1
|
Farooq MS, Shafique N, Vargas GM, Guo J, Miura JT, Lefler DS, Karakousis GC. Neoadjuvant Immunotherapy for Resectable Dedifferentiated Liposarcoma: A National Cohort Analysis. J Surg Oncol 2025. [PMID: 40358214 DOI: 10.1002/jso.28155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/03/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Neoadjuvant immunotherapy (NIT) with checkpoint blockade has been increasingly studied for soft tissue sarcomas, however, survival outcomes data are limited, and dedifferentiated liposarcoma (DDLPS) histology remains underrepresented in recent trial cohorts. We assessed the impact of NIT with or without radiation therapy (RT) on overall survival (OS) for resectable DDLPS. METHODS The National Cancer Database (NCDB) was used to identify patients diagnosed with nonmetastatic DDLPS who received NIT and underwent surgical resection between 2016 and 2022. Primary outcome was 5-year OS. RESULTS A total of 3414 patients with DDLPS met the inclusion criteria and NIT was administered to 31 (1%) patients. Factors associated with receipt of NIT were receipt of neoadjuvant RT (NRT, odds ratio [OR]: 5.75, p < 0.001) and male sex (OR: 3.33, p = 0.036). NIT was associated with a hazard ratio (HR) for mortality of 0.89 (p = 0.786). No difference was found in 5-year OS in the overall cohort (NIT 72% vs. 61% no NIT, p = 0.320) or in the propensity-matched cohort (68% vs. 65%, p = 0.848). Subanalysis between NIT with NRT versus NRT-only also did not find any significant difference in 5-year OS (88% vs. 59%, p = 0.331). CONCLUSION In this retrospective NCDB analysis of patients with resectable DDLPS, administration of NIT did not significantly affect OS.
Collapse
Affiliation(s)
- Mohammad S Farooq
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Neha Shafique
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gracia M Vargas
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jennifer Guo
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John T Miura
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel S Lefler
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Giorgos C Karakousis
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Kret ZS, Sweder RJ, Pollock R, Tinoco G. Potential Mechanisms for Immunotherapy Resistance in Adult Soft-Tissue Sarcoma. Target Oncol 2025:10.1007/s11523-025-01145-5. [PMID: 40289241 DOI: 10.1007/s11523-025-01145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
3
|
Aobo Z, Xiao Z, Chengfei X, Zhe X, Yingxue C, Chenhe Z, Fuan X, Fan Y, Mengmeng X, Feng Y, Wengang L. Combination of immune checkpoint inhibitors and anthracyclines as a potential first-line regimen for dedifferentiated liposarcoma: systematic review and meta-analysis. Cancer Immunol Immunother 2025; 74:179. [PMID: 40257618 PMCID: PMC12011665 DOI: 10.1007/s00262-025-04007-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 04/22/2025]
Abstract
INTRODUCTION Dedifferentiated liposarcoma (DDLPS) is a rare and aggressive subtype of soft tissue sarcoma, characterized by limited treatment options and poor prognosis. Despite surgical resection being the only potentially curative treatment for localized DDLPS, the recurrence rate remains high, and systemic chemotherapy, typically anthracycline-based, shows limited efficacy in advanced stages. While immune checkpoint inhibitors (ICIs) have shown promise in various sarcoma subtypes, including DDLPS, their role as a first-line treatment remains unclear. METHODS We conducted a systematic meta-analysis to evaluate the efficacy of ICIs in treating patients with DDLPS. A total of 25 studies encompassing 245 patients were included. Data on overall response rate (ORR), progression-free survival, and grade III-V treatment-related adverse events were analyzed. We assessed treatment efficacy based on the line of therapy and treatment regimens, including ICI monotherapy, dual ICI therapy, and ICI combinations with other modalities. RESULTS The pooled ORR for all ICI-based treatments was 7%. First-line ICI therapy yielded a significantly higher ORR of 22%, compared to 4% in later-line treatment. The combination of ICI with anthracyclines demonstrated the highest ORR of 52%. In contrast, ICI regimens combined with trabectedin or other agents showed limited efficacy. Sensitivity analysis confirmed the stability of results, and publication bias was not detected. CONCLUSION This meta-analysis supports the potential role of ICIs, particularly in combination with anthracyclines, as a first-line therapeutic strategy for DDLPS. These results provide a foundation for future prospective studies aimed at optimizing immunotherapy approaches for this rare and challenging malignancy.
Collapse
Affiliation(s)
- Zhuang Aobo
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhou Xiao
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xu Chengfei
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China
| | - Xi Zhe
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Chen Yingxue
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhang Chenhe
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xie Fuan
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yang Fan
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiao Mengmeng
- Department of Retroperitoneal Tumor Surgery, Peking University People's Hospital, Beijing, China.
| | - Ye Feng
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Li Wengang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
4
|
Chen A, Qiu Y, Yen Y, Wang C, Wang X, Li C, Wei Z, Li L, Yu L, Liu F, Li R. Expression of Cancer-Testis Antigens MAGE-A1, MAGE-A4, NY-ESO-1 and PRAME in Bone and Soft Tissue Sarcomas: The Experience From a Single Center in China. Cancer Med 2025; 14:e70750. [PMID: 40152485 PMCID: PMC11951172 DOI: 10.1002/cam4.70750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/01/2025] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
OBJECTIVE Sarcomas are a heterogeneous group of malignancies, low disease-control levels and the limited durability of responses have prompted the exploration of various novel immunotherapeutic approaches. To preliminarily explore the feasibility of cancer vaccines based on cancer testis antigen in the immunotherapy of sarcomas, we investigate the expression of Cancer/Testis Antigens (CTA) MAGE-A4, PRAME, MAGE-A1, KK-LC-1, and NY-ESO-1 in bone and soft tissue sarcomas, with the aim of assessing their potential for use in sarcoma immunotherapy and determining their expression levels in different subtypes. METHODS AND RESULTS We employed immunohistochemistry and multiplex immunostaining microarrays (MI chips) to assess the expression of MAGE-A4, PRAME, MAGE-A1, KK-LC-1, and NY-ESO-1 in 21 cases of undifferentiated pleomorphic sarcoma (UPS), 26 cases of smooth muscle sarcoma, 28 cases of liposarcoma, 40 cases of osteosarcoma (OS), and 13 cases of chondrosarcoma. MAGE-A1 showed the highest expression in osteosarcoma (32.50%), while it was lower in liposarcoma and undifferentiated pleomorphic sarcoma (10.71% and 10.00%) and undetectable in chondrosarcoma. MAGE-A4 expression was elevated in osteosarcoma and undifferentiated pleomorphic sarcoma (40.00% and 33.00%), but lower in liposarcoma and smooth muscle sarcoma (17.00% and 33.00%). NY-ESO-1 expression was relatively low across all sarcoma subtypes. PRAME expression was highest in undifferentiated pleomorphic sarcoma (47.62%) and low in chondrosarcoma (7.69%). None of the sarcomas expressed KK-LC-1. Additionally, while there was no statistically significant correlation between CTA expression and patient age or gender, some differences related to age and gender were observed. CONCLUSIONS CTA expression in bone and soft tissue sarcomas was correlated with both CTA type and sarcoma subtype, showing relatively high levels of expression in undifferentiated pleomorphic sarcoma (UPS) and osteosarcoma (OS). The poly-expression of MAGE-A4, PRAME, and MAGE-A1 across all subtypes suggests that these antigens may serve as potential targets for sarcoma-specific immunotherapy.
Collapse
Affiliation(s)
- Anni Chen
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Yuling Qiu
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Ying‐Tzu Yen
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Chun Wang
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Xiaolu Wang
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Chunhua Li
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Zijian Wei
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Lin Li
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Lixia Yu
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| | - Fangcen Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
- Department of Pathology, Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Rutian Li
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingChina
| |
Collapse
|
5
|
Levy A, Morel D, Texier M, Rodriguez-Ruiz ME, Bouarroudj L, Bouquet F, Bustillos A, Quevrin C, Clémenson C, Mondini M, Meziani L, Sun R, Zaghdoud N, Tselikas L, Assi T, Faron M, Honoré C, Ngo C, Verret B, Le Péchoux C, Le Cesne A, Ginhoux F, Massard C, Bahleda R, Deutsch E. Monocyte-lineage tumor infiltration predicts immunoradiotherapy response in advanced pretreated soft-tissue sarcoma: phase 2 trial results. Signal Transduct Target Ther 2025; 10:103. [PMID: 40097400 PMCID: PMC11914280 DOI: 10.1038/s41392-025-02173-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Immunoradiotherapy holds promise for improving outcomes in patients with advanced solid tumors, including in soft-tissue sarcoma (STS). However, the ideal combination of treatment modalities remains to be determined, and reliable biomarkers to predict which patients will benefit are lacking. Here, we report the results of the STS cohort of the SABR-PDL1 phase II trial that evaluated the anti-PDL1 atezolizumab combined with stereotactic body radiation therapy (SBRT) delivered concurrently with the 2nd cycle to at least one tumor site. Eligible patients received atezolizumab until progression or unmanageable toxicity, with SBRT at 45 Gy in 3 fractions). The primary endpoint was one-year progression-free survival (PFS) rate with success defined as 13 patients achieving 1-year PFS. Sixty-one heavily pretreated patients with STS (median 5 prior lines; 52% men; median age 54 years; 28% leiomyosarcoma) were enrolled across two centers (France, Spain). SBRT was delivered to 55 patients (90%), with the lung being the most commonly irradiated site (50%). After a median follow-up of 45 months, the one-year PFS rate was 8.3% [95% CI: 3.6-18.1]. Median PFS and overall survival were 2.5 and 8.6 months, respectively. Best responses included partial responses (5%) and stable disease (60%). Immune profiling revealed increased immunosuppressive tumor-associated macrophages (e.g., IL4I1, HES1) and monocyte-recruiting chemokines in non-responders. Higher monocyte/lymphocyte ratios (MonoLR) in tumor and blood correlated with progression. PD-L1 status, lymphoid infiltration, and tertiary-lymphoid structures were not predictive. Although the primary endpoint was not met, this study highlights MonoLR imbalance as a potential biomarker to identify STS patients likely to benefit from immunoradiotherapy. EudraCT No. 2015-005464-42; Clinicaltrial.gov number: NCT02992912.
Collapse
Affiliation(s)
- Antonin Levy
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France.
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France.
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France.
- Sarcoma unit, Gustave Roussy, Villejuif, France.
| | - Daphné Morel
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
| | - Matthieu Texier
- Biostatistics and Epidemiology Office, Gustave Roussy, Villejuif, France
- Oncostat 1018 Inserm, University Paris-Saclay, Villejuif, France
| | | | - Lisa Bouarroudj
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
- Bioinformatic platform, Gustave Roussy, Villejuif, France
| | - Fanny Bouquet
- Product Development Medical Affairs, F Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Alberto Bustillos
- Product Development Medical Affairs, F Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Clément Quevrin
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
| | - Céline Clémenson
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
| | - Michele Mondini
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
| | - Lydia Meziani
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
| | - Roger Sun
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Nadia Zaghdoud
- Biostatistics and Epidemiology Office, Gustave Roussy, Villejuif, France
| | - Lambros Tselikas
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Department of Interventional Radiology, Gustave Roussy, Villejuif, France
| | - Tarek Assi
- Sarcoma unit, Gustave Roussy, Villejuif, France
| | - Matthieu Faron
- Sarcoma unit, Gustave Roussy, Villejuif, France
- Oncostat 1018 Inserm, University Paris-Saclay, Villejuif, France
| | | | - Carine Ngo
- Sarcoma unit, Gustave Roussy, Villejuif, France
| | | | - Cécile Le Péchoux
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- Sarcoma unit, Gustave Roussy, Villejuif, France
| | | | - Florent Ginhoux
- Gustave Roussy, Inserm U1015, Université Paris-Saclay, Villejuif, France
| | - Christophe Massard
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France
- Drug Development Department (DITEP) Gustave Roussy-Cancer Campus, Villejuif, France
| | - Rastilav Bahleda
- Sarcoma unit, Gustave Roussy, Villejuif, France
- Drug Development Department (DITEP) Gustave Roussy-Cancer Campus, Villejuif, France
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France.
- Gustave Roussy, Inserm U1030, Université Paris-Saclay, Villejuif, France.
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France.
| |
Collapse
|
6
|
Falcao RM, de Souza JES, Gonzalez-Molina J, Mathieson W, Carlson JW, Petta TB. Deep multi-omics integration approach reveals new molecular features of uterine leiomyosarcoma. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167632. [PMID: 39708976 DOI: 10.1016/j.bbadis.2024.167632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Uterine leiomyosarcoma (uLMS) is a rare and aggressive cancer representing approximately 25 % of all uterine malignancies. The molecular heterogeneity and pathogenesis of uLMS are not well understood, and translational studies aimed at discovering the vulnerabilities of this tumor type are of high priority. We conducted an innovative comprehensive multi-omics integration study from DNA to protein using freshly frozen tumors. Here, we show that two tumors harbor actionable therapeutic targets, IDH1_p.Arg132Cys and KRAS_p.Gly12Cys, and homologous recombination deficiency (HRD) is the most predominant genomic signature. Additionally, 80 % of the samples presented a chromothripsis signature, reinforcing the aneuploidy phenotype of these tumors. Tumors with a high proliferation score and high Ki67 expression was associated with worse overall survival (OS). We observed a high frequency of balanced fusion events involving EEF1A1 with enrichment of the EGFR pathway. For the first time, uLMS proteomics analysis showed the enrichment of pathways associated with suppression of the innate immune system and ECM organization. Finally, our comprehensive multi-omics integration analysis identified amplification of the CTHRC1 gene from the matrisome, with a negative impact on OS. Interestingly, the expression of Ki67 and CTHRC1 exhibits a strong negative correlation, underscoring two distinct and mutually exclusive biological profiles in uLMS: (i) highly proliferative tumors, characterized by elevated Ki67 expression, and (ii) tumors driven by ECM remodeling, marked by high CTHRC1 levels. Taken together, this deep functional multi-omics approach contributes to the detection of new molecular features of uLMS and suggests that patients could benefit from precision oncology in clinical practice.
Collapse
Affiliation(s)
- Raul Maia Falcao
- Universidade Federal do Rio Grande do Norte, IMD, Ppg-Bioinformatica, Natal, Brazil.
| | | | - Jordi Gonzalez-Molina
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - William Mathieson
- Integrated Biobank of Luxembourg, Luxembourg Institute of Health, Dudelange, Luxembourg.
| | | | - Tirzah Braz Petta
- Universidade Federal do Rio Grande do Norte, IMD, Ppg-Bioinformatica, Natal, Brazil; University of Southern California, Keck School of Medicine, Department of Translational Genomics, 1450 Biggy St., Los Angeles, CA 90089, United States of America.
| |
Collapse
|
7
|
Zhuang AB, Xi Z, Cheng YX, Zhang CH, Li WG. Current status and future perspectives of immunotherapy for abdominal liposarcoma: From basic research to clinical practice. Shijie Huaren Xiaohua Zazhi 2025; 33:81-88. [DOI: 10.11569/wcjd.v33.i2.81] [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: 09/28/2024] [Revised: 11/06/2024] [Accepted: 12/17/2024] [Indexed: 02/28/2025] Open
Abstract
Liposarcoma is a highly heterogeneous type of soft tissue sarcoma originating from adipose tissue, characterized by complex biological behavior and invasiveness. Traditional treatments have shown limited efficacy in high-grade and metastatic liposarcoma, with unsatisfactory patient outcomes. In recent years, the breakthroughs of immunotherapy in various solid tumors have sparked interest in its potential application to liposarcoma. This review systematically examines the progress in basic research and clinical practice of immunotherapy for liposarcoma, discussing the tumor immune microenvironment, mechanisms of immune evasion, the application of immune checkpoint inhibitors, combination therapy strategies, the challenges faced, as well as the future direction, with an aim to provide a theoretical basis for personalized treatment of liposarcoma, promote the development of novel immunotherapy strategies, and ultimately improve patient prognosis and quality of life.
Collapse
Affiliation(s)
- Ao-Bo Zhuang
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Zhe Xi
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Ying-Xue Cheng
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Chen-He Zhang
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Wen-Gang Li
- Department of Hepatobiliary and Pancreatic Surgery, Xiang'an Hospital of Xiamen University, Xiamen 361102, Fujian Province, China
- Cancer Research Center of Xiamen University, Xiamen 361005, Fujian Province, China
| |
Collapse
|
8
|
Cao Y, Wang W, Xu H, Yi H, Gao Y, Wan M, Wang M, Chen T, Chen Y, Chi Y, Wei S, Jin S, Bai M, Li X, Gao Y, Niu X, Liu Y. Efficacy of immune checkpoint inhibitors in the treatment of soft tissue sarcoma: A systematic review and meta-analysis of clinical trials. Int Immunopharmacol 2025; 148:114070. [PMID: 39826454 DOI: 10.1016/j.intimp.2025.114070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Soft tissue sarcomas (STS) are a heterogeneous group of tumors with diverse clinical and molecular characteristics, characterized by limited treatment options and poor prognosis. Immune checkpoint inhibitors (ICIs) have emerged as promising therapies for STS, yet comprehensive evaluations of their efficacy, especially in combination with other treatments, are scarce. METHODS We conducted a systematic review and meta-analysis of clinical trials on ICIs in STS treatment, sourced from PubMed, Embase, and the Cochrane Central Register of Controlled Trials up to May 31, 2024. The studies included both monotherapy and combination therapies with ICIs. We assessed the methodological quality using the Cochrane Risk of Bias 2 tool and the Methodological Index for Non-Randomized Studies. Data synthesis involved random-effects meta-analysis to determine pooled proportions and 95% confidence intervals (CIs) for objective response rates (ORR), disease control rates (DCR), and high-grade treatment-related adverse events (TRAEs). RESULTS The analysis included 38 studies with 1349 patients covering 24 STS subtypes. The overall ORR was 16% (95% CI 0.12-0.21), DCR was 64% (95% CI 0.57-0.70), and the rate of Grade 3-5 TRAEs was 19% (95% CI 0.13-0.27). Treatments combining ICIs with tyrosine kinase inhibitors (TKIs) showed the highest efficacy (ORR 28%, 95% CI 0.18-0.40), albeit with increased adverse events. ORRs in first-line treatments were substantially higher (28%) compared to second-line treatments or beyond (11%). Subtypes like alveolar soft part sarcoma (ASPS), angiosarcoma (AS), and epithelioid sarcoma (ES) exhibited favorable responses exceeding 30%. CONCLUSIONS This systematic review and meta-analysis indicate that ICIs, particularly when combined with TKIs, provide substantial therapeutic benefits in treating STS, significantly enhancing response rates in specific subtypes such as ASPS and AS. The results underscore the transformative potential of ICIs in STS treatment strategies. However, the variability across subtypes and treatment lines emphasizes the need for further randomized controlled trials to refine and personalize therapeutic approaches, ensuring optimal outcomes for patients with these diverse malignancies.
Collapse
Affiliation(s)
- Yang Cao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wei Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hairong Xu
- Department of Orthopedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, No.31 Xin Jie Kou East Street, Xi Cheng District, Beijing 100035, China
| | - Hang Yi
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yinyan Gao
- Department of Epidemiology and Biostatistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Mingzhong Wan
- Shantou University Medical College, Shantou 515041, China
| | - Mingzhao Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Tong Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yanchao Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yihebali Chi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shuqing Wei
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, General Internal Medicine, Taiyuan, Shanxi 030013, China
| | - Shi Jin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Ming Bai
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Xin Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Yibo Gao
- Central Laboratory & Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China.
| | - Xiaohui Niu
- Department of Orthopedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, No.31 Xin Jie Kou East Street, Xi Cheng District, Beijing 100035, China.
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
9
|
Li J, Zhou X, Wu L, Ma J, Tan Y, Wu S, Zhu J, Wang Q, Shi Q. Optimal early endpoint for second-line or subsequent immune checkpoint inhibitors in previously treated advanced solid cancers: a systematic review. BMC Cancer 2025; 25:293. [PMID: 39966752 PMCID: PMC11837729 DOI: 10.1186/s12885-025-13712-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND The administration of second-line or subsequent immune checkpoint inhibitors (ICIs) in previously treated patients with advanced solid cancers has been clinically investigated. However, previous clinical trials lacked an appropriate primary endpoint for efficacy assessment. This systematic review aimed to explore the most optimal early efficacy endpoint for such trials. METHODS Phase 2 or 3 clinical trials involving patients with advanced solid cancers with disease progression following standard first-line therapy receiving second-line or subsequent ICI administration, with adequate survival outcome data, were included from PubMed, Embase, Web of Science, and Cochrane Library databases before February 2023. Quality assessment was conducted using the Cochrane tool and Newcastle-Ottawa Quality Assessment Scale for Cohort Studies for randomized controlled trials (RCTs) and non-randomized trials, respectively. Objective response rate (ORR) and progression-free survival (PFS) at 3, 6, and 9 months were investigated as potential early efficacy endpoint candidates for 12-month overall survival (OS), with a strong correlation defined as Pearson's correlation coefficient r ≥ 0.8. RESULTS A total of 64 RCTs comprising 22,725 patients and 106 non-randomized prospective trials involving 10,608 participants were eligible for modeling and external validation, respectively. RCTs examined 15 different cancer types, predominantly non-small-cell lung cancer (NSCLC) (17, 28%), melanoma (9, 14%), and esophageal squamous cell carcinoma (5, 8%). The median sample size of RCTs was 124 patients, and the median follow-up time was 3.2-57.7 months. The ORR (r = 0.38; 95% confidence interval [CI], 0.18-0.54) and PFS (r = 0.42; 95% CI, 0.14-0.64) exhibited weak trial-level correlations with OS. Within ICI treatment arms, the r values of ORR and 3-, 6-, and 9-month PFS with 12-month OS were 0.61 (95% CI, 0.37-0.79), 0.78 (95% CI, 0.62-0.88), 0.84 (95% CI, 0.77-0.90), and 0.86 (95% CI, 0.79-0.90), respectively. External validation of 6-month PFS indicated an acceptable discrepancy between actual and predicted 12-month OS. CONCLUSIONS In non-randomized phase 2 trials on second-line or subsequent ICI therapy in patients with advanced solid cancers, 6-month PFS could serve as an early efficacy endpoint. However, early efficacy endpoints are not recommended in RCTs to replace OS.
Collapse
Affiliation(s)
- Jingqiu Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoding Zhou
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Wu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiabao Ma
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Tan
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Songke Wu
- Department of Oncology, People'S Hospital of Cangxi County, Guangyuan, China.
| | - Jie Zhu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| | - Qifeng Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| | - Qiuling Shi
- Center for Cancer Prevention Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Gómez-Puerto D, Cicala CM, Valverde C, Serrano C. Emerging treatments for sarcoma: from 2024 onward. Expert Opin Emerg Drugs 2025:1-18. [PMID: 39874592 DOI: 10.1080/14728214.2025.2460525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 01/30/2025]
Abstract
INTRODUCTION Sarcomas are a rare and diverse group of mesenchymal-origin solid tumors, constituting only 1% of adult malignancies and classified into soft tissue and bone sarcomas. For localized diseases, surgery and radiotherapy remain the cornerstone treatments. However, systemic options for advanced stages are limited, with an overall survival of approximately 20 months. There is an urgent need to refine clinical trial designs and evaluate novel therapies to improve patient outcomes. AREAS COVERED This review summarizes recent advancements in sarcoma clinical trials, highlighting promising therapies that may reshape treatment paradigms. It also examines insights from comprehensive genomic profiling studies, which are paving the way for next-generation therapeutic approaches. EXPERT OPINION The heterogeneity and rarity of sarcomas present substantial challenges to the development and regulatory approval of effective therapies. Recent progress in the field has introduced innovative approaches, including novel chemotherapeutic agents, targeted therapies, and immunotherapeutics. However, despite these advancements, significant hurdles persist. While these strategies have demonstrated some success, the overall improvements in survival have often been modest, irrespective of the therapeutic modality. This underscores critical concerns regarding the true cost-benefit balance and the potential for adverse effects, particularly when evaluated over extended follow-up periods.
Collapse
Affiliation(s)
- Diego Gómez-Puerto
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Carlo M Cicala
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
- Sarcoma Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Claudia Valverde
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - César Serrano
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
- Sarcoma Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
11
|
D’Alessandro PR, Homanick CE, Cooper BD, Ferguson K, Rutan H, Pressey JG. "From Drowning to Treading Water": Adolescents and Young Adults Living with Incurable and Indolent Metastatic Soft Tissue Sarcoma for More than Two Years. Cancers (Basel) 2025; 17:442. [PMID: 39941809 PMCID: PMC11816224 DOI: 10.3390/cancers17030442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/21/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
INTRODUCTION Adolescent/young adult (AYA) patients with metastatic soft tissue sarcoma (STS) typically face a dismal prognosis. However, a subset of patients with incurable disease lives beyond two years. Due to the rarity of diagnoses and inherent heterogeneity within this population, a paucity of data exists regarding the experiences of AYAs with an indolent course (and how to best capture these experiences). With increasing biological insight and clinical experience, including the use of targeted or immune therapies, it is anticipated that more such patients will experience prolonged survival. Our pilot study aimed to describe the clinical characteristics and illness experiences of AYAs with incurable yet indolent metastatic STS who were living two years after their diagnoses. Our exploratory aim was to generate a conceptual framework that could subsequently be tested in a multi-center study with a larger cohort of patients. MATERIALS AND METHODS Patients with metastatic incurable STS, aged 15-39 years at diagnosis, and at least two years from diagnosis, were eligible. Patients were recruited over a two-year period at a quaternary children's hospital with a comprehensive AYA oncology program. Participants completed a demographic form and PROMIS short form questionnaires for seven domains and answered an open-ended question. Responses to open-ended questions were coded independently by two authors and utilized to generate themes. Clinical variables were collected from medical records. RESULTS Five patients completed questionnaires. Mean age was 29.4 years (18.5-39.8 years) at diagnosis and 34 years (23.2-45.7 years) at study. Three patients were female; two were male; four were White; and one was Black/African American. Diagnoses included ASPSCR1::TFE3 alveolar soft part sarcoma; WWTR1::CAMTA1 epithelioid hemangioendothelioma; INI-1 deficient epithelioid sarcoma; EWSR1::NR4A3 extra-skeletal myxoid chondrosarcoma; and low-grade ARHGAP23::FER spindle cell malignancy, a novel fusion-driven sarcoma. Mean time since diagnosis was 4.5 years (2.6-6 years), and mean treatment duration was 4.2 years (1.5-6 years). On average, patients received 4.8 lines (range 2-8 lines) of antineoplastic therapy. All patients received at least one targeted therapy or immune checkpoint inhibitor. Patients reported increased fatigue and anxiety and decreased physical function compared to the standardized US reference population. Themes emerging from qualitative responses included managing physical symptoms, navigating feelings of guilt and inadequacy, self-reflection generating gratitude, and changing illness experiences over time. CONCLUSIONS AYA patients living with incurable metastatic soft tissue sarcoma for more than two years were treated with multiple lines of antineoplastic therapy longitudinally. PROMIS data identified fatigue, anxiety, and decreased physical function within this population. Exploratory thematic analysis of qualitative responses generated concepts that could be further tested in an expanded cohort of patients.
Collapse
Affiliation(s)
- Paul R. D’Alessandro
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Caitlin E. Homanick
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Brittany D. Cooper
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Katelyn Ferguson
- College of Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - Hillary Rutan
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Joseph G. Pressey
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
12
|
Martin-Broto J, Diaz-Beveridge R, Moura D, Ramos R, Martinez-Trufero J, Carrasco I, Sebio A, González-Billalabeitia E, Gutierrez A, Fernandez-Jara J, Hernández-Vargas L, Cruz J, Valverde C, Hindi N. Phase Ib Study for the Combination of Doxorubicin, Dacarbazine, and Nivolumab as the Upfront Treatment in Patients With Advanced Leiomyosarcoma: A Study by the Spanish Sarcoma Group (GEIS). J Clin Oncol 2025; 43:297-307. [PMID: 39356980 DOI: 10.1200/jco.24.00358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/06/2024] [Accepted: 07/17/2024] [Indexed: 10/04/2024] Open
Abstract
PURPOSE Doxorubicin, alongside a select group of cytotoxic agents, is capable of inducing an adaptive immune response via a well-established peculiar type of tumor cell death called immunogenic cell death (ICD). We hypothesize that combining doxorubicin and dacarbazine with nivolumab may enhance therapeutic efficacy by exerting synergy in the ICD circuit. We hereby present a phase Ib trial with this combination. PATIENTS AND METHODS Patients with advanced leiomyosarcoma and anthracycline-naïve were eligible. The initial dose level consisted of doxorubicin 75 mg/m2 once on day 1, once every three weeks, followed by dacarbazine 400 mg/m2 once on days 1 and 2, once every three weeks, plus nivolumab 360 mg once on day 2, once every 3 weeks, for six courses and then 1 year of nivolumab. A (-1) dose level was the same regimen but with nivolumab 240 mg. A classic 3 + 3 phase-I design was used to determine the recommended phase-II dose (RP2D). Secondary end points included overall response rate, safety profile, survival, and translational research. RESULTS From January 2002 to July 2023, 24 patients were enrolled and 23 were evaluable for efficacy, excluding one patient because of noncompliant dose. All patients were treated with the initial dose level, then the RP2D. Toxicity was mild, with the most frequent being grade 4 toxicity neutropenia (16.7%) and thrombocytopenia (8.3%), while no grade 5 toxicity occurred. The centrally reviewed objective response rate was as follows: partial response 56.5%, stable disease 39.1%, and progression 4.4%. The 6-month progression-free survival (PFS) rate was 80% (95% CI, 63 to 98). Dynamic increases of HMGB1 in blood significantly correlated with longer PFS. CONCLUSION This scheme of doxorubicin, dacarbazine, and nivolumab is feasible and well tolerated. Clinical activity is encouraging and the prognostic impact of HMGB1 supports the relevance of ICD activation. Further clinical research is already underway with this concept in leiomyosarcoma.
Collapse
Affiliation(s)
- Javier Martin-Broto
- Medical Oncology Department, Fundacion Jimenez Diaz University Hospital, University Hospital General de Villalba, Madrid, Spain
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | | | - David Moura
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Rafael Ramos
- Pathology Department, Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | | | - Irene Carrasco
- Medical Oncology Department, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Ana Sebio
- Medical Oncology Department, Hospital Sant Pau, Barcelona, Spain
| | | | - Antonio Gutierrez
- Hematology Department, Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | | | | | - Josefina Cruz
- Medical Oncology Department, Hospital Universitario de Canarias, La Laguna, Spain
| | - Claudia Valverde
- Medical Oncology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Nadia Hindi
- Medical Oncology Department, Fundacion Jimenez Diaz University Hospital, University Hospital General de Villalba, Madrid, Spain
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| |
Collapse
|
13
|
Liu H, Hao Q, Wang X, Cheng M, Qiu F, Zhou B. Efficacy and safety of the combination of anlotinib and envafolimab in the treatment of unresectable or metastatic liposarcoma: findings from a single-center retrospective study. Front Oncol 2025; 14:1502945. [PMID: 39868378 PMCID: PMC11757892 DOI: 10.3389/fonc.2024.1502945] [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: 09/27/2024] [Accepted: 12/19/2024] [Indexed: 01/28/2025] Open
Abstract
Objective To evaluate the efficacy and safety of anlotinib combined with envafolimab in the treatment of unresectable or metastatic liposarcoma. Methods This single-center, retrospective study enrolled 15 patients with unresectable or metastatic liposarcoma, who were treated at the Retroperitoneal Tumor Surgery Research Center of Qingdao University Affiliated Hospital between April 2022 and November 2023. The treatment regimen consisted of anlotinib combined with envafolimab. Treatment efficacy was evaluated using the Response Evaluation Criteria in Solid Tumors version 1.1. Treatment-related adverse events (TRAEs) were assessed using Common Terminology Criteria for Adverse Events version 5.0. Results A total of 15 patients with unresectable or metastatic liposarcoma were included; among them, seven were male (46.7%) and eight were female (53.3%), with a median age of 55 years. The pathological subtype distribution was as follows: three (20.0%) patients with well-differentiated liposarcoma, 11 (73.3%) patients with dedifferentiated liposarcoma, and one (6.7%) patient with myxoid liposarcoma. At 12 weeks post-diagnosis, none of the patients achieved a complete response. The objective response rate was 6.7%, with one patient (6.7%) achieving a partial response. Disease stability was observed in 10 (66.6%) patients, which corresponded to a disease control rate of 73.3%. Disease progression occurred in four (26.7%) patients. The median follow-up time was 16.9 months and the median progression-free survival time was 14.2 months. Seven patients experienced TRAEs, of whom three (42.2%) had grade 3-4 TRAEs. The most common TRAEs were liver function abnormalities, hypertension, and fatigue. Conclusion Anlotinib combined with envafolimab demonstrates promising efficacy and manageable safety in treating unresectable or metastatic liposarcoma.
Collapse
Affiliation(s)
- Hongliang Liu
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qisheng Hao
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xi Wang
- Department of Oncology, Women and Children’s Hospital Affiliated to Qingdao University, Qingdao, China
| | - Mengxing Cheng
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fabo Qiu
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery & Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
14
|
Zeng Y, Fu BM. Angiogenesis and Microvascular Permeability. Cold Spring Harb Perspect Med 2025; 15:a041163. [PMID: 38692737 PMCID: PMC11694756 DOI: 10.1101/cshperspect.a041163] [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: 05/03/2024]
Abstract
Angiogenesis, the formation of new blood microvessels, is a necessary physiological process for tissue generation and repair. Sufficient blood supply to the tissue is dependent on microvascular density, while the material exchange between the circulating blood and the surrounding tissue is controlled by microvascular permeability. We thus begin this article by reviewing the key signaling factors, particularly vascular endothelial growth factor (VEGF), which regulates both angiogenesis and microvascular permeability. We then review the role of angiogenesis in tissue growth (bone regeneration) and wound healing. Finally, we review angiogenesis as a pathological process in tumorigenesis, intraplaque hemorrhage, cerebral microhemorrhage, pulmonary fibrosis, and hepatic fibrosis. Since the glycocalyx is important for both angiogenesis and microvascular permeability, we highlight the role of the glycocalyx in regulating the interaction between tumor cells and endothelial cells (ECs) and VEGF-containing exosome release and uptake by tumor-associated ECs, all of which contribute to tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Bingmei M Fu
- Department of Biomedical Engineering, The City College of the City University of New York, New York, New York 10031, USA
| |
Collapse
|
15
|
Zhao S, Du H, Zhang Z, Yang J, Zhou Y, Xiao G, Ma H, Lan C, Liang J, Yang K, Wen L. SMARCB1/INI-1-Deficient sinonasal carcinoma demonstrates a poor prognosis but favorable clinical outcomes after PD-1/PD-L1 inhibitor therapy: A case series. Sci Prog 2025; 108:368504251315075. [PMID: 39886754 PMCID: PMC11783480 DOI: 10.1177/00368504251315075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Despite advances in multimodal cancer therapy, such as combining radical surgery with high-intensity chemoradiotherapy, for SMARCB1/INI-1-deficient sinonasal carcinoma (SDSC), the prognosis of patients remains poor. Immunotherapy is gaining increasing popularity as a novel treatment strategy for patients with SMARCB1/INI-1-deficient tumors. Herein, we report on the management of three patients with SDSC who received PD-1/PD-L1 inhibitor therapy as a part of multimodal therapy based on surgery and chemoradiotherapy. All three patients survived and demonstrated good clinical remission and disease control. To our knowledge, this is the first case series reporting the use of immunotherapy to improve clinical outcomes in neoadjuvant, adjuvant, and late first-line stages of treatment in patients with SDSC. Furthermore, we reviewed the relevant literature and further explored the correlation between SMARCB1/INI-1 deletion and immunotherapy.
Collapse
Affiliation(s)
- Shuhan Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Hao Du
- Department of Neurosurgery, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhanjie Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Jinsong Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - You Zhou
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guixiang Xiao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Ma
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Caini Lan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Jinzi Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Lu Wen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| |
Collapse
|
16
|
Jin X, Ji R, Wu Y, Wang J, Chen X. Treatment of Retroperitoneal Well-Differentiated Liposarcoma with Combination of Penpulimab and Anlotinib: A Case Report and Literature Review. Niger J Clin Pract 2025; 28:128-133. [PMID: 40326946 DOI: 10.4103/njcp.njcp_817_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 12/16/2024] [Indexed: 05/07/2025]
Abstract
To observe the efficacy of combination therapy with Penpulimab and Anlotinib in the treatment of retroperitoneal well-differentiated liposarcoma. Retrospective analysis of clinical data of a patient with retroperitoneal well-differentiated soft tissue sarcoma admitted to Shaoxing People's Hospital, and review of relevant literature. The patient is a young male who experienced recurrence of retroperitoneal well-differentiated liposarcoma after two surgeries. After first-line treatment with Anlotinib combined with Penpulimab, the patient achieved almost complete remission with a progression free survival period of about 16 months. The first-line treatment of retroperitoneal well-differentiated soft tissue sarcoma using Anlotinib combined with Penpulimab resulted in a good prognosis.
Collapse
Affiliation(s)
- X Jin
- Medical Oncology, Shaoxing People's Hospital, Shaoxing, China
| | - R Ji
- Department of Radiation Oncology, The Second Hospital of Shaoxing, Shaoxing, China
| | - Y Wu
- College of Medicine, Shaoxing University, Shaoxing, China
| | - J Wang
- Medical Oncology, Shaoxing People's Hospital, Shaoxing, China
| | - X Chen
- Medical Oncology, Shaoxing People's Hospital, Shaoxing, China
| |
Collapse
|
17
|
Hayes AJ, Nixon IF, Strauss DC, Seddon BM, Desai A, Benson C, Judson IR, Dangoor A. UK guidelines for the management of soft tissue sarcomas. Br J Cancer 2025; 132:11-31. [PMID: 38734790 PMCID: PMC11724041 DOI: 10.1038/s41416-024-02674-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: 01/08/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 05/13/2024] Open
Abstract
Soft tissue sarcomas (STS) are rare tumours arising in mesenchymal tissues and can occur almost anywhere in the body. Their rarity, and the heterogeneity of subtype and location, means that developing evidence-based guidelines is complicated by the limitations of the data available. This makes it more important that STS are managed by expert multidisciplinary teams, to ensure consistent and optimal treatment, recruitment to clinical trials, and the ongoing accumulation of further data and knowledge. The development of appropriate guidance, by an experienced panel referring to the evidence available, is therefore a useful foundation on which to build progress in the field. These guidelines are an update of the previous versions published in 2010 and 2016 [1, 2]. The original guidelines were drawn up by a panel of UK sarcoma specialists convened under the auspices of the British Sarcoma Group (BSG) and were intended to provide a framework for the multidisciplinary care of patients with soft tissue sarcomas. This iteration of the guidance, as well as updating the general multidisciplinary management of soft tissue sarcoma, includes specific sections relating to the management of sarcomas at defined anatomical sites: gynaecological sarcomas, retroperitoneal sarcomas, breast sarcomas, and skin sarcomas. These are generally managed collaboratively by site specific multidisciplinary teams linked to the regional sarcoma specialist team, as stipulated in the recently published sarcoma service specification [3]. In the UK, any patient with a suspected soft tissue sarcoma should be referred to a specialist regional soft tissues sarcoma service, to be managed by a specialist sarcoma multidisciplinary team. Once the diagnosis has been confirmed using appropriate imaging and a tissue biopsy, the main modality of management is usually surgical excision performed by a specialist surgeon, combined with pre- or post-operative radiotherapy for tumours at higher risk for local recurrence. Systemic anti-cancer therapy (SACT) may be utilised in cases where the histological subtype is considered more sensitive to systemic treatment. Regular follow-up is recommended to assess local control, development of metastatic disease, and any late effects of treatment.
Collapse
Affiliation(s)
- Andrew J Hayes
- The Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK.
- The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Ioanna F Nixon
- Department of Clinical Oncology, The Beatson West of Scotland Cancer Center, Glasgow, G12 0YN, UK
| | - Dirk C Strauss
- The Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Beatrice M Seddon
- Department of Medical Oncology, University College London Hospital NHS Foundation Trust, London, NW1 2BU, UK
| | - Anant Desai
- The Midlands Abdominal and Retroperitoneal Sarcoma Unit, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
| | - Charlotte Benson
- The Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Ian R Judson
- The Institute of Cancer Research, London, SM2 5NG, UK
| | - Adam Dangoor
- Department of Medical Oncology, University Hospitals Bristol & Weston NHS Foundation Trust, Bristol, BS1 3NU, UK
| |
Collapse
|
18
|
Mavroeidis L, Napolitano A, Huang P, Jones RL. Novel Therapeutics in Soft Tissue Sarcoma. Cancers (Basel) 2024; 17:10. [PMID: 39796641 PMCID: PMC11718850 DOI: 10.3390/cancers17010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
There has been noteworthy progress in molecular characterisation and therapeutics in soft tissue sarcomas. Novel agents have gained regulatory approval by the FDA. Examples are the tyrosine kinase inhibitors avapritinib and ripretinib in gastrointestinal stromal tumours (GIST), the immune check point inhibitor atezolizumab in alveolar soft part tissue sarcoma, the γ-secretase inhibitor nirogacestat in desmoid tumours, the NTRK inhibitors larotrectinib and entrectinib in tumours with NTRK fusions, the mTOR inhibitor nab-sirolimus in PEComa, and the EZH-2 inhibitor tazemetostat in epithelioid sarcoma. The FDA has also recently granted accelerated approval for autologous T-cell therapy with afami-cel in patients with HLA-A*02 and MAGE-A4-expressing synovial sarcoma. There are other promising treatments that are still investigational, such as MDM2 and CDK4/6 inhibitors in well-/dedifferentiated liposarcoma, immune checkpoint inhibitors in the head and neck angiosarcoma and a subset of patients with undifferentiated pleomorphic sarcoma, and PARP inhibitors in leiomyosarcoma. The challenges in drug development in soft tissue sarcoma are due to the rarity and the molecular heterogeneity of the disease and the fact that many subtypes are associated with complex karyotypes or non-targetable molecular alterations. We believe that progress maybe possible with a better understanding of the complex biology, the development of novel compounds for difficult targets such as proteolysis targeting chimeras (Protacs), the utilisation of modern clinical trial designs, and enhanced collaboration of academia with industry to develop treatments with a strong biologic rationale.
Collapse
Affiliation(s)
- Leonidas Mavroeidis
- Sarcoma Unit, The Royal Marsden Hospital and Institute of Cancer Research, London SW3 6JZ, UK
| | | | | | | |
Collapse
|
19
|
Pan M, Zhou M, Xie L, Bui N, Ganjoo K. Recent advances in sarcoma therapy: new agents, strategies and predictive biomarkers. J Hematol Oncol 2024; 17:124. [PMID: 39696530 PMCID: PMC11656826 DOI: 10.1186/s13045-024-01650-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
Soft tissue and bone sarcomas are a heterogenous group of uncommon mesenchymal tumors with high unmet needs for novel therapeutic and diagnostic strategies. Despite many challenges that persist, innovative therapeutics are emerging. Here we provide a review of the studies presented at the 2024 American Society of Clinical Oncology annual meeting that were focused on sarcoma. There were many outstanding studies that were reported at the meeting. We begin by discussing the clinical studies on soft tissue sarcoma (STS) that included multiple histology subtypes, followed by highlighting developments in cellular therapy, before delving into specific STS histologic subtypes followed by a section covering the studies that were focused on predictive biomarkers. We conclude by discussing the studies in bone sarcomas. Some of the studies discussed here are likely to be practice changing. Some of the early-phase clinical trials have shown encouraging results.
Collapse
Affiliation(s)
- Minggui Pan
- Department of Medicine Division of Oncology, Sarcoma Program, Stanford University School of Medicine, Stanford, Palo Alto, CA, 94305, USA.
| | - Maggie Zhou
- Department of Medicine Division of Oncology, Sarcoma Program, Stanford University School of Medicine, Stanford, Palo Alto, CA, 94305, USA
| | - Lu Xie
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
| | - Nam Bui
- Department of Medicine Division of Oncology, Sarcoma Program, Stanford University School of Medicine, Stanford, Palo Alto, CA, 94305, USA
| | - Kristen Ganjoo
- Department of Medicine Division of Oncology, Sarcoma Program, Stanford University School of Medicine, Stanford, Palo Alto, CA, 94305, USA
| |
Collapse
|
20
|
Movva S, Seier K, Avutu V, Banks LB, Chan J, Chi P, Dickson MA, Gounder MM, Kelly CM, Keohan ML, Maki R, Rosenbaum E, Salcito T, Rodriguez K, Dempsey R, Meyers PA, Cohen SM, Hensley ML, Konner JA, Schram AM, Lefkowitz RA, Erinjeri JP, Qin LX, Tap WD, D'Angelo SP. Histology-Specific Clinical Trial of Lenvatinib and Pembrolizumab in Patients with Sarcoma. Clin Cancer Res 2024; 30:5612-5619. [PMID: 39405335 PMCID: PMC11730159 DOI: 10.1158/1078-0432.ccr-24-2519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/20/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
PURPOSE Survival of patients with metastatic sarcoma remains poor, and there is a pressing need for new therapies. Most sarcoma subtypes are not responsive to immune checkpoint inhibition alone. Lenvatinib, a multireceptor tyrosine kinase inhibitor targeting tumor vasculature, has an immunomodulatory activity that contributes to its antitumor effects. Therefore, we hypothesized that a combination of lenvatinib and pembrolizumab would lead to improved clinical outcomes in patients with sarcoma. PATIENTS AND METHODS This was an open-label, single-arm study of lenvatinib and pembrolizumab in the following cohorts: (A) leiomyosarcoma, (B) undifferentiated pleomorphic sarcoma (UPS), (C) vascular sarcomas (angiosarcoma and epithelioid hemangioendothelioma), (D) synovial sarcoma or malignant peripheral nerve sheath tumor (MPNST), and (E) bone sarcomas (osteosarcoma and chondrosarcoma). The primary endpoint was the best overall response (BOR) rate documented by RECIST v1.1 by 27 weeks in each cohort, with a threshold of ≥2 responses among 10 patients. Secondary endpoints included progression-free survival, overall survival, duration of response, and safety. RESULTS Forty-six patients were evaluable for the primary endpoint, which was met in the UPS and MPNST/synovial cohorts (BOR rates by 27 weeks of 25% and 30%, respectively). There were seven partial responses overall with additional responses noted in angiosarcoma and osteosarcoma. Treatment-related adverse events of any grade and grade 3 or higher occurred in 50/51 (98%) and 29/51 (57%) of patients, respectively. CONCLUSIONS We observed durable responses in MPNST, synovial sarcoma, and osteosarcoma. Patients with UPS and angiosarcoma also responded. Further exploration of this approach is warranted to confirm activity and determine optimal dosing schedules.
Collapse
Affiliation(s)
- Sujana Movva
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Kenneth Seier
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Viswatej Avutu
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lauren B. Banks
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Jason Chan
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Ping Chi
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mark A. Dickson
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Mrinal M. Gounder
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Ciara M. Kelly
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Mary L. Keohan
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert Maki
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Evan Rosenbaum
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Tiffany Salcito
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kaithleen Rodriguez
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Rebecca Dempsey
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Paul A. Meyers
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Seth M. Cohen
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Gynecology Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Martee L. Hensley
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Gynecology Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jason A. Konner
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Gynecology Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alison M. Schram
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Gynecology Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert A. Lefkowitz
- Body Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Radiology, Weill Cornell Medical College, New York, NY
| | - Joseph P. Erinjeri
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Radiology, Weill Cornell Medical College, New York, NY
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Li-Xuan Qin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - William D. Tap
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Sandra P. D'Angelo
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
21
|
Tan Z, Wu Y, Fan Z, Gao T, Guo W, Bai C, Xue R, Li S, Zhang L, Wang X, Jia L, Liu J. Anlotinib plus TQB2450, a PD-L1 Antibody, in Patients with Advanced Alveolar Soft Part Sarcoma: A Single-Arm, Phase II Trial. Clin Cancer Res 2024; 30:5577-5583. [PMID: 39453774 DOI: 10.1158/1078-0432.ccr-24-2444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/22/2024] [Accepted: 10/23/2024] [Indexed: 10/27/2024]
Abstract
PURPOSE Alveolar soft part sarcoma (ASPS) is an ultrarare soft-tissue sarcoma with a high rate of metastasis and no established treatment. This study aimed to explore the efficacy and safety of anlotinib (a tyrosine kinase inhibitor) and TQB2450 (a PD-L1 inhibitor) in patients with ASPS. PATIENTS AND METHODS This single-arm, phase II study evaluated the efficacy of TQB2450, an anti-PD-L1 agent, combined with anlotinib, a tyrosine kinase inhibitor, in adults with advanced ASPS. TQB2450 was given intravenously (1,200 mg) on day 1, and anlotinib (12 mg/day) was taken orally from day 1 to 14 every 3 weeks. The primary endpoint was overall response rate, with secondary endpoints including duration of response, progression-free survival, and overall survival. Lymphocyte infiltration and tertiary lymphoid structure (TLS) were also analyzed as potential prognostic biomarkers. RESULTS The study enrolled 29 patients, of whom 28 were evaluable (one withdrew because of acute pancreatitis). An objective response was achieved in 82.1% of patients, including 4 complete and 19 partial responses. The median time to response was 2.8 months, and the duration of response was not reached, with an estimated median progression-free survival of 35.2 months. Grade 3 to 4 treatment-related adverse events occurred in 44.8% of patients, with no study-related deaths. Responders had a higher proportion of TLS area, TLS density, and CD20-positive immune cells. CONCLUSIONS The combination of anlotinib and TQB2450 is effective and tolerable in patients with ASPS. TLS may serve as a prognostic biomarker, meriting further investigation.
Collapse
Affiliation(s)
- Zhichao Tan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhengfu Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Tian Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
| | - Chujie Bai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ruifeng Xue
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Shu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Lu Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xinyu Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ling Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiayong Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
22
|
Moura DS, Lopez-Marti JM, Benesova I, de Andrea C, di Lernia D, Lacerenza S, Mondaza-Hernandez JL, Martin-Ruiz M, Ramirez-Calvo M, Grignani G, Martinez-Trufero J, Redondo A, Valverde C, Stacchiotti S, Lopez-Pousa A, Lopez-Guerrero JA, Gutierrez A, Encinas-Tobajas V, Hindi N, Sangiolo D, Lopez-Martin JA, Strizova ZO, Martin-Broto J. Predictive and Dynamic Signature for Antiangiogenics in Combination with a PD1 Inhibitor in Soft-Tissue Sarcoma: Correlative Studies Linked to the IMMUNOSARC Trial. Clin Cancer Res 2024; 30:5192-5206. [PMID: 39283727 DOI: 10.1158/1078-0432.ccr-24-1782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/26/2024] [Accepted: 09/11/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE The IMMUNOSARC trial combined an antiangiogenic agent (sunitinib) with a PD1 inhibitor (nivolumab) in advanced sarcomas. Here, we present the first correlative studies of the soft-tissue sarcoma cohort enrolled in this trial. EXPERIMENTAL DESIGN Formalin-fixed paraffin-embedded and peripheral blood samples were collected at baseline and week 13. Formalin-fixed paraffin-embedded samples were used for transcriptomics and multiplex immunofluorescence, whereas peripheral blood samples were used for multiplexed immunoassays. Flow cytometry and Luminex assays were performed to validate translational findings in tumor-isolated cells and peripheral blood mononuclear cells derived from patients. RESULTS The density of intratumoral CD8+ T cells, measured by multiplexed immunophenotyping, was significantly increased after treatment. This augment was accompanied by the dynamic significant increase in the gene expressions of CD86, CHI3L1, CXCL10, CXCL9, LAG3, and VCAM1 and the decrease in the expression levels of NR4A1. In peripheral blood, 12 proteins were significantly modulated by treatment at week 13. A score integrating the dynamic expression of the 7 genes and the 12 soluble factors separated 2 groups with distinct progression-free survival (PFS): 4.1 months [95% confidence interval, 3.5-not reached (NR)] versus 17 months (95% confidence interval, 12.0-NR), P = 0.014. This molecular score was predictive of PFS when applied to the normalized data determined in the baseline samples. CONCLUSIONS Treatment with sunitinib and nivolumab inflamed the sarcoma microenvironment, increasing CD8+ T-cell density and the expression of several genes/proteins with relevance in the response to PD1 inhibitors. A molecular signature identified two groups of patients with distinct PFS for the combination of antiangiogenics plus PD1 inhibitor therapy.
Collapse
Affiliation(s)
- David S Moura
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Jesus M Lopez-Marti
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Iva Benesova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Carlos de Andrea
- Department of Pathology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Davide di Lernia
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Serena Lacerenza
- Institute of Biomedicine of Seville (IBiS; US, CSIC, HUVR), Seville, Spain
| | | | - Marta Martin-Ruiz
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | | | - Giovanni Grignani
- Medical Oncology Unit, Città della Salute e della Scienza di Torino, Turin, Italy
| | | | - Andres Redondo
- Department of Medical Oncology, University Hospital La Paz, Madrid, Spain
| | - Claudia Valverde
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Silvia Stacchiotti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - José A Lopez-Guerrero
- Fundación Institute Valenciano of Oncology, Valencia, Spain
- Joint Cancer Research Unit IVO-CIPF, Valencia, Spain
- Department of Pathology, Medical School of the Catholic University of Valencia, Valencia, Spain
| | - Antonio Gutierrez
- Department of Hematology, University Hospital Son Espases, Mallorca, Spain
| | | | - Nadia Hindi
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
- Department of Medical Oncology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Turin, Italy
| | - Jose A Lopez-Martin
- Department of Medical Oncology, University Hospital 12 de Octubre, Madrid, Spain
| | - Zuzana Ozaniak Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Javier Martin-Broto
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
- Department of Medical Oncology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| |
Collapse
|
23
|
Cruz-Ramos M, Cabrera-Nieto SA, Murguia-Perez M, Fajardo-Espinoza FS. The Role of Adenosine in Overcoming Resistance in Sarcomas. Int J Mol Sci 2024; 25:12209. [PMID: 39596278 PMCID: PMC11594806 DOI: 10.3390/ijms252212209] [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: 10/07/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Resistance to systemic therapies in sarcomas poses a significant challenge to improving clinical outcomes. Recent research has concentrated on the tumor microenvironment's role in sarcoma progression and treatment resistance. This microenvironment comprises a variety of cell types and signaling molecules that influence tumor behavior, including proliferation, metastasis, and resistance to therapy. Adenosine, abundant in the tumor microenvironment, has been implicated in promoting immunosuppression and chemoresistance. Targeting adenosine receptors and associated pathways offers a novel approach to enhancing immune responses against tumors, potentially improving immunotherapy outcomes in cancers, including sarcomas. Manipulating adenosine signaling also shows promise in overcoming chemotherapy resistance in these tumors. Clinical trials investigating adenosine receptor antagonists in sarcomas have fueled interest in this pathway for sarcoma treatment. Ultimately, a comprehensive understanding of the tumor and vascular microenvironments, as well as the adenosine pathway, may open new avenues for improving treatment outcomes and overcoming resistance in sarcoma. Further studies and clinical trials are crucial to validate these findings and optimize therapeutic strategies, particularly for osteosarcoma. This study provides a literature review exploring the potential role of the adenosine pathway in sarcomas.
Collapse
Affiliation(s)
- Marlid Cruz-Ramos
- Investigadora por México del Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Mexico City 03940, Mexico
- Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan 52786, Mexico; (S.A.C.-N.); (F.S.F.-E.)
| | - Sara Aileen Cabrera-Nieto
- Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan 52786, Mexico; (S.A.C.-N.); (F.S.F.-E.)
| | - Mario Murguia-Perez
- Laboratorio de Anatomía Patológica e Inmunohistoquímica Especializada DIME, Hospital Médica Campestre, León 37180, Mexico;
- Departamento de Patología Quirúrgica, UMAE Hospital de Especialidades No. 1, Centro Médico Nacional Bajío, Instituto Mexicano del Seguro Social, León 37328, Mexico
| | | |
Collapse
|
24
|
Mondaza-Hernandez JL, Hindi N, Fernandez-Serra A, Ramos R, Gonzalez-Cámpora R, Gómez-Mateo MC, Martinez-Trufero J, Lavernia J, Lopez-Pousa A, Laínez N, Martinez-Garcia J, Valverde C, Vaz-Salgado MÁ, Garcia-Plaza G, Marin-Borrero I, Carrillo-Garcia J, Martin-Ruiz M, Romero P, Gutierrez A, López-Guerrero JA, Moura DS, Martin-Broto J. Exploratory analysis of immunomodulatory factors identifies L1CAM as a prognostic marker in alveolar soft-part sarcoma. Ther Adv Med Oncol 2024; 16:17588359241293951. [PMID: 39502403 PMCID: PMC11536517 DOI: 10.1177/17588359241293951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Background Alveolar soft-part sarcoma (ASPS) is a rare tumor driven by the ASPSCR1-TFE3 fusion protein, with a propensity for metastasis. Prognostic factors remain poorly understood, and traditional chemotherapies are largely ineffective. Recent interest lies in immune checkpoint inhibitors (ICIs), yet predictive biomarkers for treatment response are lacking. Previous studies have shown promising results with ICIs in ASPS, indicating a need for further investigation into biomarkers associated with immune response. Objectives To identify prognostic biomarkers in ASPS and to explore the role of immune-related markers, particularly L1CAM, in predicting patient outcomes. Design A retrospective cohort study of 19 ASPS patients registered in the GEIS database. The study involved the collection of clinical and histopathological data, followed by an analysis of immune markers and gene expression profiles to identify potential prognostic indicators. Methods Clinical and histopathological data were retrospectively collected from the GEIS-26 study cohort of 19 ASPS patients. Immunohistochemistry was performed to evaluate immune markers programmed death-1 ligand (PD-L1), programmed death-1, FAS, FASL, CD8, CD3, and CD4. An HTG ImmunOncology panel was conducted on formalin-fixed paraffin-embedded samples to explore gene expression. Effects of differentially expressed genes on survival were explored by Kaplan-Meier. Results PD-L1 positivity was widely observed (63%) in tumors, and CD8+ lymphocytic infiltration was common. High CD8 density correlated with greater overall survival (OS) while not statistically significant. No associations were found for other immune markers. L1CAM was identified as differentially expressed in patients with low CD8 infiltration and correlated negatively with OS. Conclusion High L1CAM expression correlated with poorer OS, highlighting its potential as a prognostic marker and therapeutic target in ASPS. Immunomodulatory interventions may hold promise, as evidenced by PD-L1 expression and CD8+ infiltration. Further research, including larger cohorts and international collaborations, is needed to validate these findings and explore therapeutic strategies targeting L1CAM in ASPS.
Collapse
Affiliation(s)
- José L. Mondaza-Hernandez
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Nadia Hindi
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| | | | - Rafael Ramos
- Pathology Department, University Hospital Son Espases, Mallorca, Spain
| | | | | | | | - Javier Lavernia
- Medical Oncology Department, Fundación Instituto Valenciano de Oncologia, Valencia, Spain
| | | | - Nuria Laínez
- Department of Medical Oncology, Complejo Hospitalario de Navarra Pamplona, Spain
| | | | - Claudia Valverde
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
| | - María Ángeles Vaz-Salgado
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, CIBERONC, Madrid, Spain
| | - Gabriel Garcia-Plaza
- Department of Surgery, Hospital Universitario Insular, Las Palmas de Gran Canaria, Spain
| | - Isabel Marin-Borrero
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
| | - Jaime Carrillo-Garcia
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Marta Martin-Ruiz
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Pablo Romero
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Antonio Gutierrez
- Hematology Department, Hospital Son Espases/IdISBa, Palma de Mallorca, Spain
| | - Jose A. López-Guerrero
- Molecular Biology Department, Fundación Instituto Valenciano de Oncologia, Valencia, Spain
| | - David S. Moura
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
| | - Javier Martin-Broto
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Reyes Católicos 2, Madrid 28040, Spain
- University Hospital General de Villalba, Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| |
Collapse
|
25
|
Ishiguro N, Nakagawa M. ASPSCR1::TFE3-mediated upregulation of insulin receptor substrate 2 (IRS-2) activates PI3K/AKT signaling and promotes malignant phenotype. Int J Biochem Cell Biol 2024; 176:106676. [PMID: 39419345 DOI: 10.1016/j.biocel.2024.106676] [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: 05/24/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/19/2024]
Abstract
The ASPSCR1::TFE3 fusion gene, resulting from chromosomal translocation, is detected in alveolar soft part sarcoma (ASPS) and a subset of renal cell carcinomas (RCC). The ASPSCR1::TFE3 oncoprotein, functioning as an aberrant transcription factor, contributes to tumor development and progression by inappropriately upregulating target genes. Here, we identified insulin receptor substrate 2 (IRS-2), a cytoplasmic adaptor protein, as a novel transcriptional target of ASPSCR1::TFE3. Ectopic expression of ASPSCR1::TFE3 led to increased IRS-2 mRNA and protein levels. Chromatin immunoprecipitation and luciferase assays demonstrated that ASPSCR1::TFE3 bound to the IRS-2 promoter region and enhanced its transcription. Moreover, IRS-2 was highly expressed in the ASPSCR1::TFE3-positive RCC cell line FU-UR1, while small interfering RNA-mediated depletion of ASPSCR1::TFE3 markedly decreased IRS-2 mRNA and protein levels. Functionally, IRS-2 knockdown attenuated activation of the PI3K/AKT pathway and reduced proliferation, migration, invasion, adhesion, and clonogenicity in FU-UR1 cells. Pharmacological inhibition of IRS-2 also reduced AKT activation as well as cell viability, clonogenicity, migration, invasion, and adhesion. These findings suggest that IRS-2, regulated by ASPSCR1::TFE3, promotes tumor progression by activating PI3K/AKT signaling and enhancing the malignant phenotype.
Collapse
MESH Headings
- Humans
- Insulin Receptor Substrate Proteins/metabolism
- Insulin Receptor Substrate Proteins/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics
- Signal Transduction
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphatidylinositol 3-Kinases/genetics
- Cell Line, Tumor
- Up-Regulation
- Gene Expression Regulation, Neoplastic
- Cell Movement/genetics
- Cell Proliferation
- Phenotype
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/metabolism
- Kidney Neoplasms/pathology
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Sarcoma, Alveolar Soft Part/pathology
- Sarcoma, Alveolar Soft Part/genetics
- Sarcoma, Alveolar Soft Part/metabolism
- Intracellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Naoko Ishiguro
- Department of Pathobiological Science and Technology, Faculty of Medicine, Tottori University, Yonago, Japan.
| | - Mayumi Nakagawa
- Department of Pathobiological Science and Technology, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
26
|
Li AY, Bu J, Xiao HN, Zhao ZY, Zhang JL, Yu B, Li H, Li JP, Xiao T. Two-step consensus clustering approach to immune cell infiltration: An integrated exploration and validation of prognostic and immune implications in sarcomas. Heliyon 2024; 10:e38253. [PMID: 39492897 PMCID: PMC11531637 DOI: 10.1016/j.heliyon.2024.e38253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 11/05/2024] Open
Abstract
To conduct a comprehensive investigation of the sarcoma immune cell infiltration (ImmCI) patterns and tumoral microenvironment (TME). We utilized transcriptomic, clinical, and mutation data of sarcoma patients (training cohort) obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) server. Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) and Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) algorithms were applied to decipher the immune cell infiltration landscape and TME profiles of sarcomas. An unsupervised clustering method was utilized for classifying ImmCI clusters (initial clustering) and ImmCI-based differentially expressed gene-driven clusters (secondary clustering). Mortality rates and immune checkpoint gene levels was analyzed among the identified clusters. We calculated the ImmCI score through principal component analysis. The tumor immune dysfunction evaluation (TIDE) score was also employed to quantify immunotherapy efficacy between two ImmCI score groups. We further validated the biomarkers for ImmCI and gene-driven clusters via experimental verification and the accuracy of the ImmCI score in predicting survival outcomes and immunotherapy efficacy by external validation cohorts (testing cohort). We demonstrated that ImmCI cluster A and gene-driven cluster A, were beneficial prognostic biomarkers and indicators of immune checkpoint blockade response in sarcomas via in-silico and laboratory experiments. Additionally, the ImmCI score exhibited independent prognostic significance and was predictive of immunotherapy response. Our research underscores the clinical significance of ImmCI scores in identifying sarcoma patients likely to respond to immunotherapy.
Collapse
Affiliation(s)
- Ao-Yu Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Jie Bu
- Department of Orthopedics, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Hui-Ni Xiao
- Department of Gastroenterology, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Zi-Yue Zhao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Jia-Lin Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Bin Yu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Hui Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Jin-Ping Li
- Department of Orthopedics, Changsha Central Hospital, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Tao Xiao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| |
Collapse
|
27
|
Nie G, Liu C, Tian Z. Comprehensive analysis of prognostic and immunological role of basement membrane-related genes in soft tissue sarcoma. Immun Inflamm Dis 2024; 12:e70037. [PMID: 39392257 PMCID: PMC11467964 DOI: 10.1002/iid3.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Soft tissue sarcoma (STS) represents highly multifarious malignant tumors that often occur in adolescents and have a poor prognosis. The basement membrane, as an ancient cellular matrix, was recently proven to play a vital role in developing abundant tumors. The relationship between basement membrane-related genes and STS remains unknown. METHODS Consensus clustering was employed to identify subgroups related to differentially expressed basement membrane-related genes. Cox and least absolute shrinkage and selection operator regression analyses were utilized to construct this novel signature. Then, we established a nomogram and calibration curve, including the risk score and available clinical characteristics. Finally, we carried out functional enrichment analysis and immune microenvironment analysis to investigate enriched pathways and the tumor immune microenvironment related to the novel signature. RESULTS A prognostic predictive signature consisting of eight basement membrane-related genes was established. Kaplan-Meier survival curves demonstrated that the patients in the high-risk group had a poor prognosis. Independent analysis illustrated that this risk model could be an independent prognostic predictor. We validated the accuracy of our signature in the validation data set. In addition, gene set enrichment analysis and immune microenvironment analysis showed that patients with low-risk scores were enriched in some pathways associated with immunity. Finally, in vitro experiments showed significantly differential expression levels of these signature genes in STS cells and PSAT1 could promote the malignant behavior of STS. CONCLUSIONS The novel signature is a promising prognostic predictor for STS. The present study may improve the prognosis and enhance individualized treatment for STS in the future.
Collapse
Affiliation(s)
- Guang‐hua Nie
- Department of Foot and Ankle Surgery, Honghui HospitalXi'an Jiaotong UniversityXi'anChina
| | - Cheng‐yi Liu
- Department of Foot and Ankle Surgery, Honghui HospitalXi'an Jiaotong UniversityXi'anChina
| | - Zhao Tian
- Department of Hand Surgery, Honghui HospitalXi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
28
|
Fernandes I, Dias E Silva D, Segatelli V, Filippi RZ, Carolina de Rezende A, Campregher P, Moura F, Jesus-Garcia R, Pestana RC. Microsatellite Instability and Clinical Use in Sarcomas: Systematic Review and Illustrative Case Report. JCO Precis Oncol 2024; 8:e2400047. [PMID: 39432881 DOI: 10.1200/po.24.00047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 07/01/2024] [Accepted: 07/29/2024] [Indexed: 10/23/2024] Open
Abstract
Sarcomas, a diverse group of malignancies, exhibit substantial heterogeneity in both biological behavior and microenvironment, influencing their response to immunotherapy. Although pembrolizumab is approved for deficient mismatch repair or microsatellite instability-high (dMMR/MSI-H) tumors regardless of histology, there is a paucity of data to support its effectiveness in dMMR/MSI-H sarcomas. This study presents a case of a metastatic undifferentiated pleomorphic sarcoma of the retroperitoneum with dMMR status demonstrating a sustained complete response to pembrolizumab. Our case revealed inconsistencies in identifying dMMR/MSI-H status through next-generation sequencing, immunohistochemistry, and polymerase chain reaction methods. Therefore, we conducted a systematic review to analyze various methods for assessing dMMR/MSI-H and to explore whether pembrolizumab's indications rely on this biomarker.
Collapse
|
29
|
Genevois AL, Carton M, Jean-Denis M, Cyrta J, Corradini N, Metayer L, Chemin-Airiau C, Karanian M, Dufresne A, Pannier S, El Zein S, Defachelles AS, Bompas E, Gantzer J, Honoré C, Noal S, Héritier S, Guillemet C, Serre J, Le Loarer F, Pierron G, Merlin MS, Anract P, Gomez-Mascard A, Llacer C, Ducimetière F, Toulmonde M, Blay JY, Orbach D. Alveolar soft part sarcomas in young patients: The French national NETSARC+ network experience. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108483. [PMID: 38897095 DOI: 10.1016/j.ejso.2024.108483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/27/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND AND AIMS Alveolar soft part sarcoma (ASPS) is an ultra-rare chemo-resistant sarcoma in children, occurring preferentially in young adults. We aimed to describe and compare its clinical presentation and behaviour in children and young adults to determine whether the same therapeutic strategy should be addressed for both populations. METHODS National retrospective multicentre study of children (0-18 years) vs. young adults (19-30 years) included in the "ConticaBase" sarcoma database, treated for ASPS between 2010 and 2019 with pathology reviewed via the NETSARC + network. RESULTS Overall, 45 patients were identified, 19 children (42%) and 26 young adults (58%). All ASPS diagnoses were confirmed with TFE3 rearrangement by immunohistochemistry or FISH. All clinical characteristics were balanced between both populations with frequent metastases at diagnosis (8/19 vs. 10/26). The therapeutic strategy was based on surgery (17/19 vs. 21/26), radiotherapy (8/19 vs. 12/26) ± systemic treatment (8/19 vs. 9/26). In patients with initially localized disease, metastatic relapse occurred only in adults (8/16), whereas metastatic progression was present in both metastatic groups (5/8 vs. 8/10). After a median follow-up of 5.2 years (range, 0.2-12.2), 5-year EFS was 74% [95%CI, 56-96] vs. 47% [30-74] (p = 0.071) respectively, and 5-year OS was 95% [85-100] vs. 85% [70-100] (p = 0.84). For localized tumours, 5-year MFS was 100% [100-100] vs. 60% [39-91] (p = 0.005). The 5-year OS of all patients with metastasis at diagnosis was 80.2% (62.2%-100%). CONCLUSIONS ASPS appears to have the overall same clinical characteristics, but a more aggressive behaviour in young adults than in children. However, despite frequent metastases at diagnosis, long-term survival is high in both groups. Overall, the same therapeutic strategies may be considered for both populations.
Collapse
Affiliation(s)
- Anne-Laure Genevois
- SIREDO Oncology Centre (Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer), Institut Curie, Paris-Saclay University (PSL), Paris, France.
| | - Matthieu Carton
- Unit of Biometry, Institut Curie, DRCI, PSL Research University, Paris, France
| | | | - Joanna Cyrta
- Department of Pathology, Institut Curie, Paris, France
| | - Nadège Corradini
- Department of Paediatric Oncology, Paediatric Haematology and Oncology Institute, Léon Bérard Centre, Lyon, France
| | - Lucy Metayer
- Department of Paediatric and Adolescent Oncology, Gustave Roussy Cancer Centre, Paris-Saclay University, Villejuif, France
| | | | - Marie Karanian
- Department of Biopathology, Léon Bérard Centre, Lyon, France
| | - Armelle Dufresne
- Department of Medical Oncology, Léon Bérard Centre, Lyon, France
| | | | | | | | | | - Justine Gantzer
- Department of Medical Oncology, Strasbourg-Europe Cancer Institute (ICANS), Strasbourg, France
| | - Charles Honoré
- Department of Surgery, Gustave Roussy Cancer Centre, Villejuif, France
| | - Sabine Noal
- Department of Medical Oncology, François Baclesse Centre, Caen, France
| | - Sébastien Héritier
- Department of Paediatric Haematology and Oncology, Trousseau Hospital, Paris, France
| | - Cécile Guillemet
- Department of Medical Oncology, Henry Becquerel Centre, Rouen, France
| | - Jill Serre
- Department of Paediatric Haematology and Oncology, Trousseau Hospital, Tours, France
| | | | | | - Marie-Sophie Merlin
- Department of Paediatric Haematology and Oncology, Brabois Hospital, Nancy, France
| | - Philippe Anract
- Department of Orthopedic and Traumatology, Cochin Hospital, Paris, France
| | | | - Carmen Llacer
- Department of Radiotherapy, Montpellier Cancer Institute, Montpellier, France
| | | | - Maud Toulmonde
- Department of Medical Oncology, Bergonié Institute, Bordeaux, France
| | - Jean-Yves Blay
- Department of Medical Oncology, Léon Bérard Centre, Lyon, France
| | - Daniel Orbach
- SIREDO Oncology Centre (Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer), Institut Curie, Paris-Saclay University (PSL), Paris, France
| |
Collapse
|
30
|
Li S, Sun Q, Bai R, Wang Y, Wang H, Chen H, Dong Y. Real-world efficacy, safety data and predictive clinical parameters for treatment outcomes in advanced soft tissue sarcoma treated with combined immunotherapy and antiangiogenic therapy. BMC Cancer 2024; 24:1028. [PMID: 39164643 PMCID: PMC11337792 DOI: 10.1186/s12885-024-12810-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND The combination of immunotherapy and antiangiogenic therapy has shown potential in the treatment of numerous malignant tumors, but limited evidence was available for soft tissue sarcomas (STS). Therefore, the aim of the present study is to assess the efficacy and safety of immunotherapy in conjunction with antiangiogenic therapy in patients diagnosed with advanced STS (aSTS). METHODS The study enrolled patients with aSTS from January 2014 to October 2022. Eligible participants had previously received anthracycline-based chemotherapy, presented with an anthracycline-resistant sarcoma subtype, or were ineligible for anthracycline treatment due to medical conditions. Following enrollment, these patients received a combination of immunotherapy and antiangiogenic therapy. The primary endpoints were the objective response rate (ORR) and progression-free survival (PFS), while the secondary endpoints included the disease control rate (DCR), overall survival (OS), and the incidence of adverse events. RESULTS Fifty-one patients were included in this cohort study. The median duration of follow-up was 15.8 months. The ORR and DCR were 17.6%, and 76.5%, respectively. The median PFS (mPFS) was 5.8 months (95% CI: 4.8-6.8) for all patients, and the median OS had not been reached as of the date cutoff. Multivariate analysis indicated that Eastern Cooperative Oncology Group performance status of 0-1 and ≤ second-line treatment were positive predictors for both PFS and OS. Patients with alveolar soft part sarcoma or clear cell sarcoma had longer mPFS (16.2 months, 95% CI: 7.8-25.6) when compared to those with other subtypes of STS (4.4 months, 95% CI: 1.4-7.5, P < 0.001). Among the observed adverse events, hypertension (23.5%), diarrhea (17.6%), and proteinuria (17.6%) were the most common, with no treatment-related deaths reported. CONCLUSION The combination of immunotherapy and antiangiogenic agents showed promising efficacy and acceptable toxicity in patients with aSTS, especially those with alveolar soft part sarcoma or clear cell sarcoma.
Collapse
Affiliation(s)
- Shaoli Li
- Department of Medical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, China
| | - Qunan Sun
- Department of Medical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, China
| | - Rui Bai
- Department of Medical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, China
| | - Youping Wang
- Department of Medical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, China
| | - Hui Wang
- Department of Pathology, Institute of Basic Medicine and Cancer (IBMC), the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Haifeng Chen
- Department of Medical Oncology, Shaoxing Second Hospital, Shaoxing, 312000, China
| | - Ying Dong
- Department of Medical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Cancer Center, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
31
|
Pasquali S, Vallacchi V, Lalli L, Collini P, Barisella M, Romagosa C, Bague S, Coindre JM, Dei Tos AP, Palmerini E, Quagliuolo V, Martin-Broto J, Lopez-Pousa A, Grignani G, Blay JY, Beveridge RD, Casiraghi E, Brich S, Renne SL, Bergamaschi L, Vergani B, Sbaraglia M, Casali PG, Rivoltini L, Stacchiotti S, Gronchi A. Spatial distribution of tumour immune infiltrate predicts outcomes of patients with high-risk soft tissue sarcomas after neoadjuvant chemotherapy. EBioMedicine 2024; 106:105220. [PMID: 39018755 PMCID: PMC11287012 DOI: 10.1016/j.ebiom.2024.105220] [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: 10/18/2023] [Revised: 05/22/2024] [Accepted: 06/11/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Anthracycline-based neoadjuvant chemotherapy (NAC) may modify tumour immune infiltrate. This study characterized immune infiltrate spatial distribution after NAC in primary high-risk soft tissue sarcomas (STS) and investigate association with prognosis. METHODS The ISG-STS 1001 trial randomized STS patients to anthracycline plus ifosfamide (AI) or a histology-tailored (HT) NAC. Four areas of tumour specimens were sampled: the area showing the highest lymphocyte infiltrate (HI) at H&E; the area with lack of post-treatment changes (highest grade, HG); the area with post-treatment changes (lowest grade, LG); and the tumour edge (TE). CD3, CD8, PD-1, CD20, FOXP3, and CD163 were analyzed at immunohistochemistry and digital pathology. A machine learning method was used to generate sarcoma immune index scores (SIS) that predict patient disease-free and overall survival (DFS and OS). FINDINGS Tumour infiltrating lymphocytes and PD-1+ cells together with CD163+ cells were more represented in STS histologies with complex compared to simple karyotype, while CD20+ B-cells were detected in both these histology groups. PD-1+ cells exerted a negative prognostic value irrespectively of their spatial distribution. Enrichment in CD20+ B-cells at HI and TE areas was associated with better patient outcomes. We generated a prognostic SIS for each tumour area, having the HI-SIS the best performance. Such prognostic value was driven by treatment with AI. INTERPRETATION The different spatial distribution of immune populations and their different association with prognosis support NAC as a modifier of tumour immune infiltrate in STS. FUNDING Pharmamar; Italian Ministry of Health [RF-2019-12370923; GR-2016-02362609]; 5 × 1000 Funds-2016, Italian Ministry of Health; AIRC Grant [ID#28546].
Collapse
Affiliation(s)
- Sandro Pasquali
- Molecular Pharmacology, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy.
| | - Viviana Vallacchi
- Translational Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Luca Lalli
- Translational Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy.
| | - Paola Collini
- Soft Tissue Tumor Pathology Unit, Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | | | - Cleofe Romagosa
- Pathology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Silvia Bague
- Pathology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jean Michel Coindre
- Department of Pathology, Institut Bergonié, 33000, Bordeaux, France; INSERM U1218 ACTION, Institut Bergonié, 33000, Bordeaux, France
| | - Angelo Paolo Dei Tos
- Surgical Pathology & Cytopathology Unit, Department of Medicine - DIMED, University of Padua, Padua, Italy
| | - Emanuela Palmerini
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Javier Martin-Broto
- Oncology Department, Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Antonio Lopez-Pousa
- Medical Oncology Department, Hospital de la Santa Creu i Sant Pau, Carrer de Sant Quintí, 89, 08041, Barcelona, Spain
| | - Giovanni Grignani
- Medical Oncology Unit, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Jean-Yves Blay
- Centre Léon Bérard & Université Claude Bernard Lyon 1, Lyon, France
| | - Robert Diaz Beveridge
- Department of Cancer Medicine, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Elena Casiraghi
- AnacletoLab, Department of Computer Science "Giovanni degli Antoni", Università degli Studi di Milano, Milan, Italy
| | - Silvia Brich
- Soft Tissue Tumor Pathology Unit, Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Salvatore Lorenzo Renne
- Pathology Department, IRCCS Humanitas Research Hospital, Rozzano, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Laura Bergamaschi
- Translational Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Barbara Vergani
- School of Medicine and Surgery, University of Milano Bicocca, Monza, Italy
| | - Marta Sbaraglia
- Surgical Pathology & Cytopathology Unit, Department of Medicine - DIMED, University of Padua, Padua, Italy
| | - Paolo Giovanni Casali
- Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Licia Rivoltini
- Translational Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy.
| | - Silvia Stacchiotti
- Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Alessandro Gronchi
- Sarcoma Service, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy.
| |
Collapse
|
32
|
Sicinska E, Sudhakara Rao Kola V, Kerfoot JA, Taddei ML, Al-Ibraheemi A, Hsieh YH, Church AJ, Landesman-Bollag E, Landesman Y, Hemming ML. ASPSCR1::TFE3 Drives Alveolar Soft Part Sarcoma by Inducing Targetable Transcriptional Programs. Cancer Res 2024; 84:2247-2264. [PMID: 38657118 PMCID: PMC11250573 DOI: 10.1158/0008-5472.can-23-2115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 02/09/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Alveolar soft part sarcoma (ASPS) is a rare mesenchymal malignancy driven by the ASPSCR1::TFE3 fusion. A better understanding of the mechanisms by which this oncogenic transcriptional regulator drives cancer growth is needed to help identify potential therapeutic targets. In this study, we characterized the transcriptional and chromatin landscapes of ASPS tumors and preclinical models, identifying the essential role of ASPSCR1::TFE3 in tumor cell viability by regulating core transcriptional programs involved in cell proliferation, angiogenesis, and mitochondrial biology. ASPSCR1::TFE3 directly interacted with key epigenetic regulators at enhancers and promoters to support ASPS-associated transcription. Among the effector programs driven by ASPSCR1::TFE3, cell proliferation was driven by high levels of cyclin D1 expression. Disruption of cyclin D1/CDK4 signaling led to a loss of ASPS proliferative capacity, and combined inhibition of CDK4/6 and angiogenesis halted tumor growth in xenografts. These results define the ASPS oncogenic program, reveal mechanisms by which ASPSCR1::TFE3 controls tumor biology, and identify a strategy for therapeutically targeting tumor cell-intrinsic vulnerabilities. Significance: The ASPSCR1::TFE3 fusion propels the growth of alveolar soft part sarcoma by activating transcriptional programs that regulate proliferation, angiogenesis, mitochondrial biogenesis, and differentiation and can be therapeutically targeted to improve treatment.
Collapse
MESH Headings
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics
- Sarcoma, Alveolar Soft Part/genetics
- Sarcoma, Alveolar Soft Part/pathology
- Sarcoma, Alveolar Soft Part/metabolism
- Humans
- Animals
- Mice
- Cell Proliferation/genetics
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Gene Expression Regulation, Neoplastic
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Cyclin-Dependent Kinase 4/genetics
- Cyclin-Dependent Kinase 4/metabolism
- Cyclin-Dependent Kinase 4/antagonists & inhibitors
- Female
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/metabolism
- Intracellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Ewa Sicinska
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Vijaya Sudhakara Rao Kola
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Joseph A. Kerfoot
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Madeleine L. Taddei
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Alyaa Al-Ibraheemi
- Department of Pathology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yi-Hsuan Hsieh
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Alanna J. Church
- Department of Pathology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Esther Landesman-Bollag
- Department of Medicine, Section of Hematology and Oncology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Yosef Landesman
- Cure Alveolar Soft Part Sarcoma International, Brookline, Massachusetts, USA
| | - Matthew L. Hemming
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
33
|
Martin-Broto J, Hindi N, Moura DS. Combination treatment with PD1/PDL-1 inhibitors for sarcomas: state of the art, next questions. Curr Opin Oncol 2024; 36:269-275. [PMID: 38726845 DOI: 10.1097/cco.0000000000001050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
PURPOSE OF REVIEW Only a small fraction of sarcomas exhibit recognized parameters of immune sensitivity, such as tumor mutational burden, PDL-1 expression, or microsatellite instability. Combined strategies aimed to modulate tumor microenvironment to increase the efficacy of PD1/PDL-1 inhibitors in sarcoma. Most explored prospective studies were based on combinations of PD1/PDL-1 inhibitors with antiangiogenics, other immune checkpoints, or chemotherapy. RECENT FINDINGS Results on 6-month PFS rate, median PFS, and ORR in trials using PD1/PDL-1 inhibitors plus antiangiogenics ranged respectively as 46.9-55%, 4.7-7.8 months and 21-36.7%. In combination with other immune checkpoint inhibitors, the results of median PFS and ORR ranged from 2.8-4.1 months and 10-16%, respectively. In combination with chemotherapy, the best results were obtained with doxorubicin-based regimens compared to other agents. Duplet-based chemotherapy plus anti-PD1/PDL-1 obtained the highest ORR (56.2%) compared with doxorubicin (19-36.7%). Currently, the most robust predictive biomarker for anti-PD1/PDL-1 efficacy is the presence of tertiary lymphoid structures (TLS) with mature dendritic cells. SUMMARY Even when direct comparisons between PD1/PDL-1 inhibitor-based combinations and single agents have not been performed yet in sarcoma, some combinations appear promising. Studies controlling heterogeneity by biomarker or histotype selection contribute to an increase in efficacy or knowledge crucial for future comparative trials.
Collapse
Affiliation(s)
- Javier Martin-Broto
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital
- University Hospital General de Villalba
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - Nadia Hindi
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital
- University Hospital General de Villalba
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - David S Moura
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| |
Collapse
|
34
|
Nguyen A, Nguyen A, Fleeting C, Patel A, Bazett N, Hey G, Mandavali A, Brown NJ, Woolridge M, Foreman M, Lucke-Wold B. An Evaluation of Risk Factors for Intracranial Metastases of Sarcomas: A Systematic Review and Meta-Analysis. World Neurosurg 2024; 187:e683-e699. [PMID: 38704144 DOI: 10.1016/j.wneu.2024.04.146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
INTRODUCTION Sarcomas, a group of neoplasms comprising both tissue and bone soft tissue tumors, has an increasing prevalence in recent years. Prognosis significantly hinges on early detection, and if not detected early, may consequently metastasize. This review will be the first systematic review and meta-analysis characterizing the presentation and progression of brain metastases from bone and soft tissue cancers. METHODS The PubMed, Scopus, and Web of Science databases were queried to identify studies reporting the incidence of intracranial brain metastases from primary sarcoma to the present. Abstract and full-text screening of 1822 initial articles returned by preliminary search yielded 28 studies for inclusion and data extraction. Qualitative assessment of the studies was conducted in accordance with the Newcastle-Ottawa Scale criteria. Meta-analyses were applied to assess risk factors on outcomes. RESULTS The average age within the cohort was 27.9 years with a male and female prevalence of 59.1% and 40.9%, respectively. The odds ratio for living status (dead/alive) was calculated for several risk factors - male/female [OR 1.14, 95% CI 0.62, 2.07], single/multiple metastases [OR 0.67, 95% CI 0.35, 1.28], lung metastases/not [OR 1.63, 95% CI 0.85, 3.13], surgery/no surgery [OR 0.49, 95% CI 0.20, 1.21]. The standardized mean differences for duration from diagnoses to metastases were likewise analyzed - male/female [SMD 0.13, 95% CI -0.15, 0.42], single/multiple metastases [SMD 0.11, 95% CI -0.20, 0.42], lung metastases/not [SMD -0.03, 95% CI -0.38, 0.32], surgery/no surgery [SMD 0.45, 95% CI -0.18, 1.09]. The standardized mean differences for duration from metastases to death were analyzed - lung metastases/not [SMD 0.43, 95% CI -0.08, 0.95]. CONCLUSIONS Our study observed no statistically significant differences in mortality rate among several patient risk factors. Consequentially, there lacks a clear answer as to whether or not an association between mortality rates exists with these patient factors. As such, it is important to continue research in brain-metastasizing sarcomas despite their relative rarity.
Collapse
Affiliation(s)
| | - Andrew Nguyen
- College of Medicine, University of Florida, Gainesville, Florida, USA.
| | - Chance Fleeting
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Aashay Patel
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Nicholas Bazett
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Grace Hey
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Akhil Mandavali
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nolan J Brown
- Department of Neurosurgery, University of California-Irvine, Orange, California, USA
| | - Maxwell Woolridge
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Marco Foreman
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
35
|
Zhao S, Sun L, Zhou J, Li R, Sun Q, Wang W, Wang D. Advancements in Diagnosis and Multimodal Treatment Strategies for Retroperitoneal Tumors: A Comprehensive Review. Am J Clin Oncol 2024; 47:350-356. [PMID: 38476111 DOI: 10.1097/coc.0000000000001094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Retroperitoneal tumors (RPTs) encompass both benign and malignant entities, constituting ~0.1% to 0.2% of all malignant tumors, of which 70% to 80% manifest malignancy. Predominantly, retroperitoneal sarcomas (RPS) represent the most prevalent subtype among RPT. With over 70 histologic forms identified, liposarcomas and leiomyosarcomas emerge as the primary constituents of RPS. Accurate diagnosis of RPTs necessitates preoperative core-needle biopsy and comprehensive imaging assessment. The current staging protocol for RPS relies on the eighth edition of the American Joint Committee on Cancer/TNM classification. Surgical excision remains the established gold standard for treating RPS. Therapeutic approaches vary according to the underlying pathophysiology. Although chemotherapy and radiotherapy exhibit efficacy in managing metastatic and recurrent unresectable RPS, their role in primary RPS remains unresolved, necessitating further clinical trials for validation. Concurrently, ongoing research explores the potential of targeted therapies and immunotherapy. This literature review aims to provide a comprehensive overview of existing research, delineating diagnostic pathways and optimal therapeutic strategies for RPT.
Collapse
Affiliation(s)
- Shuai Zhao
- Department of General Surgery, Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University
| | - Longhe Sun
- Department of General Surgery, Northern Jiangsu People's Hospital, Yangzhou
| | - Jiajie Zhou
- Department of General Surgery, Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University
| | - Ruiqi Li
- Department of General Surgery, Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University
| | - Qiannan Sun
- Department of General Surgery, Taizhou Fourth People's Hospital
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou, China
| | - Wei Wang
- Department of General Surgery, Taizhou Fourth People's Hospital
| | - Daorong Wang
- Department of General Surgery, Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University
- Department of General Surgery, Taizhou Fourth People's Hospital
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou, China
| |
Collapse
|
36
|
Yang Q, Madueke-Laveaux OS, Cun H, Wlodarczyk M, Garcia N, Carvalho KC, Al-Hendy A. Comprehensive Review of Uterine Leiomyosarcoma: Pathogenesis, Diagnosis, Prognosis, and Targeted Therapy. Cells 2024; 13:1106. [PMID: 38994959 PMCID: PMC11240800 DOI: 10.3390/cells13131106] [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: 05/19/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024] Open
Abstract
Uterine leiomyosarcoma (uLMS) is the most common subtype of uterine sarcomas. They have a poor prognosis with high rates of recurrence and metastasis. The five-year survival for uLMS patients is between 25 and 76%, with survival rates approaching 10-15% for patients with metastatic disease at the initial diagnosis. Accumulating evidence suggests that several biological pathways are involved in uLMS pathogenesis. Notably, drugs that block abnormal functions of these pathways remarkably improve survival in uLMS patients. However, due to chemotherapy resistance, there remains a need for novel drugs that can target these pathways effectively. In this review article, we provide an overview of the recent progress in ascertaining the biological functions and regulatory mechanisms in uLMS from the perspective of aberrant biological pathways, including DNA repair, immune checkpoint blockade, protein kinase and intracellular signaling pathways, and the hedgehog pathway. We review the emerging role of epigenetics and epitranscriptome in the pathogenesis of uLMS. In addition, we discuss serum markers, artificial intelligence (AI) combined with machine learning, shear wave elastography, current management and medical treatment options, and ongoing clinical trials for patients with uLMS. Comprehensive, integrated, and deeper insights into the pathobiology and underlying molecular mechanisms of uLMS will help develop novel strategies to treat patients with this aggressive tumor.
Collapse
Affiliation(s)
- Qiwei Yang
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (O.S.M.-L.); (H.C.); (A.A.-H.)
| | | | - Han Cun
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (O.S.M.-L.); (H.C.); (A.A.-H.)
| | - Marta Wlodarczyk
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland;
| | - Natalia Garcia
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX 78229, USA;
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Katia Candido Carvalho
- Laboratório de Ginecologia Estrutural e Molecular (LIM 58), Disciplina de Ginecologia, Departamento deObstetricia e Ginecologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 05403-010, Brazil;
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (O.S.M.-L.); (H.C.); (A.A.-H.)
| |
Collapse
|
37
|
Wood GE, Meyer C, Petitprez F, D'Angelo SP. Immunotherapy in Sarcoma: Current Data and Promising Strategies. Am Soc Clin Oncol Educ Book 2024; 44:e432234. [PMID: 38781557 DOI: 10.1200/edbk_432234] [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: 05/25/2024]
Abstract
Traditionally sarcomas have been considered immunologically quiet tumours, with low tumour mutational burden (TMB) and an immunosuppressive tumour microenvironment (TME), consisting of decreased T-cell infiltration and elevated levels of H1F1α, macrophages and neutrophils.1,2 However, research has shown that a subset of sarcomas are immunologically 'hot' with either high TMB, PDL-1 expression, CD8+ T cells or presence of tertiary lymphoid structures (TLS) demonstrating sensitivity to immunotherapy.3,4 Here, we review the current evidence for immunotherapy use in bone sarcomas (BS) and soft tissue sarcomas (STS), with immune checkpoint inhibitors (ICI) and adoptive cellular therapies including engineered T-cell therapies, chimeric antigen receptor (CAR) T-cell therapies, tumour infiltrating lymphocytes (TILs) and cancer vaccines and biomarkers of response.
Collapse
Affiliation(s)
- Georgina E Wood
- University College Hospital of London, London, United Kingdom
| | | | | | | |
Collapse
|
38
|
Stacchiotti S, Bouche G, Herold R, Pantziarka P, Schuster K, Wilson R, Pignatti F, Kasper B. How to develop new systemic treatments in ultra-rare cancers with high unmet needs? The case of alveolar soft-part sarcoma. Eur J Cancer 2024; 202:114003. [PMID: 38479120 DOI: 10.1016/j.ejca.2024.114003] [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: 12/07/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 04/21/2024]
Abstract
Developing new drugs or generating evidence for existing drugs in new indications for ultra-rare cancers is complex and carries a high-risk of failure. This gets even harder in ultra-rare tumours, which have an annual incidence of 1 per 1,000,000 population or less. Here, we illustrate the problem of adequate evidence generation in ultra-rare tumours, using Alveolar Soft-Part Sarcomas (ASPS) - an ultra-rare sarcoma newly diagnosed in approximately 60 persons a year in the European Union - as an exemplar case showing challenges in development despite being potentially relevant for classes of agents. We discuss some possible approaches for addressing such challenges, especially focussing on constructive collaboration between academic groups, patients and advocates, drug manufacturers, and regulators to optimise drug development in ultra-rare cancers. This article, written by various European stakeholders, proposes a way forward to ultimately get better options for patients with ultra-rare cancers.
Collapse
Affiliation(s)
- Silvia Stacchiotti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.
| | | | | | | | | | - Roger Wilson
- Sarcoma Patient Advocacy Global Network, Wölfersheim, Germany
| | | | - Bernd Kasper
- Sarcoma Unit, Mannheim Cancer Center (MCC), Mannheim University Medical Center, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
39
|
Knoedler L, Huelsboemer L, Hollmann K, Alfertshofer M, Herfeld K, Hosseini H, Boroumand S, Stoegner VA, Safi AF, Perl M, Knoedler S, Pomahac B, Kauke-Navarro M. From standard therapies to monoclonal antibodies and immune checkpoint inhibitors - an update for reconstructive surgeons on common oncological cases. Front Immunol 2024; 15:1276306. [PMID: 38715609 PMCID: PMC11074450 DOI: 10.3389/fimmu.2024.1276306] [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: 08/11/2023] [Accepted: 04/05/2024] [Indexed: 05/23/2024] Open
Abstract
Malignancies represent a persisting worldwide health burden. Tumor treatment is commonly based on surgical and/or non-surgical therapies. In the recent decade, novel non-surgical treatment strategies involving monoclonal antibodies (mAB) and immune checkpoint inhibitors (ICI) have been successfully incorporated into standard treatment algorithms. Such emerging therapy concepts have demonstrated improved complete remission rates and prolonged progression-free survival compared to conventional chemotherapies. However, the in-toto surgical tumor resection followed by reconstructive surgery oftentimes remains the only curative therapy. Breast cancer (BC), skin cancer (SC), head and neck cancer (HNC), and sarcoma amongst other cancer entities commonly require reconstructive surgery to restore form, aesthetics, and functionality. Understanding the basic principles, strengths, and limitations of mAB and ICI as (neo-) adjuvant therapies and treatment alternatives for resectable or unresectable tumors is paramount for optimized surgical therapy planning. Yet, there is a scarcity of studies that condense the current body of literature on mAB and ICI for BC, SC, HNC, and sarcoma. This knowledge gap may result in suboptimal treatment planning, ultimately impairing patient outcomes. Herein, we aim to summarize the current translational endeavors focusing on mAB and ICI. This line of research may serve as an evidence-based fundament to guide targeted therapy and optimize interdisciplinary anti-cancer strategies.
Collapse
Affiliation(s)
- Leonard Knoedler
- Department of Plastic, Hand, and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Lioba Huelsboemer
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Katharina Hollmann
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Faculty of Medicine, University of Wuerzbuerg, Wuerzburg, Germany
| | - Michael Alfertshofer
- Division of Hand, Plastic and Aesthetic Surgery, Ludwig-Maximilians University Munich, Munich, Germany
| | - Konstantin Herfeld
- Department of Internal Medicine III (Oncology and Haematology), University Hospital Regensburg, Regensburg, Germany
- Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Helia Hosseini
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Sam Boroumand
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Viola A. Stoegner
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Burn Center, Hannover Medical School, Hannover, Germany
| | - Ali-Farid Safi
- Craniologicum, Center for Cranio-Maxillo-Facial Surgery, Bern, Switzerland
- Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Markus Perl
- Department of Internal Medicine III (Oncology and Haematology), University Hospital Regensburg, Regensburg, Germany
- Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Samuel Knoedler
- Department of Plastic, Hand, and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Bohdan Pomahac
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Martin Kauke-Navarro
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
40
|
Guérin R, Menard AL, Angot E, Piton N, Vera P, Schwarz L, Sabourin JC, Laé M, Thiébaut PA. An unusual case of primary splenic soft part alveolar sarcoma: case report and review of the literature with emphasis on the spectrum of TFE3-associated neoplasms. Diagn Pathol 2024; 19:62. [PMID: 38643139 PMCID: PMC11031972 DOI: 10.1186/s13000-024-01483-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 04/02/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Alveolar soft part sarcoma is a rare tumour of soft tissues, mostly localized in muscles or deep soft tissues of the extremities. In rare occasions, this tumour develops in deep tissues of the abdomen or pelvis. CASE PRESENTATION In this case report, we described the case of a 46 year old man who developed a primary splenic alveolar soft part sarcoma. The tumour displayed typical morphological alveolar aspect, as well as immunohistochemical profile notably TFE3 nuclear staining. Detection of ASPSCR1 Exon 7::TFE3 Exon 6 fusion transcript in molecular biology and TFE3 rearrangement in FISH confirmed the diagnosis. CONCLUSION We described the first case of primary splenic alveolar soft part sarcoma, which questions once again the cell of origin of this rare tumour.
Collapse
Affiliation(s)
- René Guérin
- Department of Pathology, Rouen University Hospital, Rouen, France
| | | | - Emilie Angot
- Department of Pathology, Rouen University Hospital, Rouen, France
| | - Nicolas Piton
- Department of Pathology, Rouen University Hospital, Rouen, France
| | - Pierre Vera
- Department of Nuclear Medecine, Centre Henri Becquerel, Rouen, France
| | - Lilian Schwarz
- Department of Digestive Surgery, Rouen University Hospital, Rouen, France
| | | | - Marick Laé
- Department of Pathology, Centre Henri Becquerel, Rouen, France
| | | |
Collapse
|
41
|
Bergsma EJ, Elgawly M, Mancuso D, Orr R, Vuskovich T, Seligson ND. Atezolizumab as the First Systemic Therapy Approved for Alveolar Soft Part Sarcoma. Ann Pharmacother 2024; 58:407-415. [PMID: 37466080 DOI: 10.1177/10600280231187421] [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: 07/20/2023] Open
Abstract
OBJECTIVE The objective was to review the pharmacology, efficacy, and safety of atezolizumab (Tecentriq) for the treatment of adult and pediatric patients aged 2 years and older with unresectable or metastatic alveolar soft part sarcoma (ASPS). DATA SOURCES A literature search was conducted using PubMed and MEDLINE databases, published abstracts, and ongoing studies from ClinicalTrials.gov between January 1, 1981, and May 31, 2023. Keywords included atezolizumab, Tecentriq, MPDL3280, immunotherapy, PD-L1, PD-1, pediatrics, sarcoma, and ASPS. STUDY SELECTION AND DATA EXTRACTION All English-language studies involving atezolizumab for ASPS were included and discussed. DATA SYNTHESIS Atezolizumab is an anti-programmed death-ligand 1 (PD-L1) monoclonal antibody designed to block the interaction between PD-L1 and the programmed cell death protein 1 (PD-1) receptor. Atezolizumab was granted approval by the FDA specifically for ASPS based on a phase II clinical trial in adult and pediatric patients (n = 49), which reported an overall response rate of 24% and a durable response rate at 6 and 12 months of 67% and 42%, respectively. Common grade 3/4 adverse reactions include musculoskeletal pain (8%), followed by hypertension (6%), weight gain (6%), headache (4%), and dizziness (4%). RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE IN COMPARISON WITH EXISTING DRUGS Advanced ASPS is a high-risk disease with limited treatment options. Atezolizumab appears to be a viable treatment option in ASPS demonstrating clinical efficacy and a manageable toxicity profile. CONCLUSIONS With no other treatments that are FDA approved specifically for ASPS, and few demonstrating efficacy in the advanced setting, the approval of atezolizumab, including the first approval for pediatric patients, represents a landmark improvement to the therapeutic arsenal against this rare disease.
Collapse
Affiliation(s)
- Emilie J Bergsma
- Department of Pharmacy, University of Florida Health Shands Hospital, Gainesville, FL, USA
| | - Mariam Elgawly
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL, USA
| | - David Mancuso
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL, USA
| | - Roger Orr
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL, USA
| | - Theresa Vuskovich
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL, USA
| | - Nathan D Seligson
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL, USA
- Precision Medicine, Nemours Children's Health, Jacksonville, FL, USA
| |
Collapse
|
42
|
Kyriazoglou A, Pagkali A, Kotsantis I, Economopoulou P, Kyrkasiadou M, Moutafi M, Gavrielatou N, Anastasiou M, Boulouta A, Pantazopoulos A, Giannakakou M, Digklia A, Psyrri A. Well-differentiated liposarcomas and dedifferentiated liposarcomas: Systemic treatment options for two sibling neoplasms. Cancer Treat Rev 2024; 125:102716. [PMID: 38492514 DOI: 10.1016/j.ctrv.2024.102716] [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: 10/17/2023] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
Well-differentiated liposarcomas (WDLPS) and dedifferentiated liposarcomas (DDLPS) account for 60 % of all liposarcomas, reflecting the heterogeneity of this type of sarcoma. Genetically, both types of liposarcomas are characterized by the amplification of MDM2 and CDK4 genes, which indicates an important molecular event with diagnostic and therapeutic relevance. In both localized WDLPS and DDLPS of the retroperitoneum and the extremities, between 25 % and 30 % of patients have local or distant recurrence, even when perioperatively treated, with clear margins present. The systemic treatment of WDLPS and DDLPS remains a challenge, with anthracyclines as the gold standard for first-line treatment. Several regimens have been tested with modest results regarding their efficacy. Herein we discuss the systemic treatment options for WDLPS and DDLPS and review their reported clinical efficacy results.
Collapse
Affiliation(s)
- A Kyriazoglou
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece.
| | - A Pagkali
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - I Kotsantis
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - P Economopoulou
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - M Kyrkasiadou
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - M Moutafi
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - N Gavrielatou
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - M Anastasiou
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - A Boulouta
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - A Pantazopoulos
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - M Giannakakou
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - A Digklia
- Sarcoma Center, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne University Lausanne, Switzerland
| | - A Psyrri
- Section of Medical Oncology, 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| |
Collapse
|
43
|
Starzer AM, Wolff L, Popov P, Kiesewetter B, Preusser M, Berghoff AS. The more the merrier? Evidence and efficacy of immune checkpoint- and tyrosine kinase inhibitor combinations in advanced solid cancers. Cancer Treat Rev 2024; 125:102718. [PMID: 38521009 DOI: 10.1016/j.ctrv.2024.102718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/03/2024] [Accepted: 03/09/2024] [Indexed: 03/25/2024]
Abstract
Immune checkpoint inhibitors (ICI) and tyrosine kinase inhibitors (TKI) have gained therapeutical significance in cancer therapy over the last years. Due to the high efficacy of each substance group, additive or complementary effects are considered, and combinations are the subject of multiple prospective trials in different tumor entities. The majority of available data results from clinical phase I and II trials. Although regarded as well-tolerated therapies ICI-TKI combinations have higher toxicities compared to monotherapies of one of the substance classes and some combinations were shown to be excessively toxic leading to discontinuation of trials. So far, ICI-TKI combinations with nivolumab + cabozantinib, pembrolizumab + axitinib, avelumab + axitinib, pembrolizumab + lenvatinib have been approved in advanced renal cell (RCC), with pembrolizumab + lenvatinib in endometrial carcinoma and with camrelizumab + rivoceranib in hepatocellular carcinoma (HCC). Several ICI-TKI combinations are currently investigated in phase I to III trials in various other cancer entities. Further, the optimal sequence of ICI-TKI combinations is an important subject of investigation, as cross-resistances between the substance classes were observed. This review reports on clinical trials with ICI-TKI combinations in different cancer entities, their efficacy and toxicity.
Collapse
Affiliation(s)
- Angelika M Starzer
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Ladislaia Wolff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Petar Popov
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Barbara Kiesewetter
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
44
|
Fontebasso AM, Rytlewski JD, Blay JY, Gladdy RA, Wilky BA. Precision Oncology in Soft Tissue Sarcomas and Gastrointestinal Stromal Tumors. Surg Oncol Clin N Am 2024; 33:387-408. [PMID: 38401916 DOI: 10.1016/j.soc.2023.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2024]
Abstract
Soft tissue sarcomas (STSs), including gastrointestinal stromal tumors (GISTs), are mesenchymal neoplasms with heterogeneous clinical behavior and represent broad categories comprising multiple distinct biologic entities. Multidisciplinary management of these rare tumors is critical. To date, multiple studies have outlined the importance of biological characterization of mesenchymal tumors and have identified key molecular alterations which drive tumor biology. GIST has represented a flagship for targeted therapy in solid tumors with the advent of imatinib which has revolutionized the way we treat this malignancy. Herein, the authors discuss the importance of biological and molecular diagnostics in managing STS and GIST patients.
Collapse
Affiliation(s)
- Adam M Fontebasso
- Division of Surgical Oncology, Department of Surgery, University of Toronto, 700 University Avenue, 7th Floor, Ontario Power Generation Building, Toronto, Ontario, Canada; Department of Surgery, Mount Sinai Hospital, Sinai Health Systems, 600 University Avenue Room 6-445.10 Surgery, Toronto, Ontario M5G 1X5, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jeffrey D Rytlewski
- University of Colorado School of Medicine, 12801 East 17th Avenue, Mailstop 8117, Aurora, CO 80045, USA
| | - Jean-Yves Blay
- Centre Léon Bérard, 28, rue Laennec, 69373 cedex 08. Lyon, France
| | - Rebecca A Gladdy
- Division of Surgical Oncology, Department of Surgery, University of Toronto, 700 University Avenue, 7th Floor, Ontario Power Generation Building, Toronto, Ontario, Canada; Department of Surgery, Mount Sinai Hospital, Sinai Health Systems, 600 University Avenue Room 6-445.10 Surgery, Toronto, Ontario M5G 1X5, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Breelyn A Wilky
- University of Colorado School of Medicine, 12801 East 17th Avenue, Mailstop 8117, Aurora, CO 80045, USA.
| |
Collapse
|
45
|
Zhang QS, Hayes JP, Gondi V, Pollack SM. Immunotherapy and Radiotherapy Combinations for Sarcoma. Semin Radiat Oncol 2024; 34:229-242. [PMID: 38508787 DOI: 10.1016/j.semradonc.2023.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Sarcomas are a heterogeneous group of bone and soft tissue tumors. Survival outcomes for advanced (unresectable or metastatic) disease remain poor, so therapeutic improvements are needed. Radiotherapy plays an integral role in the neoadjuvant and adjuvant treatment of localized disease as well as in the treatment of metastatic disease. Combining radiotherapy with immunotherapy to potentiate immunotherapy has been used in a variety of cancers other than sarcoma, and there is opportunity to further investigate combining immunotherapy with radiotherapy to try to improve outcomes in sarcoma. In this review, we describe the diversity of the tumor immune microenvironments for sarcomas and describe the immunomodulatory effects of radiotherapy. We discuss studies on the timing of radiotherapy relative to immunotherapy and studies on the radiotherapy dose and fractionation regimen to be used in combination with immunotherapy. We describe the impact of radiotherapy on the tumor immune microenvironment. We review completed and ongoing clinical trials combining radiotherapy with immunotherapy for sarcoma and propose future directions for studies combining immunotherapy with radiotherapy in the treatment of sarcoma.
Collapse
Affiliation(s)
- Qian S Zhang
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - John P Hayes
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Vinai Gondi
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Seth M Pollack
- Division of Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL..
| |
Collapse
|
46
|
Liao Z, Teng J, Li T, Liu H, Li T, Zhang C, Xing R, Teng S, Yang Y, Zhao J, Xiao W, Zhang G, Li MJ, Yao W, Yang J. Evaluation of the efficacy and safety of immunotherapy in sarcoma: a two-center study. Front Immunol 2024; 15:1292325. [PMID: 38585276 PMCID: PMC10995229 DOI: 10.3389/fimmu.2024.1292325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/21/2024] [Indexed: 04/09/2024] Open
Abstract
Background Sarcoma is a highly heterogeneous malignancy with a poor prognosis. Although chemotherapy and targeted therapy have improved the prognosis to some extent, the efficacy remains unsatisfactory in some patients. The efficacy and safety of immunotherapy in sarcoma need further evaluation. Methods We conducted a two-center study of sarcoma patients receiving PD-1 immunotherapy at Tianjin Medical University Cancer Institute and Hospital and Henan Provincial Cancer Hospital. The treatment regimens included PD-1 inhibitor monotherapy and combination therapy based on PD-1 inhibitors. The observed primary endpoints were median progression-free survival (mPFS) and median overall survival (mOS). Survival curves were compared using the Kaplan-Meier method. Results A total of 43 patients were included from the two centers. The median follow-up time for all patients was 13 months (range, 1-48 months). In the group of 37 patients with advanced or unresectable sarcoma, the mPFS was 6 months (95%CI: 5-12 months), and the mOS was 16 months (95%CI: 10-28 months). The ORR was 10.8% (4/37), and the DCR was 18.9% (7/37). Subgroup analysis showed no significant differences in mPFS (p=0.11) and mOS (p=0.88) between patients with PD-L1 negative/positive expression. There were also no significant differences in mPFS (p=0.13) or mOS (p=0.72) between PD-1 inhibitor monotherapy and combination therapy. Additionally, there were no significant differences in mPFS (p=0.52) or mOS (p=0.49) between osteogenic sarcoma and soft tissue sarcoma. Furthermore, the results showed no significant differences in mPFS (p=0.66) or mOS (p=0.96) between PD-1 inhibitors combined with targeted therapy and PD-1 inhibitors combined with AI chemotherapy. Among the 6 patients receiving adjuvant therapy after surgery, the mPFS was 15 months (95%CI: 6-NA months), and the mOS was not reached. In terms of safety, most adverse events were mild (grade 1-2) and manageable. The most severe grade 4 adverse events were bone marrow suppression, which occurred in 4 patients but resolved after treatment. There was also one case of a grade 4 adverse event related to hypertension. Conclusion Immunotherapy is an effective treatment modality for sarcoma with manageable safety. Further inclusion of more patients or prospective clinical trials is needed to validate these findings.
Collapse
Affiliation(s)
- Zhichao Liao
- 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 Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jianjin Teng
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Tao Li
- Department of Bone and Soft-Tissue Tumor, Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Haotian Liu
- 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 Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ting Li
- 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 Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chao Zhang
- 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 Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ruwei Xing
- 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 Medical University Cancer Institute and Hospital, Tianjin, China
| | - Sheng Teng
- 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 Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yun 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 Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jun Zhao
- 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 Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wanyi Xiao
- 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 Medical University Cancer Institute and Hospital, Tianjin, China
| | - Gengpu Zhang
- 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 Medical University Cancer Institute and Hospital, Tianjin, China
| | - Mulin Jun Li
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Weitao Yao
- Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 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 Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
47
|
Li W, Liu L, Liang Z, Lai H, Wu J, Zhang H, Fang C. Efficacy of tyrosine kinase inhibitors in patients with advanced or metastatic sarcomas after prior chemotherapy: A meta-analysis. Medicine (Baltimore) 2024; 103:e37423. [PMID: 38489731 PMCID: PMC10939701 DOI: 10.1097/md.0000000000037423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/07/2024] [Accepted: 02/07/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Sarcoma is a heterogeneous malignancy arising from interstitial tissue. Anthracycline-based therapy is the first-line treatment recommended by guidelines for patients with locally advanced or metastatic unresectable sarcoma. Recently, targeted therapies, in particular tyrosine kinase inhibitors (TKIs), have made significant progress in the treatment of sarcoma, and their efficacy has been investigated in randomized controlled trials. The aim of this meta-analysis is to evaluate the efficacy of TKIs in patients with advanced or metastatic sarcoma who have previously received chemotherapy. METHODS We completed a meta-analysis after conducting literature searches in PubMed, Embase, and Cochrane. The single-drug, placebo-controlled, randomized controlled clinical trials of TKIs in patients with advanced or progressive sarcoma who have previously received chemotherapy are available for inclusion in the study. The observation results were objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). The subgroup analysis was performed according to histological subtypes of sarcoma. RESULTS This study included 6 studies, including 1033 patients. The ORR (OR: 7.99, 95% CI: 3.62-19.61, P < .00001), DCR (OR: 2.54, 95% CI: 1.27-5.08, P = .009), PFS (HR: 0.46, 95% CI: 0.34-0.62, P < .00001), and OS (HR: 0.80, 95% CI: 0.67-0.96, P = .02) of patients treated with TKIs were better than those in the placebo group. CONCLUSIONS In patients with advanced sarcoma, TKIs have been shown to have advantages in terms of ORR, DCR and PFS and OS. Multi-targeted TKIs may be considered as one of the second-line treatment options for sarcoma patients who have received prior chemotherapy.
Collapse
Affiliation(s)
- Wenxia Li
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Liwen Liu
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Zhanpeng Liang
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Huiqin Lai
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Jiaming Wu
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Huatang Zhang
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Cantu Fang
- Department of Oncology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| |
Collapse
|
48
|
Duan XP, Qin BD, Jiao XD, Liu K, Wang Z, Zang YS. New clinical trial design in precision medicine: discovery, development and direction. Signal Transduct Target Ther 2024; 9:57. [PMID: 38438349 PMCID: PMC10912713 DOI: 10.1038/s41392-024-01760-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
In the era of precision medicine, it has been increasingly recognized that individuals with a certain disease are complex and different from each other. Due to the underestimation of the significant heterogeneity across participants in traditional "one-size-fits-all" trials, patient-centered trials that could provide optimal therapy customization to individuals with specific biomarkers were developed including the basket, umbrella, and platform trial designs under the master protocol framework. In recent years, the successive FDA approval of indications based on biomarker-guided master protocol designs has demonstrated that these new clinical trials are ushering in tremendous opportunities. Despite the rapid increase in the number of basket, umbrella, and platform trials, the current clinical and research understanding of these new trial designs, as compared with traditional trial designs, remains limited. The majority of the research focuses on methodologies, and there is a lack of in-depth insight concerning the underlying biological logic of these new clinical trial designs. Therefore, we provide this comprehensive review of the discovery and development of basket, umbrella, and platform trials and their underlying logic from the perspective of precision medicine. Meanwhile, we discuss future directions on the potential development of these new clinical design in view of the "Precision Pro", "Dynamic Precision", and "Intelligent Precision". This review would assist trial-related researchers to enhance the innovation and feasibility of clinical trial designs by expounding the underlying logic, which be essential to accelerate the progression of precision medicine.
Collapse
Affiliation(s)
- Xiao-Peng Duan
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bao-Dong Qin
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Dong Jiao
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ke Liu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhan Wang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yuan-Sheng Zang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
49
|
Denu RA, Dann AM, Keung EZ, Nakazawa MS, Nassif Haddad EF. The Future of Targeted Therapy for Leiomyosarcoma. Cancers (Basel) 2024; 16:938. [PMID: 38473300 PMCID: PMC10930698 DOI: 10.3390/cancers16050938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Leiomyosarcoma (LMS) is an aggressive subtype of soft tissue sarcoma that arises from smooth muscle cells, most commonly in the uterus and retroperitoneum. LMS is a heterogeneous disease with diverse clinical and molecular characteristics that have yet to be fully understood. Molecular profiling has uncovered possible targets amenable to treatment, though this has yet to translate into approved targeted therapies in LMS. This review will explore historic and recent findings from molecular profiling, highlight promising avenues of current investigation, and suggest possible future strategies to move toward the goal of molecularly matched treatment of LMS. We focus on targeting the DNA damage response, the macrophage-rich micro-environment, the PI3K/mTOR pathway, epigenetic regulators, and telomere biology.
Collapse
Affiliation(s)
- Ryan A. Denu
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Amanda M. Dann
- Division of Surgical Oncology, Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Emily Z. Keung
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Michael S. Nakazawa
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elise F. Nassif Haddad
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
50
|
Pan M, Zhou MY, Jiang C, Zhang Z, Bui N, Bien J, Siy A, Achacoso N, Solorzano AV, Tse P, Chung E, Hu W, Thomas S, Ganjoo K, Habel LA. PTEN pathogenic variants are associated with poor prognosis in patients with advanced soft tissue sarcoma. BJC REPORTS 2024; 2:9. [PMID: 39516677 PMCID: PMC11524139 DOI: 10.1038/s44276-023-00029-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 11/16/2024]
Abstract
BACKGROUND We aimed to examine whether PTEN pathogenic variants (mutPTEN) were associated with overall survival (OS) in patients with advanced soft tissue sarcoma (STS) with the presence of one or more of the most common genomic alterations including p53, CDKN2A, RB1, and ATRX pathogenic variants. METHODS This study included patients from Kaiser Permanente Northern California and Stanford Cancer Center with grade 2 or higher locally advanced and metastatic STS. RESULTS A total of 174 patients had leiomyosarcoma (LMS), 136 had undifferentiated pleomorphic sarcoma (UPS), 78 had Liposarcoma (LPS), and 214 had other histology subtypes (Others). Among all patients with STS, OS was worse for those with mutPTEN versus wild-type PTEN (wtPTEN, adjusted HR [aHR] = 1.58 [95% CI, 1.11-2.23]), mutCDKN2A vs wtCDKN2A (aHR = 1.33 [95% CI .99-1.80]), and mutRB1 vs wtRB1 (aHR = 1.26 [95% CI 0.93-1.70[), while OS was similar for mutp53 vs wtp53 and mutATRX vs wtATRX. MutPTEN versus wtPTEN was consistently associated with worse OS in histologic subtypes including LMS and UPS and molecular subgroups. CONCLUSION MutPTEN vs wtPTEN was associated with worse OS in advanced STS. If confirmed, our findings could be helpful for prognostic stratification in clinical practice and for further understanding the molecular mechanisms of STS.
Collapse
Affiliation(s)
- Minggui Pan
- Division of Research, Kaiser Permanente, Oakland, CA, 94612, USA.
- Sarcoma Program, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Maggie Y Zhou
- Sarcoma Program, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Chen Jiang
- Division of Research, Kaiser Permanente, Oakland, CA, 94612, USA
| | - Zheyang Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Nam Bui
- Sarcoma Program, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jeffrey Bien
- Sarcoma Program, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Amanda Siy
- Sarcoma Program, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ninah Achacoso
- Division of Research, Kaiser Permanente, Oakland, CA, 94612, USA
| | | | - Pam Tse
- Division of Research, Kaiser Permanente, Oakland, CA, 94612, USA
| | - Elaine Chung
- Division of Research, Kaiser Permanente, Oakland, CA, 94612, USA
| | - Wenwei Hu
- Rutger's Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Sachdev Thomas
- Department of Oncology and Hematology, Kaiser Permanente, Vallejo, CA, 94589, USA
| | - Kristen Ganjoo
- Sarcoma Program, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Laurel A Habel
- Division of Research, Kaiser Permanente, Oakland, CA, 94612, USA
| |
Collapse
|