue Expression Portal. Differential and prognostic analyses were performed based on known inflammatory response genes from Gene Set Enrichment Analysis (GSEA). We utilized LASSO-Cox analysis to determine hub genes and built an inflammatory score (INFscore) and risk stratification model. Furthermore, a nomogram, including the risk stratification model, was established to predict the prognosis. We further elucidated the characteristics among different risk STS patients by GSEA, gene set variation analysis, and detailed immune infiltration analysis. Finally, the INFscore and risk stratification model in predicting prognosis and depicting immune microenvironment status were verified by pan-cancer analysis. Results Five hub genes (HAS2, IL1R1, NMI, SERPINE1, and TACR1) were identified and were used to develop the INFscore. The risk stratification model distinguished the immune microenvironment status and evaluated the efficacy of immunotherapy and chemotherapy in STS. The novel nomogram had good efficacy in predicting the prognosis of STS patients. Finally, a pan-cancer investigation verified the association of INFscore with prognosis and immunity. Conclusions According to the present study, the risk stratification model can be used to evaluate STS prognosis, tumor microenvironment status, immunotherapy, and chemotherapy efficacy. The novel nomogram has an excellent predictive value. Thus, the INFscore and risk stratification model has potential value in assessing the prognosis and immune status of multiple malignancies.
Collapse
Affiliation(s)
- Zhehong Li
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde, China
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Honghong Zheng
- Department of General Surgery, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Lirui Liu
- Department of Neonatal Department, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Zhen Fen
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Haiying Cao
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Jilong Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- *Correspondence: Jilong Yang, ; Junqiang Wei,
| | - Junqiang Wei
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical University, Chengde, China
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- *Correspondence: Jilong Yang, ; Junqiang Wei,
| |
Collapse
|
40
|
Boudin L, de Nonneville A, Finetti P, Mescam L, Le Cesne A, Italiano A, Blay JY, Birnbaum D, Mamessier E, Bertucci F. CSPG4 expression in soft tissue sarcomas is associated with poor prognosis and low cytotoxic immune response. Lab Invest 2022; 20:464. [PMID: 36221119 PMCID: PMC9552405 DOI: 10.1186/s12967-022-03679-y] [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: 08/06/2022] [Accepted: 09/29/2022] [Indexed: 11/10/2022]
Abstract
Background Soft tissue sarcomas (STS) are heterogeneous and pro-metastatic tumors. Identification of accurate prognostic factors and novel therapeutic targets are crucial. CSPG4 is a cell surface proteoglycan with oncogenic functions. It recently emerged as a potential target for immunotherapy, including cell therapy based on CSPG4-specific chimeric antigen receptor (CAR)-redirected cytokine-induced killer lymphocytes (CSPG4-CAR.CIKs) in STS. However, expression of CSPG4 is poorly known in STS so far. Methods We analyzed CSPG4 gene expression in 1378 localized STS clinical samples, and searched for correlations with clinicopathological data, including disease-free survival (DFS), and with tumor immune features. Results CSPG4 expression was heterogeneous across samples. High expression was associated with younger patients’ age, more frequent undifferentiated pleomorphic sarcoma and myxofibrosarcoma pathological subtypes, more frequent internal trunk tumor site, and more CINSARC high-risk samples. No correlation existed with pathological tumor size and grade, and tumor depth. Patients with high CSPG4 expression displayed 49% (95% CI 42–57) 5-year DFS versus 61% (95% CI 56–68) in patients with low expression (p = 3.17E−03), representing a 49% increased risk of event in the “CSPG4-high” group (HR = 1.49, 95% CI 1.14–1.94). This unfavorable prognostic value persisted in multivariate analysis, independently from other variables. There were significant differences in immune variables between “CSPG4-high” and “CSPG4-low” tumors. The "CSPG4-low" tumors displayed profiles suggesting higher anti-tumor cytotoxic immune response and higher potential vulnerability to immune checkpoint inhibitors (ICI). By contrast, the "CSPG4-high" tumors displayed profiles implying an immune-excluded tumor microenvironment, potentially induced by hypoxia, resulting from an immature chaotic microvasculature, and/or the presence of contractile myofibroblasts. Conclusions Patients with “CSPG4-high” STS, theoretically candidate for CAR.CIKs, display shorter DFS and an immune environment unfavorable to vulnerability to CAR.CIKs, which could be improved by combining anti-angiogenic drugs able to normalize the tumor vasculature. By contrast, “CSPG4-low” STS are better candidates for immune therapy involving ICI. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03679-y.
Collapse
Affiliation(s)
- Laurys Boudin
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - A de Nonneville
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France.,Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Pascal Finetti
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Léna Mescam
- French Sarcoma Group, Lyon, France.,Department of Pathology, Institut Paoli-Calmettes, 232 Bd. Sainte-Marguerite, 13009, Marseille, France
| | - A Le Cesne
- French Sarcoma Group, Lyon, France.,Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Antoine Italiano
- French Sarcoma Group, Lyon, France.,Department of Medical Oncology, Institut Bergonie, Bordeaux, France
| | - Jean-Yves Blay
- French Sarcoma Group, Lyon, France.,Department of Medical Oncology, Centre Léon Bérard, UNICANCER &, Université Claude Bernard Lyon I, Lyon, France
| | - Daniel Birnbaum
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Emilie Mamessier
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - François Bertucci
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France. .,Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France. .,French Sarcoma Group, Lyon, France.
| |
Collapse
|
41
|
Rometti M, Bryczkowski C. Thigh Mass Case Report. JOURNAL OF EDUCATION & TEACHING IN EMERGENCY MEDICINE 2022; 7:V7-V9. [PMID: 37465130 PMCID: PMC10332667 DOI: 10.21980/j8qd3c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 08/16/2022] [Indexed: 07/20/2023]
Abstract
Patients commonly present to the Emergency Department for the evaluation of soft tissues masses of various etiology. Point-of-care ultrasound (POCUS) can aid in the initial evaluation of these masses to begin narrowing a given differential. Soft tissue sarcomas are a malignant neoplasm that frequently present in an extremity, and require close follow-up for the evaluation of metastasis and possible resection, among other treatment options. Being able to effectively differentiate between infectious, inflammatory, benign, or potentially malignant pathology for undifferentiated soft tissue masses is critical for Emergency Medicine clinicians to ensure patients receive appropriate treatment and referrals for definitive care. Topics Thigh mass, soft tissue mass, sarcoma, point-of-care ultrasound.
Collapse
Affiliation(s)
- Mary Rometti
- Rutgers Robert Wood Johnson Medical School, Department of Emergency Medicine, New Brunswick, NJ
| | - Christopher Bryczkowski
- Rutgers Robert Wood Johnson Medical School, Department of Emergency Medicine, New Brunswick, NJ
| |
Collapse
|
42
|
MacNevin W, Rendon RA, Colwell B, Wang C, Hache KD, Merrimen J, Mason RJ. Primary Renal Synovial Sarcoma. JOURNAL OF UROLOGICAL SURGERY 2022. [DOI: 10.4274/jus.galenos.2022.2021.0094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
43
|
Fujiwara T, Nakata E, Kunisada T, Ozaki T, Kawai A. Alveolar soft part sarcoma: progress toward improvement in survival? A population-based study. BMC Cancer 2022; 22:891. [PMID: 35971085 PMCID: PMC9377116 DOI: 10.1186/s12885-022-09968-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 08/01/2022] [Indexed: 11/18/2022] Open
Abstract
Background Alveolar soft part sarcoma (ASPS) is a rare histological subtype of soft-tissue sarcoma, which remains refractory to conventional cytotoxic chemotherapy. We aimed to characterize ASPS and investigate whether the oncological outcome has improved over the past decade. Methods One hundred and twenty patients with newly diagnosed ASPS from 2006 to 2017, identified from the Bone and Soft-Tissue Tumor Registry in Japan, were analyzed retrospectively. Results The study cohort comprised 34 (28%) patients with localized ASPS and 86 (72%) with metastatic disease at presentation. The 5-year disease-specific survival (DSS) was 68% for all patients and 86% and 62% for localized and metastatic disease, respectively (p = 0.019). Metastasis at presentation was the only adverse prognostic factor for DSS (hazard ratio [HR]: 7.65; p = 0.048). Patients who were > 25 years (80%; p = 0.023), had deep-seated tumors (75%; p = 0.002), and tumors > 5 cm (5–10 cm, 81%; > 10 cm, 81%; p < 0.001) were more likely to have metastases at presentation. In patients with localized ASPS, adjuvant chemotherapy or radiotherapy did not affect survival, and 13 patients (45%) developed distant metastases in the lung (n = 12, 92%) and brain (n = 2, 15%). In patients with metastatic ASPS (lung, n = 85 [99%]; bone, n = 12 [14%]; and brain n = 9 [11%]), surgery for the primary or metastatic site did not affect survival. Prolonged survival was seen in patients who received pazopanib treatment (p = 0.045), but not in those who received doxorubicin-based cytotoxic chemotherapy. Overall, improved DSS for metastatic ASPS has been observed since 2012 (5-year DSS, from 58 to 65%) when pazopanib was approved for advanced diseases, although without a statistically significant difference (p = 0.117). Conclusion The national study confirmed a unique feature of ASPS with frequent metastasis to the lung and brain but an indolent clinical course. An overall trend toward prolonged survival after the introduction of targeted therapy encourages continuous efforts to develop novel therapeutic options for this therapeutically resistant soft-tissue sarcoma.
Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09968-5.
Collapse
Affiliation(s)
- Tomohiro Fujiwara
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, Japan.
| | - Eiji Nakata
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, Japan
| | - Toshiyuki Kunisada
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| |
Collapse
|
44
|
Abstract
Tumors of soft tissue origin are not common but are increasing in incidence. Given the rare and heterogeneous nature of the disease, deciding on an effective treatment approach to the patient can be challenging. Approximately 20-50% of patients with sarcoma will develop metastases to the lung via hematogenous spread. Despite improvements in systemic therapy options for patients with metastatic disease to the lung, surgical resection of metastases is often the preferred option in patients who are safe surgical candidates. Clearance of metastatic disease with surgical resection has been proven to be cost-effective and can improve chances for long term survival. Deciding on who may benefit from surgical resection is best achieved in a multidisciplinary setting.
Collapse
Affiliation(s)
- Mark Hennon
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; Department of Surgery, Jacobs School of Medicine, State University of New York at Buffalo, 100 High Street, Buffalo, NY 14203, USA.
| |
Collapse
|
45
|
Huang ZD, Fu YC, Liu SY, Mao YJ, Zhang Y, Hu C, Wei RX. m6A-related metabolism molecular classification with distinct prognosis and immunotherapy response in soft tissue sarcoma. Front Immunol 2022; 13:895465. [PMID: 35967408 PMCID: PMC9374037 DOI: 10.3389/fimmu.2022.895465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 07/01/2022] [Indexed: 11/14/2022] Open
Abstract
N6-methyladenosine (m6A) methylation, one of the most crucial RNA modifications, has been proven to play a key role that affect prognosis of soft tissue sarcoma (STS). However, m6A methylation potential role in STS metabolic processes remains unknown. We comprehensively estimated the m6A metabolic molecular subtypes and corresponding survival, immunity, genomic and stemness characteristics based on 568 STS samples and m6A related metabolic pathways. Then, to quantify the m6A metabolic subtypes, machine learning algorithms were used to develop the m6A-metabolic Scores of individual patients. Finally, two distinct m6A metabolic subtypes (Cluster A and Cluster B) among the STS patients were identified. Compared to Cluster B subtype, the Cluster A subtype was mainly characterized by better survival advantages, activated anti-tumor immune microenvironment, lower gene mutation frequency and higher anti-PD-1 immunotherapy response rates. We also found that the m6A-metabolic Scores could accurately predict the molecular subtype of STS, prognosis, the abundance of immune cell infiltration, tumor metastasis status, sensitivity to chemotherapeutics and immunotherapy response. In general, this study revealed that m6A-regulated tumor metabolism processes played a key role in terms of prognosis of STS, tumor progression, and immune microenvironment. The identification of metabolic molecular subtypes and the construction of m6A-metabolic Score will help to more effectively guide immunotherapy, metabolic therapy and chemotherapy in STS.
Collapse
Affiliation(s)
- Zhen-Dong Huang
- Department of Spine and Orthopedic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yong-Cheng Fu
- The Third Clinical School, Hubei University of Medicine, Shiyan, China
| | - Shu-Yan Liu
- The Third Clinical School, Hubei University of Medicine, Shiyan, China
| | - Ya-Juan Mao
- The Third Clinical School, Hubei University of Medicine, Shiyan, China
| | - Yan Zhang
- The Third Clinical School, Hubei University of Medicine, Shiyan, China
| | - Chao Hu
- Department of Spine and Orthopedic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ren-Xiong Wei
- Department of Spine and Orthopedic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence:Ren-Xiong Wei,
| |
Collapse
|
46
|
Qiu Y, Li J, Yao J, Meng J, Huang X, Zheng X, Wen Z, Huang J, Wang H. Correlative Study on the Relationship between the Expression of m6a-Related Genes and the Prognosis and Immunotherapy of Soft Tissue Sarcoma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5439023. [PMID: 38024481 PMCID: PMC10667055 DOI: 10.1155/2022/5439023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 12/01/2023]
Abstract
Background Soft tissue sarcomas (STS) are rare malignancies arising from mesenchymal tissue and interlacing ectodermal nerve tissue. Immunotherapy plays an important role in the prognosis and survival of STS patients. However, there is insufficient evidence to confirm the prognostic value of m6A-related genes and to evaluate the efficacy of immunotherapy for STS. Methods We analyzed 23 m6A regulators from STS samples using R software and defined the modification patterns for three STS m6A regulators. Then, we constructed the m6A scores and divided the samples into high and low subgroups. Finally, we used data from the GEO database to verify the results. Results We found that the m6A clusters differed in the overall survival (OS), progression-free survival (PFS), and immune infiltration rate. Additionally, the m6A score was positively correlated with the contents of activated B cells, activated dendritic cells, CD56 bright natural killer cells, helper T cells, and regulatory T cells. The group with a higher m6A score also presented higher OS and PFS rates. Regarding immunotherapy, STS patients with a high m6A score presented better results. Consistently, we found similar results in another dataset with patients that received anti-PD-1/PD-L1 therapy. Conclusion Our current results indicated that the m6A score can be used to assess the survival rate of STS patients and guide immunotherapy and predict its effects. The analysis of different m6A patterns of STS samples contributed to the understanding of the diversity and complexity of the tumor microenvironment (TME) and provided new ideas for the clinical development of personalized immunotherapy and prediction of the prognosis of STS patients.
Collapse
Affiliation(s)
- Yue Qiu
- The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jia Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jun Yao
- The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jinzhi Meng
- The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xing Huang
- The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xifan Zheng
- The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zhenpei Wen
- The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Junpu Huang
- The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Hongtao Wang
- The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
47
|
Kaynak A, Davis HW, Kogan AB, Lee JH, Narmoneva DA, Qi X. Phosphatidylserine: The Unique Dual-Role Biomarker for Cancer Imaging and Therapy. Cancers (Basel) 2022; 14:2536. [PMID: 35626139 PMCID: PMC9139557 DOI: 10.3390/cancers14102536] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/06/2022] [Accepted: 05/17/2022] [Indexed: 01/27/2023] Open
Abstract
Cancer is among the leading causes of death worldwide. In recent years, many cancer-associated biomarkers have been identified that are used for cancer diagnosis, prognosis, screening, and early detection, as well as for predicting and monitoring carcinogenesis and therapeutic effectiveness. Phosphatidylserine (PS) is a negatively charged phospholipid which is predominantly located in the inner leaflet of the cell membrane. In many cancer cells, PS externalizes to the outer cell membrane, a process regulated by calcium-dependent flippases and scramblases. Saposin C coupled with dioleoylphosphatidylserine (SapC-DOPS) nanovesicle (BXQ-350) and bavituximab, (Tarvacin, human-mouse chimeric monoclonal antibodies) are cell surface PS-targeting drugs being tested in clinical trial for treating a variety of cancers. Additionally, a number of other PS-selective agents have been used to trigger cytotoxicity in tumor-associated endothelial cells or cancer cells in pre-clinical studies. Recent studies have demonstrated that upregulation of surface PS exposure by chemodrugs, radiation, and external electric fields can be used as a novel approach to sensitize cancer cells to PS-targeting anticancer drugs. The objectives of this review are to provide an overview of a unique dual-role of PS as a biomarker/target for cancer imaging and therapy, and to discuss PS-based anticancer strategies that are currently under active development.
Collapse
Affiliation(s)
- Ahmet Kaynak
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (A.K.); (J.-H.L.); (D.A.N.)
| | - Harold W. Davis
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Andrei B. Kogan
- Physics Department, University of Cincinnati, Cincinnati, OH 45221, USA;
| | - Jing-Huei Lee
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (A.K.); (J.-H.L.); (D.A.N.)
| | - Daria A. Narmoneva
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (A.K.); (J.-H.L.); (D.A.N.)
| | - Xiaoyang Qi
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (A.K.); (J.-H.L.); (D.A.N.)
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| |
Collapse
|
48
|
Gong S, Schopow N, Duan Y, Wu C, Kallendrusch S, Osterhoff G. PLOD Family: A Novel Biomarker for Prognosis and Personalized Treatment in Soft Tissue Sarcoma. Genes (Basel) 2022; 13:genes13050787. [PMID: 35627171 PMCID: PMC9141206 DOI: 10.3390/genes13050787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022] Open
Abstract
Despite various treatment attempts, the heterogenous group of soft tissue sarcomata (STS) with more than 100 subtypes still shows poor outcomes. Therefore, effective biomarkers for prognosis prediction and personalized treatment are of high importance. The Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase (PLOD) gene family, which is related to multiple cancer entities, consists of three members which encode important enzymes for the formation of connective tissue. The relation to STS, however, has not yet been explored. In this study, data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases were used to analyze the role of PLOD1–3 in STS. It was found that an overexpression of PLOD family members correlates with poor prognosis, which might be due to an increased infiltration of immune-related cells in the tumor microenvironment. In STS, the expression of PLOD genes could be a novel biomarker for prognosis and a personalized, more aggressive treatment in these patients.
Collapse
Affiliation(s)
- Siming Gong
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, 04103 Leipzig, Germany; (S.G.); (N.S.); (S.K.)
| | - Nikolas Schopow
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, 04103 Leipzig, Germany; (S.G.); (N.S.); (S.K.)
- Sarcoma Center, Department for Orthopedics, Trauma Surgery and Reconstructive Surgery, University Hospital Leipzig, Liebigstraße 20, 04103 Leipzig, Germany;
| | - Yingjuan Duan
- Faculty of Chemistry and Mineralogy, University of Leipzig, Johannisallee 29, 04103 Leipzig, Germany;
| | - Changwu Wu
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, 04103 Leipzig, Germany; (S.G.); (N.S.); (S.K.)
- Correspondence: or
| | - Sonja Kallendrusch
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, 04103 Leipzig, Germany; (S.G.); (N.S.); (S.K.)
- Department of Medicine, Health and Medical University Potsdam, Olympischer Weg 1, 14471 Potsdam, Germany
| | - Georg Osterhoff
- Sarcoma Center, Department for Orthopedics, Trauma Surgery and Reconstructive Surgery, University Hospital Leipzig, Liebigstraße 20, 04103 Leipzig, Germany;
| |
Collapse
|
49
|
Tu B, Jia Y, Qian J. Bioinformatics Analysis Identified Five Widely Expressed Genes Associated with Prognosis in Sarcoma. Int J Gen Med 2022; 15:3711-3725. [PMID: 35414751 PMCID: PMC8995039 DOI: 10.2147/ijgm.s352048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Bizhi Tu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Yaya Jia
- Department of Pediatrics, The Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Jun Qian
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
- Correspondence: Jun Qian, Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, People’s Republic of China, Tel +86-13955116514, Fax +86-551-62922025, Email
| |
Collapse
|
50
|
Tian J, Liu Y, Zhang T, Yue L, Xiao Y, Guo C. LAG-3 is a promising inhibitory immune checkpoint for antitumor immunotherapy. Expert Rev Anticancer Ther 2022; 22:289-296. [PMID: 35132925 DOI: 10.1080/14737140.2022.2039124] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Lymphocyte activation gene-3 (LAG-3) is a member of the immunoglobulin superfamily. Engagement of LAG-3 by its ligands to trigger downstream signaling can inhibit immune responses and regulate the pathogenesis of many diseases, including cancer and inflammatory diseases. AREAS COVERED We used keywords to search for relevant publications in PubMed and information on websites. After systematic analysis, we discuss the biological characteristics of LAG-3 and its ligands, LAG-3 related signaling, its roles in the pathogenesis of tumors, and its blockages for the treatment of cancers, as well as current challenges and future directions of research. EXPERT OPINION Although the mechanisms underlying the action of LAG-3/ligand-related signaling in tumor development are not fully understood, advances in scientific research and LAG-3-based immunotherapies are promising. Further studies to explore its biological roles and molecular mechanisms may aid in developing new LAG-3- and ligand-based therapeutic drugs to benefit patients with different types of cancers.
Collapse
Affiliation(s)
- Jin Tian
- Institute of Clinical Medicine, Qingdao University Medical College, Qingdao, China.,Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - Yang Liu
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - TengLong Zhang
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - Lu Yue
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - YaNan Xiao
- Department of Endocrinology, Qingdao Municipal Hospital West Campus, Qingdao, China
| | - ChengYe Guo
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| |
Collapse
|