8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Optimal Clinical Management and the Molecular Biology of Angiosarcomas

      review-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Simple Summary

          Angiosarcomas are a group of a rare vascular cancers transformed from endothelial cells that could occur in any body part. Most angiosarcomas have unknown aetiology but secondary angiosarcomas could occur after radiation exposure or chronic lymphedema. The optimal treatment for localized angiosarcoma is complete surgical resection but neoadjuvant therapy may be helpful for some patients. For advanced angiosarcoma, systemic treatment including chemotherapy, anti-angiogenic therapy, and immunotherapies are options. We also review the molecular alterations that are associated with the pathogenesis of angiosarcoma, including c-Myc, TP53, vascular endothelial growth factor receptors, and others. This clinical management and molecular biology review will provide both clinicians and researchers of angiosarcoma with insights to help patients and move the field forward.

          Abstract

          Angiosarcomas comprise less than 3% of all soft tissue sarcomas but have a poor prognosis. Most angiosarcomas occur without obvious risk factors but secondary angiosarcoma could arise after radiotherapy or chronic lymphedema. Surgery remains the standard treatment for localized angiosarcoma but neoadjuvant systemic treatment may improve the curability. For advanced angiosarcoma, anthracyclines and taxanes are the main chemotherapy options. Anti-angiogenic agents have a substantial role but the failure of a randomized phase 3 trial of pazopanib with or without an anti-endoglin antibody brings a challenge to future trials in angiosarcomas. Immune checkpoint inhibitors as single agents or in combination with oncolytic virus may play an important role but the optimal duration remains to be investigated. We also report the current understanding of the molecular pathways involved in angiosarcoma pathogenesis including MYC amplification, activation of angiogenic pathways and different molecular alterations that are associated with angiosarcomas of different aetiology. The success of the patient-partnered Angiosarcoma Project (ASCProject) has provided not only detailed insights into the molecular features of angiosarcomas of different origins but also offers a template for future fruitful collaborations between patients, physicians, and researchers. Lastly, we provide our perspective of future developments in optimizing the clinical management of angiosarcomas.

          Related collections

          Most cited references128

          • Record: found
          • Abstract: found
          • Article: not found

          Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer.

          Immune checkpoint inhibitors, which unleash a patient's own T cells to kill tumors, are revolutionizing cancer treatment. To unravel the genomic determinants of response to this therapy, we used whole-exome sequencing of non-small cell lung cancers treated with pembrolizumab, an antibody targeting programmed cell death-1 (PD-1). In two independent cohorts, higher nonsynonymous mutation burden in tumors was associated with improved objective response, durable clinical benefit, and progression-free survival. Efficacy also correlated with the molecular smoking signature, higher neoantigen burden, and DNA repair pathway mutations; each factor was also associated with mutation burden. In one responder, neoantigen-specific CD8+ T cell responses paralleled tumor regression, suggesting that anti-PD-1 therapy enhances neoantigen-specific T cell reactivity. Our results suggest that the genomic landscape of lung cancers shapes response to anti-PD-1 therapy. Copyright © 2015, American Association for the Advancement of Science.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Tumor mutational load predicts survival after immunotherapy across multiple cancer types

            Immune checkpoint inhibitor (ICI) treatments benefit some patients with metastatic cancers, but predictive biomarkers are needed. Findings in select cancer types suggest that tumor mutational burden (TMB) may predict clinical response to ICI.To examine this association more broadly, we analyzed the clinical and genomic data of 1662 advanced cancer patients treated with ICI, and 5371 non-ICI treated patients, whose tumors underwent targeted next-generation sequencing (MSK-IMPACT). Among all patients, higher somatic TMB (highest 20% in each histology) was associated with better OS (HR 0.52; p=1.6 ×10 −6 ). For most cancer histologies, an association between higher TMB and improved survival was observed. The TMB cutpoints associated with improved survival varied markedly between cancer types. These data indicate that TMB is associated with improved survival in patients receiving ICI across a wide variety of cancer types, but that there may not be one universal definition of high TMB.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Alpelisib for PIK3CA-Mutated, Hormone Receptor–Positive Advanced Breast Cancer

              PIK3CA mutations occur in approximately 40% of patients with hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer. The PI3Kα-specific inhibitor alpelisib has shown antitumor activity in early studies.
                Bookmark

                Author and article information

                Journal
                Cancers (Basel)
                Cancers (Basel)
                cancers
                Cancers
                MDPI
                2072-6694
                10 November 2020
                November 2020
                : 12
                : 11
                : 3321
                Affiliations
                [1 ]Department of Oncology, National Taiwan University Hospital and Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 100, Taiwan
                [2 ]Division of Molecular Pathology, The Institute of Cancer Research, London SW3 6JB, UK; jess.burns@ 123456icr.ac.uk
                [3 ]Sarcoma Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London SW3 6JJ, UK; Robin.Jones@ 123456rmh.nhs.uk
                Author notes
                [* ]Correspondence: tomwchen@ 123456ntuh.gov.tw (T.W.-W.C.); Paul.Huang@ 123456icr.ac.uk (P.H.H.)
                Author information
                https://orcid.org/0000-0003-4112-4029
                https://orcid.org/0000-0002-3508-721X
                Article
                cancers-12-03321
                10.3390/cancers12113321
                7696056
                33182685
                0c91fbe1-ef89-47d7-ac7f-199e56bb9eec
                © 2020 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 28 September 2020
                : 02 November 2020
                Categories
                Review

                angiosarcoma,angiogenesis,chemotherapy,immunotherapy,molecular biology,radiation-associated sarcoma,soft tissue sarcomas

                Comments

                Comment on this article