1
|
Robinson MJ, Davis EJ. Neoadjuvant Chemotherapy for Adults with Osteogenic Sarcoma. Curr Treat Options Oncol 2024:10.1007/s11864-024-01269-2. [PMID: 39417976 DOI: 10.1007/s11864-024-01269-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
OPINION STATEMENT Osteosarcoma is the most common primary malignant bone tumor in adolescents and adults. The 5-year survival rate is 65% when localized; however, survival drops dramatically to 10-20% in cases of metastatic disease. Therapy for osteosarcoma saw its first significant advancement in the 1970-80's, with the introduction of our current standard of care, consisting of the neo/adjuvant treatment regimen methotrexate, doxorubicin (Adriamycin), and cisplatin (collectively referred to as MAP) and surgical resection. Since MAP, development of a better therapeutic approach has stalled, creating a plateau in patient outcomes that has persisted for 40 years. Despite substantial research into a variety of pathways for novel treatment options, clinical trials have not produced sizeable improvements in outcomes. In this article, we discuss our current neoadjuvant standard of care therapy, followed by a review of contemporary therapeutic options, including tyrosine kinase inhibitors (TKIs), immune checkpoint inhibitors (ICIs), monoclonal antibodies (mAbs), and chimeric antigen receptor (CAR) T cells. Lastly, we consider the challenges hindering the success of novel treatment options and future research directions.
Collapse
Affiliation(s)
- Michael J Robinson
- Vanderbilt-Ingram Cancer Center, 2220 Pierce Ave, PRB 777, Nashville, TN, 37232, USA
| | - Elizabeth J Davis
- Vanderbilt-Ingram Cancer Center, 2220 Pierce Ave, PRB 777, Nashville, TN, 37232, USA.
| |
Collapse
|
2
|
Gaspar N, Hung GY, Strauss SJ, Campbell-Hewson Q, Dela Cruz FS, Glade Bender JL, Koh KN, Whittle SB, Chan GCF, Gerber NU, Palmu S, Morgenstern DA, Longhi A, Baecklund F, Lee JA, Locatelli F, Márquez Vega C, Janeway KA, McCowage G, McCabe MG, Bidadi B, Huang J, McKenzie J, Okpara CE, Bautista F. Lenvatinib Plus Ifosfamide and Etoposide in Children and Young Adults With Relapsed Osteosarcoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol 2024:2824985. [PMID: 39418029 DOI: 10.1001/jamaoncol.2024.4381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Importance The combination of ifosfamide and etoposide (IE) is commonly used to treat relapsed or refractory osteosarcoma; however, second-line treatment recommendations vary across guidelines. Objective To evaluate whether the addition of lenvatinib to IE (LEN-IE) improves outcomes in children and young adults with relapsed or refractory osteosarcoma. Design, Setting, and Participants The OLIE phase II, open-label, randomized clinical trial was conducted globally across Europe, Asia and the Pacific, and North America. From March 22, 2020, through November 11, 2021, the trial enrolled patients aged 2 to 25 years with high-grade osteosarcoma, measurable or evaluable disease per Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST 1.1), and 1 to 2 prior lines of systemic treatment. The data analyses were performed between March 22, 2020 (first patient in) and June 22, 2022 (data cutoff for the primary analysis), and September 29, 2023 (end of study final database lock). Interventions The OLIE trial assessed the efficacy and safety of lenvatinib (14 mg/m2 taken orally once daily) combined with up to 5 cycles of ifosfamide (3000 mg/m2 intravenously) and etoposide (100 mg/m2 intravenously) on days 1 to 3 of each cycle vs IE alone at the same doses. Patients randomized to IE could cross over to receive lenvatinib upon disease progression by independent imaging review. Main Outcomes and Measures The primary end point was progression-free survival (PFS) per RECIST 1.1 by independent imaging review. The Kaplan-Meier method was used to estimate the PFS distribution, with a prespecified 1-sided significance threshold of .025 by stratified log-rank test. Secondary end points included PFS rate at 4 months and overall survival. Adverse events were summarized using descriptive statistics. Results A total of 81 patients were enrolled (median [IQR] age, 15.0 [12.0-18.0] years; 46 males [56.8%]), with 40 in the LEN-IE arm and 41 in the IE arm. Median PFS was 6.5 months (95% CI, 5.7-8.2 months) for the LEN-IE arm and 5.5 months (95% CI, 2.9-6.5 months) for the IE arm (hazard ratio [HR], 0.54; 95% CI, 0.27-1.08; 1-sided P = .04). The rate of PFS at 4 months was 76.3% (95% CI, 59.3%-86.9%) in the LEN-IE arm and 66.0% (95% CI, 47.7%-79.2%) in the IE arm. Median overall survival was 11.9 months (95% CI, 10.1 months to not estimable) with LEN-IE and 17.4 months (95% CI, 14.2 months to not estimable) with IE (HR, 1.28; 95% CI, 0.60-2.70; 1-sided nominal P = .75). Grade 3 or higher treatment-related adverse events occurred in 35 of 39 patients (89.7%) in the LEN-IE arm and 31 of 39 patients (79.5%) in the IE arm. Conclusions and Relevance Although LEN-IE did not meet prespecified statistical significance for improved PFS vs IE, this study demonstrates the importance of international collaboration and randomized clinical trials in patients with relapsed or refractory osteosarcoma and may inform future trial design. Trial Registration ClinicalTrials.gov Identifier: NCT04154189.
Collapse
Affiliation(s)
- Nathalie Gaspar
- Department of Oncology for Child and Adolescent, Gustave Roussy Cancer Campus, Villejuif, France
| | - Giun-Yi Hung
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Sandra J Strauss
- London Sarcoma Service, University College London Hospital NHS Trust, London, United Kingdom
| | - Quentin Campbell-Hewson
- The Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, United Kingdom
| | - Filemon S Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Julia L Glade Bender
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kyung-Nam Koh
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sarah B Whittle
- Department of Pediatrics, Texas Children's Cancer Center, Baylor College of Medicine, Houston
| | - Godfrey Chi-Fung Chan
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Nicolas U Gerber
- Department of Oncology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Sauli Palmu
- Tampere Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University and University Hospital, Tampere, Finland
| | - Daniel A Morgenstern
- Division of Haematology/Oncology, Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Alessandra Longhi
- Chemotherapy Service, Istituto Ortopedico Rizzoli, Istituto di Ricovero e Cura a Carattere Scientifico, Bologna, Italy
| | - Fredrik Baecklund
- Paediatric Oncology Unit, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Jun Ah Lee
- Center for Pediatric Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Katherine A Janeway
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Geoffrey McCowage
- Cancer Centre for Children, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Martin G McCabe
- Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Behzad Bidadi
- Clinical Research, Merck & Co Inc, Rahway, New Jersey
| | - Jie Huang
- Biostatistics, Eisai Inc, Nutley, New Jersey
| | - Jodi McKenzie
- Oncology Business Group, Eisai Inc, Nutley, New Jersey
| | | | - Francisco Bautista
- Hospital del Niño Jesús, Madrid, Spain
- Now with Princess Maxima Centrum for Pediatric Cancer, Utrecht, the Netherlands
| |
Collapse
|
3
|
Young EP, Marinoff AE, Lopez-Fuentes E, Sweet-Cordero EA. Osteosarcoma through the Lens of Bone Development, Signaling, and Microenvironment. Cold Spring Harb Perspect Med 2024; 14:a041635. [PMID: 38565264 PMCID: PMC11444254 DOI: 10.1101/cshperspect.a041635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
In this work, we review the multifaceted connections between osteosarcoma (OS) biology and normal bone development. We summarize and critically analyze existing research, highlighting key areas that merit further exploration. The review addresses several topics in OS biology and their interplay with normal bone development processes, including OS cell of origin, genomics, tumor microenvironment, and metastasis. We examine the potential cellular origins of OS and how their roles in normal bone growth may contribute to OS pathogenesis. We survey the genomic landscape of OS, highlighting the developmental roles of genes frequently altered in OS. We then discuss the OS microenvironment, emphasizing the transformation of the bone niche in OS to facilitate tumor growth and metastasis. The role of stromal and immune cells is examined, including their impact on tumor progression and therapeutic response. We further provide insights into potential development-informed opportunities for novel therapeutic strategies.
Collapse
Affiliation(s)
- Elizabeth P Young
- Division of Pediatric Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, California 94158, USA
| | - Amanda E Marinoff
- Division of Pediatric Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, California 94158, USA
| | - Eunice Lopez-Fuentes
- Division of Pediatric Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, California 94158, USA
| | - E Alejandro Sweet-Cordero
- Division of Pediatric Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, California 94158, USA
| |
Collapse
|
4
|
Huang Y, Cao D, Zhang M, Yang Y, Niu G, Tang L, Shen Z, Zhang Z, Bai Y, Min D, He A. Exploring the impact of PDGFD in osteosarcoma metastasis through single-cell sequencing analysis. Cell Oncol (Dordr) 2024; 47:1715-1733. [PMID: 38652223 PMCID: PMC11467127 DOI: 10.1007/s13402-024-00949-3] [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] [Accepted: 04/08/2024] [Indexed: 04/25/2024] Open
Abstract
PURPOSE The overall survival rate for metastatic osteosarcoma hovers around 20%. Responses to second-line chemotherapy, targeted therapies, and immunotherapies have demonstrated limited efficacy in metastatic osteosarcoma. Our objective is to validate differentially expressed genes and signaling pathways between non-metastatic and metastatic osteosarcoma, employing single-cell RNA sequencing (scRNA-seq) and additional functional investigations. We aim to enhance comprehension of metastatic mechanisms and potentially unveil a therapeutic target. METHODS scRNA-seq was performed on two primary osteosarcoma lesions (1 non-metastatic and 1 metastatic). Seurat package facilitated dimensionality reduction and cluster identification. Copy number variation (CNV) was predicted using InferCNV. CellChat characterized ligand-receptor-based intercellular communication networks. Differentially expressed genes underwent GO function enrichment analysis and GSEA. Validation was achieved through the GSE152048 dataset, which identified PDGFD-PDGFRB as a common ligand-receptor pair with significant contribution. Immunohistochemistry assessed PDGFD and PDGFRB expression, while multicolor immunofluorescence and flow cytometry provided insight into spatial relationships and the tumor immune microenvironment. Kaplan-Meier survival analysis compared metastasis-free survival and overall survival between high and low levels of PDGFD and PDGFRB. Manipulation of PDGFD expression in primary osteosarcoma cells examined invasion abilities and related markers. RESULTS Ten clusters encompassing osteoblasts, osteoclasts, osteocytes, fibroblasts, pericytes, endothelial cells, myeloid cells, T cells, B cells, and proliferating cells were identified. Osteoblasts, osteoclasts, and osteocytes exhibited heightened CNV levels. Ligand-receptor-based communication networks exposed significant fibroblast crosstalk with other cell types, and the PDGF signaling pathway was activated in non-metastatic osteosarcoma primary lesion. These results were corroborated by the GSE152048 dataset, confirming the prominence of PDGFD-PDGFRB as a common ligand-receptor pair. Immunohistochemistry demonstrated considerably greater PDGFD expression in non-metastatic osteosarcoma tissues and organoids, correlating with extended metastasis-free and overall survival. PDGFRB expression showed no significant variation between non-metastatic and metastatic osteosarcoma, nor strong correlations with survival times. Multicolor immunofluorescence suggested co-localization of PDGFD with PDGFRB. Flow cytometry unveiled a highly immunosuppressive microenvironment in metastatic osteosarcoma. Manipulating PDGFD expression demonstrated altered invasive abilities and marker expressions in primary osteosarcoma cells from both non-metastatic and metastatic lesions. CONCLUSIONS scRNA-seq illuminated the activation of the PDGF signaling pathway in primary lesion of non-metastatic osteosarcoma. PDGFD displayed an inhibitory effect on osteosarcoma metastasis, likely through the suppression of the EMT signaling pathway.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Oncology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dongyan Cao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Biliary-Pancreatic Surgery, the Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Manxue Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Biliary-Pancreatic Surgery, the Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yue Yang
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu Province, China
| | | | - Lina Tang
- Department of Oncology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zan Shen
- Department of Oncology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhichang Zhang
- Department of Orthopaedic, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yueqing Bai
- Department of Pathology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Daliu Min
- Department of Oncology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Aina He
- Department of Oncology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Yang C, Li B, Dong S, Xu J, Sun X, Liang X, Liu K, Sun K, Yang Y, Ji T, Ye Z, Xie L, Tang X. Efficacy and Safety of Fruquintinib-Based Treatment in Patients with Refractory Bone and Soft Tissue Sarcoma after Developing Resistance to Several TKIs: A Multicenter Retrospective Study. Orthop Surg 2024; 16:2380-2390. [PMID: 39030807 PMCID: PMC11456709 DOI: 10.1111/os.14163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/17/2024] [Accepted: 06/22/2024] [Indexed: 07/22/2024] Open
Abstract
OBJECTIVE Multitargeted tyrosine kinase inhibitors (TKIs) have been approved as second-line therapy in refractory sarcoma, prolonging progression-free survival (PFS) but with short-lived duration of disease control. Fruquintinib is a TKI that specifically inhibits vascular endothelial growth factor receptor-1,2,3 with no metabolism by liver enzymes. In this retrospective study, we assessed the efficacy and safety of fruquintinib-based treatment in patients with refractory sarcoma after developing several lines of TKI resistance. METHODS We retrospectively analyzed the clinical data of patients with refractory sarcoma after they had developed several lines of resistance to TKIs and who received fruquintinib-based treatment from November 2021 to August 2023. The primary endpoint was the progression-free survival rate at 4 months (4m-PFSR). Secondary endpoints were the median PFS, overall survival (OS), objective response rate, disease control rate, and adverse effects (AEs). PFS and OS were estimated using the Kaplan-Meier method. A log-rank test was used to compare survival curves between different clinical and pathological factors. Cox proportional hazards analysis was performed to identify PFS-related prognostic factors. RESULTS We included 124 patients: 56 (45.2%) with osteosarcoma, 28 (22.6%) with Ewing sarcoma, seven (5.6%) with chondrosarcoma, and 33 (26.6%) with soft tissue sarcomas (STS). Only 18 (14.5%) patients received monotherapy with fruquintinib. With a median follow-up time of 6.8 (interquartile range [IQR], 4.6-9.4) months, 22 (17.7%) patients had partial response and 78 (62.9%) had stable disease. The 4m-PFSR was 58.4% (95% confidence interval [CI], 49.6%-67.1%). The median PFS and OS were 4.4 (95% CI, 3.9-5.0) months and 11.4 (95% CI, 10.3-12.5) months. In multivariate analysis, a high hazard ratio for progression was associated with target lesions located outside the lung and bone with 1.79 (95% CI, 1.10-2.93; p = 0.020). Eighty-eight AEs were recorded in 47 (37.9%) patients; the most common were pneumothorax (18/124, 14.5%), diarrhea (8/124, 6.5%), oral mucositis (7/124, 5.6%), and thrombocytopenia (7/124, 5.6%). CONCLUSIONS Fruquintinib may be a potential option for patients with refractory sarcoma after developing several lines of TKI resistance, with a satisfactory efficacy and safety profile in combination therapy. However, the degree of contribution of fruquintinib to results is unclear when combined with other effective substances. Additional prospective trials of fruquintinib should be conducted, especially involving different pathological types and combination regimens.
Collapse
Affiliation(s)
- Chenchen Yang
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
| | - Binghao Li
- Department of OrthopedicsThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Sen Dong
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
| | - Jie Xu
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
| | - Xin Sun
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
| | - Xin Liang
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
| | - Kuisheng Liu
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
| | - Kunkun Sun
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
| | - Yi Yang
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
| | - Tao Ji
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
| | - Zhaoming Ye
- Department of OrthopedicsThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Lu Xie
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
| | - Xiaodong Tang
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijingChina
| |
Collapse
|
6
|
Giordano G, Tucciarello C, Merlini A, Cutrupi S, Pignochino Y. Targeting the EphA2 pathway: could it be the way for bone sarcomas? Cell Commun Signal 2024; 22:433. [PMID: 39252029 PMCID: PMC11382444 DOI: 10.1186/s12964-024-01811-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
Bone sarcomas are malignant tumors of mesenchymal origin. Complete surgical resection is the cornerstone of multidisciplinary treatment. However, advanced, unresectable forms remain incurable. A crucial step towards addressing this challenge involves comprehending the molecular mechanisms underpinning tumor progression and metastasis, laying the groundwork for innovative precision medicine-based interventions. We previously showed that tyrosine kinase receptor Ephrin Type-A Receptor 2 (EphA2) is overexpressed in bone sarcomas. EphA2 is a key oncofetal protein implicated in metastasis, self-renewal, and chemoresistance. Molecular, genetic, biochemical, and pharmacological approaches have been developed to target EphA2 and its signaling pathway aiming to interfere with its tumor-promoting effects or as a carrier for drug delivery. This review synthesizes the main functions of EphA2 and their relevance in bone sarcomas, providing strategies devised to leverage this receptor for diagnostic and therapeutic purposes, with a focus on its applicability in the three most common bone sarcoma histotypes: osteosarcoma, chondrosarcoma, and Ewing sarcoma.
Collapse
Affiliation(s)
- Giorgia Giordano
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Oncology, University of Turin, 10043, Orbassano, TO, Italy
| | - Cristina Tucciarello
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy
| | - Alessandra Merlini
- Department of Oncology, University of Turin, 10043, Orbassano, TO, Italy
| | - Santina Cutrupi
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy
| | - Ymera Pignochino
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy.
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy.
| |
Collapse
|
7
|
Chen H, Wang W, Chang S, Huang X, Wang N. A useful mTORC1 signaling-related RiskScore model for the personalized treatment of osteosarcoma patients by using the bulk RNA-seq analysis. Discov Oncol 2024; 15:418. [PMID: 39251459 PMCID: PMC11383908 DOI: 10.1007/s12672-024-01301-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
AIMS This research developed a prognostic model for OS patients based on the Mechanistic Target of Rapamycin Complex 1 (mTORC1) signature. BACKGROUND The mTORC1 signaling pathway has a critical role in the maintenance of cellular homeostasis and tumorigenesis and development through the regulation of cell growth, metabolism and autophagy. However, the mechanism of action of this signaling pathway in Osteosarcoma (OS) remains unclear. OBJECTIVE The datasets including the TARGET-OS and GSE39058, and 200 mTORC1 genes were collected. METHODS The mTORC1 signaling-related genes were obtained based on the Molecular Signatures Database (MSigDB) database, and the single sample gene set enrichment analysis (ssGSEA) algorithm was utilized in order to calculate the mTORC1 score. Then, the WGCNA were performed for the mTORC1-correlated gene module, the un/multivariate and lasso Cox regression analysis were conducted for the RiskScore model. The immune infiltration analysis was performed by using the ssGSEA method, ESTIMATE tool and MCP-Count algorithm. KM survival and Receiver Operating Characteristic (ROC) Curve analysis were performed by using the survival and timeROC package. RESULTS The mTORC1 score and WGCNA with β = 5 screened the mTORC1 positively correlated skyblue2 module that included 67 genes, which are also associated with the metabolism and hypoxia pathways. Further narrowing of candidate genes and calculating the regression coefficient, we developed a useful and reliable RiskScore model, which can classify the patients in the training and validation set into high and low-risk groups based on the median value of RiskScore as an independent and robust prognostic factor. High-risk patients had a significantly poor prognosis, lower immune infiltration level of multiple immune cells and prone to cancer metastasis. Finally, we a nomogram model incorporating the metastasis features and RiskScore showed excellent prediction accuracy and clinical practicability. CONCLUSION We developed a useful and reliable risk prognosis model based on the mTORC1 signaling signature.
Collapse
Affiliation(s)
- Hongxia Chen
- Department of Hematology, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Wei Wang
- Department of Oncology, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Shichuan Chang
- Department of Oncology, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Xiaoping Huang
- Department of Oncology, Chongqing University Three Gorges Hospital, Chongqing, 404000, China.
| | - Ning Wang
- Department of Oncology, Chongqing University Three Gorges Hospital, Chongqing, 404000, China.
| |
Collapse
|
8
|
Reed DR, Tulpule A, Metts J, Trucco M, Robertson-Tessi M, O'Donohue TJ, Iglesias-Cardenas F, Isakoff MS, Mauguen A, Shukla N, Dela Cruz FS, Tap W, Kentsis A, Morris CD, Hameed M, Honeyman JN, Behr GG, Sulis ML, Ortiz MV, Slotkin E. Pediatric Leukemia Roadmaps Are a Guide for Positive Metastatic Bone Sarcoma Trials. J Clin Oncol 2024; 42:2955-2960. [PMID: 38843482 DOI: 10.1200/jco.23.02717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/02/2024] [Accepted: 04/11/2024] [Indexed: 08/30/2024] Open
Abstract
ALL cures require many MRD therapies. This strategy should drive experiments and trials in metastatic bone sarcomas.
Collapse
Affiliation(s)
- Damon R Reed
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Asmin Tulpule
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jonathan Metts
- Johns Hopkins All Children's Hospital, St Petersburg, FL
| | | | | | - Tara J O'Donohue
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Filemon S Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - William Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alex Kentsis
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Carol D Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Meera Hameed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Joshua N Honeyman
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gerald G Behr
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maria Luisa Sulis
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael V Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Emily Slotkin
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
9
|
Yan P, Wang J, Yue B, Wang X. Unraveling molecular aberrations and pioneering therapeutic strategies in osteosarcoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189171. [PMID: 39127243 DOI: 10.1016/j.bbcan.2024.189171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Osteosarcoma, a rare primary bone cancer, presents diverse molecular aberrations that underscore its complexity. Despite the persistent endeavors by researchers, the limited amelioration in the five-year survival rate indicates that current therapeutic strategies prove inadequate in addressing the clinical necessities. Advancements in molecular profiling have facilitated an enhanced comprehension of the biology of osteosarcoma, offering a promising outlook for treatment. There is an urgent need to develop innovative approaches to address the complex challenges of osteosarcoma, ultimately contributing to enhanced patient outcomes. This review explores the nexus between osteosarcoma and cancer predisposition syndromes, intricacies in its somatic genome, and clinically actionable alterations. This review covers treatment strategies, including surgery, chemotherapy, immune checkpoint inhibitors (ICIs), and tyrosine kinase inhibitors (TKIs). Innovative treatment modalities targeting diverse pathways, including multi-target tyrosine kinases, cell cycle, PI3K/mTOR pathway, and DNA damage repair (DDR), offer promising interventions. This review also covers promising avenues, including antibody-drug conjugates (ADCs) and immunotherapy strategies, such as cytokines, adoptive cellular therapy (ACT), ICIs, and cancer vaccines. This comprehensive exploration contributes to a holistic understanding, offering guidance for clinical applications to advance the management of osteosarcoma.
Collapse
Affiliation(s)
- Peng Yan
- Department of Orthopedic Oncology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, China
| | - Jie Wang
- Department of Orthopedic Oncology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, China
| | - Bin Yue
- Department of Orthopedic Oncology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, China.
| | - Xinyi Wang
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, China.
| |
Collapse
|
10
|
Zhang Q, Xia Y, Wang L, Wang Y, Bao Y, Zhao GS. Targeted anti-angiogenesis therapy for advanced osteosarcoma. Front Oncol 2024; 14:1413213. [PMID: 39252946 PMCID: PMC11381227 DOI: 10.3389/fonc.2024.1413213] [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: 04/06/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
To date, despite extensive research, the prognosis of advanced osteosarcoma has not improved significantly. Thus, patients experience a reduced survival rate, suggesting that a reevaluation of current treatment strategies is required. Recently, in addition to routine surgery, chemotherapy and radiotherapy, researchers have explored more effective and safer treatments, including targeted therapy, immunotherapy, anti-angiogenesis therapy, metabolic targets therapy, and nanomedicine therapy. The tumorigenesis and development of osteosarcoma is closely related to angiogenesis. Thus, anti-angiogenesis therapy is crucial to treat osteosarcoma; however, recent clinical trials found that it has insufficient efficacy. To solve this problem, the causes of treatment failure and improve treatment strategies should be investigated. This review focuses on summarizing the pathophysiological mechanisms of angiogenesis in osteosarcoma and recent advances in anti-angiogenesis treatment of osteosarcoma. We also discuss some clinical studies, with the aim of providing new ideas to improve treatment strategies for osteosarcoma and the prognosis of patients.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Pain and Rehabilitation, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuxuan Xia
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - LiYuan Wang
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Wang
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yixi Bao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guo-Sheng Zhao
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Green D, van Ewijk R, Tirtei E, Andreou D, Baecklund F, Baumhoer D, Bielack SS, Botchu R, Boye K, Brennan B, Capra M, Cottone L, Dirksen U, Fagioli F, Fernandez N, Flanagan AM, Gambarotti M, Gaspar N, Gelderblom H, Gerrand C, Gomez-Mascard A, Hardes J, Hecker-Nolting S, Kabickova E, Kager L, Kanerva J, Kester LA, Kuijjer ML, Laurence V, Lervat C, Marchais A, Marec-Berard P, Mendes C, Merks JH, Ory B, Palmerini E, Pantziarka P, Papakonstantinou E, Piperno-Neumann S, Raciborska A, Roundhill EA, Rutkauskaite V, Safwat A, Scotlandi K, Staals EL, Strauss SJ, Surdez D, Sys GM, Tabone MD, Toulmonde M, Valverde C, van de Sande MA, Wörtler K, Campbell-Hewson Q, McCabe MG, Nathrath M. Biological Sample Collection to Advance Research and Treatment: A Fight Osteosarcoma Through European Research and Euro Ewing Consortium Statement. Clin Cancer Res 2024; 30:3395-3406. [PMID: 38869831 PMCID: PMC11334773 DOI: 10.1158/1078-0432.ccr-24-0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/27/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024]
Abstract
Osteosarcoma and Ewing sarcoma are bone tumors mostly diagnosed in children, adolescents, and young adults. Despite multimodal therapy, morbidity is high and survival rates remain low, especially in the metastatic disease setting. Trials investigating targeted therapies and immunotherapies have not been groundbreaking. Better understanding of biological subgroups, the role of the tumor immune microenvironment, factors that promote metastasis, and clinical biomarkers of prognosis and drug response are required to make progress. A prerequisite to achieve desired success is a thorough, systematic, and clinically linked biological analysis of patient samples, but disease rarity and tissue processing challenges such as logistics and infrastructure have contributed to a lack of relevant samples for clinical care and research. There is a need for a Europe-wide framework to be implemented for the adequate and minimal sampling, processing, storage, and analysis of patient samples. Two international panels of scientists, clinicians, and patient and parent advocates have formed the Fight Osteosarcoma Through European Research consortium and the Euro Ewing Consortium. The consortia shared their expertise and institutional practices to formulate new guidelines. We report new reference standards for adequate and minimally required sampling (time points, diagnostic samples, and liquid biopsy tubes), handling, and biobanking to enable advanced biological studies in bone sarcoma. We describe standards for analysis and annotation to drive collaboration and data harmonization with practical, legal, and ethical considerations. This position paper provides comprehensive guidelines that should become the new standards of care that will accelerate scientific progress, promote collaboration, and improve outcomes.
Collapse
Affiliation(s)
- Darrell Green
- Biomedical Research Centre, Norwich Medical School, University of East Anglia, Norwich, United Kingdom.
| | - Roelof van Ewijk
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| | - Elisa Tirtei
- Pediatric Oncology, Regina Margherita Children’s Hospital, Turin, Italy.
- Department of Public Health and Pediatrics, University of Turin, Turin, Italy.
| | - Dimosthenis Andreou
- Department of Orthopaedics and Trauma, Medical University of Graz, Graz, Austria.
| | - Fredrik Baecklund
- Pediatric Oncology Unit, Karolinska University Hospital, Stockholm, Sweden.
| | - Daniel Baumhoer
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland.
| | - Stefan S. Bielack
- Center for Pediatric, Adolescent and Women’s Medicine, Klinikum Stuttgart—Olgahospital, Stuttgart Cancer Centre, Stuttgart, Germany.
| | - Rajesh Botchu
- Department of Musculoskeletal Radiology, Royal Orthopaedic Hospital NHS Foundation Trust, Birmingham, United Kingdom.
| | - Kjetil Boye
- Department of Oncology, Oslo University Hospital, Oslo, Norway.
| | - Bernadette Brennan
- Paediatric Oncology, Royal Manchester Children’s Hospital, Central Manchester University Hospital NHS Foundation Trust, Manchester, United Kingdom.
| | - Michael Capra
- Haematology/Oncology, Children’s Health Ireland at Crumlin, Dublin, Ireland.
| | - Lucia Cottone
- Department of Pathology, UCL Cancer Institute, University College London, London, United Kingdom.
| | - Uta Dirksen
- Pediatrics III, West German Cancer Center, University Hospital Essen, German Cancer Consortium (DKTK) Site Essen, Cancer Research Center (NCT) Cologne-Essen, University of Duisburg-Essen, Essen, Germany.
| | - Franca Fagioli
- Pediatric Oncology, Regina Margherita Children’s Hospital, Turin, Italy.
- Department of Public Health and Pediatrics, University of Turin, Turin, Italy.
| | - Natalia Fernandez
- Patient and Parent Advocacy Group, FOSTER, Washington, District of Columbia.
| | - Adrienne M. Flanagan
- Department of Pathology, UCL Cancer Institute, University College London, London, United Kingdom.
- Histopathology, The Royal National Orthopaedic Hospital NHS Trust, Stanmore, United Kingdom.
| | - Marco Gambarotti
- Department of Pathology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | - Nathalie Gaspar
- Department of Oncology for Child and Adolescent, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.
- U1015, Université Paris-Saclay, Villejuif, France.
| | - Hans Gelderblom
- Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Craig Gerrand
- Orthopaedic Oncology, The Royal National Orthopaedic Hospital NHS Trust, Stanmore, United Kingdom.
| | - Anne Gomez-Mascard
- Department of Pathology, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France.
- EQ ONCOSARC, CRCT Inserm/UT3, ERL CNRS, Toulouse, France.
| | - Jendrik Hardes
- Tumour Orthopaedics, University Hospital Essen, German Cancer Consortium (DKTK) Site Essen, Cancer Research Center (NCT) Cologne-Essen, University of Duisburg-Essen, Essen, Germany.
| | - Stefanie Hecker-Nolting
- Center for Pediatric, Adolescent and Women’s Medicine, Klinikum Stuttgart—Olgahospital, Stuttgart Cancer Centre, Stuttgart, Germany.
| | - Edita Kabickova
- Paediatric Haematology and Oncology, University Hospital Motol, Prague, Czech Republic.
| | - Leo Kager
- Pediatrics, St Anna Children’s Hospital, Medical University Vienna, Vienna, Austria.
- St Anna Children’s Cancer Research Institute, Vienna, Austria.
| | - Jukka Kanerva
- Hematology-Oncology and Stem Cell Transplantation, HUS Helsinki University Hospital, New Children’s Hospital, Helsinki, Finland.
| | - Lennart A. Kester
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| | - Marieke L. Kuijjer
- Computational Biology and Systems Medicine Group, Centre for Molecular Medicine Norway, University of Oslo, Oslo, Norway.
- Pathology, Leiden University Medical Center, Leiden, the Netherlands.
- Leiden Center for Computational Oncology, Leiden University Medical Center, Leiden, the Netherlands.
| | | | - Cyril Lervat
- Department of Pediatrics and AYA Oncology, Centre Oscar Lambret, Lille, France.
| | - Antonin Marchais
- Department of Oncology for Child and Adolescent, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.
| | - Perrine Marec-Berard
- Institute of Hematology and Pediatric Oncology, Léon Bérard Center, Lyon, France.
| | - Cristina Mendes
- Portuguese Institute of Oncology of Lisbon, Lisbon, Portugal.
| | - Johannes H.M. Merks
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands.
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Benjamin Ory
- School of Medicine, Nantes Université, Nantes, France.
| | - Emanuela Palmerini
- Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Orthopedico Rizzoli, Bologna, Italy.
| | - Pan Pantziarka
- Patient and Parent Advocacy Group, FOSTER, Washington, District of Columbia.
- Anticancer Fund, Meise, Belgium.
- The George Pantziarka TP53 Trust, London, United Kingdom.
| | - Evgenia Papakonstantinou
- Pediatric Hematology-Oncology, Ippokratio General Hospital of Thessaloniki, Thessaloniki, Greece.
| | | | - Anna Raciborska
- Oncology and Surgical Oncology for Children and Youth, Institute of Mother and Child, Warsaw, Poland.
| | - Elizabeth A. Roundhill
- Children’s Cancer Research Group, Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom.
| | - Vilma Rutkauskaite
- Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania.
| | - Akmal Safwat
- The Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark.
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | - Eric L. Staals
- Orthopaedics and Trauma, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | - Sandra J. Strauss
- Department of Oncology, University College London Hospitals NHS Foundation Trust, UCL Cancer Institute, London, United Kingdom.
| | - Didier Surdez
- Balgrist University Hospital, Faculty of Medicine, University of Zurich (UZH), Zurich, Switzerland.
| | - Gwen M.L. Sys
- Department of Orthopaedic Surgery and Traumatology, Ghent University Hospital, Belgium.
| | - Marie-Dominique Tabone
- Department of Hematology and Oncology, A. Trousseau Hospital, Sorbonne University, APHP, Paris, France.
| | - Maud Toulmonde
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France.
| | - Claudia Valverde
- Medical Oncology, Vall d’Hebron University Hospital, Barcelona, Spain.
| | | | - Klaus Wörtler
- Musculoskeletal Radiology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.
| | - Quentin Campbell-Hewson
- Great North Children’s Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.
| | - Martin G. McCabe
- Division of Cancer Sciences, School of Medical Sciences, The University of Manchester, Manchester, United Kingdom.
- The Christie NHS Foundation Trust, Manchester, United Kingdom.
| | - Michaela Nathrath
- Children’s Cancer Research Center, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.
- Pediatric Oncology, Klinikum Kassel, Kassel, Germany.
| |
Collapse
|
12
|
Pearson AD, DuBois SG, Macy ME, de Rojas T, Donoghue M, Weiner S, Knoderer H, Bernardi R, Buenger V, Canaud G, Cantley L, Chung J, Fox E, Friend J, Glade-Bender J, Gorbatchevsky I, Gore L, Gupta A, Hawkins DS, Juric D, Lang LA, Leach D, Liaw D, Lesa G, Ligas F, Lindberg G, Lindberg W, Ludwinski D, Marshall L, Mazar A, McDonough J, Nysom K, Ours C, Pappo A, Parsons DW, Rosenfeld A, Scobie N, Smith M, Taylor D, Weigel B, Weinstein A, Karres D, Vassal G. Paediatric strategy forum for medicinal product development of PI3-K, mTOR, AKT and GSK3β inhibitors in children and adolescents with cancer. Eur J Cancer 2024; 207:114145. [PMID: 38936103 DOI: 10.1016/j.ejca.2024.114145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 05/16/2024] [Accepted: 05/27/2024] [Indexed: 06/29/2024]
Abstract
Phosphatidylinositol 3-kinase (PI3-K) signalling pathway is a crucial path in cancer for cell survival and thus represents an intriguing target for new paediatric anti-cancer drugs. However, the unique clinical toxicities of targeting this pathway (resulting in hyperglycaemia) difficulties combining with chemotherapy, rarity of mutations in childhood tumours and concomitant mutations have resulted in major barriers to clinical translation of these inhibitors in treating both adults and children. Mutations in PIK3CA predict response to PI3-K inhibitors in adult cancers. The same mutations occur in children as in adults, but they are significantly less frequent in paediatrics. In children, high-grade gliomas, especially diffuse midline gliomas (DMG), have the highest incidence of PIK3CA mutations. New mutation-specific PI3-K inhibitors reduce toxicity from on-target PI3-Kα wild-type activity. The mTOR inhibitor everolimus is approved for subependymal giant cell astrocytomas. In paediatric cancers, mTOR inhibitors have been predominantly evaluated by academia, without an overall strategy, in empiric, mutation-agnostic clinical trials with very low response rates to monotherapy. Therefore, future trials of single agent or combination strategies of mTOR inhibitors in childhood cancer should be supported by very strong biological rationale and preclinical data. Further preclinical evaluation of glycogen synthase kinase-3 beta inhibitors is required. Similarly, even where there is an AKT mutation (∼0.1 %), the role of AKT inhibitors in paediatric cancers remains unclear. Patient advocates strongly urged analysing and conserving data from every child participating in a clinical trial. A priority is to evaluate mutation-specific, central nervous system-penetrant PI3-K inhibitors in children with DMG in a rational biological combination. The choice of combination, should be based on the genomic landscape e.g. PTEN loss and resistance mechanisms supported by preclinical data. However, in view of the very rare populations involved, innovative regulatory approaches are needed to generate data for an indication.
Collapse
Affiliation(s)
| | - Steven G DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, USA
| | | | | | | | | | | | - Ronald Bernardi
- Genentech, A Member of the Roche Group, South San Francisco, CA USA
| | - Vickie Buenger
- Coalition Against Childhood Cancer (CAC2), Philadelphia, USA
| | | | | | - John Chung
- Bayer Healthcare Pharmaceuticals, Whippany, NJ, USA
| | | | | | | | | | | | - Abha Gupta
- The Hospital for Sick Children (SickKids), Princess Margaret Hospital Toronto, Canada
| | | | | | - Leigh Anna Lang
- Rally Foundation for Childhood Cancer Research, Atlanta, GA, USA
| | | | | | - Giovanni Lesa
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Division, European Medicines Agency (EMA), the Netherlands
| | - Franca Ligas
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Division, European Medicines Agency (EMA), the Netherlands
| | | | | | | | - Lynley Marshall
- The Royal Marsden Hospital, London, UK; The Institute of Cancer Research, London, UK
| | | | - Joe McDonough
- The Andrew McDonough B+ Foundation, Wilmington, DE, USA
| | | | - Christopher Ours
- National Human Genome Research Institute/National Institutes of Health, MD, USA
| | | | | | | | | | | | | | | | - Amy Weinstein
- Pediatric Brain Tumor Foundation of the US, Atlanta, USA
| | - Dominik Karres
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Division, European Medicines Agency (EMA), the Netherlands
| | - Gilles Vassal
- ACCELERATE, Europe, Belgium; Gustave Roussy Cancer Centre, Paris, France
| |
Collapse
|
13
|
Gao M, Liu W, Li T, Song Z, Wang X, Zhang X. Identifying Genetic Signatures Associated with Oncogene-Induced Replication Stress in Osteosarcoma and Screening for Potential Targeted Drugs. Biochem Genet 2024; 62:1690-1715. [PMID: 37672187 DOI: 10.1007/s10528-023-10497-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 08/07/2023] [Indexed: 09/07/2023]
Abstract
Osteosarcoma is the most common type of primary malignant bone tumor. Due to the lack of selectivity and sensitivity of chemotherapy drugs to tumor cells, coupled with the use of large doses, chemotherapy drugs often have systemic toxicity. The use of modern sequencing technology to screen tumor markers in a large number of tumor samples is a common method for screening highly specific and selective anti-tumor drugs. This study aims to identify potential biomarkers using the latest reported gene expression signatures of oncogene-induced replication stress (ORS) in aggressive cancers, and potential anti-osteosarcoma drugs were screened in different drug databases. In this study, we obtained 89 osteosarcoma-related samples in the TARGET database, all of which included survival information. According to the median expression of each of six reported ORS gene markers (NAT10/DDX27/ZNF48/C8ORF33/MOCS3/MPP6), we divided 89 osteosarcoma gene expression datasets into a high expression group and a low expression group and then performed a differentially expressed gene (DEG) analysis. The coexisting genes of 6 groups of DEGs were used as replication stress-related genes (RSGs) of osteosarcoma. Then, key RSGs were screened using LASSO regression, a Cox risk proportional regression prognostic model and a tenfold cross-validation test. GSE21257 datasets collected from the Gene Expression Omnibus (GEO) database were used to verify the prognostic model. The final key RSGs selected were used in the L1000PWD and DGIdb databases to mine potential drugs. After further validation by the prognostic model, we identified seven genes associated with ORS in osteosarcoma as key RSGs, including transcription factor 7 like 2 (TCF7L2), solute carrier family 27 member 4 (SLC27A4), proprotein convertase subtilisin/kexin type 5 (PCSK5), nucleolar protein 6 (NOL6), coiled-coil-coil-coil-coil-helix domain containing 4 (CHCHD4), eukaryotic translation initiation factor 3 subunit B (EIF3B), and synthesis of cytochrome C oxidase 1 (SCO1). Then, we screened the seven key RSGs in two drug databases and found six potential anti-osteosarcoma drugs (D GIdb database: repaglinide, tacrolimus, sirolimus, cyclosporine, and hydrochlorothiazide; L1000PWD database: the small molecule VU-0365117-1). Seven RSGs (TCF7L2, SLC27A4, PCSK5, NOL6, CHCHD4, EIF3B, and SCO1) may be associated with the ORS gene signatures in osteosarcoma. Repaglinide, tacrolimus, sirolimus, cyclosporine, hydrochlorothiazide and the small molecule VU-0365117-1 are potential therapeutic drugs for osteosarcoma.
Collapse
Affiliation(s)
- Meng Gao
- School of Medicine, Nankai University, Tianjin, China
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Haidian District, 51 Fucheng Road, Beijing, 100048, China
| | - Weibo Liu
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Haidian District, 51 Fucheng Road, Beijing, 100048, China
| | - Teng Li
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Haidian District, 51 Fucheng Road, Beijing, 100048, China
| | - ZeLong Song
- School of Medicine, Nankai University, Tianjin, China
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Haidian District, 51 Fucheng Road, Beijing, 100048, China
| | - XiangYu Wang
- Department of Pain Medicine, First Medical Center, PLA General Hospital, Beijing, 100000, China.
| | - XueSong Zhang
- School of Medicine, Nankai University, Tianjin, China.
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Haidian District, 51 Fucheng Road, Beijing, 100048, China.
| |
Collapse
|
14
|
Dogan I, Paksoy N, Basaran M. Efficacy and safety of sorafenib in adult metastatic osteosarcoma patients. J Cancer Res Ther 2024; 20:979-983. [PMID: 39023606 DOI: 10.4103/jcrt.jcrt_2581_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/04/2023] [Indexed: 07/20/2024]
Abstract
BACKGROUND There are limited data on the efficacy of targeted therapy in metastatic osteosarcoma. The goal of this study was to assess the effectiveness of sorafenib in adult patients with heavily pretreated metastatic osteosarcoma. METHOD Patients with metastatic osteosarcoma aged more than 18 years were assessed retrospectively. The patients' clinical, pathological, and therapeutic data were collected. For survival analysis, Kaplan-Meier models were used. RESULTS The research involved 15 patients. The ratio of male and female patients was 2/1, with a median age of 25 years (range: 19-64 years). The most common primary tumor localization was the extremities (66.6%). Fourteen (93.3%) patients had previously received palliative chemotherapy and six (40%) patients had palliative radiotherapy. The median progression-free survival was found as 5.5 months (95% confidence interval, 1.3-9.7). A stable response was observed in seven (46.6%) patients and progressive disease in eight (53.4%) patients. Grade 1-2 toxicities were detected in 50% of the patients, while grade 3-4 toxicities were observed in 14.3% of the patients. CONCLUSIONS We demonstrated real-life results of sorafenib for disease management in pretreated adult patients with metastatic osteosarcoma in the study. Sorafenib was effective for disease control and well tolerated in the patients. Sorafenib may be a treatment option for disease control after the disease progresses with chemotherapy in patients with metastatic osteosarcoma.
Collapse
Affiliation(s)
- Izzet Dogan
- Medical Oncology, Istanbul University Institute of Oncology, Istanbul, Turkey
| | | | | |
Collapse
|
15
|
Mettmann VL, Blattmann C, Friedel G, Harrabi S, von Kalle T, Kager L, Kevric M, Kühne T, Nathrath M, Sorg B, Werner M, Bielack SS, Hecker-Nolting S. Primary Multi-Systemic Metastases in Osteosarcoma: Presentation, Treatment, and Survival of 83 Patients of the Cooperative Osteosarcoma Study Group. Cancers (Basel) 2024; 16:275. [PMID: 38254767 PMCID: PMC10813782 DOI: 10.3390/cancers16020275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/02/2024] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND To evaluate patient and tumour characteristics, treatment, and their impact on survival in patients with multi-systemic metastases at initial diagnosis of high-grade osteosarcoma. Precedure: Eighty-three consecutive patients who presented with multi-systemic metastases at initial diagnosis of high-grade osteosarcoma were retrospectively reviewed. In cases of curative intent, the Cooperative Osteosarcoma Study Group recommended surgical removal of all detectable metastases in addition to complete resection of the primary tumour and chemotherapy. RESULTS Eighty-three eligible patients (1.8%) were identified among a total of 4605 individuals with high-grade osteosarcoma. Nine (10.8%) of these achieved complete surgical remission, of whom seven later had recurrences. The median follow-up time was 12 (range, 1-165) months for all patients. Actuarial event-free survival after 1, 2, and 5 years was 9.6 ± 3.2%, 1.4 ± 1.4%, and 1.4 ± 1.4%, and overall survival was 54.0 ± 5.6%, 23.2 ± 4.9%, and 8.7 ± 3.3%. In univariate analyses, elevated alkaline phosphatase before chemotherapy, pleural effusion, distant bones as metastatic sites, and more than one bone metastasis were negative prognostic factors. Among treatment-related factors, the microscopically complete resection of the primary tumour, a good response to first-line chemotherapy, the macroscopically complete resection of all affected tumour sites, and local treatment (surgery ± radiotherapy) of all bone metastases were associated with better outcomes. Tumour progression under first-line treatment significantly correlated with shorter survival times. CONCLUSION The outlook for patients with multi-systemic primary metastases from osteosarcoma remains very poor. The utmost importance of surgical resection of all tumour sites was confirmed. For unresectable bone metastases, radiotherapy might be considered. In the patient group studied, standard chemotherapy was often insufficiently effective. In the case of such advanced disease, alternative treatment options are urgently required.
Collapse
Affiliation(s)
- Vanessa L. Mettmann
- Cooperative Osteosarcoma Study Group, Paediatrics 5 (Oncology, Haematology, Immunology), Centre for Paediatric, Adolescent and Women’s Medicine, and Stuttgart Cancer Centre, Klinikum Stuttgart–Olgahospital, 70174 Stuttgart, Germany
| | - Claudia Blattmann
- Cooperative Osteosarcoma Study Group, Paediatrics 5 (Oncology, Haematology, Immunology), Centre for Paediatric, Adolescent and Women’s Medicine, and Stuttgart Cancer Centre, Klinikum Stuttgart–Olgahospital, 70174 Stuttgart, Germany
| | - Godehard Friedel
- Department of Thoracic Surgery, Faculty of Science, University of Tubingen, 72076 Tubingen, Germany
| | - Semi Harrabi
- Heidelberg Ion Beam Therapy Centre (HIT), Department of Radiation Oncology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Thekla von Kalle
- Radiologic Institute, Centre for Paediatric, Adolescent and Women’s Medicine, Stuttgart Cancer Centre, Klinikum Stuttgart–Olgahospital, 70174 Stuttgart, Germany
| | - Leo Kager
- St. Anna Children’s Hospital, University Hospital for Paediatric and Adolescent Medicine of the Medical University and St. Anna Children’s Cancer Research Institute (CCRI), 1090 Vienna, Austria
| | - Matthias Kevric
- Cooperative Osteosarcoma Study Group, Paediatrics 5 (Oncology, Haematology, Immunology), Centre for Paediatric, Adolescent and Women’s Medicine, and Stuttgart Cancer Centre, Klinikum Stuttgart–Olgahospital, 70174 Stuttgart, Germany
| | - Thomas Kühne
- University Children’s Hospital Basel, 4031 Basel, Switzerland
| | - Michaela Nathrath
- Department of Paediatrics and Children’s Cancer Research Centre, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Paediatric Haematology and Oncology, Klinikum Kassel, 34125 Kassel, Germany
| | - Benjamin Sorg
- Cooperative Osteosarcoma Study Group, Paediatrics 5 (Oncology, Haematology, Immunology), Centre for Paediatric, Adolescent and Women’s Medicine, and Stuttgart Cancer Centre, Klinikum Stuttgart–Olgahospital, 70174 Stuttgart, Germany
| | - Mathias Werner
- Osteopathology Reference Centre, Institute of Pathology, Vivantes Klinikum im Friedrichshein, 10249 Berlin, Germany
| | - Stefan S. Bielack
- Cooperative Osteosarcoma Study Group, Paediatrics 5 (Oncology, Haematology, Immunology), Centre for Paediatric, Adolescent and Women’s Medicine, and Stuttgart Cancer Centre, Klinikum Stuttgart–Olgahospital, 70174 Stuttgart, Germany
- Department for Paediatric Haematology and Oncology, University’s Children’s Hospital Muenster, 48149 Muenster, Germany
| | - Stefanie Hecker-Nolting
- Cooperative Osteosarcoma Study Group, Paediatrics 5 (Oncology, Haematology, Immunology), Centre for Paediatric, Adolescent and Women’s Medicine, and Stuttgart Cancer Centre, Klinikum Stuttgart–Olgahospital, 70174 Stuttgart, Germany
| |
Collapse
|
16
|
Assi A, Farhat M, Hachem MCR, Zalaquett Z, Aoun M, Daher M, Sebaaly A, Kourie HR. Tyrosine kinase inhibitors in osteosarcoma: Adapting treatment strategiesa. J Bone Oncol 2023; 43:100511. [PMID: 38058514 PMCID: PMC10696463 DOI: 10.1016/j.jbo.2023.100511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/19/2023] [Accepted: 11/01/2023] [Indexed: 12/08/2023] Open
Abstract
Osteosarcoma (OS) is an aggressive primary bone malignancy that metastasizes rapidly. The standard of care has changed little over the previous four decades, and survival rates have plateaued. In this context, tyrosine kinase inhibitors (TKIs) emerge as potential treatments. A literature search was conducted to collect data related to receptor tyrosine kinase genetic alterations and expression in OS specimens. Gene amplification and protein expression of these receptors were linked to prognosis and tumor behavior. Relevant TKIs were evaluated as monotherapies and as parts of combination therapies. Certain TKIs, such as apatinib, regorafenib, and cabozantinib, present a potential therapeutic avenue for OS patients, especially when combined with chemotherapy. Producing long-lasting responses and enhancing quality of life remain key goals in OS treatment. To this effect, optimizing the use of TKIs by identifying biomarkers predictive of response and assessing promising TKIs in larger-scale trials to validate the efficacy and safety outcomes relative to these drugs reported in phase II clinical trials. To this effect, it is necessary to identify biomarkers predictive of response to TKIs in larger-scale trials and to validate the efficacy and safety of these drugs reported in phase II clinical trials.
Collapse
Affiliation(s)
- Ahmad Assi
- Hematology-Oncology Department, Hotel Dieu de France, Beirut, Lebanon
| | - Mohamad Farhat
- Hematology-Oncology Department, Hotel Dieu de France, Beirut, Lebanon
| | | | - Ziad Zalaquett
- Hematology-Oncology Department, Hotel Dieu de France, Beirut, Lebanon
| | - Marven Aoun
- Orthopedics Department, Hotel Dieu de France, Beirut, Lebanon
| | - Mohammad Daher
- Orthopedics Department, Hotel Dieu de France, Beirut, Lebanon
- Orthopedics Department, Brown University, Providence, RI, USA
| | - Amer Sebaaly
- Orthopedics Department, Hotel Dieu de France, Beirut, Lebanon
| | | |
Collapse
|
17
|
Morakote W, Adams LC, Ramasamy SK, Spunt SL, Baratto L, Liang T, Daldrup-Link HE. Tyrosine kinase inhibitor therapy in pediatric sarcoma: Prognostic implications of pulmonary metastatic cavitation. Pediatr Blood Cancer 2023; 70:e30629. [PMID: 37580891 PMCID: PMC10947454 DOI: 10.1002/pbc.30629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/08/2023] [Accepted: 08/06/2023] [Indexed: 08/16/2023]
Abstract
PURPOSES This study aims to ascertain the prevalence of cavitations in pulmonary metastases among pediatric and young adult patients with sarcoma undergoing tyrosine kinase inhibitor (TKI) therapy, and assess whether cavitation can predict clinical response and survival outcomes. METHODS In a single-center retrospective analysis, we examined chest computed tomography (CT) scans of 17 patients (median age 16 years; age range: 4-25 years) with histopathologically confirmed bone (n = 10) or soft tissue (n = 7) sarcoma who underwent TKI treatment for lung metastases. The interval between TKI initiation and the onset of lung nodule cavitation and tumor regrowth were assessed. The combination of all imaging studies and clinical data served as the reference standard for clinical responses. Progression-free survival (PFS) was compared between patients with cavitating and solid nodules using Kaplan-Meier survival analysis and log-rank test. RESULTS Five out of 17 patients (29%) exhibited cavitation of pulmonary nodules during TKI therapy. The median time from TKI initiation to the first observed cavitation was 79 days (range: 46-261 days). At the time of cavitation, all patients demonstrated stable disease. When the cavities began to fill with solid tumor, 60% (3/5) of patients exhibited progression in other pulmonary nodules. The median PFS for patients with cavitated pulmonary nodules after TKI treatment (6.7 months) was significantly longer compared to patients without cavitated nodules (3.8 months; log-rank p-value = .03). CONCLUSIONS Cavitation of metastatic pulmonary nodules in sarcoma patients undergoing TKI treatment is indicative of non-progressive disease, and significantly correlates with PFS.
Collapse
Affiliation(s)
- Wipawee Morakote
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California, USA
- Department of Radiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Lisa C Adams
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California, USA
| | - Shakthi K Ramasamy
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California, USA
| | - Sheri L Spunt
- Department of Pediatrics, Division of Hematology and Oncology, Stanford University, Stanford, California, USA
| | - Lucia Baratto
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California, USA
| | - Tie Liang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California, USA
| | - Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California, USA
- Department of Pediatrics, Division of Hematology and Oncology, Stanford University, Stanford, California, USA
| |
Collapse
|
18
|
van Ewijk R, Cleirec M, Herold N, le Deley MC, van Eijkelenburg N, Boudou-Rouquette P, Risbourg S, Strauss SJ, Palmerini E, Boye K, Kager L, Hecker-Nolting S, Marchais A, Gaspar N. A systematic review of recent phase-II trials in refractory or recurrent osteosarcoma: Can we inform future trial design? Cancer Treat Rev 2023; 120:102625. [PMID: 37738712 DOI: 10.1016/j.ctrv.2023.102625] [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: 07/22/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND/OBJECTIVE To analyze changes in recurrent/refractory osteosarcoma phase II trials over time to inform future trials in this population with poor prognosis. METHODS A systematic review of trials registered on trial registries between 01/01/2017-14/02/2022. Comparison of 98 trials identified between 2003 and 2016. Publication search/analysis for both periods, last update on 01/12/2022. RESULTS Between 2017 and 2022, 71 phase-II trials met our selection criteria (19 osteosarcoma-specific trials, 14 solid tumor trials with and 38 trials without an osteosarcoma-specific stratum). The trial number increased over time: 13.9 versus 7 trials/year (p = 0.06). Monotherapy remained the predominant treatment (62% vs. 62%, p = 1). Targeted therapies were increasingly evaluated (66% vs. 41%, P = 0.001). Heterogeneity persisted in the trial characteristics. The inclusion criteria were measurable disease (75%), evaluable disease (14%), and surgical remission (11%). 82% of the trials included pediatric or adolescent patients. Biomarker-driven trials accounted for 25% of the total trials. The survival endpoint use (rather than response) slightly increased (40% versus 31%), but the study H1/H0 hypotheses remained heterogeneous. Single-arm designs predominated over multiarm trials (n = 7). Available efficacy data on 1361 osteosarcoma patients in 58 trials remained disappointing, even though 21% of these trials were considered positive, predominantly those evaluating multi-targeted kinase inhibitors. CONCLUSION Despite observed changes in trial design and an increased number of trials investigating new therapies, high heterogeneity remained with respect to patient selection, study design, primary endpoints, and statistical hypotheses in recently registered phase II trials for osteosarcoma. Continued optimization of trial design informed by a deeper biological understanding should strengthen the development of new therapies.
Collapse
Affiliation(s)
- Roelof van Ewijk
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Morgane Cleirec
- Department of Pediatric Oncology, CHU Nantes, Nantes, France
| | - Nikolas Herold
- Paediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden, and Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Marie-Cécile le Deley
- Unité de Méthodologie et Biostatistiques, Centre Oscar Lambret, Lille, France; Université Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, U1018 ONCOSTAT, F-94085 Villejuif, France
| | | | - Pascaline Boudou-Rouquette
- Department of Medical Oncology, Cochin Hospital, Cochin Institute, INSERMU1016, Paris Cancer Institute, CARPEM, AP-HP, Paris, France
| | - Séverine Risbourg
- Unité de Méthodologie et Biostatistiques, Centre Oscar Lambret, Lille, France
| | - Sandra J Strauss
- Department of Oncology, University College London Cancer Institute, London, UK
| | - Emanuela Palmerini
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Kjetil Boye
- Department of Oncology, Oslo University Hospital, Norway
| | - Leo Kager
- St. Anna Children's Hospital, Department of Pediatrics, Medical University Vienna, Vienna, Austria; St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | | | - Antonin Marchais
- Department of Oncology for Child and Adolescents, Gustave Roussy Cancer Center, Paris-Saclay University, Villejuif, France; National Institute for Health and Medical Research (INSERM) U1015, BiiOSTeam, Gustave Roussy Institute, Villejuif, France
| | - Nathalie Gaspar
- Department of Oncology for Child and Adolescents, Gustave Roussy Cancer Center, Paris-Saclay University, Villejuif, France; National Institute for Health and Medical Research (INSERM) U1015, BiiOSTeam, Gustave Roussy Institute, Villejuif, France.
| |
Collapse
|
19
|
Olsen HE, Liu KX, Frazier AL, O’Neill AF, Janeway KA, DuBois SG, Shulman DS. Evaluation of prevalence and outcomes of serial tyrosine kinase inhibitor use in pediatric patients with advanced solid tumors. Pediatr Blood Cancer 2023; 70:e30652. [PMID: 37644664 PMCID: PMC10528491 DOI: 10.1002/pbc.30652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/31/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023]
Abstract
PURPOSE Multitargeted tyrosine kinase inhibitors (mTKIs) are increasingly utilized in the treatment of pediatric sarcomas and other solid tumors. It is unknown whether serial treatment with multiple TKIs provides a benefit and which patients are most likely to benefit from mTKI rechallenge. METHODS We performed a retrospective cohort study of pediatric cancer patients who received serial mTKI therapy off-study between 2007 and 2020 as either monotherapy or combination therapy. We report patient characteristics, clinical outcomes, dosing patterns, and treatment-associated toxicity. RESULTS The study cohort included 25 patients. The overall prevalence of serial mTKI therapy among all patients treated for sarcoma at our institution was 3.7%, and the response rate to second mTKI was 9%. Median 6-month progression-free survival (PFS) and overall survival (OS) from start of second mTKI were 42.1% (95% CI: 20.4%-62.5%) and 79.1% (95% CI: 57.0%-90.8%), respectively. Patients who had received 4 months or more (n = 11) of therapy with first mTKI had significantly longer PFS versus those who received less than 4 months (n = 11; p = .001). Thirty-three percent of patients discontinued second mTKI due to toxicity. Six (40%) of 15 patients who discontinued the first mTKI due to progression had either a partial response or stable disease on the second mTKI. CONCLUSIONS We observed a low response rate to mTKI rechallenge. However, we identified patients who had been treated with first mTKI for ≥4 months as more likely to have prolonged stable disease with second mTKI. Several patients had a response or stable disease on the second mTKI despite having progressed on the first mTKI. Though toxicity was common, only a minority of patients discontinued the second mTKI due to toxicity.
Collapse
Affiliation(s)
| | - Kevin X. Liu
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston Children's Hospital, Boston, MA, USA
| | - A. Lindsay Frazier
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Allison F. O’Neill
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Katherine A. Janeway
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Steven G. DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - David S. Shulman
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| |
Collapse
|
20
|
Li H, Li Y, Song L, Ai Q, Zhang S. Retrospective review of safety and efficacy of anlotinib in advanced osteosarcoma with metastases after failure of standard multimodal therapy. Asia Pac J Clin Oncol 2023; 19:e314-e319. [PMID: 36658675 DOI: 10.1111/ajco.13916] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/21/2022] [Accepted: 12/05/2022] [Indexed: 01/21/2023]
Abstract
AIM To study the safety and efficacy of anlotinib, a multitargeted tyrosine kinase inhibitor, in the treatment of advanced osteosarcoma (OSS) with metastases. METHODS We retrospectively studied patients with advanced OSS and metastases who received anlotinib treatment in our hospital from June 2018 to April 2020. All patients had received standard multimodal therapies, before taking anlotinib. Therapeutic doses of anlotinib were 12 mg for adults and 10 mg for children and adolescents once a day for 2 consecutive weeks, followed by a week of withdrawal. This 3-week cycle of treatment was continued until the tumor progressed rapidly or the patients failed to tolerate the side effects. Adverse drug reactions were recorded, and therapeutic efficacy was evaluated based on progression-free survival (PFS), disease control rate (DCR), overall survival (OS), and objective response rate (ORR). RESULTS The median PFS was 9.8 ± .9 months, and the 6- and 10-month PFS rates were 73% and 33%, respectively. The median OS was 11.4 ± .6 months. No patients achieved complete response. After 6 months of treatment, the DCR and ORR were 80% and 13%, respectively. No drug-related deaths or Grade 4 adverse events occurred in the patients. Five patients (33%) had Grade 3 adverse events. The most common drug-related adverse events were hand-food syndrome, fatigue, high blood pressure, anorexia, and pneumothorax. CONCLUSIONS Anlotinib had a modest therapeutic effect in patients with advanced OSS after the failure of standard treatment. The adverse events were mostly tolerable or relieved after treatment.
Collapse
Affiliation(s)
- Hanqing Li
- Orthopedics Department, Southwest Hospital, The Army Military Medical University (The Third Military Medical University), Chongqing, China
| | - Yang Li
- Orthopedics Department, Southwest Hospital, The Army Military Medical University (The Third Military Medical University), Chongqing, China
| | - Lei Song
- Orthopedics Department, Southwest Hospital, The Army Military Medical University (The Third Military Medical University), Chongqing, China
| | - Qiuchi Ai
- Orthopedics Department, Southwest Hospital, The Army Military Medical University (The Third Military Medical University), Chongqing, China
| | - Shuai Zhang
- Orthopedics Department, Southwest Hospital, The Army Military Medical University (The Third Military Medical University), Chongqing, China
| |
Collapse
|
21
|
Reed DR, Grohar P, Rubin E, Binitie O, Krailo M, Davis J, DuBois SG, Janeway KA. Children's Oncology Group's 2023 blueprint for research: Bone tumors. Pediatr Blood Cancer 2023; 70 Suppl 6:e30583. [PMID: 37501549 PMCID: PMC10499366 DOI: 10.1002/pbc.30583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/29/2023]
Abstract
The Children's Oncology Group (COG) Bone Tumor Committee is responsible for clinical trials and biological research on localized, metastatic, and recurrent osteosarcoma and Ewing sarcoma (EWS). Results of clinical trials in localized disease completed and published in the past 10 years have led to international standard-of-care chemotherapy for osteosarcoma and EWS. A recent focus on identifying disease subgroups has led to the identification of biological features associated with poor outcomes including the presence of circulating tumor DNA (ctDNA) at diagnosis, and specific genomic alterations-MYC amplification for osteosarcoma and STAG2 and TP53 mutation for EWS. Studies validating these potential biomarkers are under way. Clinical trials evaluating the addition of multitargeted kinase inhibitors, which are active in relapsed bone sarcomas, to standard chemotherapy are under way in osteosarcoma and planned in EWS. In addition, the Committee has data analyses and a clinical trial under way to evaluate approaches to local management of the primary tumor and metastatic sites. Given the rarity of bone sarcomas, we have prioritized international interactions and are in the process of forming an international data-sharing consortium to facilitate refinement of risk stratification and study of rare disease subtypes.
Collapse
Affiliation(s)
- Damon R Reed
- Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, Florida, USA
| | - Patrick Grohar
- Division of Oncology, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elyssa Rubin
- Department of Oncology, Children's Hospital of Orange County, Orange, California, USA
| | - Odion Binitie
- Department of Sarcoma, Moffitt Cancer Center, Tampa, Florida, USA
| | - Mark Krailo
- Keck School of Medicine, University of Southern California and Children's Oncology Group, Monrovia, California, USA
| | - Jessica Davis
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Steven G DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Katherine A Janeway
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| |
Collapse
|
22
|
Peretz Soroka H, Vora T, Noujaim J, Marcoux N, Cohen‐Gogo S, Alcindor T, Holloway C, Rodrigues C, Karachiwala H, Alvi S, Lee U, Cheng S, Banerji S, Oberoi S, Feng X, Smrke A, Simmons C, Razak AA, Gupta AA. Real-world experience of tyrosine kinase inhibitors in children, adolescents and adults with relapsed or refractory bone tumours: A Canadian Sarcoma Research and Clinical Collaboration (CanSaRCC) study. Cancer Med 2023; 12:18872-18881. [PMID: 37724607 PMCID: PMC10557866 DOI: 10.1002/cam4.6515] [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: 03/21/2023] [Revised: 08/26/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023] Open
Abstract
OBJECTIVES We conducted a retrospective multi-centre study to assess the real-world outcome of regorafenib (REGO) and cabozantinib (CABO) in recurrent/refractory bone tumours (BTs) including osteosarcoma (OST), Ewing sarcoma (EWS) and chondrosarcoma (CS)/extra-skeletal mesenchymal CS (ESMC). METHODS After regulatory approval, data from patients with recurrent BT (11 institutions) were extracted from CanSaRCC (Canadian Sarcoma Research and Clinical Collaboration) database. Patient characteristics, treatment and outcomes were collected. Progression-free survival (PFS) and overall survival (OS) were estimated using the Kaplan-Meier method. RESULTS From July 2018 to May 2022, 66 patients received REGO or CABO; 39 OST, 18 EWS, 4 CS and 5 ESMC. Median age was 27.8 years (range 12-76); median starting dose was 60 mg for CABO (n = 37, range 40-60) and 120 mg for REGO (n = 29, range 40-160). Twenty-eight (42.4%) patients required dose reduction: hand-foot syndrome 7 (10.6%), nausea/vomiting 1 (1.5%), diarrhoea 1 (1.5%), 2 elevated LFTs (3%), elevated bilirubin 1 (1.5%) and mucositis 1 (1.5%). The median OS for patients with OST, EWS, CS and ESMC was 8.5 months (n = 39, 95% CI 7-13.1); 13.4 months (n = 18, 95% CI 3.4-27.2), 8.1 (n = 4, 95% CI 4.1-9.3) and 18.2 (n = 5, 95% CI (10.4-na), respectively. Median PFS for OST, EWS, CS and ECMS was 3.5 (n = 39, 95% CI 2.8-5), 3.9 (n = 18, 95% CI 2.1-5.9), 5.53 (n = 4. 95% CI 2.13-NA) and 11.4 (n = 5, 95% CI 1.83-14.7), respectively. Age, line of therapy, REGO versus CABO, or time from diagnosis to initiation of TKI were not associated with PFS on univariable analysis. CONCLUSION Our real-world data show that TKIs have meaningful activity in recurrent BT with acceptable toxicities when started at modified dosing. Inclusion of TKIs in earlier lines of treatment and/or maintenance therapy could be questions for future research.
Collapse
Affiliation(s)
- Hagit Peretz Soroka
- Division of Medical Oncology, Princess Margaret Cancer CentreUniversity of TorontoTorontoOntarioCanada
| | - Tushar Vora
- Division of Hematology/Oncology, Hospital for Sick ChildrenUniversity of TorontoTorontoOntarioCanada
| | - Jonathan Noujaim
- Division of Medical Oncology, Hôpital Maisonneuve RosemontUniversity of MontrealMontrealQuebecCanada
| | - Nicolas Marcoux
- Division of Hematology‐OncologyCentre Hospitalier Universitaire de QuébecQuebecCanada
| | - Sarah Cohen‐Gogo
- Division of Hematology/Oncology, Hospital for Sick ChildrenUniversity of TorontoTorontoOntarioCanada
| | - Thierry Alcindor
- Division of Medical OncologyMcGill University Health CentreMontrealQuebecCanada
| | - Caroline Holloway
- Division of Radiation Oncology, BC CancerUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Caroline Rodrigues
- Division of Medical Oncology, Princess Margaret Cancer CentreUniversity of TorontoTorontoOntarioCanada
| | - Hatim Karachiwala
- Division of Medical Oncology, Cross Cancer InstituteAlberta Health ServicesEdmontonAlbertaCanada
| | - Saima Alvi
- Division of Pediatric Hematology/OncologyJim Pattison Children's Hospital SaskatoonSaskatoonSaskatchewanCanada
| | - Ursula Lee
- Division of Medical Oncology, BC CancerUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Sylvia Cheng
- Division of Pediatric Hematology/Oncology/BMTB.C. Children's Hospital, BC CancerVancouverBritish ColumbiaCanada
| | - Shantanu Banerji
- Department of Pediatric Hematology‐Oncology, CancerCare Manitoba Research Institute, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Sapna Oberoi
- Department of Pediatric Hematology‐Oncology, CancerCare Manitoba Research Institute, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Xiaolan Feng
- Division of Medical Oncology, BC CancerUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Alannah Smrke
- Division of Medical Oncology, BC CancerUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Christine Simmons
- Division of Medical Oncology, BC CancerUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Albiruni Abdul Razak
- Division of Medical Oncology, Princess Margaret Cancer CentreUniversity of TorontoTorontoOntarioCanada
- Division of Medical Oncology, Mount Sinai HospitalUniversity of TorontoTorontoOntarioCanada
| | - Abha A. Gupta
- Division of Medical Oncology, Princess Margaret Cancer CentreUniversity of TorontoTorontoOntarioCanada
- Division of Hematology/Oncology, Hospital for Sick ChildrenUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
23
|
Ji Z, Shen J, Lan Y, Yi Q, Liu H. Targeting signaling pathways in osteosarcoma: Mechanisms and clinical studies. MedComm (Beijing) 2023; 4:e308. [PMID: 37441462 PMCID: PMC10333890 DOI: 10.1002/mco2.308] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 07/15/2023] Open
Abstract
Osteosarcoma (OS) is a highly prevalent bone malignancy among adolescents, accounting for 40% of all primary malignant bone tumors. Neoadjuvant chemotherapy combined with limb-preserving surgery has effectively reduced patient disability and mortality, but pulmonary metastases and OS cells' resistance to chemotherapeutic agents are pressing challenges in the clinical management of OS. There has been an urgent need to identify new biomarkers for OS to develop specific targeted therapies. Recently, the continued advancements in genomic analysis have contributed to the identification of clinically significant molecular biomarkers for diagnosing OS, acting as therapeutic targets, and predicting prognosis. Additionally, the contemporary molecular classifications have revealed that the signaling pathways, including Wnt/β-catenin, PI3K/AKT/mTOR, JAK/STAT3, Hippo, Notch, PD-1/PD-L1, MAPK, and NF-κB, have an integral role in OS onset, progression, metastasis, and treatment response. These molecular classifications and biological markers have created new avenues for more accurate OS diagnosis and relevant treatment. We herein present a review of the recent findings for the modulatory role of signaling pathways as possible biological markers and treatment targets for OS. This review also discusses current OS therapeutic approaches, including signaling pathway-based therapies developed over the past decade. Additionally, the review covers the signaling targets involved in the curative effects of traditional Chinese medicines in the context of expression regulation of relevant genes and proteins through the signaling pathways to inhibit OS cell growth. These findings are expected to provide directions for integrating genomic, molecular, and clinical profiles to enhance OS diagnosis and treatment.
Collapse
Affiliation(s)
- Ziyu Ji
- School of Integrated Traditional Chinese and Western MedicineSouthwest Medical UniversityLuzhouSichuanChina
| | - Jianlin Shen
- Department of OrthopaedicsAffiliated Hospital of Putian UniversityPutianFujianChina
| | - Yujian Lan
- School of Integrated Traditional Chinese and Western MedicineSouthwest Medical UniversityLuzhouSichuanChina
| | - Qian Yi
- Department of PhysiologySchool of Basic Medical ScienceSouthwest Medical UniversityLuzhouSichuanChina
| | - Huan Liu
- Department of OrthopaedicsThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouSichuanChina
| |
Collapse
|
24
|
Liao Z, Li M, Wen G, Wang K, Yao D, Chen E, Liang Y, Xing T, Su K, Liang C, Che Z, Ning Q, Tang J, Yan W, Li Y, Huang L. Comprehensive analysis of angiogenesis pattern and related immune landscape for individual treatment in osteosarcoma. NPJ Precis Oncol 2023; 7:62. [PMID: 37386055 DOI: 10.1038/s41698-023-00415-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/15/2023] [Indexed: 07/01/2023] Open
Abstract
Postoperative recurrence and metastasis are the main reasons for the poor prognosis of osteosarcoma (OS). Currently, an ideal predictor for not only prognosis but also drug sensitivity and immunotherapy responses in OS patients is urgently needed. Angiogenesis plays a crucial role in tumour progression, which suggests its immense potential for predicting prognosis and responses to immunotherapy for OS. Angiogenesis patterns in OS were explored in depth in this study to construct a prognostic model called ANGscore and clarify the underlying mechanism involved in the immune microenvironment. The efficacy and robustness of the model were validated in multiple datasets, including bulk RNA-seq datasets (TARGET-OS, GSE21257), a single-cell RNA-seq dataset (GSE152048) and immunotherapy-related datasets (GSE91061, GSE173839). OS patients with a high ANGscore had a worse prognosis, accompanied by the immune desert phenotype. Pseudotime and cellular communication analyses in scRNA-seq data revealed that as the ANGscore increased, the malignant degree of cells increased, and IFN-γ signalling was involved in tumour progression and regulation of the tumour immune microenvironment. Furthermore, the ANGscore was associated with immune cell infiltration and the response rate to immunotherapy. OS patients with high ANGscore might be resistant to uprosertib, and be sensitive to VE821, AZD6738 and BMS.345541. In conclusion, we established a novel ANGscore system by comprehensively analysing the expression pattern of angiogenesis genes, which can accurately differentiate the prognosis and immune characteristics of OS populations. Additionally, the ANGscore can be used for patient stratification during immunotherapy, and guide individualized treatment strategies.
Collapse
Affiliation(s)
- Zhuangyao Liao
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ming Li
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guoming Wen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Kun Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Dengbo Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Enming Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuwei Liang
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tong Xing
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Kaihui Su
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Changchun Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhen Che
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing Ning
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Tang
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenbin Yan
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuxi Li
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Lin Huang
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
25
|
Xu J, Chen C, Sun K, Shi Q, Wang B, Huang Y, Ren T, Tang X. Tocilizumab (monoclonal anti-IL-6R antibody) reverses anlotinib resistance in osteosarcoma. Front Oncol 2023; 13:1192472. [PMID: 37404767 PMCID: PMC10315670 DOI: 10.3389/fonc.2023.1192472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/05/2023] [Indexed: 07/06/2023] Open
Abstract
Purpose Anlotinib, a tyrosine kinase inhibitor (TKI) has been in clinical application to inhibit malignant cell growth and lung metastasis in osteosarcoma (OS). However, a variety of drug resistance phenomena have been observed in the treatment. We aim to explore the new target to reverse anlotinib resistance in OS. Materials and Methods In this study, we established four OS anlotinib-resistant cell lines, and RNA-sequence was performed to evaluate differentially expressed genes. We verified the results of RNA-sequence by PCR, western blot and ELISA assay. We further explored the effects of tocilizumab (anti- IL-6 receptor), either alone or in combined with anlotinib, on the inhibition of anlotinib-resistant OS cells malignant viability by CCK8, EDU, colony formation, apoptosis, transwell, wound healing, Cytoskeletal stain assays, and xenograft nude mouse model. The expression of IL-6 in 104 osteosarcoma samples was tested by IHC. Results We found IL-6 and its downstream pathway STAT3 were activated in anlotinib-resistant osteosarcoma. Tocilizumab impaired the tumor progression of anlotinib-resistant OS cells, and combined treatment with anlotinib augmented these effects by inhibiting STAT3 expressions. IL-6 was highly expressed in patients with OS and correlated with poor prognosis. Conclusion Tocilizumab could reverse anlotinib resistance in OS by IL-6/STAT3 pathway and the combination treatment with anlotinib rationalized further studies and clinical treatment of OS.
Collapse
Affiliation(s)
- Jiuhui Xu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Chenglong Chen
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Department of Orthopedics, Beijing Jishuitan Hospital, Beijing, China
| | - Kunkun Sun
- Department of Pathology, Peking University People’s Hospital, Beijing, China
| | - Qianyu Shi
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Boyang Wang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Yi Huang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Tingting Ren
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Xiaodong Tang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
26
|
Zarghooni K, Bratke G, Landgraf P, Simon T, Maintz D, Eysel P. The Diagnosis and Treatment of Osteosarcoma and Ewing's Sarcoma in Children and Adolescents. DEUTSCHES ARZTEBLATT INTERNATIONAL 2023; 120:405-412. [PMID: 37097079 PMCID: PMC10437036 DOI: 10.3238/arztebl.m2023.0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 06/24/2022] [Accepted: 03/22/2023] [Indexed: 04/26/2023]
Abstract
BACKGROUND Osteosarcoma and Ewing's sarcoma in children and adolescents require age-specific interdisciplinary diagnosis and treatment to achieve optimal therapeutic outcomes. METHODS The diagnosis and treatment of malignant bone tumors in childhood and adolescence are presented in the light of publications retrieved by a selective search, pertinent guidelines, and the authors' extensive experience in an interdisciplinary cancer center. RESULTS Bone sarcomas make up approximately 5% of all malignancies in children and adolescents; the most common types are Ewing's sarcoma and osteosarcoma. Patients are often not referred to a specialized center until long after the onset of symptoms, as they and their physicians rarely consider the possibility of a bone tumor, and the symptoms are often trivialized. Bone pain of unknown origin, swelling, and functional limitations should be investigated with conventional x-rays. Lesions of unclear origin should be biopsied after a meticulous clinical and radiologic evaluation. Multimodal treatment consists of neo - adjuvant chemotherapy, limb-preserving resection if possible, and radiotherapy where indicated. In multicenter studies, patients with osteosarcoma achieve event-free survival in 64% of cases if their disease is localized, and 28% if it is metastatic; the corresponding figures for patients with Ewing's sarcoma are 80% and 27%, respectively. CONCLUSION With implementation of the current treatment recommendations, most children and adolescents with malignant bone tumors can be treated successfully with curative intent. These patients should be referred to a sarcoma center for diagnosis and treatment.
Collapse
Affiliation(s)
- Kourosh Zarghooni
- Department of Orthopedics and Trauma Surgery, Helios Hospital Hildesheim, Germany
- Department of Orthopedics and Trauma Surgery, University Hospital Cologne, Germany
| | - Grischa Bratke
- Institute of Diagnostic and Interventional Radiology, University Hospital of Cologne, Germany
| | - Pablo Landgraf
- Department of Pediatric Oncology, University Hospital Cologne, Germany
| | - Thorsten Simon
- Department of Pediatric Oncology, University Hospital Cologne, Germany
| | - David Maintz
- Institute of Diagnostic and Interventional Radiology, University Hospital of Cologne, Germany
| | - Peer Eysel
- Department of Orthopedics and Trauma Surgery, University Hospital Cologne, Germany
| |
Collapse
|
27
|
Liu D, Peng Y, Li X, Zhu Z, Mi Z, Zhang Z, Fan H. Comprehensive landscape of TGFβ-related signature in osteosarcoma for predicting prognosis, immune characteristics, and therapeutic response. J Bone Oncol 2023; 40:100484. [PMID: 37234254 PMCID: PMC10205544 DOI: 10.1016/j.jbo.2023.100484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
Osteosarcoma (OS) is a highly heterogeneous malignant bone tumor, and its tendency to metastasize leads to a poor prognosis. TGFβ is an important regulator in the tumor microenvironment and is closely associated with the progression of various types of cancer. However, the role of TGFβ-related genes in OS is still unclear. In this study, we identified 82 TGFβ DEGs based on RNA-seq data from the TARGET and GETx databases and classified OS patients into two TGFβ subtypes. The KM curve showed that the Cluster 2 patients had a substantially poorer prognosis than the Cluster 1 patients. Subsequently, a novel TGFβ prognostic signatures (MYC and BMP8B) were developed based on the results of univariate, LASSO, and multifactorial Cox analyses. These signatures showed robust and reliable predictive performance for the prognosis of OS in the training and validation cohorts. To predict the three-year and five-year survival rate of OS, a nomogram that integrated clinical features and risk scores was also developed. The GSEA analysis showed that the different subgroups analyzed had distinct functions, particularly, the low-risk group was associated with high immune activity and a high infiltration abundance of CD8 T cells. Moreover, our results indicated that low-risk cases had higher sensitivity to immunotherapy, while high-risk cases were more sensitive to sorafenib and axitinib. scRNA-Seq analysis further revealed that MYC and BMP8B were strongly expressed mainly in tumor stromal cells. Finally, in this study, we confirmed the expression of MYC and BMP8B by performing qPCR, WB, and IHC analyses. To conclude, we developed and validated a TGFβ-related signature to accurately predict the prognosis of OS. Our findings might contribute to personalized treatment and making better clinical decisions for OS patients.
Collapse
Affiliation(s)
- Dong Liu
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Ye Peng
- Department of Orthopaedics, Air Force Medical Center, PLA, Beijing 100142, China
| | - Xian Li
- Department of Orthopaedics, Shenzhen University General Hospital, Shenzhen, China
| | - Zhijie Zhu
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Zhenzhou Mi
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Zhao Zhang
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Hongbin Fan
- Department of Orthopaedic Surgery, Xi-jing Hospital, The Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
28
|
Brown LM, Ekert PG, Fleuren EDG. Biological and clinical implications of FGFR aberrations in paediatric and young adult cancers. Oncogene 2023:10.1038/s41388-023-02705-7. [PMID: 37130917 DOI: 10.1038/s41388-023-02705-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/16/2023] [Accepted: 04/20/2023] [Indexed: 05/04/2023]
Abstract
Rare but recurrent mutations in the fibroblast growth factor receptor (FGFR) pathways, most commonly in one of the four FGFR receptor tyrosine kinase genes, can potentially be targeted with broad-spectrum multi-kinase or FGFR selective inhibitors. The complete spectrum of these mutations in paediatric cancers is emerging as precision medicine programs perform comprehensive sequencing of individual tumours. Identification of patients most likely to benefit from FGFR inhibition currently rests on identifying activating FGFR mutations, gene fusions, or gene amplification events. However, the expanding use of transcriptome sequencing (RNAseq) has identified that many tumours overexpress FGFRs, in the absence of any genomic aberration. The challenge now presented is to determine when this indicates true FGFR oncogenic activity. Under-appreciated mechanisms of FGFR pathway activation, including alternate FGFR transcript expression and concomitant FGFR and FGF ligand expression, may mark those tumours where FGFR overexpression is indicative of a dependence on FGFR signalling. In this review, we provide a comprehensive and mechanistic overview of FGFR pathway aberrations and their functional consequences in paediatric cancer. We explore how FGFR over expression might be associated with true receptor activation. Further, we discuss the therapeutic implications of these aberrations in the paediatric setting and outline current and emerging therapeutic strategies to treat paediatric patients with FGFR-driven cancers.
Collapse
Affiliation(s)
- Lauren M Brown
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| | - Paul G Ekert
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia.
- University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia.
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Parkville, VIC, Australia.
| | - Emmy D G Fleuren
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
- University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
29
|
Wood GE, Graves LA, Rubin EM, Reed DR, Riedel RF, Strauss SJ. Bad to the Bone: Emerging Approaches to Aggressive Bone Sarcomas. Am Soc Clin Oncol Educ Book 2023; 43:e390306. [PMID: 37220319 DOI: 10.1200/edbk_390306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Bone sarcomas are rare heterogeneous tumors that affect patients of all ages including children, adolescent young adults, and older adults. They include many aggressive subtypes and patient groups with poor outcomes, poor access to clinical trials, and lack of defined standard therapeutic strategies. Conventional chondrosarcoma remains a surgical disease, with no defined role for cytotoxic therapy and no approved targeted systemic therapies. Here, we discuss promising novel targets and strategies undergoing evaluation in clinical trials. Multiagent chemotherapy has greatly improved outcomes for patients with Ewing sarcoma (ES) and osteosarcoma, but management of those with high-risk or recurrent disease remains challenging and controversial. We describe the impact of international collaborative trials, such as the rEECur study, that aim to define optimal treatment strategies for those with recurrent, refractory ES, and evidence for high-dose chemotherapy with stem-cell support. We also discuss current and emerging strategies for other small round cell sarcomas, such as CIC-rearranged, BCOR-rearranged tumors, and the evaluation of emerging novel therapeutics and trial designs that may offer a new paradigm to improve survival in these aggressive tumors with notoriously bad (to the bone) outcomes.
Collapse
Affiliation(s)
- Georgina E Wood
- Department of Oncology, University College London Hospitals NHS Trust, UCL Cancer Institute, London, United Kingdom
| | - Laurie A Graves
- Division of Hematology/Oncology, Department of Pediatrics, Duke University, Durham, NC
| | - Elyssa M Rubin
- Division of Oncology, Children's Hospital of Orange County, Orange, CA
| | - Damon R Reed
- Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, FL
| | - Richard F Riedel
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Durham, NC
| | - Sandra J Strauss
- Department of Oncology, University College London Hospitals NHS Trust, UCL Cancer Institute, London, United Kingdom
| |
Collapse
|
30
|
Pilavaki P, Gahanbani Ardakani A, Gikas P, Constantinidou A. Osteosarcoma: Current Concepts and Evolutions in Management Principles. J Clin Med 2023; 12:jcm12082785. [PMID: 37109122 PMCID: PMC10143544 DOI: 10.3390/jcm12082785] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/08/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Osteosarcoma is a rare malignancy arising from mesenchymal tissue, and represents the most common bone sarcoma. The management of osteosarcoma is challenging, and requires a multidisciplinary approach. In daily clinical practice, surgery, radiotherapy, and conventional chemotherapy constitute the therapeutic armamentarium against the disease. However, a significant number of patients with initially localized osteosarcoma will experience local or distant recurrence, and the prognosis for metastatic disease remains dismal. There is a pressing need to identify novel therapeutic strategies to better manage osteosarcoma and improve survival outcomes. In this study, we present recent advances in the therapeutic management of osteosarcoma, including surgical and medical advances. The role of immunotherapy (immune checkpoint inhibitors, adoptive cellular therapy, cancer vaccines) and other targeted therapies including tyrosine kinase inhibitors is discussed; however, additional studies are required to delineate their roles in clinical practice.
Collapse
Affiliation(s)
- Pampina Pilavaki
- Medical School, University of Cyprus, Nicosia 1678, Cyprus
- Medical Oncology, Bank of Cyprus Oncology Center, Nicosia 2006, Cyprus
| | | | - Panagiotis Gikas
- Department of Orthopaedics, Cleveland Clinic London, London SW1X 7HY, UK
| | - Anastasia Constantinidou
- Medical School, University of Cyprus, Nicosia 1678, Cyprus
- Medical Oncology, Bank of Cyprus Oncology Center, Nicosia 2006, Cyprus
- Cyprus Cancer Research Institute, Nicosia 2109, Cyprus
| |
Collapse
|
31
|
Umeda K, Sakamoto A, Noguchi T, Uchihara Y, Kobushi H, Akazawa R, Ogata H, Saida S, Kato I, Hiramatsu H, Uto M, Mizowaki T, Haga H, Date H, Okamoto T, Watanabe K, Adachi S, Toguchida J, Matsuda S, Takita J. Clinical Outcomes of Patients With Osteosarcoma Experiencing Relapse or Progression: A Single-institute Experience. J Pediatr Hematol Oncol 2023; 45:e356-e362. [PMID: 35973000 DOI: 10.1097/mph.0000000000002521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/28/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Patients with osteosarcoma who experience relapse or progression [R/P] have a poor prognosis. METHODS Data from 30 patients who experienced R/P among 59 with a diagnosis of high-grade osteosarcoma, who were younger than 40 years old between 2000 and 2019, were retrospectively analyzed to identify prognostic and therapeutic factors influencing their outcomes. RESULTS The 5-year overall survival [OS] rates after the last R/P of patients experiencing first [n=30], second [n=14], and third [n=9] R/P were 50.3%, 51.3%, and 46.7%, respectively. Multivariate analysis did not identify any independent risk factors affecting OS. The 5-year PFS rate of the 30 patients after first R/P was 22.4%, and multivariate analysis identified histologic subtype and curative local surgery as independent risk factors influencing PFS. Long [>6 mo] partial response was observed in three patients treated using temozolomide+etoposide, irinotecan+carboplatin, or regorafenib. CONCLUSIONS OS rate in the patients with osteosarcoma experiencing R/P included in this study was markedly higher than that reported previously, mainly due to the surgical total removal of tumors, even after subsequent R/P. The recent establishment of salvage chemotherapy or molecular targeted therapy may also increase survival rates in a subgroup of patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Megumi Uto
- Radiation Oncology and Image-Applied Therapy
| | | | | | | | - Takeshi Okamoto
- Department of Orthopedic Surgery, Otsu Red Cross Hospital, Otsu, Japan
| | - Kenichiro Watanabe
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Souichi Adachi
- Department of Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junya Toguchida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Japan
| | | | | |
Collapse
|
32
|
Fleuren EDG, Vlenterie M, van der Graaf WTA. Recent advances on anti-angiogenic multi-receptor tyrosine kinase inhibitors in osteosarcoma and Ewing sarcoma. Front Oncol 2023; 13:1013359. [PMID: 36994209 PMCID: PMC10040783 DOI: 10.3389/fonc.2023.1013359] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 02/06/2023] [Indexed: 03/16/2023] Open
Abstract
Osteosarcoma (OS) and Ewing sarcoma (ES) are the two most common types of primary bone cancer that predominantly affect the young. Despite aggressive multimodal treatment, survival has not improved significantly over the past four decades. Clinical efficacy has historically been observed for some mono-Receptor Tyrosine Kinase (RTK) inhibitors, albeit in small subsets of OS and ES patients. Clinical efficacy in larger groups of OS or ES patients was reported recently with several newer generation multi-RTK inhibitors. All these inhibitors combine a strong anti-angiogenic (VEGFRs) component with simultaneous inhibition of other key RTKs implicated in OS and ES progression (PDGFR, FGFR, KIT and/or MET). However, despite interesting clinical data, none of these agents have obtained a registration for these indications and are thus difficult to implement in routine OS and ES patient care. It is at present also unclear which of these drugs, with largely overlapping molecular inhibition profiles, would work best for which patient or subtype, and treatment resistance almost uniformly occurs. Here, we provide a critical assessment and systemic comparison on the clinical outcomes to the six most tested drugs in this field in OS and ES to date, including pazopanib, sorafenib, regorafenib, anlotinib, lenvatinib and cabozantinib. We pay special attention to clinical response evaluations in bone sarcomas and provide drug comparisons, including drug-related toxicity, to put these drugs into context for OS and ES patients, and describe how future trials utilizing anti-angiogenic multi-RTK targeted drugs could be designed to ultimately improve response rates and decrease toxicity.
Collapse
Affiliation(s)
- Emmy D. G. Fleuren
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
- Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
- *Correspondence: Emmy D. G. Fleuren,
| | - Myrella Vlenterie
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Winette T. A. van der Graaf
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
33
|
Li S, Zhang H, Liu J, Shang G. Targeted therapy for osteosarcoma: a review. J Cancer Res Clin Oncol 2023:10.1007/s00432-023-04614-4. [PMID: 36807762 DOI: 10.1007/s00432-023-04614-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/27/2023] [Indexed: 02/21/2023]
Abstract
BACKGROUND Osteosarcoma is a common primary malignant tumour of the bone that usually occurs in children and adolescents. It is characterised by difficult treatment, recurrence and metastasis, and poor prognosis. Currently, the treatment of osteosarcoma is mainly based on surgery and auxiliary chemotherapy. However, for recurrent and some primary osteosarcoma cases, owing to the rapid progression of disease and chemotherapy resistance, the effects of chemotherapy are poor. With the rapid development of tumour-targeted therapy, molecular-targeted therapy for osteosarcoma has shown promise. PURPOSE In this paper, we review the molecular mechanisms, related targets, and clinical applications of targeted osteosarcoma therapy. In doing this, we provide a summary of recent literature on the characteristics of targeted osteosarcoma therapy, the advantages of its clinical application, and development of targeted therapy in future. We aim to provide new insights into the treatment of osteosarcoma. CONCLUSION Targeted therapy shows potential in the treatment of osteosarcoma and may offer an important means of precise and personalised treatment in the future, but drug resistance and adverse effects may limit its application.
Collapse
Affiliation(s)
- Shizhe Li
- Department of Bone and Soft Tissue Oncology, Shengjing Hospital Affiliated to China Medical University, Shenyang, 110022, Liaoning Province, China.,Graduate School, Jinzhou Medical University, Jinzhou, 121001, Liaoning Province, China
| | - He Zhang
- Department of Bone and Soft Tissue Oncology, Shengjing Hospital Affiliated to China Medical University, Shenyang, 110022, Liaoning Province, China
| | - Jinxin Liu
- Department of Bone and Soft Tissue Oncology, Shengjing Hospital Affiliated to China Medical University, Shenyang, 110022, Liaoning Province, China
| | - Guanning Shang
- Department of Bone and Soft Tissue Oncology, Shengjing Hospital Affiliated to China Medical University, Shenyang, 110022, Liaoning Province, China.
| |
Collapse
|
34
|
Candido MF, Medeiros M, Veronez LC, Bastos D, Oliveira KL, Pezuk JA, Valera ET, Brassesco MS. Drugging Hijacked Kinase Pathways in Pediatric Oncology: Opportunities and Current Scenario. Pharmaceutics 2023; 15:pharmaceutics15020664. [PMID: 36839989 PMCID: PMC9966033 DOI: 10.3390/pharmaceutics15020664] [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: 12/15/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Childhood cancer is considered rare, corresponding to ~3% of all malignant neoplasms in the human population. The World Health Organization (WHO) reports a universal occurrence of more than 15 cases per 100,000 inhabitants around the globe, and despite improvements in diagnosis, treatment and supportive care, one child dies of cancer every 3 min. Consequently, more efficient, selective and affordable therapeutics are still needed in order to improve outcomes and avoid long-term sequelae. Alterations in kinases' functionality is a trademark of cancer and the concept of exploiting them as drug targets has burgeoned in academia and in the pharmaceutical industry of the 21st century. Consequently, an increasing plethora of inhibitors has emerged. In the present study, the expression patterns of a selected group of kinases (including tyrosine receptors, members of the PI3K/AKT/mTOR and MAPK pathways, coordinators of cell cycle progression, and chromosome segregation) and their correlation with clinical outcomes in pediatric solid tumors were accessed through the R2: Genomics Analysis and Visualization Platform and by a thorough search of published literature. To further illustrate the importance of kinase dysregulation in the pathophysiology of pediatric cancer, we analyzed the vulnerability of different cancer cell lines against their inhibition through the Cancer Dependency Map portal, and performed a search for kinase-targeted compounds with approval and clinical applicability through the CanSAR knowledgebase. Finally, we provide a detailed literature review of a considerable set of small molecules that mitigate kinase activity under experimental testing and clinical trials for the treatment of pediatric tumors, while discuss critical challenges that must be overcome before translation into clinical options, including the absence of compounds designed specifically for childhood tumors which often show differential mutational burdens, intrinsic and acquired resistance, lack of selectivity and adverse effects on a growing organism.
Collapse
Affiliation(s)
- Marina Ferreira Candido
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Mariana Medeiros
- Regional Blood Center, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Luciana Chain Veronez
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - David Bastos
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Karla Laissa Oliveira
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Julia Alejandra Pezuk
- Departament of Biotechnology and Innovation, Anhanguera University of São Paulo, UNIAN/SP, São Paulo 04119-001, SP, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - María Sol Brassesco
- Departament of Biotechnology and Innovation, Anhanguera University of São Paulo, UNIAN/SP, São Paulo 04119-001, SP, Brazil
- Correspondence: ; Tel.: +55-16-3315-9144; Fax: +55-16-3315-4886
| |
Collapse
|
35
|
Macrophage Repolarization as a Therapeutic Strategy for Osteosarcoma. Int J Mol Sci 2023; 24:ijms24032858. [PMID: 36769180 PMCID: PMC9917837 DOI: 10.3390/ijms24032858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Macrophages are versatile immune cells and can adapt to both external stimuli and their surrounding environment. Macrophages are categorized into two major categories; M1 macrophages release pro-inflammatory cytokines and produce protective responses that lead to antimicrobial or antitumor activity. M2 or tumor-associated macrophages (TAM) release anti-inflammatory cytokines that support tumor growth, invasion capacity, and metastatic potential. Since macrophages can be re-polarized from an M2 to an M1 phenotype with a variety of strategies, this has emerged as an innovative anti-cancer approach. Osteosarcoma (OS) is a kind of bone cancer and consists of a complex niche, and immunotherapy is not very effective. Therefore, immediate attention to new strategies is required. We incorporated the recent studies that have used M2-M1 repolarization strategies in the aspect of treating OS cancer.
Collapse
|
36
|
Zeng C, Zhong L, Liu W, Zhang Y, Yu X, Wang X, Zhang R, Kang T, Liao D. Targeting the Lysosomal Degradation of Rab22a-NeoF1 Fusion Protein for Osteosarcoma Lung Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205483. [PMID: 36529692 PMCID: PMC9929137 DOI: 10.1002/advs.202205483] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/03/2022] [Indexed: 06/17/2023]
Abstract
Rab22a-NeoF fusion protein has recently been reported as a promising target for osteosarcoma lung metastasis. However, how this fusion protein is regulated in cells remains unknown. Here, using multiple screenings, it is reported that Rab22a-NeoF1 fusion protein is degraded by an E3 ligase STUB1 via the autophagy receptor NDP52-mediated lysosome pathway, which is facilitated by PINK1 kinase. Mechanistically, STUB1 catalyzes the K63-linked ubiquitin chains on lysine112 of Rab22a-NeoF1, which is responsible for the binding of Rab22a-NeoF1 to NDP52, resulting in lysosomal degradation of Rab22a-NeoF1. PINK1 is able to phosphorylate Rab22a-NeoF1 at serine120, which promotes ubiquitination and degradation of Rab22a-NeoF1. Consistently, by upregulating PINK1, Sorafenib and Regorafenib can inhibit osteosarcoma lung metastasis induced by Rab22a-NeoF1. These findings reveal that the lysosomal degradation of Rab22a-NeoF1 fusion protein is targetable for osteosarcoma lung metastasis, proposing that Sorafenib and Regorafenib may benefit cancer patients who are positive for the RAB22A-NeoF1 fusion gene.
Collapse
Affiliation(s)
- Cuiling Zeng
- State Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Li Zhong
- Center of Digestive DiseasesThe Seventh Affiliated Hospital, Sun Yat‐sen UniversityShenzhen518107China
- Scientific Research CenterThe Seventh Affiliated Hospital, Sun Yat‐sen UniversityShenzhenChina
| | - Wenqiang Liu
- State Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
- Department of OncologyThe Fifth Affiliated Hospital, Sun Yat‐sen UniversityZhuhai519000China
| | - Yu Zhang
- State Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Xinhao Yu
- State Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Xin Wang
- State Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Ruhua Zhang
- State Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Tiebang Kang
- State Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Dan Liao
- State Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| |
Collapse
|
37
|
Kokkali S, Kyriazoglou A, Mangou E, Economopoulou P, Panousieris M, Psyrri A, Ardavanis A, Vassos N, Boukovinas I. Real-World Data on Cabozantinib in Advanced Osteosarcoma and Ewing Sarcoma Patients: A Study from the Hellenic Group of Sarcoma and Rare Cancers. J Clin Med 2023; 12:jcm12031119. [PMID: 36769769 PMCID: PMC9918141 DOI: 10.3390/jcm12031119] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/04/2023] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Advanced osteosarcomas (OSs) and Ewing sarcomas (ESs) tend to have poor prognosis with limited therapeutic options beyond first-line therapy. Aberrant angiogenesis and MET signaling play an important role in preclinical models. The anti-angiogenic drug cabozantinib was tested in a phase 2 trial of advanced OS and ES and was associated with clinical benefits. We retrospectively analyzed the off-label use of cabozantinib in adult patients with advanced OS and ES/primitive neuroectodermal tumors (PNETs) in three centers of the Hellenic Group of Sarcoma and Rare Cancers (HGSRC). Between April 2019 and January 2022, 16 patients started taking 60 mg of cabozantinib for advanced bone sarcoma or PNET. Median age at cabozantinib initiation was 31 years (17-83). All patients had received peri-operative chemotherapy for primary sarcoma and between 0 and 4 lines of treatment (median; 2.5) for advanced disease. The most common adverse effects included fatigue, anorexia, hypertransaminasemia, weight loss, and diarrhea. One toxic death was noted (cerebral hemorrhage). Dose reduction to 40 mg was required in 31.3% of the patients. No objective response was noted, and 9/16 patients exhibited stable disease outcomes. Progression-free survival varied from 1 to 8 (median; 5) months. Our study demonstrates that cabozantinib has antitumor activity in this population. In the real-life setting, we observed similar adverse events as in the CABONE study and in other neoplasms.
Collapse
Affiliation(s)
- Stefania Kokkali
- Department of Medical Oncology, Saint-Savvas Anticancer Hospital, 11522 Athens, Greece
- Medical Oncology Unit, Department of Internal Medicine, Hippocratio General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Correspondence: ; Tel.: +30-2132089511
| | - Anastasios Kyriazoglou
- Medical Oncology Unit, Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Elpida Mangou
- Department of Medical Oncology, Saint-Savvas Anticancer Hospital, 11522 Athens, Greece
| | - Panagiota Economopoulou
- Medical Oncology Unit, Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Michail Panousieris
- Department of Medical Oncology, Saint-Savvas Anticancer Hospital, 11522 Athens, Greece
| | - Amanda Psyrri
- Medical Oncology Unit, Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Alexandros Ardavanis
- Department of Medical Oncology, Saint-Savvas Anticancer Hospital, 11522 Athens, Greece
| | - Nikolaos Vassos
- Medical Faculty Mannheim, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Ioannis Boukovinas
- Department of Medical Oncology, Bioclinic Hospital, 54622 Thessaloniki, Greece
| |
Collapse
|
38
|
Tarone L, Mareschi K, Tirtei E, Giacobino D, Camerino M, Buracco P, Morello E, Cavallo F, Riccardo F. Improving Osteosarcoma Treatment: Comparative Oncology in Action. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122099. [PMID: 36556464 PMCID: PMC9783386 DOI: 10.3390/life12122099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Osteosarcoma (OSA) is the most common pediatric malignant bone tumor. Although surgery together with neoadjuvant/adjuvant chemotherapy has improved survival for localized OSA, most patients develop recurrent/metastatic disease with a dismally poor outcome. Therapeutic options have not improved for these OSA patients in recent decades. As OSA is a rare and "orphan" tumor, with no distinct targetable driver antigens, the development of new efficient therapies is still an unmet and challenging clinical need. Appropriate animal models are therefore critical for advancement in the field. Despite the undoubted relevance of pre-clinical mouse models in cancer research, they present some intrinsic limitations that may be responsible for the low translational success of novel therapies from the pre-clinical setting to the clinic. From this context emerges the concept of comparative oncology, which has spurred the study of pet dogs as a uniquely valuable model of spontaneous OSA that develops in an immune-competent system with high biological and clinical similarities to corresponding human tumors, including in its metastatic behavior and resistance to conventional therapies. For these reasons, the translational power of studies conducted on OSA-bearing dogs has seen increasing recognition. The most recent and relevant veterinary investigations of novel combinatorial approaches, with a focus on immune-based strategies, that can most likely benefit both canine and human OSA patients have been summarized in this commentary.
Collapse
Affiliation(s)
- Lidia Tarone
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Katia Mareschi
- Department of Public Health and Paediatrics, University of Torino, Piazza Polonia 94, 10126 Torino, Italy
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Department, Regina Margherita Children’s Hospital, City of Health and Science of Torino, 10126 Torino, Italy
| | - Elisa Tirtei
- Department of Public Health and Paediatrics, University of Torino, Piazza Polonia 94, 10126 Torino, Italy
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Department, Regina Margherita Children’s Hospital, City of Health and Science of Torino, 10126 Torino, Italy
| | - Davide Giacobino
- Department of Veterinary Sciences, University of Torino, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Mariateresa Camerino
- Department of Veterinary Sciences, University of Torino, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Torino, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Emanuela Morello
- Department of Veterinary Sciences, University of Torino, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Federica Cavallo
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
- Correspondence: (F.C.); (F.R.)
| | - Federica Riccardo
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
- Correspondence: (F.C.); (F.R.)
| |
Collapse
|
39
|
Vasella M, Gousopoulos E, Guidi M, Storti G, Song SY, Grieb G, Pauli C, Lindenblatt N, Giovanoli P, Kim BS. Targeted therapies and checkpoint inhibitors in sarcoma. QJM 2022; 115:793-805. [PMID: 33486519 DOI: 10.1093/qjmed/hcab014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
Sarcomas are defined as a group of mesenchymal malignancies with over 100 heterogeneous subtypes. As a rare and difficult to diagnose entity, micrometastasis is already present at the time of diagnosis in many cases. Current treatment practice of sarcomas consists mainly of surgery, (neo)adjuvant chemo- and/or radiotherapy. Although the past decade has shown that particular genetic abnormalities can promote the development of sarcomas, such as translocations, gain-of-function mutations, amplifications or tumor suppressor gene losses, these insights have not led to established alternative treatment strategies so far. Novel therapeutic concepts with immunotherapy at its forefront have experienced some remarkable success in different solid tumors while their impact in sarcoma remains limited. In this review, the most common immunotherapy strategies in sarcomas, such as immune checkpoint inhibitors, targeted therapy and cytokine therapy are concisely discussed. The programmed cell death (PD)-1/PD-1L axis and apoptosis-inducing cytokines, such as TNF-related apoptosis-inducing ligand (TRAIL), have not yielded the same success like in other solid tumors. However, in certain sarcoma subtypes, e.g. liposarcoma or undifferentiated pleomorphic sarcoma, encouraging results in some cases when employing immune checkpoint inhibitors in combination with other treatment options were found. Moreover, newer strategies such as the targeted therapy against the ancient cytokine macrophage migration inhibitory factor (MIF) may represent an interesting approach worth investigation in the future.
Collapse
Affiliation(s)
- M Vasella
- From the Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - E Gousopoulos
- From the Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - M Guidi
- From the Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - G Storti
- Department of Surgical Sciences, Plastic and Reconstructive Surgery, University of Rome-'Tor Vergata', Via Montepellier, 1, 00133 Rome, Italy
| | - S Y Song
- Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, Korea
| | - G Grieb
- Department of Plastic Surgery and Hand Surgery, Gemeinschaftskrankenhaus Havelhoehe, Kladower Damm 221, 14089 Berlin, Germany
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - C Pauli
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - N Lindenblatt
- From the Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - P Giovanoli
- From the Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - B-S Kim
- From the Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
40
|
Chen C, Shi Q, Xu J, Ren T, Huang Y, Guo W. Current progress and open challenges for applying tyrosine kinase inhibitors in osteosarcoma. Cell Death Dis 2022; 8:488. [PMID: 36509754 PMCID: PMC9744866 DOI: 10.1038/s41420-022-01252-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/06/2022] [Accepted: 11/10/2022] [Indexed: 12/15/2022]
Abstract
Osteosarcoma (OS) is a mesenchymal-origin tumor that constitutes the most common primary malignant bone tumor. The survival rate of the patients has significantly improved since the introduction of neoadjuvant chemotherapy and extensive resection, but it has stagnated in recent 40 years. Tyrosine kinase inhibitors (TKIs) have played a key part in the treatment of malignant tumors. In advanced OS, TKIs including anlotinib, apatinib, sorafenib, etc. have significantly improved the progression-free survival of patients, while the overall survival remains unchanged. The main reason is the rapid and inevitable progress of acquired drug resistance of OS. However, as the application of TKIs in OS and other tumors is still in the exploratory phase, its drug resistance mechanism and corresponding solutions are rarely reported. Hence, in this review, we summarize knowledge of the applications of TKIs, the mechanism of TKIs resistance, and the attempts to overcome TKIs resistance in OS, which are the three potentially novel insights of TKIs in OS. Because most evidence is derived from studies using animal and cell models, we also reviewed clinical trials and related bioinformatics data available in public databases, which partially improved our understanding of TKIs applications.
Collapse
Affiliation(s)
- Chenglong Chen
- grid.414360.40000 0004 0605 7104Department of Orthopedics, Beijing Jishuitan Hospital, Beijing, People’s Republic of China ,grid.411634.50000 0004 0632 4559Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Qianyu Shi
- grid.411634.50000 0004 0632 4559Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, People’s Republic of China ,grid.411634.50000 0004 0632 4559Musculoskeletal Tumor Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Jiuhui Xu
- grid.411634.50000 0004 0632 4559Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, People’s Republic of China ,grid.411634.50000 0004 0632 4559Musculoskeletal Tumor Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Tingting Ren
- grid.411634.50000 0004 0632 4559Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, People’s Republic of China ,grid.411634.50000 0004 0632 4559Musculoskeletal Tumor Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Yi Huang
- grid.411634.50000 0004 0632 4559Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, People’s Republic of China ,grid.411634.50000 0004 0632 4559Musculoskeletal Tumor Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Wei Guo
- grid.411634.50000 0004 0632 4559Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, People’s Republic of China ,grid.411634.50000 0004 0632 4559Musculoskeletal Tumor Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| |
Collapse
|
41
|
Beird HC, Bielack SS, Flanagan AM, Gill J, Heymann D, Janeway KA, Livingston JA, Roberts RD, Strauss SJ, Gorlick R. Osteosarcoma. Nat Rev Dis Primers 2022; 8:77. [PMID: 36481668 DOI: 10.1038/s41572-022-00409-y] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2022] [Indexed: 12/13/2022]
Abstract
Osteosarcoma is the most common primary malignant tumour of the bone. Osteosarcoma incidence is bimodal, peaking at 18 and 60 years of age, and is slightly more common in males. The key pathophysiological mechanism involves several possible genetic drivers of disease linked to bone formation, causing malignant progression and metastasis. While there have been significant improvements in the outcome of patients with localized disease, with event-free survival outcomes exceeding 60%, in patients with metastatic disease, event-free survival outcomes remain poor at less than 30%. The suspicion of osteosarcoma based on radiographs still requires pathological evaluation of a bone biopsy specimen for definitive diagnosis and CT imaging of the chest should be performed to identify lung nodules. So far, population-based screening and surveillance strategies have not been implemented due to the rarity of osteosarcoma and the lack of reliable markers. Current screening focuses only on groups at high risk such as patients with genetic cancer predisposition syndromes. Management of osteosarcoma requires a multidisciplinary team of paediatric and medical oncologists, orthopaedic and general surgeons, pathologists, radiologists and specialist nurses. Survivors of osteosarcoma require specialized medical follow-up, as curative treatment consisting of chemotherapy and surgery has long-term adverse effects, which also affect the quality of life of patients. The development of osteosarcoma model systems and related research as well as the evaluation of new treatment approaches are ongoing to improve disease outcomes, especially for patients with metastases.
Collapse
Affiliation(s)
- Hannah C Beird
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stefan S Bielack
- Pediatric Oncology, Hematology, Immunology, Klinikum Stuttgart - Olgahospital, Stuttgart Cancer Center, Stuttgart, Germany
| | - Adrienne M Flanagan
- Research Department of Pathology, Cancer Institute, University College London, London, UK
| | - Jonathan Gill
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dominique Heymann
- Nantes Université, CNRS, UMR6286, US2B, Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Katherine A Janeway
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - J Andrew Livingston
- Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ryan D Roberts
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Sandra J Strauss
- University College London Hospitals NHS Foundation Trust, University College London, London, UK
| | - Richard Gorlick
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA. .,Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
42
|
Mittal A, Pushpam D, Ganguly S, Kumar VS, Khan SA, Bakhshi S. Controversies and Challenges in the Management of Osteosarcoma-an Indian Perspective. Indian J Surg Oncol 2022; 13:939-955. [PMID: 36687236 PMCID: PMC9845467 DOI: 10.1007/s13193-021-01486-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 11/30/2021] [Indexed: 01/25/2023] Open
Abstract
Osteosarcoma (OGS) is the most common primary bone tumor in children and adolescents which requires a multidisciplinary approach to management. Although chemotherapy and surgery can cure more than half of localized OGS cases, the unique challenges faced by resource-limited countries like India make this outcome difficult to achieve. Various questions in the management of OGS including role of high-dose methotrexate (HDMTX) in neoadjuvant setting, triplet vs doublet chemotherapy, intensification of chemotherapy based on response in setting of doublet, and indigenous prosthesis in setting of limb salvage need to be defined. Similarly, in the metastatic and recurrent setting, questions regarding intent of treatment, indications of chemotherapy, timing of surgery, and role of targeted therapies need clarification. Lack of randomized trials from India makes definite conclusions difficult, but an attempt can be made to define the best approach in the Indian scenario from available evidence. Hence, a critical review of literature from India and the West was done to define possible management approaches and highlight the lacuna for future research.
Collapse
Affiliation(s)
- Abhenil Mittal
- Department of Medical Oncology, DR BRAIRCH, AIIMS, New Delhi, 110029 India
| | - Deepam Pushpam
- Department of Medical Oncology, DR BRAIRCH, AIIMS, New Delhi, 110029 India
| | - Shuvadeep Ganguly
- Department of Medical Oncology, DR BRAIRCH, AIIMS, New Delhi, 110029 India
| | | | - Shah Alam Khan
- Department of Orthopedics, AIIMS, New Delhi, 110029 India
| | - Sameer Bakhshi
- Department of Medical Oncology, DR BRAIRCH, AIIMS, New Delhi, 110029 India
| |
Collapse
|
43
|
Yan Q, Yang J, Yao Y, Jia Z, Wang Y, Cheng M, Yan X, Xu Y. Research of the Active Components and Potential Mechanisms of Qingfei Gujin Decoction in the Treatment of Osteosarcoma Based on Network Pharmacology and Molecular Docking Technology. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7994425. [PMID: 36466554 PMCID: PMC9713469 DOI: 10.1155/2022/7994425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/09/2022] [Accepted: 11/03/2022] [Indexed: 09/10/2024]
Abstract
Aim Qingfei Gujin Decoction (QGD) has been shown to be effective against osteosarcoma. This research was aimed at investigating the main active ingredients and potential mechanisms of QGD acting on osteosarcoma through network pharmacology and molecular docking techniques. Methods The active ingredients and targets of QGD were screened from the TCMSP database, and the predicted targets were obtained from the PharmMapper database. Meanwhile, the targets of osteosarcoma were collected using OMIM, PharmGKB, and DisGeNET databases. Then, GO and KEGG enrichment analyses were performed by RStudio. PPI and drug-ingredient-target networks were constructed using Cytoscape 3.2.1 to screen the major active ingredients, key networks, and targets. Finally, molecular docking of key genes and their regulatory active ingredients was performed using AutoDockTools 1.5.6 software. Results 38 active ingredients were collected, generating 89 cross-targets; quercetin, luteolin, β-sitosterol, and kaempferol were the main active ingredients of QGD acting on osteosarcoma, and major signaling pathways such as PI3K-Akt signaling pathway, MAPK signaling pathway, and IL-17 signaling pathway were observed. TP53, SRC, and ESR1 were identified as key proteins that docked well with their regulated compounds. Conclusion QGD is effective against osteosarcoma through multicomponent, multitarget, and multipathway. This study was helpful for finding effective targets and compounds for osteosarcoma treatment.
Collapse
Affiliation(s)
- Qingying Yan
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
- Department of Oncology, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiewen Yang
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
| | - Yongwei Yao
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
| | - Zhen Jia
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
| | - Yiqing Wang
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
| | - Miao Cheng
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
| | - Xiaobo Yan
- Department of Orthopedics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yefeng Xu
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
| |
Collapse
|
44
|
Albarrán V, Villamayor ML, Chamorro J, Rosero DI, Pozas J, San Román M, Calvo JC, Pérez de Aguado P, Moreno J, Guerrero P, González C, García de Quevedo C, Álvarez-Ballesteros P, Vaz MÁ. Receptor Tyrosine Kinase Inhibitors for the Treatment of Recurrent and Unresectable Bone Sarcomas. Int J Mol Sci 2022; 23:13784. [PMID: 36430263 PMCID: PMC9697271 DOI: 10.3390/ijms232213784] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Bone sarcomas are a heterogeneous group of rare tumors with a predominance in the young population. Few options of systemic treatment are available once they become unresectable and resistant to conventional chemotherapy. A better knowledge of the key role that tyrosine kinase receptors (VEGFR, RET, MET, AXL, PDGFR, KIT, FGFR, IGF-1R) may play in the pathogenesis of these tumors has led to the development of multi-target inhibitors (TKIs) that are progressively being incorporated into our therapeutic arsenal. Osteosarcoma (OS) is the most frequent primary bone tumor and several TKIs have demonstrated clinical benefit in phase II clinical trials (cabozantinib, regorafenib, apatinib, sorafenib, and lenvatinib). Although the development of TKIs for other primary bone tumors is less advanced, preclinical data and early trials have begun to show their potential benefit in advanced Ewing sarcoma (ES) and rarer bone tumors (chondrosarcoma, chordoma, giant cell tumor of bone, and undifferentiated pleomorphic sarcoma). Previous reviews have mainly provided information on TKIs for OS and ES. We aim to summarize the existing knowledge regarding the use of TKIs in all bone sarcomas including the most recent studies as well as the potential synergistic effects of their combination with other systemic therapies.
Collapse
Affiliation(s)
- Víctor Albarrán
- Department of Medical Oncology, Ramon y Cajal University Hospital, 28034 Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Sorafenib inhibits doxorubicin-induced PD-L1 upregulation to improve immunosuppressive microenvironment in Osteosarcoma. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04458-4. [DOI: 10.1007/s00432-022-04458-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022]
|
46
|
Blay JY, Duffaud F, George S, Maki RG, Penel N. Regorafenib for the Treatment of Sarcoma. Curr Treat Options Oncol 2022; 23:1477-1502. [PMID: 36178573 DOI: 10.1007/s11864-022-00990-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2022] [Indexed: 01/30/2023]
Abstract
OPINION STATEMENT Sarcomas are a rare group of tumors with many subtypes, conventionally classified into soft-tissue sarcomas and bone sarcomas. Chemotherapeutic regimens form the mainstay of systemic therapy but are not well defined beyond the first-line setting and clinical outcomes are variable. Tyrosine kinase inhibitors (TKIs), with a broad inhibition profile which have been shown to target tumor angiogenesis, have an established role in the treatment of sarcomas without characteristic driver alterations. One such TKI, regorafenib, has been evaluated in sarcomas and clinical data are discussed in this review. An overview of regorafenib data from five phase 2 and one phase 1b clinical trials in over 10 sarcoma subtypes (both soft-tissue and bone) in adult and pediatric patients is reviewed. Regorafenib demonstrated clinical benefit in patients with non-adipocytic soft-tissue sarcomas, osteosarcoma and Ewing sarcoma who had progressed on prior therapy. Patients with otherwise limited treatment options may therefore benefit from regorafenib therapy.
Collapse
Affiliation(s)
- Jean-Yves Blay
- Department of Medicine, Léon Bérard Center, Lyon, France.
| | - Florence Duffaud
- Medical Oncology Unit, La Timone University Hospital, Marseille, France.,Aix Marseille University (AMU), Marseille, France
| | - Suzanne George
- Dana-Farber Cancer Institute, Harvard Medical School, Cambridge, MA, USA
| | - Robert G Maki
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolas Penel
- Medical Oncology Department, Oscar Lambret Cancer Center and Lille University, Lille, France
| |
Collapse
|
47
|
Gong T, Huang Q, Tang F, Wang Y, Li Z, Luo Y, Min L, Zhou Y, Tu C. Activity and safety of apatinib monotherapy or apatinib combined with chemotherapy for patients with metastatic or unresectable osteosarcoma over the age of 40 years: A retrospective analysis. Front Oncol 2022; 12:1031787. [PMID: 36387068 PMCID: PMC9664205 DOI: 10.3389/fonc.2022.1031787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/17/2022] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Osteosarcoma commonly develops during childhood and adolescence. Only one-third of osteosarcoma patients have been clinically detected over the age of 40 years, and the survivorship of those patients is quite dismal. Apatinib, a novel multitarget angiogenesis inhibitor, has shown a short-term efficacy in advanced or metastatic osteosarcoma. However, the data for apatinib in the older patients with osteosarcoma are limited. We aim to evaluate the efficacy and safety of apatinib combined with chemotherapy versus apatinib monotherapy in the treatment of patients over 40 years old with metastatic or unresectable osteosarcoma. METHODS We retrospectively analyzed the patients with metastatic osteosarcoma who were treated with apatinib monotherapy or apatinib combined with chemotherapy between May 2015 and December 2018 in the Department of Orthopedics at West China Hospital. Apatinib was initially administered with a dose of 500 mg daily, and the dose was adjusted according to toxicity. The objective response rate (ORR), disease control rate (DCR), duration of response (DOR), progression-free survival (PFS), and overall survival (OS) were investigated. The treatment-related adverse events and the safety of apatinib were also documented. RESULTS A total of 45 patients (28 men, 17 women) with metastatic or unresectable osteosarcoma were finally included, and 41 patients received at least one cycle of treatment and were evaluable for efficacy. Of 41 patients, 24 who were intolerant to intensive chemotherapy or have failed standard chemotherapy received apatinib monotherapy, and 17 patients were treated with apatinib plus chemotherapy. The median PFS and median OS were longer in the group treated with apatinib combined with chemotherapy than those of the apatinib monotherapy group (5.6 months vs. 2.6 months; 15.1 months vs. 9.7 months). Moreover, the median DOR was significantly prolonged in the group treated with apatinib combined with chemotherapy compared with that in the monotherapy group. CONCLUSION Apatinib demonstrated promising activity in patients over 40 years old with metastatic or unresectable osteosarcoma. The combination of apatinib and chemotherapy conferred a durable response compared with apatinib monotherapy, which might be an alternative therapeutic strategy for the management of osteosarcoma in older patients.
Collapse
Affiliation(s)
- Taojun Gong
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qi Huang
- Operating Room, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Fan Tang
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yitian Wang
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhuangzhuang Li
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Luo
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Min
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yong Zhou
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chongqi Tu
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
48
|
Yu L, Zhang J, Li Y. Effects of microenvironment in osteosarcoma on chemoresistance and the promise of immunotherapy as an osteosarcoma therapeutic modality. Front Immunol 2022; 13:871076. [PMID: 36311748 PMCID: PMC9608329 DOI: 10.3389/fimmu.2022.871076] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 09/28/2022] [Indexed: 12/02/2022] Open
Abstract
Osteosarcoma (OS) is one of the most common primary malignant tumors originating in bones. Its high malignancy typically manifests in lung metastasis leading to high mortality. Although remarkable advances in surgical resection and neoadjuvant chemotherapy have lengthened life expectancy and greatly improved the survival rate among OS patients, no further breakthroughs have been achieved. It is challenging to treat patients with chemoresistant tumors and distant metastases. Recent studies have identified a compelling set of links between hypoxia and chemotherapy failure. Here, we review the evidence supporting the positive effects of hypoxia in the tumor microenvironment (TME). In addition, certain anticancer effects of immune checkpoint inhibitors have been demonstrated in OS preclinical models. Continued long-term observation in clinical trials is required. In the present review, we discuss the mutualistic effects of the TME in OS treatment and summarize the mechanisms of immunotherapy and their interaction with TME when used to treat OS. We also suggest that immunotherapy, a new comprehensive and potential antitumor approach that stimulates an immune response to eliminate tumor cells, may represent an innovative approach for the development of a novel treatment regimen for OS patients.
Collapse
|
49
|
Wang T, Lin F, Huang Y, Qian G, Yu W, Hu H, Ji T, Tang L, Yao Y. The Combination of Anlotinib and Gemcitabine/Docetaxel in Patients with Metastatic Osteosarcoma Who Have Failed Standard Chemotherapy. Cancer Manag Res 2022; 14:2945-2952. [PMID: 36217441 PMCID: PMC9547547 DOI: 10.2147/cmar.s378264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose The options for the second-line treatment of metastatic osteosarcoma are still limited. Anlotinib is a multi-kinase inhibitor which has shown promising efficacy and good tolerability in various cancer types. This retrospective study was conducted to evaluate the efficacy and safety of anlotinib combined with gemcitabine/docetaxel (GD) in patients with metastatic osteosarcoma who have failed first-line chemotherapy. Patients and Methods The data of patients who received anlotinib combined with GD or GD were collected. The primary endpoint was progression-free survival. Secondary endpoints included objective response rate and safety. Results From July 2013 to November 2020, a total of 32 patients were enrolled, 13 received anlotinib combined with GD and 19 received GD. Median PFS was 9.0 months (95% CI 6.7-39.1) in the combination group and 5.0 months (95% CI 1.2-6.7) in the chemotherapy group. ORR were 38.4% and 15.8%, DCR were 69.2% and 38.1% in the combination and chemotherapy group, respectively. The most common adverse events included fatigue (78.9% in the combination group vs 69.2% in the chemotherapy group), hypertension (46.2% vs 10.5%), diarrhea (38.5% vs 21.1%), hypothyroidism (38.5% vs 15.8%), neutropenia (23.1% vs 36.8%) and AST elevation (30.8% vs 21.1%). The most common grade 3 or worse adverse events included hand-foot reaction (7.7% vs 5.3%), hypothyroidism (15.4% vs 0), neutropenia (0 vs 10.5%). Conclusion The combination of anlotinib and GD showed favorable efficacy with manageable toxicities compared with GD in the second-line treatment for metastatic osteosarcoma. This combination therapy deserves further investigations in patients with osteosarcoma.
Collapse
Affiliation(s)
- Tian Wang
- The Eighth People’s Hospital of Shanghai, Shanghai, People’s Republic of China
| | - Feng Lin
- Shanghai Sixth People’s Hospital, Shanghai Jiaotong University, Shanghai, People’s Republic of China
| | - Yujing Huang
- Shanghai Sixth People’s Hospital, Shanghai Jiaotong University, Shanghai, People’s Republic of China
| | - Guowei Qian
- Shanghai Sixth People’s Hospital, Shanghai Jiaotong University, Shanghai, People’s Republic of China
| | - Wenxi Yu
- Shanghai Sixth People’s Hospital, Shanghai Jiaotong University, Shanghai, People’s Republic of China
| | - Haiyan Hu
- Shanghai Sixth People’s Hospital, Shanghai Jiaotong University, Shanghai, People’s Republic of China
| | - Tong Ji
- Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Lina Tang
- Shanghai Sixth People’s Hospital, Shanghai Jiaotong University, Shanghai, People’s Republic of China,Correspondence: Yang Yao; Lina Tang, Shanghai Sixth People’s Hospital, Shanghai Jiaotong University, No. 600 Yishan Road, Xuhui Distinct, Shanghai, 200233, People’s Republic of China, Tel +86 2164369181; +86 2164701361, Email ;
| | - Yang Yao
- Shanghai Sixth People’s Hospital, Shanghai Jiaotong University, Shanghai, People’s Republic of China
| |
Collapse
|
50
|
Bielack SS. Systemic treatment for primary malignant sarcomas arising in craniofacial bones. Front Oncol 2022; 12:966073. [PMID: 36158667 PMCID: PMC9492845 DOI: 10.3389/fonc.2022.966073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/22/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction Craniofacial bones may be the site of origin of various sarcomas. We review the various malignancies affecting this region of the body and attempt to put systemic treatment approaches into perspective. Material and methods Non-systematic literature review Results Conventional types of osteosarcoma, Ewing sarcoma, and chondrosarcoma are the most frequent bone sarcomas occurring in craniofacial region, but variants may occur. The tumors’ biologies and the resulting treatment strategies vary distinctly. As a general rule, local control remains paramount regardless of histology. The efficacy of antineoplastic chemotherapy varies by type of malignancy. It is clearly indicated in Ewing sarcoma and related tumors, potentially of benefit in high-grade osteosarcoma, undifferentiated pleomorphic sarcoma, dedifferentiated and mesenchymal chondrosarcoma, and of no proven benefit in the others. Conclusions Various histologies demand various and distinct treatment approaches, with local control remaining paramount in all. The efficacy of systemic treatments varies by type of tumor. Prospective trials would help in all of these to better define systemic treatment strategies.
Collapse
Affiliation(s)
- Stefan S. Bielack
- Pediatrics 5 (Oncology, Hematology, Immunology), Center for Pediatric, Adolescent and Women’s Medicine, Stuttgart Cancer Center, Klinikum Stuttgart–Olgahospital, Stuttgart, Germany
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Muenster, Muenster, Germany
- *Correspondence: Stefan S. Bielack,
| |
Collapse
|