1
|
Sunbul FS, Almuqbil RM, Zhang H, Alhudaithi SS, Fernandez ME, Aldaqqa RR, Garcia VA, Robila V, Halquist MS, Gordon SW, Bos PD, da Rocha SRP. An improved experimental model of osteosarcoma lung metastases to investigate innovative therapeutic interventions and sex as a biological variable. Int J Pharm 2025; 673:125372. [PMID: 39971171 DOI: 10.1016/j.ijpharm.2025.125372] [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: 11/15/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy, with OS lung metastasis (OSLM) being the leading cause of death in OS patients. No curative pharmacotherapies for OSLM are available, highlighting the clinical need for new therapies. Improved and rigorous preclinical models of OSLM are key in supporting advancements in this field. We aimed to develop an immunocompetent mouse model of OSLM that allows monitoring pharmacotherapies' effect on the lung metastatic burden over time and assessing the impact of sex as a biological variable in tumor growth and response to therapy. We transformed K7M2 cells to express bioluminescence and fluorescence, enabling real-time tracking of OSLM in BALB/c mice following tail vein injection. Metastasis was confined to the lungs and exhibited exponential growth with typical downregulated Fas receptor expression. In vivo bioluminescence correlated strongly with ex vivo, suggesting its reliability for evaluating metastatic progression and therapy response. Fluorescence from tdT was stable upon tissue processing, providing unique opportunities to probe the tumor characteristics ex vivo. We also assessed the effect of local lung-delivered gemcitabine, which was well-tolerated and significantly reduced OSLM burden without causing pulmonary toxicity. However, treatment did not resolve metastatic disease. We also explored the effect of sex on tumor growth and response to therapy; while no difference was observed in tumor growth between male and female mice, females showed a better response to local gemcitabine administration. In sum, we established a robust and rigorous immunocompetent mouse model of OSLM that will facilitate exploring new pharmacotherapies for OSLM.
Collapse
Affiliation(s)
- Fatemah S Sunbul
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Rashed M Almuqbil
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Hanming Zhang
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Sulaiman S Alhudaithi
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Matthew E Fernandez
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Raneem R Aldaqqa
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Victoria A Garcia
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Valentina Robila
- Department of Pathology - School of Medicine, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Matthew S Halquist
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Sarah W Gordon
- Department of Medical Oncology, Sidney Kimmel Comprehensive Cancer Center, Thomas JeffersonUniversity, Philadelphia, PA, the United States of America
| | - Paula D Bos
- Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Department of Pathology - School of Medicine, Virginia Commonwealth University, Richmond, VA, the United States of America; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Sandro R P da Rocha
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, the United States of America.
| |
Collapse
|
2
|
Smith HL, Beers SA, Kanczler JM, Gray JC. Developing a 3D bone model of osteosarcoma to investigate cancer mechanisms and evaluate treatments. FASEB J 2024; 38:e70274. [PMID: 39724514 DOI: 10.1096/fj.202402011r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/27/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Osteosarcoma is the most common primary bone cancer, occurring frequently in children and young adults. Patients are treated with surgery and multi-agent chemotherapy, and despite the introduction of mifamurtide in 2011, there has been little improvement in survival for decades. 3-dimensional models offer the potential to understand the complexity of the osteosarcoma tumor microenvironment and aid in developing new treatment approaches. An osteosarcoma 3D bone core model was developed using human trabecular bone and the chorioallantoic membrane (CAM), to form a functioning vasculature. A tri-culture of cells, stromal cells, macrophages, and the Saos-2 osteosarcoma cell line, were implanted into this model to simulate components of the tumor microenvironment, and mifamurtide was tested in this context. Immunohistochemistry and micro-CT were performed to assess phenotypic and structural effects of implantation. Successful integration and angiogenesis of the bone cores were observed after incubation on the CAM. The 3D bone model also showed similar characteristics to osteosarcoma patient samples including CD68 and CD105 expression. Incubating bone cores with mifamurtide induced a reduction of cellular markers and an increase in bone volume. This 3D bone core model has the potential to investigate osteosarcoma tumor microenvironment and provides a representative model for evaluation of novel therapies.
Collapse
Affiliation(s)
- Hannah L Smith
- Antibody and Vaccine Group, Faculty of Medicine, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton, Southampton, UK
- Bone and Joint Research Group, Human Development and Health, Faculty of Medicine, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Stephen A Beers
- Antibody and Vaccine Group, Faculty of Medicine, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Janos M Kanczler
- Bone and Joint Research Group, Human Development and Health, Faculty of Medicine, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Juliet C Gray
- Antibody and Vaccine Group, Faculty of Medicine, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
3
|
Giusti V, Miserocchi G, Sbanchi G, Pannella M, Hattinger CM, Cesari M, Fantoni L, Guerrieri AN, Bellotti C, De Vita A, Spadazzi C, Donati DM, Torsello M, Lucarelli E, Ibrahim T, Mercatali L. Xenografting Human Musculoskeletal Sarcomas in Mice, Chick Embryo, and Zebrafish: How to Boost Translational Research. Biomedicines 2024; 12:1921. [PMID: 39200384 PMCID: PMC11352184 DOI: 10.3390/biomedicines12081921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Musculoskeletal sarcomas pose major challenges to researchers and clinicians due to their rarity and heterogeneity. Xenografting human cells or tumor fragments in rodents is a mainstay for the generation of cancer models and for the preclinical trial of novel drugs. Lately, though, technical, intrinsic and ethical concerns together with stricter regulations have significantly curbed the employment of murine patient-derived xenografts (mPDX). In alternatives to murine PDXs, researchers have focused on embryonal systems such as chorioallantoic membrane (CAM) and zebrafish embryos. These systems are time- and cost-effective hosts for tumor fragments and near-patient cells. The CAM of the chick embryo represents a unique vascularized environment to host xenografts with high engraftment rates, allowing for ease of visualization and molecular detection of metastatic cells. Thanks to the transparency of the larvae, zebrafish allow for the tracking of tumor development and metastatization, enabling high-throughput drug screening. This review will focus on xenograft models of musculoskeletal sarcomas to highlight the intrinsic and technically distinctive features of the different hosts, and how they can be exploited to elucidate biological mechanisms beneath the different phases of the tumor's natural history and in drug development. Ultimately, the review suggests the combination of different models as an advantageous approach to boost basic and translational research.
Collapse
Affiliation(s)
- Veronica Giusti
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Giacomo Miserocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (G.M.); (A.D.V.); (C.S.)
| | - Giulia Sbanchi
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Micaela Pannella
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Claudia Maria Hattinger
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Marilena Cesari
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Leonardo Fantoni
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Ania Naila Guerrieri
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Chiara Bellotti
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Alessandro De Vita
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (G.M.); (A.D.V.); (C.S.)
| | - Chiara Spadazzi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (G.M.); (A.D.V.); (C.S.)
| | - Davide Maria Donati
- Orthopaedic Oncology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Monica Torsello
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Enrico Lucarelli
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| | - Laura Mercatali
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (V.G.); (G.S.); (M.P.); (C.M.H.); (M.C.); (L.F.); (A.N.G.); (C.B.); (T.I.); (L.M.)
| |
Collapse
|
4
|
Gupta S, Qayoom I, Gupta P, Gupta A, Singh P, Singh S, Kumar A. Exosome-Functionalized, Drug-Laden Bone Substitute along with an Antioxidant Herbal Membrane for Bone and Periosteum Regeneration in Bone Sarcoma. ACS APPLIED MATERIALS & INTERFACES 2023; 15:8824-8839. [PMID: 36749176 DOI: 10.1021/acsami.2c18308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Developing advanced methods for effective bone reconstructive strategies in case of critical bone defects caused by tumor resection, trauma, and other implant-related complications remains a challenging problem in orthopedics. In the clinical management of bone diseases, there is a paradigm shift in using local drugs at the injury site; however, the dead space created during the surgical debridement of necrotic bone and soft tissues (periosteum and underlying muscle) leads to ineffective bone formation, thereby leading to secondary complications, and thus calls for better regenerative approaches. In this study, we have utilized an exosome-functionalized doxorubicin-loaded biodegradable nanocement (NC)-based carrier along with a Cissus quadrangularis (CQ) extract-laden antioxidant herbal membrane for simultaneously managing the periosteum as well as bone formation in the tumor resection model of osteosarcoma. We initially evaluated the efficacy of scaffolds for in vitro mineralization and bone formation. To examine the in vivo effectiveness, we developed a human osteosarcoma cell line (Saos-2)-induced tumor xenograft model with a critical-sized bone defect. The findings revealed that doxorubicin released from NC was successful in killing the tumor cells and was present even after 30 days of implantation. Additionally, the incorporation of exosomes aided the bone formation, resulting in around a 2.6-fold increase in the bone volume compared to the empty group as evaluated by micro-CT. The herbal membrane assisted in the development of periosteum and mineralizing bone callous as validated through histological and immunofluorescence analysis. Thus, our findings describe a one-step biomaterial-based cell-free approach to regenerate bone in osteosarcoma and prevent further fracture due to the complete development of periosteum and lost bone.
Collapse
Affiliation(s)
- Sneha Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Irfan Qayoom
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Purva Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Archita Gupta
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Prerna Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Sneha Singh
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
- Centre for Environmental Sciences and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, India
- Centre for Nanosciences, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Centre of Excellence for Orthopedics and Prosthetics, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur 208016, India
| |
Collapse
|
5
|
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
|
6
|
Inhibiting Endothelin Receptors with Macitentan Strengthens the Bone Protective Action of RANKL Inhibition and Reduces Metastatic Dissemination in Osteosarcoma. Cancers (Basel) 2022; 14:cancers14071765. [PMID: 35406536 PMCID: PMC8997105 DOI: 10.3390/cancers14071765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary The efficacy of current osteosarcoma therapy is diminished by two adverse events, namely resistance to chemotherapy and metastatic dissemination. In recent decades, research has been devoted to reducing these adverse events. Inhibiting bone resorption has shown promising effects on metastatic dissemination and tumor growth, with, however, the formation of significant tumoral mineralized tissue. Endothelin signaling is implicated in activating the cell that forms the mineralized tissues, consequently the impact of inhibiting it alone and in combination with the inhibition of bone resorption was evaluated using osteosarcoma models. The results obtained showed that inhibiting endothelin signaling significantly reduced the formation of mineralized tumor tissue concomitantly to metastatic dissemination without affecting sensitivity to chemotherapy. This inhibition appears to be a promising new therapeutic tool in the fight against osteosarcoma. Abstract Current treatments for osteosarcoma, combining conventional polychemotherapy and surgery, make it possible to attain a five-year survival rate of 70% in affected individuals. The presence of chemoresistance and metastases significantly shorten the patient’s lifespan, making identification of new therapeutic tools essential. Inhibiting bone resorption has been shown to be an efficient adjuvant strategy impacting the metastatic dissemination of osteosarcoma, tumor growth, and associated bone destruction. Unfortunately, over-apposition of mineralized matrix by normal and tumoral osteoblasts was associated with this inhibition. Endothelin signaling is implicated in the functional differentiation of osteoblasts, raising the question of the potential value of inhibiting it alone, or in combination with bone resorption repression. Using mouse models of osteosarcoma, the impact of macitentan, an endothelin receptor inhibitor, was evaluated regarding tumor growth, metastatic dissemination, matrix over-apposition secondary to RANKL blockade, and safety when combined with chemotherapy. The results showed that macitentan has no impact on tumor growth or sensitivity to ifosfamide, but significantly reduces tumoral osteoid tissue formation and the metastatic capacity of the osteosarcoma. To conclude, macitentan appears to be a promising therapeutic adjuvant for osteosarcoma alone or associated with bone resorption inhibitors.
Collapse
|
7
|
Martella E, Dozza B, Ferroni C, Obeyok CO, Guerrini A, Tedesco D, Manet I, Sotgiu G, Columbaro M, Ballestri M, Martini L, Fini M, Lucarelli E, Varchi G, Duchi S. Two Beats One: Osteosarcoma Therapy with Light-Activated and Chemo-Releasing Keratin Nanoformulation in a Preclinical Mouse Model. Pharmaceutics 2022; 14:pharmaceutics14030677. [PMID: 35336051 PMCID: PMC8950553 DOI: 10.3390/pharmaceutics14030677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma treatment is moving towards more effective combination therapies. Nevertheless, these approaches present distinctive challenges that can complicate the clinical translation, such as increased toxicity and multi-drug resistance. Drug co-encapsulation within a nanoparticle formulation can overcome these challenges and improve the therapeutic index. We previously synthetized keratin nanoparticles functionalized with Chlorin-e6 (Ce6) and paclitaxel (PTX) to combine photo (PDT) and chemotherapy (PTX) regimens, and the inhibition of osteosarcoma cells growth in vitro was demonstrated. In the current study, we generated an orthotopic osteosarcoma murine model for the preclinical evaluation of our combination therapy. To achieve maximum reproducibility, we systematically established key parameters, such as the number of cells to generate the tumor, the nanoparticles dose, the design of the light-delivery device, the treatment schedule, and the irradiation settings. A 60% engrafting rate was obtained using 10 million OS cells inoculated intratibial, with the tumor model recapitulating the histological hallmarks of the human counterpart. By scheduling the treatment as two cycles of injections, a 32% tumor reduction was obtained with PTX mono-therapy and a 78% reduction with the combined PTX-PDT therapy. Our findings provide the in vivo proof of concept for the subsequent clinical development of a combination therapy to fight osteosarcoma.
Collapse
Affiliation(s)
- Elisa Martella
- Institute for the Organic Synthesis and Photoreactivity (ISOF), National Research Council (CNR), 40129 Bologna, Italy; (E.M.); (C.F.); (C.O.O.); (A.G.); (D.T.); (I.M.); (G.S.); (M.B.)
| | - Barbara Dozza
- Rizzoli Laboratory Unit, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40123 Bologna, Italy;
| | - Claudia Ferroni
- Institute for the Organic Synthesis and Photoreactivity (ISOF), National Research Council (CNR), 40129 Bologna, Italy; (E.M.); (C.F.); (C.O.O.); (A.G.); (D.T.); (I.M.); (G.S.); (M.B.)
| | - Clement Osuru Obeyok
- Institute for the Organic Synthesis and Photoreactivity (ISOF), National Research Council (CNR), 40129 Bologna, Italy; (E.M.); (C.F.); (C.O.O.); (A.G.); (D.T.); (I.M.); (G.S.); (M.B.)
| | - Andrea Guerrini
- Institute for the Organic Synthesis and Photoreactivity (ISOF), National Research Council (CNR), 40129 Bologna, Italy; (E.M.); (C.F.); (C.O.O.); (A.G.); (D.T.); (I.M.); (G.S.); (M.B.)
| | - Daniele Tedesco
- Institute for the Organic Synthesis and Photoreactivity (ISOF), National Research Council (CNR), 40129 Bologna, Italy; (E.M.); (C.F.); (C.O.O.); (A.G.); (D.T.); (I.M.); (G.S.); (M.B.)
| | - Ilse Manet
- Institute for the Organic Synthesis and Photoreactivity (ISOF), National Research Council (CNR), 40129 Bologna, Italy; (E.M.); (C.F.); (C.O.O.); (A.G.); (D.T.); (I.M.); (G.S.); (M.B.)
| | - Giovanna Sotgiu
- Institute for the Organic Synthesis and Photoreactivity (ISOF), National Research Council (CNR), 40129 Bologna, Italy; (E.M.); (C.F.); (C.O.O.); (A.G.); (D.T.); (I.M.); (G.S.); (M.B.)
| | - Marta Columbaro
- Electron Microscopy Platform, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Marco Ballestri
- Institute for the Organic Synthesis and Photoreactivity (ISOF), National Research Council (CNR), 40129 Bologna, Italy; (E.M.); (C.F.); (C.O.O.); (A.G.); (D.T.); (I.M.); (G.S.); (M.B.)
| | - Lucia Martini
- Complex Structure Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (L.M.); (M.F.)
| | - Milena Fini
- Complex Structure Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (L.M.); (M.F.)
| | - Enrico Lucarelli
- Regenerative Therapies in Oncology of the Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Greta Varchi
- Institute for the Organic Synthesis and Photoreactivity (ISOF), National Research Council (CNR), 40129 Bologna, Italy; (E.M.); (C.F.); (C.O.O.); (A.G.); (D.T.); (I.M.); (G.S.); (M.B.)
- Correspondence: (G.V.); (S.D.); Tel.: +39-051-6398283 (G.V.)
| | - Serena Duchi
- Institute for the Organic Synthesis and Photoreactivity (ISOF), National Research Council (CNR), 40129 Bologna, Italy; (E.M.); (C.F.); (C.O.O.); (A.G.); (D.T.); (I.M.); (G.S.); (M.B.)
- Department of Surgery, St. Vincent’s Hospital Melbourne, University of Melbourne, Fitzroy, VIC 3065, Australia
- Correspondence: (G.V.); (S.D.); Tel.: +39-051-6398283 (G.V.)
| |
Collapse
|
8
|
Sabei FY, Taratula O, Albarqi HA, Al-Fatease AM, Moses AS, Demessie AA, Park Y, Vogel WK, Esfandiari Nazzaro E, Davare MA, Alani A, Leid M, Taratula O. A targeted combinatorial therapy for Ewing's sarcoma. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 37:102446. [PMID: 34303840 PMCID: PMC8464505 DOI: 10.1016/j.nano.2021.102446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/23/2021] [Accepted: 07/04/2021] [Indexed: 02/08/2023]
Abstract
Ewing's sarcoma (EwS) is the second most common bone cancer in children and adolescents. Current chemotherapy regimens are mainly ineffective in patients with relapsed disease and cause long-term effects in survivors. Therefore, we have developed a combinatorial therapy based on a novel drug candidate named ML111 that exhibits selective activity against EwS cells and synergizes with vincristine. To increase the aqueous solubility of hydrophobic ML111, polymeric nanoparticles (ML111-NP) were developed. In vitro data revealed that ML111-NP compromise viability of EwS cells without affecting non-malignant cells. Furthermore, ML111-NP exhibit strong synergistic effects in a combination with vincristine on EwS cells, while this drug pair exhibits antagonistic effects towards normal cells. Finally, animal studies validated that ML111-NP efficiently accumulate in orthotopic EwS xenografts after intravenous injection and provide superior therapeutic outcomes in a combination with vincristine without evident toxicity. These results support the potential of the ML111-based combinatorial therapy for EwS.
Collapse
Affiliation(s)
- Fahad Y Sabei
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA; Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Olena Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Hassan A Albarqi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA; Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Adel M Al-Fatease
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA; Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Abraham S Moses
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Ananiya A Demessie
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Youngrong Park
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Walter K Vogel
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Ellie Esfandiari Nazzaro
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Monika A Davare
- Papé Pediatric Research Institute, Oregon Health & Science University, Portland, OR, USA; Division of Pediatric Hematology & Oncology, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Adam Alani
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Mark Leid
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA; Department of Integrative Biomedical and Diagnostic Sciences, Oregon Health & Science University, Portland, OR, USA.
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA.
| |
Collapse
|
9
|
Khattab MS, AbuBakr HO, El Iraqi KG, AbdElKader NA, Kamel MM, Salem KH, Steitz J, Afify M. Intra-iliac bone marrow injection as a novel alternative to intra-tibial inoculation in rat model. Stem Cell Res Ther 2021; 12:336. [PMID: 34112243 PMCID: PMC8194056 DOI: 10.1186/s13287-021-02413-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/24/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Intra-bone marrow injection (IBMI) in rats is adopted in many studies for stem cell and hematopoietic cell transplantation. IBMI in the tibia or the femur results in severe distress to the animal. Therefore, this study aims to evaluate intra-iliac injections as an alternative approach for IBMI. METHODS Twenty-seven Sprague Dawley rats were assigned into 3 groups, 9 rats each, for 4 weeks. The control group rats were not injected. Tibia group rats were injected intra-tibial and the iliac group rats were injected intra-iliac with saline. Behavioral, radiological, histopathological, and stress evaluation was performed. Total bilirubin, cortisol, and insulin-like growth factor-1 (IGF1) were measured. RESULTS Behavioral measurements revealed deviation compared to control, in both injected groups, on the 1st and 2nd week. By the 3rd week, it was equivalent to control in the iliac group only. Bilirubin and cortisol levels were increased by intra-tibial injection compared to intra-iliac injection. The IGF-1 gene expression increased compared to control at 1st and 2nd weeks in intra-iliac injection and decreased by intra-tibial injection at 2nd week. The thickness of the iliac crest was not different from the control group, whereas there were significant differences between the control and tibia groups. Healing of the iliac crest was faster compared to the tibia. In the 3rd week, the tibia showed fibrosis at the site of injection whereas the iliac crest showed complete bone reconstruction. CONCLUSION Intra-iliac injections exert less distress on animals, and by 3 weeks, they regained their normal activity in comparison to intra-tibial injections.
Collapse
Affiliation(s)
- Marwa S Khattab
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Huda O AbuBakr
- Department of Biochemistry and Molecular Biology, Cairo University, Giza, Egypt
| | - Kassem G El Iraqi
- Department of Veterinary Hygiene and Management, Cairo University, Giza, Egypt
| | - Naglaa A AbdElKader
- Department of Surgery, Anesthesiology & Radiology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Mervat M Kamel
- Department of Veterinary Hygiene and Management, Cairo University, Giza, Egypt
| | - Khaled Hamed Salem
- Department of Orthopedic Surgery, Faculty of Medicine, Cairo University, Giza, Egypt
- Department of Orthopedic Surgery, RWTH Aachen University Faculty of Medicine, Aachen, Germany
| | - Julia Steitz
- Institute for Laboratory Animal Science, RWTH Aachen University Faculty of Medicine, Aachen, Germany
| | - Mamdouh Afify
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| |
Collapse
|
10
|
Sakolish C, House JS, Chramiec A, Liu Y, Chen Z, Halligan SP, Vunjak-Novakovic G, Rusyn I. Tissue-Engineered Bone Tumor as a Reproducible Human in Vitro Model for Studies of Anticancer Drugs. Toxicol Sci 2021; 173:65-76. [PMID: 31626302 DOI: 10.1093/toxsci/kfz220] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Studies of anticancer therapies in traditional cell culture models can demonstrate efficacy of direct-acting compounds but lack the 3-dimensional arrangement of the tumor cells and their tissue-specific microenvironments, both of which are important modulators of treatment effects in vivo. Bone cells reside in complex environments that regulate their fate and function. A bioengineered human bone-tumor model has been shown to provide a microphysiological niche for studies of cancer cell behavior. Here, we demonstrate successful transfer between 2 laboratories and utility of this model in efficacy studies using well-established chemotherapeutic agents. The bioengineered human bone-tumor model consisted of Ewing sarcoma (RD-ES) cancer cell aggregates infused into tissue-engineered bone that was grown from human mesenchymal stem cell-derived differentiated into osteoblasts within mineralized bone scaffolds. The tumor model was maintained in culture for over 5 weeks and subjected to clinically relevant doses of linsitinib, doxorubicin, cisplatin, methotrexate, vincristine, dexamethasone, or MAP (methotrexate, doxorubicin, and cisplatin combination). Drug administration cycles were designed to mimic clinical treatment regimens. The bioengineered tumors were evaluated days to weeks after the cessation of treatment to monitor the potential for relapse, using bioengineered bone and ES cell monolayers as controls. Drug binding to the scaffolds and media proteins and gene expression were also evaluated. We show that a bioengineered human bone tumor can be used as a microphysiological model for preclinical studies of anticancer drugs. We found that anticancer efficacy was achieved at concentrations approximating the human Cmax, in contrast to traditional ES cell monolayers. These studies show that the bone-tumor model can be successfully transferred between laboratories and has predictive power in preclinical studies. The effects of drugs on the bone tumors and healthy bone were studied in parallel, in support of the utility of this model for identification of new therapeutic targets.
Collapse
Affiliation(s)
- Courtney Sakolish
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843
| | - John S House
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27695
| | - Alan Chramiec
- Department of Biomedical Engineering, Columbia University, New York, New York 10032
| | - Yizhong Liu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843
| | - Zunwei Chen
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843
| | - Susan P Halligan
- Department of Biomedical Engineering, Columbia University, New York, New York 10032
| | | | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843
| |
Collapse
|
11
|
Preclinical In Vivo Modeling of Pediatric Sarcoma-Promises and Limitations. J Clin Med 2021; 10:jcm10081578. [PMID: 33918045 PMCID: PMC8069549 DOI: 10.3390/jcm10081578] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 02/07/2023] Open
Abstract
Pediatric sarcomas are an extremely heterogeneous group of genetically distinct diseases. Despite the increasing knowledge on their molecular makeup in recent years, true therapeutic advancements are largely lacking and prognosis often remains dim, particularly for relapsed and metastasized patients. Since this is largely due to the lack of suitable model systems as a prerequisite to develop and assess novel therapeutics, we here review the available approaches to model sarcoma in vivo. We focused on genetically engineered and patient-derived mouse models, compared strengths and weaknesses, and finally explored possibilities and limitations to utilize these models to advance both biological understanding as well as clinical diagnosis and therapy.
Collapse
|
12
|
Shimada Y, Naito T, Hayashi T, Saito T, Suehara Y, Kakinuma C, Nozaki Y, Takagi H, Yao T. Establishment of a patient-derived xenograft mouse model of pleomorphic leiomyosarcoma. J Toxicol Pathol 2020; 34:89-93. [PMID: 33627948 PMCID: PMC7890165 DOI: 10.1293/tox.2020-0061] [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: 09/03/2020] [Accepted: 10/29/2020] [Indexed: 11/19/2022] Open
Abstract
Soft tissue sarcomas are difficult to treat using chemotherapy owing to a current deficiency in candidate drugs for specific targets. Screening candidate compounds and analyzing therapeutic targets in sarcomas is insufficient, given the lack of an appropriate human sarcoma animal model to accurately evaluate their efficacy, as well as the lack of an adequate technical protocol for efficient transplantation and engraftment of sarcoma specimens in patient-derived xenograft (PDX) models. Accordingly, in this study, we sought to identify the optimal type of sarcoma and develop a protocol for generating a PDX model. We characterized a PDX mouse model using histopathological and immunohistochemical analyses to determine whether it would show pathological characteristics similar to those of human sarcomas. We achieved engraftment of one of the 10 transplanted sarcoma specimens, the xenografted tumor of which exhibited massive proliferation. Histologically, the engrafted sarcoma foci resembled a primary tumor of pleomorphic leiomyosarcoma and maintained their histological structure in all passages. Moreover, immunohistochemical analysis revealed the expression of specific markers of differentiation to smooth muscle, which is consistent with the features of leiomyosarcoma. We thus demonstrated that our pleomorphic leiomyosarcoma PDX mouse model mimics at least one aspect of human sarcomas, and we believe that this model will facilitate the development of novel therapies for sarcomas.
Collapse
Affiliation(s)
- Yasuhiro Shimada
- Department of Human Pathology, School of Medicine, Juntendo University, 1-1-19 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tomoharu Naito
- Department of Human Pathology, School of Medicine, Juntendo University, 1-1-19 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takuo Hayashi
- Department of Human Pathology, School of Medicine, Juntendo University, 1-1-19 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, School of Medicine, Juntendo University, 1-1-19 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yoshiyuki Suehara
- Department of Orthopaedic Surgery, School of Medicine, Juntendo University, 1-1-19 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Chihaya Kakinuma
- Department of Human Pathology, School of Medicine, Juntendo University, 1-1-19 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuji Nozaki
- Biotechnical Center, Japan SLC, Inc., 3-5-1 Aoihigashi, Naka-ku, Hamamatsu, Shizuoka 433-8114, Japan
| | - Hisayoshi Takagi
- Biotechnical Center, Japan SLC, Inc., 3-5-1 Aoihigashi, Naka-ku, Hamamatsu, Shizuoka 433-8114, Japan
| | - Takashi Yao
- Department of Human Pathology, School of Medicine, Juntendo University, 1-1-19 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
13
|
Grünewald TGP, Alonso M, Avnet S, Banito A, Burdach S, Cidre‐Aranaz F, Di Pompo G, Distel M, Dorado‐Garcia H, Garcia‐Castro J, González‐González L, Grigoriadis AE, Kasan M, Koelsche C, Krumbholz M, Lecanda F, Lemma S, Longo DL, Madrigal‐Esquivel C, Morales‐Molina Á, Musa J, Ohmura S, Ory B, Pereira‐Silva M, Perut F, Rodriguez R, Seeling C, Al Shaaili N, Shaabani S, Shiavone K, Sinha S, Tomazou EM, Trautmann M, Vela M, Versleijen‐Jonkers YMH, Visgauss J, Zalacain M, Schober SJ, Lissat A, English WR, Baldini N, Heymann D. Sarcoma treatment in the era of molecular medicine. EMBO Mol Med 2020; 12:e11131. [PMID: 33047515 PMCID: PMC7645378 DOI: 10.15252/emmm.201911131] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Sarcomas are heterogeneous and clinically challenging soft tissue and bone cancers. Although constituting only 1% of all human malignancies, sarcomas represent the second most common type of solid tumors in children and adolescents and comprise an important group of secondary malignancies. More than 100 histological subtypes have been characterized to date, and many more are being discovered due to molecular profiling. Owing to their mostly aggressive biological behavior, relative rarity, and occurrence at virtually every anatomical site, many sarcoma subtypes are in particular difficult-to-treat categories. Current multimodal treatment concepts combine surgery, polychemotherapy (with/without local hyperthermia), irradiation, immunotherapy, and/or targeted therapeutics. Recent scientific advancements have enabled a more precise molecular characterization of sarcoma subtypes and revealed novel therapeutic targets and prognostic/predictive biomarkers. This review aims at providing a comprehensive overview of the latest advances in the molecular biology of sarcomas and their effects on clinical oncology; it is meant for a broad readership ranging from novices to experts in the field of sarcoma.
Collapse
Affiliation(s)
- Thomas GP Grünewald
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
- Division of Translational Pediatric Sarcoma ResearchGerman Cancer Research Center (DKFZ), Hopp Children's Cancer Center (KiTZ), German Cancer Consortium (DKTK)HeidelbergGermany
- Institute of PathologyHeidelberg University HospitalHeidelbergGermany
| | - Marta Alonso
- Program in Solid Tumors and BiomarkersFoundation for the Applied Medical ResearchUniversity of Navarra PamplonaPamplonaSpain
| | - Sofia Avnet
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Ana Banito
- Pediatric Soft Tissue Sarcoma Research GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Stefan Burdach
- Department of Pediatrics and Children's Cancer Research Center (CCRC)Technische Universität MünchenMunichGermany
| | - Florencia Cidre‐Aranaz
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
| | - Gemma Di Pompo
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | | | | | | | | | | | - Merve Kasan
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
| | | | | | - Fernando Lecanda
- Division of OncologyAdhesion and Metastasis LaboratoryCenter for Applied Medical ResearchUniversity of NavarraPamplonaSpain
| | - Silvia Lemma
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Dario L Longo
- Institute of Biostructures and Bioimaging (IBB)Italian National Research Council (CNR)TurinItaly
| | | | | | - Julian Musa
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | - Shunya Ohmura
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
| | | | - Miguel Pereira‐Silva
- Department of Pharmaceutical TechnologyFaculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Francesca Perut
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Rene Rodriguez
- Instituto de Investigación Sanitaria del Principado de AsturiasOviedoSpain
- CIBER en oncología (CIBERONC)MadridSpain
| | | | - Nada Al Shaaili
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Shabnam Shaabani
- Department of Drug DesignUniversity of GroningenGroningenThe Netherlands
| | - Kristina Shiavone
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Snehadri Sinha
- Department of Oral and Maxillofacial DiseasesUniversity of HelsinkiHelsinkiFinland
| | | | - Marcel Trautmann
- Division of Translational PathologyGerhard‐Domagk‐Institute of PathologyMünster University HospitalMünsterGermany
| | - Maria Vela
- Hospital La Paz Institute for Health Research (IdiPAZ)MadridSpain
| | | | | | - Marta Zalacain
- Institute of Biostructures and Bioimaging (IBB)Italian National Research Council (CNR)TurinItaly
| | - Sebastian J Schober
- Department of Pediatrics and Children's Cancer Research Center (CCRC)Technische Universität MünchenMunichGermany
| | - Andrej Lissat
- University Children′s Hospital Zurich – Eleonoren FoundationKanton ZürichZürichSwitzerland
| | - William R English
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Nicola Baldini
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
| | - Dominique Heymann
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
- Université de NantesInstitut de Cancérologie de l'OuestTumor Heterogeneity and Precision MedicineSaint‐HerblainFrance
| |
Collapse
|
14
|
Endo-Munoz L, Bennett TC, Topkas E, Wu SY, Thamm DH, Brockley L, Cooper M, Sommerville S, Thomson M, O'Connell K, Lane A, Bird G, Peaston A, Matigian N, Straw RC, Saunders NA. Auranofin improves overall survival when combined with standard of care in a pilot study involving dogs with osteosarcoma. Vet Comp Oncol 2019; 18:206-213. [PMID: 31441983 DOI: 10.1111/vco.12533] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 08/11/2019] [Accepted: 08/16/2019] [Indexed: 12/29/2022]
Abstract
Osteosarcoma is the most common paediatric primary bone malignancy. The major cause of death in osteosarcoma is drug-resistant pulmonary metastasis. Previous studies have shown that thioredoxin reductase 2 is a driver of metastasis in osteosarcoma and can be inhibited by auranofin (AF). Moreover, studies have shown that AF significantly reduces pulmonary metastases in xenotransplant models. Here, we describe a phase I/II study of AF in canine osteosarcoma, a well-recognized spontaneous model of human osteosarcoma. We performed a single-arm multicentre pilot study of AF in combination with standard of care (SOC) (amputation + carboplatin). We recruited 40 dogs to the trial and used a historical SOC-only control group (n = 26). Dogs >15 kg received 9 mg AF q3d PO and dogs <15 kg received 6 mg q3d. Follow-up occurred over at least a 3-year period. Auranofin plus SOC improved overall survival (OS) (P = .036) in all dogs treated. The improved outcome was attributable entirely to improved OS in male dogs (P = .009). At the time of writing, 10 dogs (25%) survive without measurable disease in the treatment group with survival times ranging between 806 and 1525 days. Our study shows that AF improves OS in male dogs when combined with SOC. Our findings have translational relevance for the management of canine and human osteosarcoma. Our data justify a larger multicentre phase 2 trial in dogs and a phase I/II trial in human patients with refractory disease at the time of initial surgery.
Collapse
Affiliation(s)
- Liliana Endo-Munoz
- Translational Research Institute, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia.,Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | | | - Eleni Topkas
- Translational Research Institute, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Sherry Y Wu
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Douglas H Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| | - Laura Brockley
- Victorian Animal Cancer Care, Melbourne, Victoria, Australia
| | - Maureen Cooper
- Victorian Animal Cancer Care, Melbourne, Victoria, Australia
| | - Scott Sommerville
- Department of Orthopedic Oncology, Princess Alexandra Hospital and The Wesley Hospital, Brisbane, Queensland, Australia
| | - Maurine Thomson
- Veterinary Specialist Services, Brisbane, Queensland, Australia
| | | | - Amy Lane
- Small Animal Oncology, Newcastle, New South Wales, Australia
| | - Guy Bird
- Veterinary Emergency Centre and Hospital, James Cook University School of Veterinary and Biomedical Science, Townsville, Queensland, Australia
| | - Anne Peaston
- School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Nicholas Matigian
- QFAB Bioinformatics, BIODATA Institute of Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Rodney C Straw
- Brisbane Veterinary Specialist Centre, Brisbane, Queensland, Australia.,Australian Consortium of Comparative Oncology of the Australian Animal Cancer Foundation, Brisbane, Queensland, Australia
| | - Nicholas A Saunders
- Translational Research Institute, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
15
|
Schneider J, Volkmer I, Engel K, Emmer A, Staege MS. Expression of A New Endogenous Retrovirus-Associated Transcript in Hodgkin Lymphoma Cells. Int J Mol Sci 2019; 20:ijms20215320. [PMID: 31731509 PMCID: PMC6862598 DOI: 10.3390/ijms20215320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 12/20/2022] Open
Abstract
During characterization of a cDNA library from the Hodgkin lymphoma (HL) cell line L-1236, we discovered a new transcript derived from chromosome 1 at the long intergenic non-protein coding RNA 1768 (LINC01768)/colony stimulating factor 1 (CSF1) region. The first exon of this transcript from Hodgkin lymphoma cells (THOLE) starts in the predicted exon 4 of LINC01768 and is part of an endogenous retrovirus (ERV) from the HUERS-P1/LTR8 family. High expression of THOLE was only detectable in HL cell line L-1236. The expression of THOLE in L-1236 cell is another example for ERV/LTR-associated gene expression in HL cells. At the genome level, the HUERS-P1/LTR8 region including THOLE is only present in Hominoidea. The influence of ERV/LTRs on gene expression might explain the characteristic phenotype of human HL.
Collapse
Affiliation(s)
- Jana Schneider
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Martin Luther University Halle-Wittenberg, 06097 Halle, Germany; (J.S.); (I.V.); (K.E.)
| | - Ines Volkmer
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Martin Luther University Halle-Wittenberg, 06097 Halle, Germany; (J.S.); (I.V.); (K.E.)
| | - Kristina Engel
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Martin Luther University Halle-Wittenberg, 06097 Halle, Germany; (J.S.); (I.V.); (K.E.)
| | - Alexander Emmer
- Department of Neurology, Martin Luther University Halle-Wittenberg, 06097 Halle, Germany
| | - Martin S. Staege
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Martin Luther University Halle-Wittenberg, 06097 Halle, Germany; (J.S.); (I.V.); (K.E.)
- Correspondence: ; Tel.: +49-345-557-7280; Fax: +49-345-557-7275
| |
Collapse
|
16
|
Houghton PJ, Kurmasheva RT. Challenges and Opportunities for Childhood Cancer Drug Development. Pharmacol Rev 2019; 71:671-697. [PMID: 31558580 PMCID: PMC6768308 DOI: 10.1124/pr.118.016972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer in children is rare with approximately 15,700 new cases diagnosed in the United States annually. Through use of multimodality therapy (surgery, radiation therapy, and aggressive chemotherapy), 70% of patients will be "cured" of their disease, and 5-year event-free survival exceeds 80%. However, for patients surviving their malignancy, therapy-related long-term adverse effects are severe, with an estimated 50% having chronic life-threatening toxicities related to therapy in their fourth or fifth decade of life. While overall intensive therapy with cytotoxic agents continues to reduce cancer-related mortality, new understanding of the molecular etiology of many childhood cancers offers an opportunity to redirect efforts to develop effective, less genotoxic therapeutic options, including agents that target oncogenic drivers directly, and the potential for use of agents that target the tumor microenvironment and immune-directed therapies. However, for many high-risk cancers, significant challenges remain.
Collapse
Affiliation(s)
- Peter J Houghton
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| |
Collapse
|
17
|
Shekhar TM, Burvenich IJG, Harris MA, Rigopoulos A, Zanker D, Spurling A, Parker BS, Walkley CR, Scott AM, Hawkins CJ. Smac mimetics LCL161 and GDC-0152 inhibit osteosarcoma growth and metastasis in mice. BMC Cancer 2019; 19:924. [PMID: 31521127 PMCID: PMC6744692 DOI: 10.1186/s12885-019-6103-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/28/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Current therapies fail to cure over a third of osteosarcoma patients and around three quarters of those with metastatic disease. "Smac mimetics" (also known as "IAP antagonists") are a new class of anti-cancer agents. Previous work revealed that cells from murine osteosarcomas were efficiently sensitized by physiologically achievable concentrations of some Smac mimetics (including GDC-0152 and LCL161) to killing by the inflammatory cytokine TNFα in vitro, but survived exposure to Smac mimetics as sole agents. METHODS Nude mice were subcutaneously or intramuscularly implanted with luciferase-expressing murine 1029H or human KRIB osteosarcoma cells. The impacts of treatment with GDC-0152, LCL161 and/or doxorubicin were assessed by caliper measurements, bioluminescence, 18FDG-PET and MRI imaging, and by weighing resected tumors at the experimental endpoint. Metastatic burden was examined by quantitative PCR, through amplification of a region of the luciferase gene from lung DNA. ATP levels in treated and untreated osteosarcoma cells were compared to assess in vitro sensitivity. Immunophenotyping of cells within treated and untreated tumors was performed by flow cytometry, and TNFα levels in blood and tumors were measured using cytokine bead arrays. RESULTS Treatment with GDC-0152 or LCL161 suppressed the growth of subcutaneously or intramuscularly implanted osteosarcomas. In both models, co-treatment with doxorubicin and Smac mimetics impeded average osteosarcoma growth to a greater extent than either drug alone, although these differences were not statistically significant. Co-treatments were also more toxic. Co-treatment with LCL161 and doxorubicin was particularly effective in the KRIB intramuscular model, impeding primary tumor growth and delaying or preventing metastasis. Although the Smac mimetics were effective in vivo, in vitro they only efficiently killed osteosarcoma cells when TNFα was supplied. Implanted tumors contained high levels of TNFα, produced by infiltrating immune cells. Spontaneous osteosarcomas that arose in genetically-engineered immunocompetent mice also contained abundant TNFα. CONCLUSIONS These data imply that Smac mimetics can cooperate with TNFα secreted by tumor-associated immune cells to kill osteosarcoma cells in vivo. Smac mimetics may therefore benefit osteosarcoma patients whose tumors contain Smac mimetic-responsive cancer cells and TNFα-producing infiltrating cells.
Collapse
Affiliation(s)
- Tanmay M. Shekhar
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086 Australia
| | - Ingrid J. G. Burvenich
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Michael A. Harris
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086 Australia
| | - Angela Rigopoulos
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Damien Zanker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086 Australia
| | - Alex Spurling
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086 Australia
| | - Belinda S. Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086 Australia
| | - Carl R. Walkley
- St. Vincent’s Institute, Fitzroy, Victoria 3065 Australia
- Department of Medicine, St. Vincent’s Hospital, University of Melbourne, Fitzroy, Victoria 3065 Australia
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria 3000 Australia
| | - Andrew M. Scott
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research and Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
- Departments of Medical Oncology and Molecular Imaging & Therapy, Austin Health, Heidelberg, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Christine J. Hawkins
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086 Australia
| |
Collapse
|
18
|
Kim JM, Wong AJ, Lu AJ, Pointdujour-Lim R. Chondrosarcoma Metastasis to the Choroid. Ocul Oncol Pathol 2019; 5:234-237. [PMID: 31367583 DOI: 10.1159/000494979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/29/2018] [Indexed: 11/19/2022] Open
Abstract
We report a rare case of chondrosarcoma metastatic to the choroid. A 64-year-old male with a history of chondrosarcoma metastatic to the lungs and to the spine presented with blurred vision. A choroidal tumor was found. Fine-needle biopsy confirmed the histologic identity of the tumor as chondrosarcoma. Metastatic spread of chondrosarcoma to the eye is extremely rare. When present, lesions may grow rapidly, and systemic prognosis is poor. Co-management with medical oncology is of utmost importance. This is the third case of chondrosarcoma metastatic to the choroid in the literature and the first with bilateral involvement.
Collapse
Affiliation(s)
- Jenna May Kim
- Department of Ophthalmology and Visual Science, Yale New Haven Hospital, Yale School of Medicine, New Haven, Connecticut, USA
| | - Amanda J Wong
- Department of Ophthalmology and Visual Science, Yale New Haven Hospital, Yale School of Medicine, New Haven, Connecticut, USA
| | - Amanda J Lu
- Department of Ophthalmology and Visual Science, Yale New Haven Hospital, Yale School of Medicine, New Haven, Connecticut, USA
| | - Renelle Pointdujour-Lim
- Department of Ophthalmology and Visual Science, Yale New Haven Hospital, Yale School of Medicine, New Haven, Connecticut, USA.,Ophthalmic Oncology, Smilow Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
19
|
Abstract
Although the investigation into biomarkers specific for pulmonary metastasis within osteosarcoma (OS) has recently expanded, their usage within the clinic remains sparse. The current screening protocol after any OS diagnosis includes a chest CT scan; however, metastatic lung nodules frequently go undetected and remain the primary cause of death in OS. Recently, screening technologies such as liquid biopsy and next-generation sequencing have revealed a promising array of biomarkers with predictive and diagnostic value for the pulmonary metastasis associated with OS. These biomarkers draw from genomics, transcriptomics, epigenetics, and metabolomics. When assessed in concert, their utility is most promising as OS is a highly heterogeneous cancer. Accordingly, there has been an expansion of clinical trials not only aimed at further demonstrating the significance of these individual biomarkers but to also reveal which therapies resolve the pulmonary metastasis once detected. This review will focus on the recently discovered and novel metastatic biomarkers within OS, their molecular and cellular mechanisms, the expansion of humanized OS mouse models amenable to their testing, and the associated clinical trials aimed at managing the metastatic phase of OS.
Collapse
|