1
|
Metts J, Rodriguez-Valentin M, Hensel J, Alfaro A, Snyder CW, Binitie O, Chebli C, Monforte H, Pilon-Thomas S, Mullinax J. Expansion of tumor-infiltrating and marrow-infiltrating lymphocytes from pediatric malignant solid tumors. Cytotherapy 2024:S1465-3249(24)00825-9. [PMID: 39243253 DOI: 10.1016/j.jcyt.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 09/09/2024]
Abstract
INTRODUCTION The expansion of tumor-infiltrating lymphocytes (TIL) for adoptive cellular therapy is under investigation in many solid tumors of adulthood. Marrow-infiltrating lymphocytes (MIL) have demonstrated antitumor reactivity preclinically. Successful expansion of TIL/MIL has not been reported across pediatric solid tumor histologies. The objective of this study was to demonstrate successful expansion of TIL from pediatric solid tumors for translation in an adoptive cell therapy (ACT) treatment strategy. METHODS A prospective study of TIL/MIL expansion was performed on solid tumors of pediatric patients undergoing standard-of-care procedures. TIL/MIL expansions were performed in the presence of high-dose interleukin 2. To demonstrate a full-scale expansion to clinically-relevant cell doses for TIL therapy, initial TIL culture was followed by a rapid expansion protocol for select patients. Expanded specimens were analyzed for phenotype by flow cytometry and for anti-tumor reactivity by the interferon-gamma release assay. RESULTS Eighteen tumor samples were obtained. Initial TIL cultures were successfully generated from 14/18 samples (77.7%). A median of 5.52 × 107 (range: 2.5 × 106-3.23 × 108) cells were produced from initial cultures, with 46.9% expressing a CD3 phenotype (46.9%). Eight samples underwent rapid expansion, demonstrating a median 458-fold expansion and a CD3 phenotype of 98%. Initial MIL cultures were successfully generated from five samples, with a predominantly CD3 phenotype (45.2%). Sufficient tumor tissue was only available for seven TIL samples to be tested for reactivity; none demonstrated responsiveness to autologous tumor. CONCLUSIONS TIL and MIL expansion from pediatric solid tumors was successful, including the full-scale expansion process. This data supports translation to an ACT-TIL treatment strategy in the pediatric population and thus a Phase I trial of ACT-TIL in pediatric high-risk solid tumors is planned.
Collapse
Affiliation(s)
- Jonathan Metts
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA; Departments of Sarcoma, Immunology, and Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA.
| | | | | | - Alex Alfaro
- Departments of Sarcoma, Immunology, and Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Christopher W Snyder
- Division of Pediatric Surgery, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| | - Odion Binitie
- Departments of Sarcoma, Immunology, and Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Caroline Chebli
- Department of Orthopedic Surgery, James A Haley Veteran's Administration Hospital, Tampa, Florida, USA
| | - Hector Monforte
- Section of Anatomic Pathology, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| | - Shari Pilon-Thomas
- Departments of Sarcoma, Immunology, and Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - John Mullinax
- Departments of Sarcoma, Immunology, and Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
2
|
Yang L, Long Y, Xiao S. Osteosarcoma-Associated Immune Genes as Potential Immunotherapy and Prognosis Biomarkers. Biochem Genet 2024; 62:798-813. [PMID: 37452172 DOI: 10.1007/s10528-023-10444-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023]
Abstract
Immune-modulating therapies exhibit abundant promise compared to traditional treatment for osteosarcoma. We aim to establish an immune-related prognostic signatures in osteosarcoma. We identified the differentially expressed genes in osteosarcoma compared with normal controls using the GEO dataset. The intersection with immune-related genes was considered as differentially expressed immune genes. Potential prognosis-related differentially expressed genes were first analyzed with the multifactor Cox regression and then the step function performed the iteration. The best model was finally chosen as the immunological risk score signature model. And finally, we evaluated the correlation of genes in the prognostic model with immune cells, common immune checkpoints, and immune checkpoint blockade responses. We identified 1527 significantly upregulated and 2407 significantly downregulated genes in osteosarcoma compared to normal samples. In the 258 differentially expressed immune genes, 20 genes with independent prognostic significance were included in the step function. Finally, we constructed a prognostic signature for overall survival based on five immune genes (JAG2, PLXNB1, CMKLR1, NUDT6, and PSMC4) in osteosarcoma. These five genes are closely related to immune infiltration. Osteosarcoma with high JAG2 expression or low CMKLR1 expression may be associated with better immune checkpoint blockade response. JAG2 overexpression or CMKLR1 inhibition induced sensitivity to PD-1 antibody in osteosarcoma cells. We constructed a prognosis prediction signature containing five immune-related genes (JAG2, PLXNB1, CMKLR1, NUDT6, and PSMC4) with a significant prognostic value in osteosarcoma. Significant differences in immune infiltration and immunotherapy responses were identified between groups with different levels of these genes.
Collapse
Affiliation(s)
- Li Yang
- Key Laboratory of Geriatric Diseases of Xinyang, Institute of Inspection Technology, Xinyang Vocational and Technical College, Xinyang, 464000, China
| | - Yi Long
- Department of Joint Surgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, 412000, China.
| | - Shengshi Xiao
- Department of Joint Surgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, 412000, China.
| |
Collapse
|
3
|
Chen L, He L, Liu B, Zhou Y, Lv L, Wang Z. Intelligent structure prediction and visualization analysis of non-coding RNA in osteosarcoma research. Front Oncol 2024; 14:1255061. [PMID: 38532928 PMCID: PMC10964489 DOI: 10.3389/fonc.2024.1255061] [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: 07/11/2023] [Accepted: 02/23/2024] [Indexed: 03/28/2024] Open
Abstract
Background Osteosarcoma (OS) is the most common bone malignant tumor in children and adolescents. Recent research indicates that non-coding RNAs (ncRNAs) have been associated with OS occurrence and development, with significant progress made in this field. However, there is no intelligent structure prediction and literature visualization analysis in this research field. From the perspective of intelligent knowledge structure construction and bibliometrics, this study will comprehensively review the role of countries, institutions, journals, authors, literature citation relationships and subject keywords in the field of ncRNAs in OS. Based on this analysis, we will systematically analyze the characteristics of the knowledge structure of ncRNAs in OS disease research and identify the current research hotspots and trends. Methods The Web of Science Core Collection (WoSCC) database was searched for articles on ncRNAs in OS between 2001 and 2023. This bibliometric analysis was performed using VOSviewers, CiteSpace, and Pajek. Results This study involved 15,631 authors from 2,631 institutions across 57 countries/regions, with a total of 3,642 papers published in 553 academic journals. China has the highest number of published papers in this research field. The main research institutions include Nanjing Medical University (n = 129, 3.54%), Shanghai Jiao Tong University (n = 128, 3.51%), Zhengzhou University (n = 110, 3.02%), and China Medical University (n = 109, 2.99%). Oncology Letters (n =139, 3.82%), European Review for Medical Pharmacological Sciences (120, 3.31%), and Molecular Medicine Reports (n = 95, 2.61%) are the most popular journals in this field, with Oncotarget being the most co-cited journal (Co-Citation = 4,268). Wei Wang, Wei Liu, and Zhenfeng Duan published the most papers, with Wang Y being the most co-cited author. "miRNA", "lncRNA" and "circRNA" are the main focuses of ncRNAs in OS studies. Key themes include "migration and invasion", "apoptosis and proliferation", "prognosis", "biomarkers" and "chemoresistance". Since 2020, hotspots and trends in ncRNA research in OS include "tumor microenvironment", "immune" and "exosome". Conclusion This study represents the first comprehensive bibliometric analysis of the knowledge structure and development of ncRNAs in OS. These findings highlight current research hotspots and frontier directions, offering valuable insights for future studies on the role of ncRNAs in OS.
Collapse
Affiliation(s)
- Longhao Chen
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The Third Clinical Medical College, Zhejiang University of Chinese Medicine, Hangzhou, Zhejiang, China
| | - Liuji He
- Faculty of Orthopedics and Traumatology, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Baijie Liu
- Faculty of Orthopedics and Traumatology, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yinghua Zhou
- First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Lijiang Lv
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The Third Clinical Medical College, Zhejiang University of Chinese Medicine, Hangzhou, Zhejiang, China
| | - Zhiguang Wang
- First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
4
|
Hong Y, Zhang L, Lin W, Yang Y, Cao Z, Feng X, Yu Z, Gao Y. Transcriptome Sequencing Unveils a Molecular-Stratification-Predicting Prognosis of Sarcoma Associated with Lipid Metabolism. Int J Mol Sci 2024; 25:1643. [PMID: 38338920 PMCID: PMC10855378 DOI: 10.3390/ijms25031643] [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: 10/14/2023] [Revised: 01/05/2024] [Accepted: 01/16/2024] [Indexed: 02/12/2024] Open
Abstract
Sarcomas are heterogeneous connective tissue malignancies that have been historically categorized into soft tissue and bone cancers. Although multimodal therapies are implemented, many sarcoma subtypes are still difficult to treat. Lipids play vital roles in cellular activities; however, ectopic levels of lipid metabolites have an impact on tumor recurrence, metastasis, and drug resistance. Thus, precision therapies targeting lipid metabolism in sarcoma need to be explored. In this study, we performed a comprehensive analysis of molecular stratification based on lipid metabolism-associated genes (LMAGs) using both public datasets and the data of patients in our cohort and constructed a novel prognostic model consisting of squalene epoxidase (SQLE) and tumor necrosis factor (TNF). We first integrated information on gene expression profile and survival outcomes to divide TCGA sarcoma patients into high- and low-risk subgroups and further revealed the prognosis value of the metabolic signature and immune infiltration of patients in both groups, thus proposing various therapeutic recommendations for sarcoma. We observed that the low-risk sarcoma patients in the TCGA-SARC cohort were characterized by high proportions of immune cells and increased expression of immune checkpoint genes. Subsequently, this lipid metabolic signature was validated in four external independent sarcoma datasets including the CHCAMS cohort. Notably, SQLE, a rate-limiting enzyme in cholesterol biosynthesis, was identified as a potential therapeutic target for sarcoma. Knockdown of SQLE substantially inhibited cell proliferation and colony formation while promoting the apoptosis of sarcoma cells. Terbinafine, an inhibitor of SQLE, displayed similar tumor suppression capacity in vitro. The prognostic predictive model and the potential drug target SQLE might serve as valuable hints for further in-depth biological, diagnostic, and therapeutic exploration of sarcoma.
Collapse
Affiliation(s)
- Yuheng Hong
- 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
| | - Lin Zhang
- Department of Thoracic Surgery, 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
| | - Weihao Lin
- 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
| | - Yannan Yang
- 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
| | - Zheng Cao
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaoli Feng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhentao Yu
- Department of Thoracic Surgery, 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
| | - 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
- Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular 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
|
5
|
Jumaniyazova E, Lokhonina A, Dzhalilova D, Kosyreva A, Fatkhudinov T. Immune Cells in the Tumor Microenvironment of Soft Tissue Sarcomas. Cancers (Basel) 2023; 15:5760. [PMID: 38136307 PMCID: PMC10741982 DOI: 10.3390/cancers15245760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Soft tissue sarcomas (STSs) are a rare heterogeneous group of malignant neoplasms characterized by their aggressive course and poor response to treatment. This determines the relevance of research aimed at studying the pathogenesis of STSs. By now, it is known that STSs is characterized by complex relationships between the tumor cells and immune cells of the microenvironment. Dynamic interactions between tumor cells and components of the microenvironment enhance adaptation to changing environmental conditions, which provides the high aggressive potential of STSs and resistance to antitumor therapy. Today, active research is being conducted to find effective antitumor drugs and to evaluate the possibility of using therapy with immune cells of STS. The difficulty in assessing the efficacy of new antitumor options is primarily due to the high heterogeneity of this group of malignant neoplasms. Studying the role of immune cells in the microenvironment in the progression STSs and resistance to antitumor therapies will provide the discovery of new biomarkers of the disease and the prediction of response to immunotherapy. In addition, it will help to initially divide patients into subgroups of good and poor response to immunotherapy, thus avoiding wasting precious time in selecting the appropriate antitumor agent.
Collapse
Affiliation(s)
- Enar Jumaniyazova
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia (T.F.)
| | - Anastasiya Lokhonina
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia (T.F.)
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997 Moscow, Russia
| | - Dzhuliia Dzhalilova
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia (T.F.)
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Anna Kosyreva
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia (T.F.)
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Timur Fatkhudinov
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia (T.F.)
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| |
Collapse
|
6
|
Zhang X, Shao Y, Zhou Y, Zhu Z, Wang X. Comprehensive analysis of immune implication and prognostic value of DHX33 in sarcoma. Medicine (Baltimore) 2023; 102:e33654. [PMID: 37115050 PMCID: PMC10145805 DOI: 10.1097/md.0000000000033654] [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: 12/21/2022] [Revised: 03/29/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
DEAH-box helicase 33 (DHX33) is an RNA helicase that has been identified to promote the progression of a variety of cancers. However, the relationship between DHX33 and sarcoma remains unknown. RNA expression data with clinical information for the sarcoma project was collected from TCGA database. The association between the differential expression of DHX33 and the prognosis for sarcoma was assessed using survival analysis. CIBERSORT was used to evaluate the immune cell infiltration in sarcoma sample tissues. We then further investigated the association between DHX33 and tumor-infiltrating immune cells in sarcoma using the TIMER database. Finally, the immune/cancer-related signaling pathways involved in DHX33 were analyzed using gene set enrichment analysis. High DHX33 expression was discovered to be a poor prognostic indicator in TCGA-SARC. Immune subpopulations in the TCGA-SARC microenvironment are dramatically altered compared to normal tissues. The tumor immune estimation resource analysis revealed a strong correlation between the expression of DHX33 and the abundance of CD8+ T cells and dendritic cells. Changes in copy number also affected neutrophils, macrophages, and CD4+ T cells. According to gene set enrichment analysis, DHX33 may be involved in a number of cancer- and immune-related pathways, such as the JAK/STAT signaling pathway, P53 signaling pathway, chemokine signaling pathway, T cell receptor signaling pathway, complement and coagulation cascades, and cytokine-cytokine receptor interaction. Our study emphasized that DHX33 may be involved in the immune microenvironment of sarcoma and play an important role. As a result, it is possible that DHX33 might serve as an immunotherapeutic target for sarcoma.
Collapse
Affiliation(s)
- Xinan Zhang
- Department of Orthopedics, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yiming Shao
- Department of Orthopedics, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yaqi Zhou
- Department of Orthopedics, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Zhi Zhu
- Department of Orthopedics, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xiaohu Wang
- Department of Orthopedics, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
The Roles of Exosomes in Metastasis of Sarcoma: From Biomarkers to Therapeutic Targets. Biomolecules 2023; 13:biom13030456. [PMID: 36979391 PMCID: PMC10046038 DOI: 10.3390/biom13030456] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
Sarcoma is a heterogeneous group of mesenchymal neoplasms with a high rate of lung metastasis. The cellular mechanisms responsible for sarcoma metastasis remain poorly understood. Furthermore, there are limited efficacious therapeutic strategies for treating metastatic sarcoma. Improved diagnostic and therapeutic modalities are of increasing importance for the treatment of sarcoma due to their high mortality in the advanced stages of the disease. Recent evidence demonstrates that the exosome, a type of extracellular vesicle released by virtually all cells in the body, is an important facilitator of intercellular communication between the cells and the surrounding environment. The exosome is gaining significant attention among the medical research community, but there is little knowledge about how the exosome affects sarcoma metastasis. In this review, we summarize the multifaceted roles of sarcoma-derived exosomes in promoting the process of metastasis via the formation of pre-metastatic niche (PMN), the regulation of immunity, angiogenesis, vascular permeability, and the migration of sarcoma cells. We also highlight the potential of exosomes as innovative diagnostic and prognostic biomarkers as well as therapeutic targets in sarcoma metastasis.
Collapse
|
8
|
Tian H, Cao J, Li B, Nice EC, Mao H, Zhang Y, Huang C. Managing the immune microenvironment of osteosarcoma: the outlook for osteosarcoma treatment. Bone Res 2023; 11:11. [PMID: 36849442 PMCID: PMC9971189 DOI: 10.1038/s41413-023-00246-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/17/2022] [Accepted: 12/29/2022] [Indexed: 03/01/2023] Open
Abstract
Osteosarcoma, with poor survival after metastasis, is considered the most common primary bone cancer in adolescents. Notwithstanding the efforts of researchers, its five-year survival rate has only shown limited improvement, suggesting that existing therapeutic strategies are insufficient to meet clinical needs. Notably, immunotherapy has shown certain advantages over traditional tumor treatments in inhibiting metastasis. Therefore, managing the immune microenvironment in osteosarcoma can provide novel and valuable insight into the multifaceted mechanisms underlying the heterogeneity and progression of the disease. Additionally, given the advances in nanomedicine, there exist many advanced nanoplatforms for enhanced osteosarcoma immunotherapy with satisfactory physiochemical characteristics. Here, we review the classification, characteristics, and functions of the key components of the immune microenvironment in osteosarcoma. This review also emphasizes the application, progress, and prospects of osteosarcoma immunotherapy and discusses several nanomedicine-based options to enhance the efficiency of osteosarcoma treatment. Furthermore, we examine the disadvantages of standard treatments and present future perspectives for osteosarcoma immunotherapy.
Collapse
Affiliation(s)
- Hailong Tian
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Jiangjun Cao
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Bowen Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Edouard C. Nice
- grid.1002.30000 0004 1936 7857Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800 Australia
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, 315020, People's Republic of China.
| | - Yi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
9
|
Antitumor Therapy Targeting the Tumor Microenvironment. JOURNAL OF ONCOLOGY 2023; 2023:6886135. [PMID: 36908706 PMCID: PMC10005879 DOI: 10.1155/2023/6886135] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023]
Abstract
The development and progression of tumors in human tissues extensively rely on its surrounding environment, that is, tumor microenvironment which includes a variety of cells, molecules, and blood vessels. These components are modified, organized, and integrated to support and facilitate the growth, invasion, and metabolism of tumor cells, suggesting them as potential therapeutic targets in anticancer treatment. An increasing number of pharmacological agents have been developed and clinically applied to target the oncogenic components in the tumor microenvironment, and in this review, we will summarize these pharmacological agents that directly or indirectly target the cellular or molecular components in the tumor microenvironment. However, difficulties and challenges still exist in this field, which will also be reported in this literature.
Collapse
|
10
|
Mercatali L, Vanni S, Miserocchi G, Liverani C, Spadazzi C, Cocchi C, Calabrese C, Gurrieri L, Fausti V, Riva N, Genovese D, Lucarelli E, Focarete ML, Ibrahim T, Calabrò L, De Vita A. The emerging role of cancer nanotechnology in the panorama of sarcoma. Front Bioeng Biotechnol 2022; 10:953555. [PMID: 36324885 PMCID: PMC9618700 DOI: 10.3389/fbioe.2022.953555] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/28/2022] [Indexed: 11/21/2022] Open
Abstract
In the field of nanomedicine a multitude of nanovectors have been developed for cancer application. In this regard, a less exploited target is represented by connective tissue. Sarcoma lesions encompass a wide range of rare entities of mesenchymal origin affecting connective tissues. The extraordinary diversity and rarity of these mesenchymal tumors is reflected in their classification, grading and management which are still challenging. Although they include more than 70 histologic subtypes, the first line-treatment for advanced and metastatic sarcoma has remained unchanged in the last fifty years, excluding specific histotypes in which targeted therapy has emerged. The role of chemotherapy has not been completely elucidated and the outcomes are still very limited. At the beginning of the century, nano-sized particles clinically approved for other solid lesions were tested in these neoplasms but the results were anecdotal and the clinical benefit was not substantial. Recently, a new nanosystem formulation NBTXR3 for the treatment of sarcoma has landed in a phase 2-3 trial. The preliminary results are encouraging and could open new avenues for research in nanotechnology. This review provides an update on the recent advancements in the field of nanomedicine for sarcoma. In this regard, preclinical evidence especially focusing on the development of smart materials and drug delivery systems will be summarized. Moreover, the sarcoma patient management exploiting nanotechnology products will be summed up. Finally, an overlook on future perspectives will be provided.
Collapse
Affiliation(s)
- Laura Mercatali
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Silvia Vanni
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giacomo Miserocchi
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Chiara Liverani
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Chiara Spadazzi
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Claudia Cocchi
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Chiara Calabrese
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lorena Gurrieri
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Valentina Fausti
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Nada Riva
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Damiano Genovese
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Bologna, Italy
| | - Enrico Lucarelli
- Osteoncologia, Sarcomi dell’osso e dei tessuti molli, e Terapie Innovative, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Toni Ibrahim
- Osteoncologia, Sarcomi dell’osso e dei tessuti molli, e Terapie Innovative, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Luana Calabrò
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Alessandro De Vita
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- *Correspondence: Alessandro De Vita,
| |
Collapse
|
11
|
Hashimoto K, Nishimura S, Ito T, Oka N, Kakinoki R, Akagi M. Clinicopathological assessment of cancer/testis antigens NY‑ESO‑1 and MAGE‑A4 in osteosarcoma. Eur J Histochem 2022; 66:3377. [PMID: 35736245 PMCID: PMC9251608 DOI: 10.4081/ejh.2022.3377] [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: 12/28/2021] [Accepted: 06/16/2022] [Indexed: 11/23/2022] Open
Abstract
The cancer/testis antigens (CTAs), New York esophageal squamous cell carcinoma-1 (NY-ESO-1) and melanoma antigen gene (MAGE)-A4 are normally restricted to male germ cells but are aberrantly expressed in several cancers. Considering the limited information regarding their significance in osteosarcoma (OS), the purpose of this study was to determine the clinical significance of NY-ESO-1 and MAGE-A4 expression in OS. Nine patients with OS treated at Kindai University Hospital were included in the study. The median age was 27 years, and median follow-up period was 40 months. The specimens obtained at the time of biopsy were used to perform immunostaining for NY-ESO, MAGE-A4, p53, and Ki-67. The positive cell rates and positive case rates of NY-ESO, MAGE-A4, p53, and Ki-67 were calculated. The correlation between the positive cell rate of immunohistochemical markers was also calculated. The correlation between the positive cell rate of NY-ESO-1 or MAGE-A4 and tumor size or maximum standardized uptake (SUV-max) was also determined. The positive cell rates of NY-ESO-1 or MAGE-A4 in continuous disease-free (CDF) cases were also compared with those in alive with disease (AWD) or dead of disease (DOD) cases. The average positive cell rates of NY-ESO, MAGEA4, p53, and Ki-67 were 71.7%, 85.1%, 16.2%, and 14.7%, and their positive case rates were 33.3%, 100%, 44.4%, and 100%, respectively. The positivity rates of NY-ESO-1 and p53 were strongly correlated, whereas those of NY-ESO-1 and Ki-67 were moderately correlated. The MAGE-A4 and p53 positivity rates and the MAGE-A4 and Ki-67 positive cell rates were both strongly correlated. The NY-ESO-1 and MAGE-A4 positivity rates were moderately correlated. The positive correlation between the NY-ESO-1 positive cell rate and tumor size was medium, and that between the MAGE-A4 positivity rate and SUV-max was very strong. There was no significant difference in the positive cell rates of NY-ESO-1 or MAGE-A4 between CDF cases and AWD or DOD cases. Overall, our results suggest that NY-ESO-1 and MAGE-A4 may be involved in the aggressiveness of OS.
Collapse
Affiliation(s)
- Kazuhiko Hashimoto
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama City, Osaka.
| | - Shunji Nishimura
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama City, Osaka.
| | - Tomohiko Ito
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama City, Osaka.
| | - Naohiro Oka
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama City, Osaka.
| | - Ryosuke Kakinoki
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama City, Osaka.
| | - Masao Akagi
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama City, Osaka.
| |
Collapse
|
12
|
Hashimoto K, Nishimura S, Ito T, Kakinoki R, Akagi M. Immunohistochemical expression and clinicopathological assessment of PD-1, PD-L1, NY-ESO-1, and MAGE-A4 expression in highly aggressive soft tissue sarcomas. Eur J Histochem 2022; 66. [PMID: 35448937 PMCID: PMC9046686 DOI: 10.4081/ejh.2022.3393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/16/2022] [Indexed: 12/05/2022] Open
Abstract
Immunotherapy has altered the treatment paradigm for soft tissue sarcomas (STSs). Considering the limited information regarding the clinical significance of immunohistochemical markers in STS, the purpose of this study was to determine the clinical significance of programmed cell death-1 (PD-1), PD ligand-1(PD-L1), New York esophageal squamous cell carcinoma-1 (NY-ESO-1), and melanoma-associated antigen-A4 (MAGE-A4) expression in STSs. Twenty-two patients (median age, 72.5 years) with STSs treated at our hospital were included in this study. The specimens obtained at the time of biopsy were used to perform immunostaining for PD-1, PD-L1, NY-ESO, and MAGE-A4. The rates of PD-1-, PD-L1-, NY-ESO-, and MAGE-A4-positive cells and cases were calculated. The correlations among the positive cell rates of the immunohistochemical markers as well as their correlations with the histological grade, tumor size, or maximum standardized uptake (SUVmax) value were also determined. The average rates of PD-1-, PD-L1-, NY-ESO-, and MAGE-A4-positive cells were 4.39%, 28.0%, 18.2%, and 39.4%, respectively. PD-1-, PD-L1-, NY-ESO-1-, and MAGE-A4- positive cell rates showed weak to strong correlations with the SUVmax value. Thus, PD-1, PD-L1, NY-ESO, and MAGE-A4 expressions might be involved in the aggressive elements of STSs.
Collapse
Affiliation(s)
- Kazuhiko Hashimoto
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama City, Osaka.
| | - Shunji Nishimura
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama City, Osaka.
| | - Tomohiko Ito
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama City, Osaka.
| | - Ryosuke Kakinoki
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama City, Osaka.
| | - Masao Akagi
- Department of Orthopedic Surgery, Kindai University Hospital, Osaka-Sayama City, Osaka.
| |
Collapse
|
13
|
Lu Y, Zhang J, Chen Y, Kang Y, Liao Z, He Y, Zhang C. Novel Immunotherapies for Osteosarcoma. Front Oncol 2022; 12:830546. [PMID: 35433427 PMCID: PMC9012135 DOI: 10.3389/fonc.2022.830546] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/28/2022] [Indexed: 02/05/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone sarcoma mainly affecting adolescents and young adults, which often progresses to pulmonary metastasis and leads to the death of OS patients. OS is characterized as a highly heterogeneous cancer type and the underlying pathologic mechanisms triggering tumor progress and metastasis are incompletely recognized. Surgery combined with neoadjuvant and postoperative chemotherapy has elevated 5-year survival to over 70% for patients with localized OS tumors, as opposed to only 20% of patients with recurrence and/or metastasis. Therefore, novel therapeutic strategies are needed to overcome the drawbacks of conventional treatments. Immunotherapy is gaining momentum for the treatment of OS with an increasing number of FDA-approved therapies for malignancies resistant to conventional therapies. Here, we review the OS tumor microenvironment and appraise the promising immunotherapies available in the management of OS.
Collapse
Affiliation(s)
- Yubao Lu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiahe Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yutong Chen
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuchen Kang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Zhipeng Liao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuanqi He
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Cangyu Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- *Correspondence: Cangyu Zhang,
| |
Collapse
|
14
|
Clinicopathological Assessment of Cancer/Testis Antigens NY-ESO-1 and MAGE-A4 in Highly Aggressive Soft Tissue Sarcomas. Diagnostics (Basel) 2022; 12:diagnostics12030733. [PMID: 35328286 PMCID: PMC8946957 DOI: 10.3390/diagnostics12030733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/04/2022] [Accepted: 03/16/2022] [Indexed: 11/17/2022] Open
Abstract
We aimed to investigate the clinical significance of the expression of NY-ESO-1 and MAGE-A4 in soft tissue sarcoma (STS). Immunostaining for NY-ESO-1, MAGE-A4, and Ki67 was performed using pathological specimens harvested from 10 undifferentiated pleomorphic sarcoma (UPS), nine myxofibrosarcoma (MFS), and three malignant peripheral nerve sheath tumor (MPNST) patients treated at our hospital. We examined the correlation of NY-ESO-1 and MAGE-A4 expression levels with tumor size, histological grade, and SUVmax values. Positive cell rates of various markers were also compared between patients in remission and those who were not in remission. The rates of cases positive for NY-ESO, MAGE-A4, and Ki67 were 50%, 63.6%, and 90.9%, respectively. The average rates of cells positive for NY-ESO, MAGE-A4, and Ki67 in all STS types were 18.2%, 39.4%, and 16.8%, respectively. A positive correlation was observed between rates of cells positive for NY-ESO-1 and MAGE-A4 and between NY-ESO-1 and MAGE-A4 expression levels and clinical features. There was no significant difference in the positive cell rate of NY-ESO-1 or MAGE-A4 between remission and non-remission cases. Our results suggest that NY-ESO-1 and MAGE-A4 expression may be useful for the diagnosis and prognostication of UPS, MFS, and MPNST.
Collapse
|
15
|
Odri GA, Tchicaya-Bouanga J, Yoon DJY, Modrowski D. Metastatic Progression of Osteosarcomas: A Review of Current Knowledge of Environmental versus Oncogenic Drivers. Cancers (Basel) 2022; 14:cancers14020360. [PMID: 35053522 PMCID: PMC8774233 DOI: 10.3390/cancers14020360] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Osteosarcomas are heterogeneous bone tumors with complex genetic and chromosomic alterations. The numerous patients with metastatic osteosarcoma have a very poor prognosis, and only those who can have full surgical resection of the primary tumor and of all the macro metastasis can survive. Despite the recent improvements in prediction and early detection of metastasis, big efforts are still required to understand the specific mechanisms of osteosarcoma metastatic progression, in order to reveal novel therapeutic targets. Abstract Metastases of osteosarcomas are heterogeneous. They may grow simultaneously with the primary tumor, during treatment or shortly after, or a long time after the end of the treatment. They occur mainly in lungs but also in bone and various soft tissues. They can have the same histology as the primary tumor or show a shift towards a different differentiation path. However, the metastatic capacities of osteosarcoma cells can be predicted by gene and microRNA signatures. Despite the identification of numerous metastasis-promoting/predicting factors, there is no efficient therapeutic strategy to reduce the number of patients developing a metastatic disease or to cure these metastatic patients, except surgery. Indeed, these patients are generally resistant to the classical chemo- and to immuno-therapy. Hence, the knowledge of specific mechanisms should be extended to reveal novel therapeutic approaches. Recent studies that used DNA and RNA sequencing technologies highlighted complex relations between primary and secondary tumors. The reported results also supported a hierarchical organization of the tumor cell clones, suggesting that cancer stem cells are involved. Because of their chemoresistance, their plasticity, and their ability to modulate the immune environment, the osteosarcoma stem cells could be important players in the metastatic process.
Collapse
Affiliation(s)
- Guillaume Anthony Odri
- INSERM UMR 1132, Biologie de l’os et du Cartilage (BIOSCAR), Lariboisière Hospital, UFR de Médecine, Faculté de Santé, University of Paris, 75010 Paris, France; (J.T.-B.); (D.J.Y.Y.); (D.M.)
- Service de Chirurgie Orthopédique et Traumatologique, DMU Locomotion, Lariboisière Hospital, 75010 Paris, France
- Correspondence:
| | - Joëlle Tchicaya-Bouanga
- INSERM UMR 1132, Biologie de l’os et du Cartilage (BIOSCAR), Lariboisière Hospital, UFR de Médecine, Faculté de Santé, University of Paris, 75010 Paris, France; (J.T.-B.); (D.J.Y.Y.); (D.M.)
| | - Diane Ji Yun Yoon
- INSERM UMR 1132, Biologie de l’os et du Cartilage (BIOSCAR), Lariboisière Hospital, UFR de Médecine, Faculté de Santé, University of Paris, 75010 Paris, France; (J.T.-B.); (D.J.Y.Y.); (D.M.)
- Service de Chirurgie Orthopédique et Traumatologique, DMU Locomotion, Lariboisière Hospital, 75010 Paris, France
| | - Dominique Modrowski
- INSERM UMR 1132, Biologie de l’os et du Cartilage (BIOSCAR), Lariboisière Hospital, UFR de Médecine, Faculté de Santé, University of Paris, 75010 Paris, France; (J.T.-B.); (D.J.Y.Y.); (D.M.)
| |
Collapse
|
16
|
The Value of Immune-Related Genes Signature in Osteosarcoma Based on Weighted Gene Co-expression Network Analysis. J Immunol Res 2021. [DOI: 10.1155/2021/9989321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background. Osteosarcoma (OS) is a serious malignant tumor that is more common in adolescents or children under 20 years of age. This study is aimed at obtaining immune-related genes (IRGs) associated with the progression and prognosis of OS. Method. Expression profiling data and clinical data for OS were downloaded from the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database. ESTIMATE calculates immune scores and stromal scores of samples and performs the prognostic analysis. Weighted gene coexpression network analysis (WGCNA) was used to find modules correlated with immune and stromal scores. Cox regression analysis and least absolute shrinkage and selection operator (LASSO) analysis were used to explore IRGs associated with OS prognosis and construct and validate a hazard score model. Finally, we verified the expression and function of EVI2B in OS. Results. WGCNA selected twenty-eight IRGs, 10 of which were associated with OS prognosis, and LASSO further obtained three key prognostic genes. A prognostic model of EVI2B was constructed, and according to the risk score model, patients in the high-risk group had a worse prognosis than those in the low-risk group, and the prognosis was statistically significant in the high- and low-risk groups. Receiver operating characteristic (ROC) curves were used to assess the prognostic model’s accuracy and externally validate the independent GSE21257 cohort. The results of immunohistochemical staining and qPCR showed that EVI2B was a tumor suppressor gene. The differential genes in the high- and low-risk groups were analyzed by enrichment analysis of GO and KEGG, indicating that the EVI2B model is associated with immune response. Conclusion. In this study, IRG EVI2B is closely related to OS’s prognosis and can be used as a potential biomarker for prognosis and treatment of OS.
Collapse
|
17
|
Wang J, Gong M, Xiong Z, Zhao Y, Xing D. Immune-related prognostic genes signatures in the tumor microenvironment of sarcoma. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:2243-2257. [PMID: 33892543 DOI: 10.3934/mbe.2021113] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Sarcomas are a heterogeneous group of malignant mesenchymal neoplasms. This study aimed to investigate the immune-related prognostic gene signatures in the tumor microenvironment of sarcoma. The RNA-sequencing data and clinical phenotype data of 260 sarcoma samples and two normal samples were downloaded from The Cancer Genome Atla (TCGA) database. Tumor purity and immune cells infiltration were evaluated by Estimation of Stromal and Immune cells in Malignant Tumors using Expression data (ESTIMATE) deconvolution algorithm. Differentially expressed genes (DEGs) were screened in high vs. low immune score groups. Survival analysis was performed using Kaplan-Meier curve with log-rank test. Tumor infiltrating of immune cells was analyzed by Tumor Immune Estimation Resource (TIMER). High immune score and ESTIMATE score were associated with favorable prognosis. A total of 623 immune DEGs were screened. The majority of these genes (532 genes accounting for 85% of the DEGs) were up-regulated, and these genes were significantly enriched in various immune related biological processed and pathways, such as neutrophil activation, T cell activation, antigen processing and presentation. A total of 146 prognosis-related immune DEGs, and seven hub genes were identified, including B2M, HLA-DRB1, HLA-DRA, HLA-E, LCK, HLA-DPA1, and VAV1. Survival analysis showed that high expression of these genes was associated with a favorable prognosis. There were negative correlations between the expression of these hub genes and tumor purity, while positive correlations between expression of these hub genes and f infiltration levels of B cells, CD4+ T cells, CD8+ T cells, neutrophils, macrophages and dendritic cells. These results help to stratify patients with different immune subtypes and help to design immunotherapy strategies for these patients in sarcoma.
Collapse
Affiliation(s)
- Jun Wang
- Department of Orthopedics and Trauma, The Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan, 250033, China
| | - Mingzhi Gong
- Department of Orthopedics and Trauma, The Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan, 250033, China
| | - Zhenggang Xiong
- Department of Orthopedics and Trauma, The Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan, 250033, China
| | - Yangyang Zhao
- Department of Orthopedics and Trauma, The Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan, 250033, China
| | - Deguo Xing
- Department of Orthopedics and Trauma, The Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan, 250033, China
| |
Collapse
|
18
|
Immunotherapy for osteosarcoma: Fundamental mechanism, rationale, and recent breakthroughs. Cancer Lett 2020; 500:1-10. [PMID: 33359211 DOI: 10.1016/j.canlet.2020.12.024] [Citation(s) in RCA: 228] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Osteosarcoma (OS) is the most common primary malignancy of the bone and has a high propensity for local invasion and metastasis. Although combining surgery with chemotherapy has immensely improved the outcomes of osteosarcoma patients, the prognosis of metastatic or recurrent osteosarcomas is still unsatisfactory. Immunotherapy has proven to be a promising therapeutic strategy against human malignancies and improved understanding of the immune response to OS, and biomarker development has increased the number of patients who benefit from immunotherapies in recent years. Here, we review recent advances in immunotherapy in osteosarcoma and discuss the mechanisms and status of immunotherapies in both preclinical and clinical trials as well as future therapies on the horizon. These advances may pave the way for novel treatments requisite for patients with osteosarcoma in need of new therapies.
Collapse
|
19
|
Prospects for NK Cell Therapy of Sarcoma. Cancers (Basel) 2020; 12:cancers12123719. [PMID: 33322371 PMCID: PMC7763692 DOI: 10.3390/cancers12123719] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Sarcomas are a group of aggressive tumors originating from mesenchymal tissues. Patients with advanced disease have poor prognosis due to the ineffectiveness of current treatment protocols. A subset of lymphocytes called natural killer (NK) cells is capable of effective surveillance and clearance of sarcomas, constituting a promising tool for immunotherapeutic treatment. However, sarcomas can cause impairment in NK cell function, associated with enhanced tumor growth and dissemination. In this review, we discuss the molecular mechanisms of sarcoma-mediated suppression of NK cells and their implications for the design of novel NK cell-based immunotherapies against sarcoma. Abstract Natural killer (NK) cells are innate lymphoid cells with potent antitumor activity. One of the most NK cell cytotoxicity-sensitive tumor types is sarcoma, an aggressive mesenchyme-derived neoplasm. While a combination of radical surgery and radio- and chemotherapy can successfully control local disease, patients with advanced sarcomas remain refractory to current treatment regimens, calling for novel therapeutic strategies. There is accumulating evidence for NK cell-mediated immunosurveillance of sarcoma cells during all stages of the disease, highlighting the potential of using NK cells as a therapeutic tool. However, sarcomas display multiple immunoevasion mechanisms that can suppress NK cell function leading to an uncontrolled tumor outgrowth. Here, we review the current evidence for NK cells’ role in immune surveillance of sarcoma during disease initiation, promotion, progression, and metastasis, as well as the molecular mechanisms behind sarcoma-mediated NK cell suppression. Further, we apply this basic understanding of NK–sarcoma crosstalk in order to identify and summarize the most promising candidates for NK cell-based sarcoma immunotherapy.
Collapse
|
20
|
Jafari F, Javdansirat S, Sanaie S, Naseri A, Shamekh A, Rostamzadeh D, Dolati S. Osteosarcoma: A comprehensive review of management and treatment strategies. Ann Diagn Pathol 2020; 49:151654. [PMID: 33130384 DOI: 10.1016/j.anndiagpath.2020.151654] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/09/2020] [Accepted: 10/20/2020] [Indexed: 01/25/2023]
Abstract
Osteosarcoma, a bone cancer usually seen in children and young adults, is generally a high-grade malignancy presented by extreme metastases to the lungs. Osteosarcoma has a tendency for appearing in bones with rapid growth rate. The etiology of osteosarcoma is multifaceted and poorly understood. A molecular consideration of this disease will lead to a directed tumor treatment. The present treatment for osteosarcoma comprises of an arrangement of systemic chemotherapy and wide surgical resection. Survival rate is increased by the progress of destructive systemic chemotherapies. So, the development of new treatment approaches for metastatic osteosarcoma is essential. Immunomodulation has been used in clinical settings. Through targeting surface antigens expressed on tumor cells, particular antibodies and exploitation of cellular immunotherapy against sarcomas have been confirmed to be effective as cancer therapeutics. In this article, we have reviewed epidemiology, etiology, pathogenesis, diagnosis, and treatment of osteosarcoma and we have focused on different methods of immunotherapy including vaccines, cell-based immunotherapy, cytokines, and monoclonal antibodies.
Collapse
Affiliation(s)
- Farzaneh Jafari
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Javdansirat
- Clinical Research development unit Center, Beheshti Hospital, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Sarvin Sanaie
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirreza Naseri
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davood Rostamzadeh
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
The Evolving Role of Radiation Therapy in Patients with Metastatic Soft Tissue Sarcoma. Curr Oncol Rep 2020; 22:79. [DOI: 10.1007/s11912-020-00936-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Houghton PJ, Kurmasheva RT. Challenges and Opportunities for Childhood Cancer Drug Development. Pharmacol Rev 2019; 71:671-697. [PMID: 31558580 PMCID: PMC6768308 DOI: 10.1124/pr.118.016972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer in children is rare with approximately 15,700 new cases diagnosed in the United States annually. Through use of multimodality therapy (surgery, radiation therapy, and aggressive chemotherapy), 70% of patients will be "cured" of their disease, and 5-year event-free survival exceeds 80%. However, for patients surviving their malignancy, therapy-related long-term adverse effects are severe, with an estimated 50% having chronic life-threatening toxicities related to therapy in their fourth or fifth decade of life. While overall intensive therapy with cytotoxic agents continues to reduce cancer-related mortality, new understanding of the molecular etiology of many childhood cancers offers an opportunity to redirect efforts to develop effective, less genotoxic therapeutic options, including agents that target oncogenic drivers directly, and the potential for use of agents that target the tumor microenvironment and immune-directed therapies. However, for many high-risk cancers, significant challenges remain.
Collapse
Affiliation(s)
- Peter J Houghton
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| |
Collapse
|
23
|
Siozopoulou V, Marcq E, Jacobs J, Zwaenepoel K, Hermans C, Brauns J, Pauwels S, Huysentruyt C, Lammens M, Somville J, Smits E, Pauwels P. Desmoid tumors display a strong immune infiltration at the tumor margins and no PD-L1-driven immune suppression. Cancer Immunol Immunother 2019; 68:1573-1583. [DOI: 10.1007/s00262-019-02390-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 09/01/2019] [Indexed: 12/11/2022]
|
24
|
Anwar MA, El-Baba C, Elnaggar MH, Elkholy YO, Mottawea M, Johar D, Al Shehabi TS, Kobeissy F, Moussalem C, Massaad E, Omeis I, Darwiche N, Eid AH. Novel therapeutic strategies for spinal osteosarcomas. Semin Cancer Biol 2019; 64:83-92. [PMID: 31152785 DOI: 10.1016/j.semcancer.2019.05.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 05/28/2019] [Accepted: 05/28/2019] [Indexed: 12/24/2022]
Abstract
At the dawn of the third millennium, cancer has become the bane of twenty-first century man, and remains a predominant public health burden, affecting welfare and life expectancy globally. Spinal osteogenic sarcoma, a primary spinal malignant tumor, is a rare and challenging neoplastic disease to treat. After the conventional therapeutic modalities of chemotherapy, radiation and surgery have been exhausted, there is currently no available alternative therapy in managing cases of spinal osteosarcoma. The defining signatures of tumor survival are characterised by cancer cell ability to stonewall immunogenic attrition and apoptosis by various means. Some of these biomarkers, namely immune-checkpoints, have recently been exploited as druggable targets in osteosarcoma and many other different cancers. These promising strides made by the use of reinvigorated immunotherapeutic approaches may lead to significant reduction in spinal osteosarcoma disease burden and corresponding reciprocity in increase of survival rates. In this review, we provide the background to spinal osteosarcoma, and proceed to elaborate on contribution of the complex ecology within tumor microenvironment giving arise to cancerous immune escape, which is currently receiving considerable attention. We follow this section on the tumor microenvironment by a brief history of cancer immunity. Also, we draw on the current knowledge of treatment gained from incidences of osteosarcoma at other locations of the skeleton (long bones of the extremities in close proximity to the metaphyseal growth plates) to make a case for application of immunity-based tools, such as immune-checkpoint inhibitors and vaccines, and draw attention to adverse upshots of immune-checkpoint blockers as well. Finally, we describe the novel biotechnique of CRISPR/Cas9 that will assist in treatment approaches for personalized medication.
Collapse
Affiliation(s)
- M Akhtar Anwar
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Chirine El-Baba
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Muhammed H Elnaggar
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Yasmeen O Elkholy
- Microbiology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Mohamed Mottawea
- Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Dina Johar
- Biomedical Sciences Program, Zewail University of Science and Technology, Giza, Egypt
| | | | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Charbel Moussalem
- Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Elie Massaad
- Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ibrahim Omeis
- Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon.
| | - A H Eid
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon; Department of Biomedical Sciences, Qatar University, Doha, Qatar.
| |
Collapse
|
25
|
Dufresne A, Brahmi M, Karanian M, Blay JY. Using biology to guide the treatment of sarcomas and aggressive connective-tissue tumours. Nat Rev Clin Oncol 2019; 15:443-458. [PMID: 29666441 DOI: 10.1038/s41571-018-0012-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sarcomas are a heterogeneous group of malignancies that arise from cells of a mesenchymal origin. Surgery forms the mainstay of the treatment of most patients with localized sarcoma and might be followed or preceded by chemotherapy and/or radiotherapy. In the metastatic setting, systemic treatments tend to improve survival and control symptoms. However, the adverse events and sometimes disappointing outcomes associated with these empirical approaches to treatment indicate a need for new approaches. The advent of next-generation sequencing (NGS) has enabled more targeted treatment of many malignancies based on the presence of specific alterations. NGS analyses of sarcomas have revealed the presence of many alterations that can be targeted using therapies that are already used in patients with other forms of cancer. In this Review, we describe the genomic alterations considered to define specific nosological subgroups of sarcoma and whose contribution to oncogenesis provides a biological rationale for the use of a specific targeted therapy. We also report several less successful examples that should guide researchers and clinicians to better define the extent to which the identification of driver molecular alterations should influence the development of novel treatments.
Collapse
Affiliation(s)
- Armelle Dufresne
- Department of Medical Oncology, Centre Leon Berard, Lyon, France.
| | - Mehdi Brahmi
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| | - Marie Karanian
- Department of Pathology, Centre Leon Berard, Lyon, France
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| |
Collapse
|
26
|
The Utility of NKX2.2 and TLE1 Immunohistochemistry in the Differentiation of Ewing Sarcoma and Synovial Sarcoma. Appl Immunohistochem Mol Morphol 2019; 27:174-179. [DOI: 10.1097/pai.0000000000000573] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Kakimoto T, Matsumine A, Kageyama S, Asanuma K, Matsubara T, Nakamura T, Iino T, Ikeda H, Shiku H, Sudo A. Immunohistochemical expression and clinicopathological assessment of the cancer testis antigens NY-ESO-1 and MAGE-A4 in high-grade soft-tissue sarcoma. Oncol Lett 2019; 17:3937-3943. [PMID: 30881511 DOI: 10.3892/ol.2019.10044] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/31/2019] [Indexed: 12/25/2022] Open
Abstract
The aim of the present study was to explore the expression of the cancer testis antigens New York-esophageal squamous cell carcinoma (NY-ESO)-1 and melanoma-associated antigen (MAGE)-A4 in high-grade soft-tissue sarcoma and to evaluate their association with the standard clinical-pathological features of surgically treated high-grade sarcoma patients. The study included 82 patients, and NY-ESO-1 and MAGE-A4 antigen expression was analyzed immunohistochemically. The results revealed NY-ESO-1- and MAGE-A4-positive staining in 58.8 and 52.9% of synovial sarcomas, and 55.6 and 0% of myxoid liposarcomas, respectively. In patients with synovial sarcoma, NY-ESO-1 and MAGE-A4 were expressed in 7 patients, only NY-ESO-1 was expressed in 3 patients, and only MAGE-A4 was expressed in 2 patients. Univariate analysis indicated that a significantly higher MAGE-A4 expression was observed in younger patients (P<0.001) and those with synovial sarcoma (P<0.001). Multivariate analysis indicated that significantly higher NY-ESO-1 expression was observed in patients with synovial sarcoma (P<0.01) and myxoid liposarcoma (P<0.01), and significantly higher MAGE-A4 expression was observed in patients with synovial sarcoma (P<0.01). In high-grade sarcomas, the 2- and 5-year overall survival rates based on Kaplan-Meier estimates were 100 and 81.3% in the NY-ESO-1-positive group, and 69.7 and 53.0% in the NY-ESO-1-negative group, respectively (P=0.049). It was also demonstrated that either NY-ESO-1 or MAGE-A4 was positive in 70.6% of synovial sarcomas. These results indicate that NY-ESO-1 and MAGE-A4 may be useful for the diagnosis of synovial sarcoma. The independent expression of NY-ESO-1 and MAGE-A4, which may help expand the pool of candidates for molecular-targeted immunotherapy, will be beneficial for synovial sarcoma patients.
Collapse
Affiliation(s)
- Takuya Kakimoto
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Mie 514-8507, Japan
| | - Akihiko Matsumine
- Department of Orthopedics and Rehabilitation Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Shinichi Kageyama
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie 514-8507, Japan
| | - Kunihiro Asanuma
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Mie 514-8507, Japan
| | - Takao Matsubara
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Mie 514-8507, Japan
| | - Tomoki Nakamura
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Mie 514-8507, Japan
| | - Takahiro Iino
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Mie 514-8507, Japan
| | - Hiroaki Ikeda
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| | - Hiroshi Shiku
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie 514-8507, Japan
| | - Akihiro Sudo
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Mie 514-8507, Japan
| |
Collapse
|
28
|
HDAC2-mediated upregulation of IL-6 triggers the migration of osteosarcoma cells. Cell Biol Toxicol 2019; 35:423-433. [DOI: 10.1007/s10565-019-09459-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 01/02/2019] [Indexed: 12/14/2022]
|
29
|
Naghavi AO, Yang GQ, Latifi K, Gillies R, McLeod H, Harrison LB. The Future of Radiation Oncology in Soft Tissue Sarcoma. Cancer Control 2018. [PMCID: PMC6291881 DOI: 10.1177/1073274818815504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Radiotherapy (RT) is an important component of the treatment of soft tissue sarcomas (STS) and has been traditionally incorporated with a homogenous approach despite the reality that STS displays a known heterogeneity in clinicopathologic features and treatment outcomes. In this article, we explore the principle components of personalized medicine, including genomics, radiomics, and treatment response, along with their impact on the future of radiation therapy for STS. We propose a shift in the treatment paradigm for STS from a one-size-fits-all technique to one that implements the tenets of personalized medicine and includes the framework for a potential clinical trial technique in this heterogeneous disease.
Collapse
Affiliation(s)
- Arash O. Naghavi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- These authors contributed equally to this work
| | - George Q. Yang
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- These authors contributed equally to this work
| | - Kujtim Latifi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Robert Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Howard McLeod
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Louis B. Harrison
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
30
|
Keremu A, Aimaiti A, Liang Z, Zou X. Role of the HDAC6/STAT3 pathway in regulating PD-L1 expression in osteosarcoma cell lines. Cancer Chemother Pharmacol 2018; 83:255-264. [PMID: 30430228 DOI: 10.1007/s00280-018-3721-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/24/2018] [Indexed: 12/20/2022]
Abstract
Histone deacetylases (HDACs), initially described as histone modifiers, have more recently been verified to target various other proteins unrelated to the chromatin environment. On this basis, findings of the current study demonstrates that the pharmacological or genetic abrogation of HDAC6 in osteosarcoma cell lines down-regulates the expression of program death receptor ligand-1 (PD-L1), an important co-stimulatory molecule expressed in cancer cells, which activates the inhibitory regulatory pathway PD-1 in T cells. As shown by our results, the mechanism by which HDAC6 regulated PD-L1 expression was mediated by the transcription factor STAT3. In addition, we observed that selective HDAC6 inhibitors could inhibit tumor progression in vivo. Crucially, these results provide an essential pre-clinical rationale and justification for the necessity of further research on HDAC6 inhibitors as potential immuno-modulatory agents in osteosarcoma.
Collapse
Affiliation(s)
- Ajimu Keremu
- Orthopedic Center, First People's Hospital of Kashgar, 120 Yingbin Road, Kashgar, 844000, Xinjiang, People's Republic of China
| | - Abudusaimi Aimaiti
- Orthopedic Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, People's Republic of China
| | - Zhilin Liang
- Orthopedic Center, First People's Hospital of Kashgar, 120 Yingbin Road, Kashgar, 844000, Xinjiang, People's Republic of China
| | - Xiaoguang Zou
- Orthopedic Center, First People's Hospital of Kashgar, 120 Yingbin Road, Kashgar, 844000, Xinjiang, People's Republic of China.
| |
Collapse
|
31
|
Keung EZ, Lazar AJ, Torres KE, Wang WL, Cormier JN, Ashleigh Guadagnolo B, Bishop AJ, Lin H, Hunt KK, Bird J, Lewis VO, Patel SR, Wargo JA, Somaiah N, Roland CL. Phase II study of neoadjuvant checkpoint blockade in patients with surgically resectable undifferentiated pleomorphic sarcoma and dedifferentiated liposarcoma. BMC Cancer 2018; 18:913. [PMID: 30249211 PMCID: PMC6154892 DOI: 10.1186/s12885-018-4829-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 09/18/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Soft tissue sarcomas are a heterogeneous and rare group of solid tumors of mesenchymal origin that can arise anywhere in the body. Although surgical resection is the mainstay of treatment for patients with localized disease, disease recurrence is common and 5-year overall survival is poor (~ 65%). Both radiation therapy and conventional chemotherapy are used to reduce local and distant recurrence. However, the utility of radiation therapy is often limited by disease location (in the case of retroperitoneal sarcomas, for instance) while systemic therapy with conventional lines of chemotherapy offer limited efficacy and are often poorly tolerated and associated with significant toxicity. Within the past decade, major advances have been made in the treatment of other malignancies including melanoma, renal cell carcinoma, and non-small cell lung carcinoma with the advent of immune-checkpoint inhibitors such as ipilimumab (anti-CTLA4), pembrolizumab (anti-PD1), and nivolumab (anti-PD1). The recently published SARC028 (NCT02301039), an open label, phase II, multicenter trial of pembrolizumab in patients with advanced bone and soft tissue sarcomas reported promising activity in select histologic subtypes of advanced STS, including undifferentiated pleomorphic sarcoma and dedifferentiated liposarcoma. METHODS There is a clear need for novel and effective adjuncts in the treatment of STS. We hypothesize that immune checkpoint blockade will be effective in patients with surgically resectable primary or locally recurrent dedifferentiated liposarcoma and undifferentiated pleomorphic sarcoma when administered in the neoadjuvant setting. The primary aim of this phase II, single-center, open label, randomized non-comparative trial is to determine the pathologic response to neoadjuvant nivolumab monotherapy and combination nivolumab/ipilimumab in patients with resectable dedifferentiated liposarcoma of the retroperitoneum or undifferentiated pleomorphic sarcoma of the trunk or extremity treated with concurrent standard of care neoadjuvant radiation therapy. DISCUSSION This study will help define the role of single agent anti-PD1 and combination anti-CTLA4 and anti-PD1 therapy in patients with surgically resectable dedifferentiated liposarcoma and undifferentiated pleomorphic sarcoma. TRIAL REGISTRATION ClinicalTrials.gov NCT03307616 , registered October 12, 2017.
Collapse
Affiliation(s)
- Emily Z Keung
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., FCT17.6054, Unit 1484, Houston, TX, 77030, USA
| | - Alexander J Lazar
- Departments of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Departments of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keila E Torres
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., FCT17.6054, Unit 1484, Houston, TX, 77030, USA
- Departments of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei-Lien Wang
- Departments of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Janice N Cormier
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., FCT17.6054, Unit 1484, Houston, TX, 77030, USA
| | - B Ashleigh Guadagnolo
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew J Bishop
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heather Lin
- Departments of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kelly K Hunt
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., FCT17.6054, Unit 1484, Houston, TX, 77030, USA
| | - Justin Bird
- Departments of Orthopaedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Valerae O Lewis
- Departments of Orthopaedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shreyaskumar R Patel
- Departments of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer A Wargo
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., FCT17.6054, Unit 1484, Houston, TX, 77030, USA
- Departments of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neeta Somaiah
- Departments of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christina L Roland
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., FCT17.6054, Unit 1484, Houston, TX, 77030, USA.
| |
Collapse
|
32
|
Bulbul A, Fahy BN, Xiu J, Rashad S, Mustafa A, Husain H, Hayes-Jordan A. Desmoplastic Small Round Blue Cell Tumor: A Review of Treatment and Potential Therapeutic Genomic Alterations. Sarcoma 2017; 2017:1278268. [PMID: 29225486 PMCID: PMC5687144 DOI: 10.1155/2017/1278268] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/17/2017] [Indexed: 02/07/2023] Open
Abstract
Desmoplastic small round blue cell tumors (DSRCTs) originate from a cell with multilineage potential. A molecular hallmark of DSRCT is the EWS-WT1 reciprocal translocation. Ewing sarcoma and DSRCT are treated similarly due to similar oncogene activation pathways, and DSRCT has been represented in very limited numbers in sarcoma studies. Despite aggressive therapy, median survival ranges from 17 to 25 months, and 5-year survival rates remain around 15%, with higher survival reported among those undergoing removal of at least 90% of tumor in the absence of extraperitoneal metastasis. Almost 100% of these tumors contain t(11;22) (p13;q12) translocation, and it is likely that EWS-WT1 functions as a transcription factor possibly through WT1 targets. While there is no standard protocol for this aggressive disease, treatment usually includes the neoadjuvant HD P6 regimen (high-dose cyclophosphamide, doxorubicin, and vincristine (HD-CAV) alternating with ifosfamide and etoposide (IE) chemotherapy combined with aggressively attempted R0 resection). We aimed to review the molecular characteristics of DSRCTs to explore therapeutic opportunities for this extremely rare and aggressive cancer type.
Collapse
Affiliation(s)
- Ajaz Bulbul
- Department of Hematology/Oncology, Kymera Independent Physicians, Carlsbad, NM, USA
- Division of Internal Medicine, Department of Hematology/Oncology, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, USA
| | - Bridget Noel Fahy
- Department of Surgery, University of New Mexico, Albuquerque, NM, USA
| | | | - Sadaf Rashad
- All Saints University School of Medicine, Roseau, Dominica
| | - Asrar Mustafa
- Acharya Shri Chander College of Medical Sciences and Hospital, Jammu, India
| | - Hatim Husain
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Andrea Hayes-Jordan
- Department of Pediatric Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
33
|
Keung EZ, Tsai JW, Ali AM, Cormier JN, Bishop AJ, Guadagnolo BA, Torres KE, Somaiah N, Hunt KK, Wargo JA, Lazar AJ, Wang WL, Roland CL. Analysis of the immune infiltrate in undifferentiated pleomorphic sarcoma of the extremity and trunk in response to radiotherapy: Rationale for combination neoadjuvant immune checkpoint inhibition and radiotherapy. Oncoimmunology 2017; 7:e1385689. [PMID: 29308306 PMCID: PMC5749668 DOI: 10.1080/2162402x.2017.1385689] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/20/2017] [Accepted: 09/24/2017] [Indexed: 02/03/2023] Open
Abstract
Background: Undifferentiated pleomorphic sarcoma of the extremity and trunk (ET-UPS) presents a unique therapeutic challenge. Although immunotherapy has recently been employed in advanced soft tissue sarcoma, there is limited data characterizing the immune infiltrate in ET-UPS. Radiotherapy (RT) has been shown in other tumor types to promote tumor antigen release and enhance tumor-specific targeting by the adaptive immune system. The aim of this study was to 1) characterize the baseline immune infiltrate and 2) evaluate the effect of preoperative RT on the histologic appearance of and the immune infiltrate in ET-UPS. Methods: We identified 17 matched ET-UPS samples before and after RT. Immunohistochemistry was performed with CD8, CD4, PD-L1, PD1, CD3, CD163 and FoxP3 positive cells identified in all samples. Changes in the immune infiltrate following RT were examined. Results: There was a trend towards increased density of tumor infiltrating immune cells in ET-UPS following RT, with increases in median number of CD3 (158 vs 219 cells/mm2, p = 0.06), CD4 (3 vs 13 cells/mm2, p = 0.01), CD8 (55 vs 111 cells/mm2, p = 0.17), and FOXP3 (14 vs 25 cells/mm2, p = 0.23) positive cells. Interestingly, although PD-L1 was not expressed in any ET-UPS tumor at baseline, positive PD-L1 expression was observed in 21% (3/14) of tumors after RT (p = 0.07). Conclusion: An immune infiltrate is present in ET-UPS at the time of diagnosis, with a trend towards increased density of immune infiltrate and PD-L1 expression after RT. These data support prospectively evaluating immune checkpoint inhibitors with standard of care RT in the treatment of ET-UPS.
Collapse
Affiliation(s)
- Emily Z. Keung
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jen-Wei Tsai
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ali M. Ali
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Janice N. Cormier
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrew J. Bishop
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - B. Ashleigh Guadagnolo
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Keila E. Torres
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kelly K. Hunt
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer A. Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alexander J. Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei-Lien Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Christina L. Roland
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
34
|
Nathenson MJ, Conley AP, Sausville E. Immunotherapy: A New (and Old) Approach to Treatment of Soft Tissue and Bone Sarcomas. Oncologist 2017; 23:71-83. [PMID: 28935774 DOI: 10.1634/theoncologist.2016-0025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 07/14/2017] [Indexed: 12/11/2022] Open
Abstract
Soft tissue and bone sarcomas are a rare and heterogeneous form of cancer. With standard of care treatment options including surgery, radiation, and chemotherapy, the long-term survival is still low for high-risk soft tissue sarcoma patients. New treatment strategies are needed. Immunotherapy offers a new potential treatment paradigm with great promise. Immunotherapy of soft tissue sarcomas dates back to Dr. Coley's first use of toxins in the late 1800s. A variety of strategies of immunotherapy have been tried in soft tissue and bone sarcomas, including various vaccines and cytokines, with limited success. Results of these early clinical trials with vaccines and cytokines were disappointing, but there are reasons to be optimistic. Recent advances, particularly with the use of adoptive T-cell therapy and immune checkpoint inhibitors, have led to a resurgence of this field for all cancer patients. Clinical trials utilizing adoptive T-cell therapy and immune checkpoint inhibitors in soft tissue and bone sarcomas are under way. This paper reviews the current state of evidence for the use of immunotherapy, as well as current immunotherapy strategies (vaccines, adopative T-cell therapy, and immune checkpoint blockade), in soft tissue and bone sarcomas. By understanding the tumor microenviroment of sarcomas and how it relates to their immunoresponsiveness, better immunotherapy clinical trials can be designed, hopefully with improved outcomes for soft tissue and bone sarcoma patients. IMPLICATIONS FOR PRACTICE Immunotherapy is a promising treatment paradigm that is gaining acceptance for the management of several cancers, including melanoma, renal cell carcinoma, prostate cancer, and lung cancer. There is a long history of immunotherapy in the treatment of soft tissue and bone sarcomas, although with little success. It is important to understand past failures to develop future immunotherapy treatment strategies with an improved possibility of success. This article reviews the history of and current state of immunotherapy research in the treatment of soft tissue and bone sarcomas, with particular regard to vaccine trials, adoptive T-cell therapy, and immune checkpoint blockade.
Collapse
Affiliation(s)
- Michael J Nathenson
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Anthony P Conley
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Edward Sausville
- Department of Medicine and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
35
|
Costa Arantes DA, Gonçalves AS, Jham BC, Duarte ECB, de Paula ÉC, de Paula HM, Mendonça EF, Batista AC. Evaluation of HLA-G, HLA-E, and PD-L1 proteins in oral osteosarcomas. Oral Surg Oral Med Oral Pathol Oral Radiol 2017; 123:e188-e196. [PMID: 28159587 DOI: 10.1016/j.oooo.2016.12.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 11/26/2016] [Accepted: 12/01/2016] [Indexed: 01/16/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the expression of human leukocyte antigens (HLAs) G and E and programmed death-ligand 1 (PD-L1) in oral osteosarcoma (OO) (n = 13). The relationship between the expression of these molecules and histologic grading and metastasis was also evaluated. STUDY DESIGN HLA-G, HLA-E, and PD-L1 were identified by immunohistochemistry. Samples of normal bone tissue (n = 6) were used as controls. The sections were evaluated using a semiquantitative scoring system with an immunoreactive score, where a score of 0 was considered absent, ≤2 was low, and >2 was high expression. RESULTS We identified high expression of HLA-G, HLA-E, and PD-L1 by malignant osteoblastic cells in 69.2% of OO cases, which was statistically higher than that in controls (P < .05). Overexpression of these proteins was identified in 8 of 11 samples of high-grade and 1 of 2 samples of low-grade OO. Additionally, 66.6% of patients with metastases (n = 4) and 71.4% of patients without metastases (n = 5) had high expression of HLA-G, HLA-E, and PD-L1 in tumor samples (P > .05). CONCLUSION OO had high expression of HLA-G, HLA-E, and PD-L1 irrespective of clinicopathologic parameters, including histologic grading and metastasis.
Collapse
Affiliation(s)
| | - Andréia Souza Gonçalves
- Department of Stomatology (Oral Pathology), Dental School, Federal University of Goiás, Goiânia, Brazil
| | - Bruno Correia Jham
- College of Dental Medicine-Illinois, Midwestern University, Downers Grove, IL, USA
| | | | - Élbio Candido de Paula
- Division of Anatomic Pathology, Araújo Jorge Hospital, Association of Cancer Combat of Goiás, Goiânia, Brazil
| | - Henrique Moura de Paula
- Division of Anatomic Pathology, Araújo Jorge Hospital, Association of Cancer Combat of Goiás, Goiânia, Brazil; Department of Pathology and Medicine Laboratory, Medicine School, Federal University of Goiás, Goiânia, Brazil
| | - Elismauro Francisco Mendonça
- Department of Stomatology (Oral Pathology), Dental School, Federal University of Goiás, Goiânia, Brazil; Head and Neck Division, Araújo Jorge Hospital, Association of Cancer Combat of Goiás, Goiânia, Brazil
| | - Aline Carvalho Batista
- Department of Stomatology (Oral Pathology), Dental School, Federal University of Goiás, Goiânia, Brazil.
| |
Collapse
|
36
|
Tan Y, Trent JC, Wilky BA, Kerr DA, Rosenberg AE. Current status of immunotherapy for gastrointestinal stromal tumor. Cancer Gene Ther 2017; 24:130-133. [DOI: 10.1038/cgt.2016.58] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/03/2016] [Accepted: 10/04/2016] [Indexed: 12/12/2022]
|
37
|
Budczies J, Mechtersheimer G, Denkert C, Klauschen F, Mughal SS, Chudasama P, Bockmayr M, Jöhrens K, Endris V, Lier A, Lasitschka F, Penzel R, Dietel M, Brors B, Gröschel S, Glimm H, Schirmacher P, Renner M, Fröhling S, Stenzinger A. PD-L1 (CD274) copy number gain, expression, and immune cell infiltration as candidate predictors for response to immune checkpoint inhibitors in soft-tissue sarcoma. Oncoimmunology 2017; 6:e1279777. [PMID: 28405504 DOI: 10.1080/2162402x.2017.1279777] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 12/29/2016] [Accepted: 01/04/2017] [Indexed: 12/25/2022] Open
Abstract
Soft-tissue sarcomas (STS) are rare malignancies that account for 1% of adult cancers and comprise more than 50 entities. Current therapeutic options for advanced-stage STS are limited. Immune checkpoint inhibitors targeting the PD-1/PD-L1 signaling axis are being explored as new treatment modality in STS; however, the determinants of response to these agents are largely unknown. Using the sarcoma data set of The Cancer Genome Altas (TCGA) and an independent cohort of untreated high-grade STS, we analyzed DNA copy number status and mRNA expression of PD-L1 in a total of 335 STS cases. Copy number gains (CNG) were detected in 54 TCGA cases (21.1%), of which 21 (8.2%) harbored focal PD-L1 CNG and that were most prevalent in myxofibrosarcoma (35%) and undifferentiated pleomorphic sarcoma (34%). In the untreated high-grade STS cohort, we detected CNG in six cases (7.6%). Analysis of co-amplified genes identified a 5.6-Mb core region comprising 27 genes, including JAK2. Patients with PD-L1 CNG had higher PD-L1 expression compared with STS without CNG (fold change, 1.8; p = 0.02), an effect that was most pronounced in the setting of focal PD-L1 CNG (fold change, 3.0; p = 0.0027). STS with PD-L1 CNG showed a significantly higher mutational load compared with tumors with a diploid PD-L1 locus (median number of mutated genes; 58 vs. 40; p = 3.6E-06), and PD-L1 CNG were associated with inferior survival (HR = 1.82; p = 0.025). In contrast, T-cell infiltrates quantified by mRNA expression of CD3Z were associated with improved survival (HR = 0.88; p = 0.024) and consequently influenced the prognostic power of PD-L1 CNG, with low CD3Z levels conferring poor survival in cases with PD-L1 CNG (HR = 1.8; p = 0.049). These data demonstrate that PD-L1 GNG and elevated expression of PD-L1 occur in a substantial proportion of STS, have prognostic impact that is modulated by T-cell infiltrates, and thus warrant investigation as response predictors for immune checkpoint inhibition.
Collapse
Affiliation(s)
- Jan Budczies
- Institute of Pathology, Charité University Hospital, Berlin, Germany; German Cancer Consortium (DKTK), partner sites Heidelberg and Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Carsten Denkert
- Institute of Pathology, Charité University Hospital, Berlin, Germany; German Cancer Consortium (DKTK), partner sites Heidelberg and Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Sadaf S Mughal
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Disease (NCT) Heidelberg , Heidelberg, Germany
| | - Priya Chudasama
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Michael Bockmayr
- Institute of Pathology, Charité University Hospital , Berlin, Germany
| | - Korinna Jöhrens
- Institute of Pathology, Charité University Hospital , Berlin, Germany
| | - Volker Endris
- Institute of Pathology, University Hospital Heidelberg , Heidelberg, Germany
| | - Amelie Lier
- Institute of Pathology, University Hospital Heidelberg , Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, University Hospital Heidelberg , Heidelberg, Germany
| | - Roland Penzel
- Institute of Pathology, University Hospital Heidelberg , Heidelberg, Germany
| | - Manfred Dietel
- Institute of Pathology, Charité University Hospital, Berlin, Germany; German Cancer Consortium (DKTK), partner sites Heidelberg and Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Benedikt Brors
- German Cancer Consortium (DKTK), partner sites Heidelberg and Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Disease (NCT) Heidelberg, Heidelberg, Germany
| | - Stefan Gröschel
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hanno Glimm
- German Cancer Consortium (DKTK), partner sites Heidelberg and Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Schirmacher
- German Cancer Consortium (DKTK), partner sites Heidelberg and Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Marcus Renner
- Institute of Pathology, University Hospital Heidelberg , Heidelberg, Germany
| | - Stefan Fröhling
- German Cancer Consortium (DKTK), partner sites Heidelberg and Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Albrecht Stenzinger
- German Cancer Consortium (DKTK), partner sites Heidelberg and Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
38
|
Paoluzzi L, Cacavio A, Ghesani M, Karambelkar A, Rapkiewicz A, Weber J, Rosen G. Response to anti-PD1 therapy with nivolumab in metastatic sarcomas. Clin Sarcoma Res 2016; 6:24. [PMID: 28042471 PMCID: PMC5200964 DOI: 10.1186/s13569-016-0064-0] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/26/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Manipulation of immune checkpoints such as CTLA4 or PD-1 with targeted antibodies has recently emerged as an effective anticancer strategy in multiple malignancies. Sarcomas are a heterogeneous group of diseases in need of more effective treatments. Different subtypes of soft tissue and bone sarcomas have been shown to express PD-1 ligand. METHODS We retrospectively analyzed a cohort of patients (pts) with relapsed metastatic/unresectable sarcomas, who were treated with nivolumab provided under a patient assistance program from the manufacturer. Pts underwent CT or PET/CT imaging at baseline and after at least four doses of nivolumab; RECIST 1.1 criteria were used for response assessment. RESULTS Twenty-eight pts with soft tissue (STS, N = 24) or bone sarcoma (N = 4), received IV nivolumab 3 mg/kg every 2 weeks from July 2015. Median age was 57 (24-78), male:female ratio was 14:14; the median number of nivolumab cycles was eight. Eighteen pts concomitantly received pazopanib at 400-800 mg daily. The most common side effect was grade 1-2 LFT elevations; grade 3-4 toxicity occurred in five patients (colitis, LFT elevations, pneumonitis). Twenty-four pts received at least four cycles. We observed three partial responses: one dedifferentiated chondrosarcoma, one epithelioid sarcoma and one maxillary osteosarcoma (last two patients on pazopanib); nine patients had stable disease including three leiomyosarcomas; 12 patients had progression of disease including 4 leiomyosarcoma. Clinical benefit (response + stability) was observed in 50% of the evaluable patients. CONCLUSIONS These data provide a rationale for further exploring the efficacy of nivolumab and other checkpoint inhibitors in soft tissue and bone sarcoma.
Collapse
Affiliation(s)
- L Paoluzzi
- Department of Medicine, NYU Langone Medical Center, New York, NY USA
| | - A Cacavio
- Department of Medicine, NYU Langone Medical Center, New York, NY USA
| | - M Ghesani
- Department of Radiology, NYU Langone Medical Center, New York, NY USA
| | - A Karambelkar
- Department of Radiology, NYU Langone Medical Center, New York, NY USA
| | - A Rapkiewicz
- Department of Pathology, New York University School of Medicine, Laura and Isaac Perlmutter Cancer Center, 10th floor, Room 1041, 160 East 34th street, New York, NY USA
| | - J Weber
- Department of Medicine, NYU Langone Medical Center, New York, NY USA
| | - G Rosen
- Department of Medicine, NYU Langone Medical Center, New York, NY USA
| |
Collapse
|
39
|
Gatalica Z, Vranic S, Ghazalpour A, Xiu J, Ocal IT, McGill J, Bender RP, Discianno E, Schlum A, Sanati S, Palazzo J, Reddy S, Pockaj B. Multiplatform molecular profiling identifies potentially targetable biomarkers in malignant phyllodes tumors of the breast. Oncotarget 2016; 7:1707-16. [PMID: 26625196 PMCID: PMC4811491 DOI: 10.18632/oncotarget.6421] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 11/17/2015] [Indexed: 12/27/2022] Open
Abstract
Malignant phyllodes tumor is a rare breast malignancy with sarcomatous overgrowth and with limited effective treatment options for recurrent and metastatic cases. Recent clinical trials indicated a potential for anti-angiogenic, anti-EGFR and immunotherapeutic approaches for patients with sarcomas, which led us to investigate these and other targetable pathways in malignant phyllodes tumor of the breast. Thirty-six malignant phyllodes tumors (including 8 metastatic tumors with two cases having matched primary and metastatic tumors) were profiled using gene sequencing, gene copy number analysis, whole genome expression, and protein expression. Whole genome expression analysis demonstrated consistent over-expression of genes involved in angiogenesis including VEGFA, Angiopoietin-2, VCAM1, PDGFRA, and PTTG1. EGFR protein overexpression was observed in 26/27 (96%) of cases with amplification of the EGFR gene in 8/24 (33%) cases. Two EGFR mutations were identified including EGFRvIII and a presumed pathogenic V774M mutation, respectively. The most common pathogenic mutations included TP53 (50%) and PIK3CA (15%). Cases with matched primary and metastatic tumors harbored identical mutations in both sites (PIK3CA/KRAS and RB1 gene mutations, respectively). Tumor expression of PD-L1 immunoregulatory protein was observed in 3/22 (14%) of cases. Overexpression of molecular biomarkers of increased angiogenesis, EGFR and immune checkpoints provides novel targeted therapy options in malignant phyllodes tumors of the breast.
Collapse
Affiliation(s)
- Zoran Gatalica
- Caris Life Sciences, Phoenix, AZ, United States of America
| | - Semir Vranic
- Department of Pathology, University Clinical Center Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Joanne Xiu
- Caris Life Sciences, Phoenix, AZ, United States of America
| | | | - John McGill
- Miraca Life Sciences, Phoenix, AZ, United States of America
| | - Ryan P Bender
- Caris Life Sciences, Phoenix, AZ, United States of America
| | - Erin Discianno
- Caris Life Sciences, Phoenix, AZ, United States of America
| | - Aaron Schlum
- Caris Life Sciences, Phoenix, AZ, United States of America
| | - Souzan Sanati
- Division of Anatomic and Molecular Pathology, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Juan Palazzo
- Department of Pathology, Anatomy, and Cell Biology, Jefferson Medical College, Philadelphia, PA, United States of America
| | - Sandeep Reddy
- Caris Life Sciences, Phoenix, AZ, United States of America
| | | |
Collapse
|
40
|
Current Immunotherapies for Sarcoma: Clinical Trials and Rationale. Sarcoma 2016; 2016:9757219. [PMID: 27703409 PMCID: PMC5039267 DOI: 10.1155/2016/9757219] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/24/2016] [Indexed: 12/21/2022] Open
Abstract
Sarcoma tumors are rare and heterogeneous, yet they possess many characteristics that may facilitate immunotherapeutic responses. Both active strategies including vaccines and passive strategies involving cellular adoptive immunotherapy have been applied clinically. Results of these clinical trials indicate a distinct benefit for select patients. The recent breakthrough of immunologic checkpoint inhibition is being rapidly introduced to a variety of tumor types including sarcoma. It is anticipated that these emerging immunotherapies will exhibit clinical efficacy for a variety of sarcomas. The increasing ability to tailor immunologic therapies to sarcoma patients will undoubtedly generate further enthusiasm and clinical research for this treatment modality.
Collapse
|
41
|
Adriani G, Pavesi A, Tan AT, Bertoletti A, Thiery JP, Kamm RD. Microfluidic models for adoptive cell-mediated cancer immunotherapies. Drug Discov Today 2016; 21:1472-1478. [PMID: 27185084 PMCID: PMC5035566 DOI: 10.1016/j.drudis.2016.05.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/07/2016] [Accepted: 05/09/2016] [Indexed: 01/02/2023]
Abstract
Current adoptive T cell therapies have shown promising results in clinical trials but need further development as an effective cancer treatment. Here, we discuss how 3D microfluidic tumour models mimicking the tumour microenvironment could help in testing T cell immunotherapies by assessing engineered T cells and identifying combinatorial therapy to improve therapeutic efficacy. We propose that 3D microfluidic systems can be used to screen different patient-specific treatments, thereby reducing the burden of in vivo testing and facilitating the rapid translation of successful T cell cancer immunotherapies to the clinic.
Collapse
Affiliation(s)
- Giulia Adriani
- Singapore-MIT Alliance for Research and Technology, BioSyM IRG, 1 Create Way, 138602, Singapore
| | - Andrea Pavesi
- Singapore-MIT Alliance for Research and Technology, BioSyM IRG, 1 Create Way, 138602, Singapore
| | - Anthony T Tan
- DUKE-NUS Graduate Medical School Singapore, Emerging Infectious Disease Program, 8 College Road, 169857, Singapore
| | - Antonio Bertoletti
- DUKE-NUS Graduate Medical School Singapore, Emerging Infectious Disease Program, 8 College Road, 169857, Singapore
| | - Jean Paul Thiery
- National University of Singapore, Department of Biochemistry, Yong Loo Lin School of Medicine MD7, 8 Medical Drive, 117597, Singapore
| | - Roger D Kamm
- Singapore-MIT Alliance for Research and Technology, BioSyM IRG, 1 Create Way, 138602, Singapore; Massachusetts Institute of Technology, Department of Biological Engineering, 77 Massachusetts Avenue, 02139 Cambridge, MA, USA.
| |
Collapse
|
42
|
Inagaki Y, Hookway E, Williams KA, Hassan AB, Oppermann U, Tanaka Y, Soilleux E, Athanasou NA. Dendritic and mast cell involvement in the inflammatory response to primary malignant bone tumours. Clin Sarcoma Res 2016; 6:13. [PMID: 27482375 PMCID: PMC4968446 DOI: 10.1186/s13569-016-0053-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 07/13/2016] [Indexed: 12/11/2022] Open
Abstract
Background A chronic inflammatory cell infiltrate is commonly seen in response to primary malignant tumours of bone. This is known to contain tumour-associated macrophages (TAMs) and lymphocytes; dendritic cells (DCs) and mast cells (MCs) have also been identified but whether these and other inflammatory cells are seen commonly in specific types of bone sarcoma is uncertain. Methods In this study we determined the nature of the inflammatory cell infiltrate in 56 primary bone sarcomas. Immunohistochemistry using monoclonal antibodies was employed to assess semiquantitatively CD45+ leukocyte infiltration and the extent of the DC, MC, TAM and T and B lymphocyte infiltrate. Results The extent of the inflammatory infiltrate in individual sarcomas was very variable. A moderate or heavy leukocyte infiltrate was more commonly seen in conventional high-grade osteosarcoma, undifferentiated pleomorphic sarcoma and giant cell tumour of bone (GCTB) than in Ewing sarcoma, chordoma and chondrosarcoma. CD14+/CD68+ TAMs and CD3+ T lymphocytes were the major components of the inflammatory cell response but (DC-SIGN/CD11c+) DCs were also commonly noted when there was a significant TAM and T lymphocyte infiltrate. MCs were identified mainly at the periphery of sarcomas, including the osteolytic tumour-bone interface. Discussion Our findings indicate that, although variable, some malignant bone tumours (e.g. osteosarcoma, GCTB) are more commonly associated with a pronounced inflammatory cell infiltrate than others (e.g. chondrosarcoma. Ewing sarcoma); the infiltrate is composed mainly of TAMs but includes a significant DC, T lymphocyte and MC infiltrate. Conclusion Tumours that contain a heavy inflammatory cell response, which includes DCs, TAMs and T lymphocytes, may be more amenable to immunomodulatory therapy. MCs are present mainly at the tumour edge and are likely to contribute to osteolysis and tumour invasion.
Collapse
Affiliation(s)
- Y Inagaki
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal and Sciences, University of Oxford, Nuffield Orthopaedic Centre, Oxford, OX3 7HE UK ; Department of Orthopaedic Surgery, Nara Medical University, Kashihara, Japan
| | - E Hookway
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal and Sciences, University of Oxford, Nuffield Orthopaedic Centre, Oxford, OX3 7HE UK
| | - K A Williams
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal and Sciences, University of Oxford, Nuffield Orthopaedic Centre, Oxford, OX3 7HE UK
| | - A B Hassan
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal and Sciences, University of Oxford, Nuffield Orthopaedic Centre, Oxford, OX3 7HE UK
| | - U Oppermann
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal and Sciences, University of Oxford, Nuffield Orthopaedic Centre, Oxford, OX3 7HE UK
| | - Y Tanaka
- Department of Orthopaedic Surgery, Nara Medical University, Kashihara, Japan
| | - E Soilleux
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal and Sciences, University of Oxford, Nuffield Orthopaedic Centre, Oxford, OX3 7HE UK
| | - N A Athanasou
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal and Sciences, University of Oxford, Nuffield Orthopaedic Centre, Oxford, OX3 7HE UK
| |
Collapse
|
43
|
Tsukahara T, Emori M, Murata K, Mizushima E, Shibayama Y, Kubo T, Kanaseki T, Hirohashi Y, Yamashita T, Sato N, Torigoe T. The future of immunotherapy for sarcoma. Expert Opin Biol Ther 2016; 16:1049-57. [PMID: 27158940 DOI: 10.1080/14712598.2016.1188075] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION The use of immunotherapeutic challenges for sarcoma has a long history. Despite the existence of objective responses, immunotherapy has been overshadowed by the results of chemotherapy, especially for osteosarcoma. However, the prognosis for non-responders to chemotherapy is still poor and immunotherapy is now focused on again. AREAS COVERED We reviewed the following types of clinical trials of immunotherapy for sarcoma: (i) vaccination with autologous tumor cells, (ii) vaccination with peptides derived from tumor-associated antigens, (iii) adoptive cell transfer using engineered T cells expressing T cell receptor directed at NY-ESO-1 and (iv) immune checkpoint inhibitors targeting CTLA-4 and PD1/PDL1. EXPERT OPINION The immunogenicity of sarcoma might be lower than that of melanoma. Patients with small lesions who have not received any chemotherapy are good candidates for peptide-based immunotherapy. Combining peptide vaccination and immune checkpoint inhibitors is an attractive option, and long-lived memory T cells are attracting attention. Memory T stem cells defined by CD95+ are long-lived and have the capacity for self-renewal and multidifferentiation. We also identified a novel memory T cell population, young memory T cells defined by CD73+CXCR3+. Regulation of such memory T stem cells will be useful for peptide vaccination and adoptive cell transfer.
Collapse
Affiliation(s)
- Tomohide Tsukahara
- a Department of Pathology , Sapporo Medical University School of Medicine , Sapporo , Japan
| | - Makoto Emori
- b Department of Orthopaedic Surgery , Sapporo Medical University School of Medicine , Sapporo , Japan
| | - Kenji Murata
- a Department of Pathology , Sapporo Medical University School of Medicine , Sapporo , Japan.,b Department of Orthopaedic Surgery , Sapporo Medical University School of Medicine , Sapporo , Japan
| | - Emi Mizushima
- a Department of Pathology , Sapporo Medical University School of Medicine , Sapporo , Japan.,b Department of Orthopaedic Surgery , Sapporo Medical University School of Medicine , Sapporo , Japan
| | - Yuji Shibayama
- a Department of Pathology , Sapporo Medical University School of Medicine , Sapporo , Japan.,b Department of Orthopaedic Surgery , Sapporo Medical University School of Medicine , Sapporo , Japan
| | - Terufumi Kubo
- a Department of Pathology , Sapporo Medical University School of Medicine , Sapporo , Japan
| | - Takayuki Kanaseki
- a Department of Pathology , Sapporo Medical University School of Medicine , Sapporo , Japan
| | - Yoshihiko Hirohashi
- a Department of Pathology , Sapporo Medical University School of Medicine , Sapporo , Japan
| | - Toshihiko Yamashita
- b Department of Orthopaedic Surgery , Sapporo Medical University School of Medicine , Sapporo , Japan
| | - Noriyuki Sato
- a Department of Pathology , Sapporo Medical University School of Medicine , Sapporo , Japan
| | - Toshihiko Torigoe
- a Department of Pathology , Sapporo Medical University School of Medicine , Sapporo , Japan
| |
Collapse
|
44
|
Abstract
Sarcomas are rare malignant tumors affecting all age groups. They are typically classified according to their resemblance to corresponding normal tissue. Their heterogeneous features, for example, in terms of disease-driving genetic aberrations and body location, complicate both disease classification and development of novel treatment regimens. Many years of failure of improved patient outcome in clinical trials has led to the conclusion that novel targeted therapies are likely needed in combination with current multimodality regimens. Sarcomas have not, in contrast to the common carcinomas, been the subject of larger systematic studies on how tumor behavior relates to characteristics of the tumor microenvironment. There is consequently an urgent need for identifying suitable molecular targets, not only in tumor cells but also in the tumor microenvironment. This review discusses preclinical and clinical data about potential molecular targets in sarcomas. Studies on targeted therapies involving the tumor microenvironment are prioritized. A greater understanding of the biological context is expected to facilitate more successful design of future clinical trials in sarcoma.
Collapse
Affiliation(s)
- Monika Ehnman
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Karolinska University Hospital , Stockholm , Sweden
| | - Olle Larsson
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Karolinska University Hospital , Stockholm , Sweden
| |
Collapse
|
45
|
SINKOVICS JOSEPHG. The cnidarian origin of the proto-oncogenes NF-κB/STAT and WNT-like oncogenic pathway drives the ctenophores (Review). Int J Oncol 2015; 47:1211-29. [PMID: 26239915 PMCID: PMC4583530 DOI: 10.3892/ijo.2015.3102] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/26/2015] [Indexed: 01/09/2023] Open
Abstract
The cell survival pathways of the diploblastic early multicellular eukaryotic hosts contain and operate the molecular machinery resembling those of malignantly transformed individual cells of highly advanced multicellular hosts (including Homo). In the present review, the STAT/NF-κB pathway of the cnidarian Nematostella vectensis is compared with that of human tumors (malignant lymphomas, including Reed-Sternberg cells) pointing out similarities, including possible viral initiation in both cases. In the ctenophore genome and proteome, β-catenin gains intranuclear advantages due to a physiologically weak destructive complex in the cytoplasm, and lack of natural inhibitors (the dickkopfs). Thus, a scenario similar to what tumor cells initiate and achieve is presented through several constitutive loss-of-function type mutations in the destructive complex and in the elimination of inhibitors. Vice versa, malignantly transformed individual cells of advanced multicellular hosts assume pheno-genotypic resemblance to cells of unicellular or early multicellular hosts, and presumably to their ancient predecessors, by returning to the semblance of immortality and to the resumption of the state of high degree of resistance to physicochemical insults. Human leukemogenic and oncogenic pathways are presented for comparisons. The supreme bioengineers RNA/DNA complex encoded both the malignantly transformed immortal cell and the human cerebral cortex. The former generates molecules for the immortality of cellular life in the Universe. The latter invents the inhibitors of the process in order to gain control over it.
Collapse
Affiliation(s)
- JOSEPH G. SINKOVICS
- St. Joseph Hospital's Cancer Institute Affiliated with the H.L. Moffitt Comprehensive Cancer Center; Department of Molecular Medicine, The University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
46
|
Small bowel sarcoma: Tumor biology and advances in therapeutics. Surg Oncol 2015; 24:136-44. [DOI: 10.1016/j.suronc.2015.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 07/17/2015] [Accepted: 08/04/2015] [Indexed: 12/26/2022]
|