1
|
Suzuki M, Kobayashi H, Hanaoka H. Near-infrared photoimmunotherapy for osteosarcoma targeting epidermal growth factor receptor. Transl Oncol 2024; 50:102132. [PMID: 39357464 PMCID: PMC11471228 DOI: 10.1016/j.tranon.2024.102132] [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: 06/05/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
Osteosarcoma is the most common bone tumor, and it possesses high metastatic propensity. Although systemic chemotherapy has improved its prognosis, improvements in survival rates have stalled in recent years. Moreover, the prognosis of patients with metastatic osteosarcoma remains poor. Near-infrared photoimmunotherapy (NIR-PIT) is a highly selective cancer therapy that induces immunogenic cell death (ICD), and the therapeutic effects spread to distant metastatic sites. Therefore, NIR-PIT could be useful in both primary and metastatic osteosarcoma treatment. In this study, we investigated the efficacy of NIR-PIT targeting epidermal growth factor receptor (EGFR) in osteosarcoma. The cytotoxic effects of NIR-PIT in osteosarcoma cell lines with different EGFR expression levels (MG63; high, Saos-2; low) were evaluated. NIR-PIT-induced cell death was dependent on the EGFR expression level. After NIR-PIT, swelling and bleb formation, the characteristic morphological changes induced by NIR-PIT associated with necrosis caused by the influx of extracellular fluid, were observed. In addition, the release of the ICD markers lactate dehydrogenase and ATP was detected after NIT-PIT. NIR-PIT significantly suppressed tumor growth in tumor-bearing mice. This study revealed that NIR-PIT targeting EGFR has therapeutic effects and induces ICD in osteosarcoma; thus, it is potentially a novel therapeutic strategy for primary and metastatic osteosarcoma.
Collapse
Affiliation(s)
- Motofumi Suzuki
- Division of Fundamental Technology Development, Near InfraRed Photo-ImmunoTherapy Research Institute at Kansai Medical University, 2-5-1, Shin-Machi, Hirakata, Osaka, 573-1010, Japan
| | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1088, United States
| | - Hirofumi Hanaoka
- Division of Fundamental Technology Development, Near InfraRed Photo-ImmunoTherapy Research Institute at Kansai Medical University, 2-5-1, Shin-Machi, Hirakata, Osaka, 573-1010, Japan.
| |
Collapse
|
2
|
McAloney CA, Makkawi R, Budhathoki Y, Cannon MV, Franz EM, Gross AC, Cam M, Vetter TA, Duhen R, Davies AE, Roberts RD. Host-derived growth factors drive ERK phosphorylation and MCL1 expression to promote osteosarcoma cell survival during metastatic lung colonization. Cell Oncol (Dordr) 2024; 47:259-282. [PMID: 37676378 PMCID: PMC10899530 DOI: 10.1007/s13402-023-00867-w] [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: 08/23/2023] [Indexed: 09/08/2023] Open
Abstract
PURPOSE For patients with osteosarcoma, disease-related mortality most often results from lung metastasis-a phenomenon shared with many solid tumors. While established metastatic lesions behave aggressively, very few of the tumor cells that reach the lung will survive. By identifying mechanisms that facilitate survival of disseminated tumor cells, we can develop therapeutic strategies that prevent and treat metastasis. METHODS We analyzed single cell RNA-sequencing (scRNAseq) data from murine metastasis-bearing lungs to interrogate changes in both host and tumor cells during colonization. We used these data to elucidate pathways that become activated in cells that survive dissemination and identify candidate host-derived signals that drive activation. We validated these findings through live cell reporter systems, immunocytochemistry, and fluorescent immunohistochemistry. We then validated the functional relevance of key candidates using pharmacologic inhibition in models of metastatic osteosarcoma. RESULTS Expression patterns suggest that the MAPK pathway is significantly elevated in early and established metastases. MAPK activity correlates with expression of anti-apoptotic genes, especially MCL1. Niche cells produce growth factors that increase ERK phosphorylation and MCL1 expression in tumor cells. Both early and established metastases are vulnerable to MCL1 inhibition, but not MEK inhibition in vivo. Combining MCL1 inhibition with chemotherapy both prevented colonization and eliminated established metastases in murine models of osteosarcoma. CONCLUSION Niche-derived growth factors drive MAPK activity and MCL1 expression in osteosarcoma, promoting metastatic colonization. Although later metastases produce less MCL1, they remain dependent on it. MCL1 is a promising target for clinical trials in both human and canine patients.
Collapse
Affiliation(s)
- Camille A McAloney
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Center for Childhood Cancers and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Rawan Makkawi
- Knight Cancer Institute's, Cancer Early Detection Advanced Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Yogesh Budhathoki
- Center for Childhood Cancers and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH, USA
| | - Matthew V Cannon
- Center for Childhood Cancers and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Emily M Franz
- Center for Childhood Cancers and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH, USA
| | - Amy C Gross
- Center for Childhood Cancers and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Maren Cam
- Center for Childhood Cancers and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Tatyana A Vetter
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Rebekka Duhen
- Knight Cancer Institute's, Cancer Early Detection Advanced Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Alexander E Davies
- Knight Cancer Institute's, Cancer Early Detection Advanced Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA.
| | - Ryan D Roberts
- Center for Childhood Cancers and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
- Division of Pediatric Hematology, Oncology, and BMT, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA.
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
3
|
Facchin C, Fraga-Timiraos AB, Schmitt J, Babaa N, Pannu N, Aliaga A, Larroque AL, Jean-Claude BJ. Molecular Analysis of the Superior Efficacy of a Dual Epidermal Growth Factor Receptor (EGFR)-DNA-Targeting Combi-Molecule in Comparison with Its Putative Prodrugs 6-Mono-Alkylamino- and 6,6-Dialkylaminoquinazoline in a Human Osteosarcoma Xenograft Model. Cells 2023; 12:914. [PMID: 36980255 PMCID: PMC10046901 DOI: 10.3390/cells12060914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023] Open
Abstract
Background: ZR2002 is a dual EGFR-DNA-targeting combi-molecule that carries a chloroethyl group at the six-position of the quinazoline ring designed to alkylate DNA. Despite its good pharmacokinetics, ZR2002 is metabolized in vivo into dechlorinated metabolites, losing the DNA-alkylating function required to damage DNA. To increase the DNA damage activity in tumor cells in vivo, we compared ZR2002 with two of its 6-N,N-disubstituted analogs: "JS61", with a nitrogen mustard function at the six-position of the quinazoline ring, and "JS84", with an N-methyl group. Methods: Tumor xenografts were performed with the human Saos-2 osteosarcoma cell line expressing EGFR. Mice were treated with ZR2002, JS84 or JS61, and the tumor burden was measured with a caliper and CT/PET imaging. Drug metabolism was analyzed with LC-MS. EGFR and ɣ-H2AX phosphorylation were quantified via Western blot analysis and immunohistochemistry. Results: In vivo analysis showed that significant tumor growth inhibition was only achieved when ZR2002 was administered in its naked form. The metabolic dealkylation of JS61 and JS84 did not release sufficient concentrations of ZR2002 for the intratumoral inhibition of P-EGFR or enhanced levels of P-H2AX. Conclusions: The results in toto suggest that intratumoral concentrations of intact ZR2002 are correlated with the highest inhibition of P-EGFR and induction of DNA damage in vivo. ZR2002 may well represent a good drug candidate for the treatment of EGFR-expressing osteosarcoma.
Collapse
Affiliation(s)
- Caterina Facchin
- Cancer Drug Research Laboratory, The Research Institute of the McGill University Health Center (RI-MUHC), Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
| | | | | | | | | | | | | | - Bertrand J. Jean-Claude
- Cancer Drug Research Laboratory, The Research Institute of the McGill University Health Center (RI-MUHC), Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
4
|
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
|
5
|
Mangiavini L, Peretti GM, Canciani B, Maffulli N. Epidermal growth factor signalling pathway in endochondral ossification: an evidence-based narrative review. Ann Med 2022; 54:37-50. [PMID: 34955078 PMCID: PMC8725985 DOI: 10.1080/07853890.2021.2015798] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
During endochondral bone development, a complex process that leads to the formation of the majority of skeletal elements, mesenchymal cells condense, differentiating into chondrocytes and producing the foetal growth plate. Chondrocytes progressively hypertrophy, induce angiogenesis and are then gradually replaced by bone. Epidermal Growth Factor (EGF), one of many growth factors, is the prototype of the EGF-ligand family, which comprises several proteins involved in cell proliferation, migration and survival. In bone, EGF pathway signalling finely tunes the first steps of chondrogenesis by maintaining mesenchymal cells in an undifferentiated stage, and by promoting hypertrophic cartilage replacement. Moreover, EGF signalling modulates bone homeostasis by stimulating osteoblast and osteoclast proliferation, and by regulating osteoblast differentiation under specific spatial and temporal conditions. This evidence-based narrative review describes the EGF pathway in bone metabolism and endochondral bone development. This comprehensive description may be useful in light of possible clinical applications in orthopaedic practice. A deeper knowledge of the role of EGF in bone may be useful in musculoskeletal conditions which may benefit from the modulation of this signalling pathway.Key messagesThe EGF pathway is involved in bone metabolism.EGF signalling is essential in the very early stages of limb development by maintaining cells in an undifferentiated stage.EGF pathway positively regulates chondrocyte proliferation, negatively modulates hypertrophy, and favours cartilage replacement by bone.EGF and EGF-like proteins finely tune the proliferation and differentiation of bone tissue cells, and they also regulate the initial phases of endochondral ossification.
Collapse
Affiliation(s)
- L Mangiavini
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milan, Italy
| | - G M Peretti
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milan, Italy
| | - B Canciani
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - N Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy.,Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Queen Mary University of London, London, UK.,School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, Stoke on Trent, UK
| |
Collapse
|
6
|
Feng Z, Ou Y, Hao L. The roles of glycolysis in osteosarcoma. Front Pharmacol 2022; 13:950886. [PMID: 36059961 PMCID: PMC9428632 DOI: 10.3389/fphar.2022.950886] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/25/2022] [Indexed: 12/02/2022] Open
Abstract
Metabolic reprogramming is of great significance in the progression of various cancers and is critical for cancer progression, diagnosis, and treatment. Cellular metabolic pathways mainly include glycolysis, fat metabolism, glutamine decomposition, and oxidative phosphorylation. In cancer cells, reprogramming metabolic pathways is used to meet the massive energy requirement for tumorigenesis and development. Metabolisms are also altered in malignant osteosarcoma (OS) cells. Among reprogrammed metabolisms, alterations in aerobic glycolysis are key to the massive biosynthesis and energy demands of OS cells to sustain their growth and metastasis. Numerous studies have demonstrated that compared to normal cells, glycolysis in OS cells under aerobic conditions is substantially enhanced to promote malignant behaviors such as proliferation, invasion, metastasis, and drug resistance of OS. Glycolysis in OS is closely related to various oncogenes and tumor suppressor genes, and numerous signaling pathways have been reported to be involved in the regulation of glycolysis. In recent years, a vast number of inhibitors and natural products have been discovered to inhibit OS progression by targeting glycolysis-related proteins. These potential inhibitors and natural products may be ideal candidates for the treatment of osteosarcoma following hundreds of preclinical and clinical trials. In this article, we explore key pathways, glycolysis enzymes, non-coding RNAs, inhibitors, and natural products regulating aerobic glycolysis in OS cells to gain a deeper understanding of the relationship between glycolysis and the progression of OS and discover novel therapeutic approaches targeting glycolytic metabolism in OS.
Collapse
|
7
|
Nanamiya R, Takei J, Ohishi T, Asano T, Tanaka T, Sano M, Nakamura T, Yanaka M, Handa S, Tateyama N, Harigae Y, Saito M, Suzuki H, Kawada M, Kaneko MK, Kato Y. Defucosylated Anti-Epidermal Growth Factor Receptor Monoclonal Antibody (134-mG 2a-f) Exerts Antitumor Activities in Mouse Xenograft Models of Canine Osteosarcoma. Monoclon Antib Immunodiagn Immunother 2022; 41:1-7. [PMID: 35225663 DOI: 10.1089/mab.2021.0036] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is a transmembrane glycoprotein. Although EGFR is physiologically essential in normal cells, it contributes to tumor malignancy through gene amplification and/or protein overexpression, which augment signaling cascades in tumor cells. We previously developed an anti-human EGFR (hEGFR) monoclonal antibody (mAb), EMab-134 (mouse IgG1, kappa), which detects hEGFR and dog EGFR (dEGFR) with high sensitivity and specificity. The mouse IgG2a version of EMab-134 (134-mG2a) has antitumor effects toward mouse xenografts of hEGFR-expressing oral squamous cell carcinomas. Furthermore, 134-mG2a-f, the defucosylated version of 134-mG2a, exhibits antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) in dEGFR-overexpressed CHO-K1 (CHO/dEGFR) cells and antitumor activities in mouse xenografts of CHO/dEGFR cells. Herein, the reactivity of 134-mG2a-f against canine cancer cells with endogenous dEGFR was first examined by flow cytometry and immunocytochemistry. In vitro analysis demonstrated that 134-mG2a-f highly exerted ADCC and CDC for a canine osteosarcoma cell line, D-17, which expresses endogenous dEGFR. Moreover, in vivo administration of 134-mG2a-f significantly suppressed the development of D-17 compared with the results in response to control mouse IgG. These results suggest that 134-mG2a-f exerts antitumor effects against dEGFR-expressing canine cancers, and could be valuable as part of an antibody treatment regimen for them.
Collapse
Affiliation(s)
- Ren Nanamiya
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junko Takei
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomokazu Ohishi
- Microbial Chemistry Research Foundation, Institute of Microbial Chemistry (BIKAKEN), Numazu-shi, Japan
| | - Teizo Asano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masato Sano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takuro Nakamura
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Miyuki Yanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Saori Handa
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nami Tateyama
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Harigae
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masaki Saito
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroyuki Suzuki
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Manabu Kawada
- Microbial Chemistry Research Foundation, Institute of Microbial Chemistry (BIKAKEN), Numazu-shi, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
8
|
Li G, Ohishi T, Kaneko MK, Takei J, Mizuno T, Kawada M, Saito M, Suzuki H, Kato Y. Defucosylated Mouse-Dog Chimeric Anti-EGFR Antibody Exerts Antitumor Activities in Mouse Xenograft Models of Canine Tumors. Cells 2021; 10:cells10123599. [PMID: 34944112 PMCID: PMC8700185 DOI: 10.3390/cells10123599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) contributes to tumor malignancy via gene amplification and protein overexpression. Previously, we developed an anti-human EGFR (hEGFR) monoclonal antibody, namely EMab-134, which detects hEGFR and dog EGFR (dEGFR) with high sensitivity and specificity. In this study, we produced a defucosylated mouse–dog chimeric anti-EGFR monoclonal antibody, namely E134Bf. In vitro analysis revealed that E134Bf highly exerted antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity against a canine osteosarcoma cell line (D-17) and a canine fibroblastic cell line (A-72), both of which express endogenous dEGFR. Moreover, in vivo administration of E134Bf significantly suppressed the development of D-17 and A-72 compared with the control dog IgG in mouse xenografts. These results indicate that E134Bf exerts antitumor effects against dEGFR-expressing canine cancers and could be valuable as part of an antibody treatment regimen for dogs.
Collapse
Affiliation(s)
- Guanjie Li
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (G.L.); (M.S.); (H.S.)
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, 18-24 Miyamoto, Numazu-shi 410-0301, Japan;
- Correspondence: (T.O.); (Y.K.); Tel.: +81-55-924-0601 (T.O.); +81-22-717-8207 (Y.K.)
| | - Mika K. Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (M.K.K.); (J.T.)
| | - Junko Takei
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (M.K.K.); (J.T.)
| | - Takuya Mizuno
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan;
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, 18-24 Miyamoto, Numazu-shi 410-0301, Japan;
| | - Masaki Saito
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (G.L.); (M.S.); (H.S.)
| | - Hiroyuki Suzuki
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (G.L.); (M.S.); (H.S.)
| | - Yukinari Kato
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (G.L.); (M.S.); (H.S.)
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (M.K.K.); (J.T.)
- Correspondence: (T.O.); (Y.K.); Tel.: +81-55-924-0601 (T.O.); +81-22-717-8207 (Y.K.)
| |
Collapse
|
9
|
Terry RL, Meyran D, Fleuren EDG, Mayoh C, Zhu J, Omer N, Ziegler DS, Haber M, Darcy PK, Trapani JA, Neeson PJ, Ekert PG. Chimeric Antigen Receptor T cell Therapy and the Immunosuppressive Tumor Microenvironment in Pediatric Sarcoma. Cancers (Basel) 2021; 13:cancers13184704. [PMID: 34572932 PMCID: PMC8465026 DOI: 10.3390/cancers13184704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/15/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary This review explores the current trials using cellular immunotherapies in pediatric sarcoma and describes examples of promising new CAR T targets in sarcoma that are in preclinical development. We provide insights into the ways in which the immunosuppressive tumor immune microenvironment can impact on CAR T cell therapy, highlighting specific mechanisms by which the tumor microenvironment may limit CAR T efficacy. Appreciation of these mechanisms may lead to rational combinations of immunotherapies, for example, the combination of CAR T cells with checkpoint inhibitor drugs. We also describe innovations in CAR T cell generation and combination therapies that may pave the way to better clinical outcomes for these patients. Abstract Sarcomas are a diverse group of bone and soft tissue tumors that account for over 10% of childhood cancers. Outcomes are particularly poor for children with refractory, relapsed, or metastatic disease. Chimeric antigen receptor T (CAR T) cells are an exciting form of adoptive cell therapy that potentially offers new hope for these children. In early trials, promising outcomes have been achieved in some pediatric patients with sarcoma. However, many children do not derive benefit despite significant expression of the targeted tumor antigen. The success of CAR T cell therapy in sarcomas and other solid tumors is limited by the immunosuppressive tumor microenvironment (TME). In this review, we provide an update of the CAR T cell therapies that are currently being tested in pediatric sarcoma clinical trials, including those targeting tumors that express HER2, NY-ESO, GD2, EGFR, GPC3, B7-H3, and MAGE-A4. We also outline promising new CAR T cells that are in pre-clinical development. Finally, we discuss strategies that are being used to overcome tumor-mediated immunosuppression in solid tumors; these strategies have the potential to improve clinical outcomes of CAR T cell therapy for children with sarcoma.
Collapse
Affiliation(s)
- Rachael L. Terry
- Children’s Cancer Institute, Randwick 2031, Australia; (R.L.T.); (E.D.G.F.); (C.M.); (D.S.Z.); (M.H.)
- School of Women and Children’s Health, University of New South Wales, Randwick 2052, Australia
| | - Deborah Meyran
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne 3000, Australia; (D.M.); (J.Z.); (P.K.D.); (J.A.T.); (P.J.N.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3000, Australia
- Inserm, Université de Paris, U976 HIPI Unit, Institut de Recherche Saint-Louis, 75475 Paris, France
| | - Emmy D. G. Fleuren
- Children’s Cancer Institute, Randwick 2031, Australia; (R.L.T.); (E.D.G.F.); (C.M.); (D.S.Z.); (M.H.)
- School of Women and Children’s Health, University of New South Wales, Randwick 2052, Australia
| | - Chelsea Mayoh
- Children’s Cancer Institute, Randwick 2031, Australia; (R.L.T.); (E.D.G.F.); (C.M.); (D.S.Z.); (M.H.)
- School of Women and Children’s Health, University of New South Wales, Randwick 2052, Australia
| | - Joe Zhu
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne 3000, Australia; (D.M.); (J.Z.); (P.K.D.); (J.A.T.); (P.J.N.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3000, Australia
| | - Natacha Omer
- Translational Innate Immunotherapy, University of Queensland Diamantina Institute (UQDI), Brisbane 4102, Australia;
- Oncology Services Group, Queensland Children Hospital, Brisbane 4101, Australia
| | - David S. Ziegler
- Children’s Cancer Institute, Randwick 2031, Australia; (R.L.T.); (E.D.G.F.); (C.M.); (D.S.Z.); (M.H.)
- School of Women and Children’s Health, University of New South Wales, Randwick 2052, Australia
- Kids Cancer Centre, Sydney Children’s Hospital, Randwick 2145, Australia
| | - Michelle Haber
- Children’s Cancer Institute, Randwick 2031, Australia; (R.L.T.); (E.D.G.F.); (C.M.); (D.S.Z.); (M.H.)
- School of Women and Children’s Health, University of New South Wales, Randwick 2052, Australia
| | - Phillip K. Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne 3000, Australia; (D.M.); (J.Z.); (P.K.D.); (J.A.T.); (P.J.N.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3000, Australia
| | - Joseph A. Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne 3000, Australia; (D.M.); (J.Z.); (P.K.D.); (J.A.T.); (P.J.N.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3000, Australia
| | - Paul J. Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne 3000, Australia; (D.M.); (J.Z.); (P.K.D.); (J.A.T.); (P.J.N.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3000, Australia
| | - Paul G. Ekert
- Children’s Cancer Institute, Randwick 2031, Australia; (R.L.T.); (E.D.G.F.); (C.M.); (D.S.Z.); (M.H.)
- School of Women and Children’s Health, University of New South Wales, Randwick 2052, Australia
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne 3000, Australia; (D.M.); (J.Z.); (P.K.D.); (J.A.T.); (P.J.N.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3000, Australia
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne 3052, Australia
- Correspondence:
| |
Collapse
|
10
|
He M, Yan G, Wang Y, Gong R, Lei H, Yu S, He X, Li G, Du W, Ma T, Gao M, Yu M, Liu S, Xu Z, Idiiatullina E, Zagidullin N, Pavlov V, Cai B, Yuan Y, Yang L. Blue LED causes autophagic cell death in human osteosarcoma by increasing ROS generation and dephosphorylating EGFR. J Cell Mol Med 2021; 25:4962-4973. [PMID: 33960631 PMCID: PMC8178260 DOI: 10.1111/jcmm.16412] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/29/2021] [Accepted: 02/08/2021] [Indexed: 12/16/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumour in adolescence. Lately, light‐emitting diodes (LED)‐based therapy has emerged as a new promising approach for several diseases. However, it remains unknown in human OS. Here, we found that the blue LED irradiation significantly suppressed the proliferation, migration and invasion of human OS cells, while we observed blue LED irradiation increased ROS production through increased NADPH oxidase enzymes NOX2 and NOX4, as well as decreased Catalase (CAT) expression levels. Furthermore, we revealed blue LED irradiation‐induced autophagy characterized by alterations in autophagy protein markers including Beclin‐1, LC3‐II/LC3‐I and P62. Moreover, we demonstrated an enhanced autophagic flux. The blockage of autophagy displayed a remarkable attenuation of anti‐tumour activities of blue LED irradiation. Next, ROS scavenger N‐acetyl‐L‐cysteine (NAC) and NOX inhibitor diphenyleneiodonium (DPI) blocked suppression of OS cell growth, indicating that ROS accumulation might play an essential role in blue LED‐induced autophagic OS cell death. Additionally, we observed blue LED irradiation decreased EGFR activation (phosphorylation), which in turn led to Beclin‐1 release and subsequent autophagy activation in OS cells. Analysis of EGFR colocalization with Beclin‐1 and EGFR‐immunoprecipitation (IP) assay further revealed the decreased interaction of EGFR and Beclin‐1 upon blue LED irradiation in OS cells. In addition, Beclin‐1 down‐regulation abolished the effects of blue LED irradiation on OS cells. Collectively, we concluded that blue LED irradiation exhibited anti‐tumour effects on OS by triggering ROS and EGFR/Beclin‐1‐mediated autophagy signalling pathway, representing a potential approach for human OS treatment.
Collapse
Affiliation(s)
- Mingyu He
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Gege Yan
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Yang Wang
- Department of Orthopedics, Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Rui Gong
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Hong Lei
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Shuting Yu
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Xiaoqi He
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Guanghui Li
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Weijie Du
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China.,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, China
| | - Tianshuai Ma
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Manqi Gao
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Meixi Yu
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Shenzhen Liu
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Zihang Xu
- Department of Pharmacology, College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Elina Idiiatullina
- Central Laboratory of Scientific Research, Bashkir State Medical University, Ufa, Russia
| | - Naufal Zagidullin
- Central Laboratory of Scientific Research, Bashkir State Medical University, Ufa, Russia
| | - Valentin Pavlov
- Central Laboratory of Scientific Research, Bashkir State Medical University, Ufa, Russia
| | - Benzhi Cai
- Department of Orthopedics, Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China.,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, China
| | - Ye Yuan
- Department of Orthopedics, Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China.,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, China
| | - Lei Yang
- Department of Orthopedics, Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
11
|
Hernández IB, Kromhout JZ, Teske E, Hennink WE, van Nimwegen SA, Oliveira S. Molecular targets for anticancer therapies in companion animals and humans: what can we learn from each other? Theranostics 2021; 11:3882-3897. [PMID: 33664868 PMCID: PMC7914358 DOI: 10.7150/thno.55760] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
Despite clinical successes in the treatment of some early stage cancers, it is undeniable that novel and innovative approaches are needed to aid in the fight against cancer. Targeted therapies offer the desirable feature of tumor specificity while sparing healthy tissues, thereby minimizing side effects. However, the success rate of translation of these therapies from the preclinical setting to the clinic is dramatically low, highlighting an important point of necessary improvement in the drug development process in the oncology field. The practice of a comparative oncology approach can address some of the current issues, by introducing companion animals with spontaneous tumors in the linear drug development programs. In this way, animals from the veterinary clinic get access to novel/innovative therapies, otherwise inaccessible, while generating robust data to aid therapy refinement and increase translational success. In this review, we present an overview of targetable membrane proteins expressed in the most well-characterized canine and feline solid cancers, greatly resembling the counterpart human malignancies. We identified particular areas in which a closer collaboration between the human and veterinary clinic would benefit both human and veterinary patients. Considerations and challenges to implement comparative oncology in the development of anticancer targeted therapies are also discussed.
Collapse
|
12
|
Lilienthal I, Herold N. Targeting Molecular Mechanisms Underlying Treatment Efficacy and Resistance in Osteosarcoma: A Review of Current and Future Strategies. Int J Mol Sci 2020; 21:ijms21186885. [PMID: 32961800 PMCID: PMC7555161 DOI: 10.3390/ijms21186885] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma is the most common primary malignant bone tumour in children and adolescents. Due to micrometastatic spread, radical surgery alone rarely results in cure. Introduction of combination chemotherapy in the 1970s, however, dramatically increased overall survival rates from 20% to approximately 70%. Unfortunately, large clinical trials aiming to intensify treatment in the past decades have failed to achieve higher cure rates. In this review, we revisit how the heterogenous nature of osteosarcoma as well as acquired and intrinsic resistance to chemotherapy can account for stagnation in therapy improvement. We summarise current osteosarcoma treatment strategies focusing on molecular determinants of treatment susceptibility and resistance. Understanding therapy susceptibility and resistance provides a basis for rational therapy betterment for both identifying patients that might be cured with less toxic interventions and targeting resistance mechanisms to sensitise resistant osteosarcoma to conventional therapies.
Collapse
Affiliation(s)
- Ingrid Lilienthal
- Division of Paediatric Oncology, Department of Women’s and Children’s Health, Karolinska Institutet, SE-171 76 Stockholm, Sweden
- Correspondence: (I.L.); (N.H.); Tel.: +46-(0)8-52483204 (I.L. & N.H.)
| | - Nikolas Herold
- Division of Paediatric Oncology, Department of Women’s and Children’s Health, Karolinska Institutet, SE-171 76 Stockholm, Sweden
- Paediatric Oncology, Astrid Lindgren’s Children Hospital, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
- Correspondence: (I.L.); (N.H.); Tel.: +46-(0)8-52483204 (I.L. & N.H.)
| |
Collapse
|
13
|
Targeted MEK inhibition by cobimetinib enhances doxorubicin's efficacy in osteosarcoma models. Biochem Biophys Res Commun 2020; 529:622-628. [PMID: 32736683 DOI: 10.1016/j.bbrc.2020.06.082] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 12/31/2022]
Abstract
The limited effectiveness and high toxicity of current treatments in osteosarcoma necessitate new therapeutic strategy. Cobimetinib is a FDA-approved MEK inhibitor and is clinically used in combination with standard of care to treat melanomas. Here, we report that targeted MEK inhibition by cobimetinib enhances doxorubicin's efficacy in osteosarcoma models. We found that cobimetinib potently inhibited growth and survival of osteosarcoma cells. We revealed that cobimetinib had anti-metastasis activity as it inhibited osteosarcoma cell migration. Notably, the effective concentrations of cobimetinib are clinically achievable. We further found that cells with the most sensitivity had highest p-ERK and cells with the least sensitivity had lowest p-ERK, suggesting the possible correlation of ERK activation with cobimetinib sensitivity in osteosarcoma. We further confirmed that inhibition of MEK/ERK signaling pathway is the mechanism of cobimetinib's action in osteosarcoma, leading to inhibition of focal adhesion kinase (FAK) and anti-apoptotic pathway, as well as activation of pro-apoptotic pathway. Using xenograft mice model, we found that cobimetinib at the tolerable dose significantly inhibited osteosarcoma formation and growth. In addition, the combination of cobimetinib and doxorubicin at sublethal dose completely arrested tumor growth without further progression. The ability of cobimetinib in enhancing doxorubicin's efficacy in osteosarcoma models makes cobimetinib as a useful addition to the treatment armamentarium for osteosarcoma. Our findings also emphasize the therapeutic value of MEK/ERK pathway to improve the clinical management of osteosarcoma.
Collapse
|
14
|
Afatinib is active in osteosarcoma in osteosarcoma cell lines. J Cancer Res Clin Oncol 2020; 146:1693-1700. [PMID: 32333142 DOI: 10.1007/s00432-020-03220-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/15/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Osteosarcoma is the most common bone tumor, mainly affecting adolescents and young adults, and metastatic disease has poor outcomes with a dismal overall survival. Currently, chemotherapy is the standard of care with limited results, finding that new therapies could improve these outcomes. Preclinical and clinical studies have suggested a possible important role of ErbB pathway aberrations in osteosarcoma etiology. The present study shows the effect of afatinib, an irreversible ErbB family blocker in osteosarcoma cell lines. METHODS Within a panel of human osteosarcoma cell lines, we addressed cell viability assay using afatinib at increasing concentrations. Motility was measured in wound-healing assays and invasion capacity was assessed in Transwell chamber assays. Finally, to monitor ErbB pathway modulation by afatinib and related compounds, we used Western blot analyses. RESULTS Cell viability inhibition, as well as a reduction of motility and migration of osteosarcoma cell line were observed after treatment with afatinib. Likewise, in the HOS cell line, afatinib decreased phosphorylation of key components in the ErbB signaling pathway. CONCLUSIONS Afatinib shows relevant antitumor effect in several osteosarcoma cell lines, as it causes a significant impact on cell viability, motility, and migration with a significant decrease in the activation of ErbB pathway activity.
Collapse
|
15
|
Breen L, Gaule PB, Canonici A, Walsh N, Collins DM, Cremona M, Hennessy BT, Duffy MJ, Crown J, Donovan NO, Eustace AJ. Targeting c-Met in triple negative breast cancer: preclinical studies using the c-Met inhibitor, Cpd A. Invest New Drugs 2020; 38:1365-1372. [PMID: 32318883 DOI: 10.1007/s10637-020-00937-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/07/2020] [Indexed: 11/26/2022]
Abstract
Introduction Triple negative breast cancer (TNBC) represents a heterogeneous subtype of breast cancer that carries a poorer prognosis. There remains a need to identify novel drivers of TNBC, which may represent targets to treat the disease. c-Met overexpression is linked with decreased survival and is associated with the basal subtype of breast cancer. Cpd A, a kinase inhibitor selective/specific for Met kinase has demonstrated preclinical anti-cancer efficacy in TNBC. We aimed to assess the anti-cancer efficacy of Cpd A when combined with Src kinase, ErbB-family or hepatocyte growth factor (HGF) inhibitors in TNBC cell lines. Methods We determined the anti-proliferative effects of Cpd A, rilotumumab, neratinib and saracatinib tested alone and in combination in a panel of TNBC cells by acid phosphatase assays. We performed reverse phase protein array analysis of c-Met and IGF1Rβ expression and phosphorylation of c-Met (Y1234/1235) in TNBC cells and correlated their expression/phosphorylation with Cpd A sensitivity. We examined the impact of Cpd A, neratinib and saracatinib tested alone and in combination on invasive potential and colony formation.Results TNBC cells are not inherently sensitive to Cpd A, and neither c-Met expression nor phosphorylation are biomarkers of sensitivity to Cpd A. Cpd A enhanced the anti-proliferative effects of neratinib in vitro; however, this effect was limited to cell lines with innate sensitivity to Cpd A. Cpd A had limited anti-invasive effects but it reduced colony formation in the TNBC cell line panel.Conclusions Despite Cpd A having a potential role in reducing cancer cell metastasis, identification of strong predictive biomarkers of c-Met sensitivity would be essential to the development of a c-Met targeted treatment for an appropriately selected cohort of TNBC patients.
Collapse
Affiliation(s)
- Laura Breen
- Molecular Therapeutics for Cancer in Ireland, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Patricia B Gaule
- Molecular Therapeutics for Cancer in Ireland, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Alexandra Canonici
- Molecular Therapeutics for Cancer in Ireland, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Naomi Walsh
- Molecular Therapeutics for Cancer in Ireland, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Denis M Collins
- Molecular Therapeutics for Cancer in Ireland, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Mattia Cremona
- Medical Oncology Group, Department of Molecular Medicine, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Bryan T Hennessy
- Medical Oncology Group, Department of Molecular Medicine, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Michael J Duffy
- UCD Clinical Research Centre, St. Vincent's University Hospital, Dublin, Ireland
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - John Crown
- Molecular Therapeutics for Cancer in Ireland, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Norma O' Donovan
- Molecular Therapeutics for Cancer in Ireland, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Alex J Eustace
- Molecular Therapeutics for Cancer in Ireland, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland.
| |
Collapse
|
16
|
Folkert IW, Devalaraja S, Linette GP, Weber K, Haldar M. Primary Bone Tumors: Challenges and Opportunities for CAR-T Therapies. J Bone Miner Res 2019; 34:1780-1788. [PMID: 31441962 DOI: 10.1002/jbmr.3852] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/10/2019] [Accepted: 07/27/2019] [Indexed: 12/22/2022]
Abstract
Primary malignant bone tumors are rare, occur in all age groups, and include distinct entities such as osteosarcoma, Ewing sarcoma, and chondrosarcoma. Traditional treatment with some combination of chemotherapy, surgery, and radiation has reached the limit of efficacy, with substantial room for improvement in patient outcome. Furthermore, genomic characterization of these tumors reveals a paucity of actionable molecular targets. Against this backdrop, recent advances in cancer immunotherapy represent a silver lining in the treatment of primary bone cancer. Major strategies in cancer immunotherapy include stimulating naturally occurring anti-tumor T cells and adoptive transfer of tumor-specific cytotoxic T cells. Chimeric antigen receptor T cells (CAR-T cells) belong to the latter strategy and are an impressive application of both insights into T cell biology and advances in genetic engineering. In this review, we briefly describe the CAR-T approach and discuss its applications in primary bone tumors. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ian W Folkert
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samir Devalaraja
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gerald P Linette
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristy Weber
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Penn Sarcoma Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Malay Haldar
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Penn Sarcoma Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
17
|
Linder M, Glitzner E, Srivatsa S, Bakiri L, Matsuoka K, Shahrouzi P, Dumanic M, Novoszel P, Mohr T, Langer O, Wanek T, Mitterhauser M, Wagner EF, Sibilia M. EGFR is required for FOS-dependent bone tumor development via RSK2/CREB signaling. EMBO Mol Med 2019; 10:emmm.201809408. [PMID: 30361264 PMCID: PMC6220323 DOI: 10.15252/emmm.201809408] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma (OS) is a rare tumor of the bone occurring mainly in young adults accounting for 5% of all childhood cancers. Because of the limited therapeutic options, there has been no survival improvement for OS patients in the past 40 years. The epidermal growth factor receptor (EGFR) is highly expressed in OS; however, its clinical relevance is unclear. Here, we employed an autochthonous c‐Fos‐dependent OS mouse model (H2‐c‐fosLTR) and human OS tumor biopsies for preclinical studies aimed at identifying novel biomarkers and therapeutic benefits of anti‐EGFR therapies. We show that EGFR deletion/inhibition results in reduced tumor formation in H2‐c‐fosLTR mice by directly inhibiting the proliferation of cancer‐initiating osteoblastic cells by a mechanism involving RSK2/CREB‐dependent c‐Fos expression. Furthermore, OS patients with co‐expression of EGFR and c‐Fos exhibit reduced overall survival. Preclinical studies using human OS xenografts revealed that only tumors expressing both EGFR and c‐Fos responded to anti‐EGFR therapy demonstrating that c‐Fos can be considered as a novel biomarker predicting response to anti‐EGFR treatment in OS patients.
Collapse
Affiliation(s)
- Markus Linder
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Glitzner
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Sriram Srivatsa
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Latifa Bakiri
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | | | - Parastoo Shahrouzi
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Monika Dumanic
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Philipp Novoszel
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Thomas Mohr
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.,Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.,Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Wanek
- Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Markus Mitterhauser
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.,LBI Applied Diagnostics, Vienna, Austria
| | - Erwin F Wagner
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Maria Sibilia
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Yuan Y, Wang Y, Liu Z, Sun Y, Yao Y, Yu W, Shen Z. MAT2B promotes proliferation and inhibits apoptosis in osteosarcoma by targeting epidermal growth factor receptor and proliferating cell nuclear antigen. Int J Oncol 2019; 54:2019-2029. [PMID: 30942439 PMCID: PMC6521932 DOI: 10.3892/ijo.2019.4764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/12/2018] [Indexed: 12/16/2022] Open
Abstract
Osteosarcoma (OS) is the most commonly diagnosed bone tumor in young people with poor prognosis. At present, the mechanisms underlying tumorigenesis in OS are not well understood. The methionine adnosyltransferase 2B (MAT2B) gene encodes the regulatory subunit of methionine adenosyltransferase (MAT). Recent studies demonstrated that it is highly expressed in a number of human malignancies; however, is undefined in OS. In the present study, MAT2B expression was investigated in tumor samples and cell lines. In vivo and in vitro, lentivirus‑mediated small hairpin RNA was constructed to target the MAT2B gene and examine the role of MAT2B in OS proliferation. Microarray analysis was performed to examine the possible downstream molecular target of MAT2B in OS. MAT2B was markedly increased in OS specimens compared with the normal bone tissues, and it was additionally abundantly expressed in OS cell lines. Inhibition of MAT2B expression caused a marked decrease in proliferation and significant increase in apoptosis. In vivo, MAT2B silencing significantly inhibited OS cell growth. Microarray analysis suggested that epidermal growth factor receptor (EGFR) and proliferating cell nuclear antigen (PCNA) may function as downstream targets of MAT2B in OS, as confirmed by reverse transcription‑quantitative polymerase chain reaction assays and western blotting. Collectively, these results suggested that MAT2B serves a critical role in the proliferation of OS by regulating EGFR and PCNA and that it may be a potential therapeutic target and prognostic factor of OS.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yonggang Wang
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Zimei Liu
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yong Sun
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yang Yao
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Wenxi Yu
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Zan Shen
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
19
|
Advanced development of ErbB family-targeted therapies in osteosarcoma treatment. Invest New Drugs 2018; 37:175-183. [DOI: 10.1007/s10637-018-0684-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 10/16/2018] [Indexed: 01/06/2023]
|
20
|
Rho A Regulates Epidermal Growth Factor-Induced Human Osteosarcoma MG63 Cell Migration. Int J Mol Sci 2018; 19:ijms19051437. [PMID: 29751634 PMCID: PMC5983621 DOI: 10.3390/ijms19051437] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/04/2018] [Accepted: 05/05/2018] [Indexed: 11/17/2022] Open
Abstract
Osteosarcoma, the most common primary bone tumor, occurs most frequently in children and adolescents and has a 5-year survival rate, which is unsatisfactory. As epidermal growth factor receptor (EGFR) positively correlates with TNM (tumor-node-metastasis) stage in osteosarcoma, EGFR may play an important role in its progression. The purpose of this study was to explore potential mechanisms underlying this correlation. We found that EGF promotes MG63 cell migration and invasion as well as stress fiber formation via Rho A activation and that these effects can be reversed by inhibiting Rho A expression. In addition, molecules downstream of Rho A, including ROCK1, LIMK2, and Cofilin, are activated by EGF in MG63 cells, leading to actin stress fiber formation and cell migration. Moreover, inhibition of ROCK1, LIMK2, or Cofilin in MG63 cells using known inhibitors or short hairpin RNA (shRNA) prevents actin stress fiber formation and cell migration. Thus, we conclude that Rho A/ROCK1/LIMK2/Cofilin signaling mediates actin microfilament formation in MG63 cells upon EGFR activation. This novel pathway provides a promising target for preventing osteosarcoma progression and for treating this cancer.
Collapse
|
21
|
Sheng J, Yin M, Sun Z, Kang X, Liu D, Jiang K, Xu J, Zhao F, Guo Q, Zheng W. SPC24 promotes osteosarcoma progression by increasing EGFR/MAPK signaling. Oncotarget 2017; 8:105276-105283. [PMID: 29285250 PMCID: PMC5739637 DOI: 10.18632/oncotarget.22167] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/29/2017] [Indexed: 12/18/2022] Open
Abstract
In this study, we investigated the role of the spindle checkpoint protein SPC24 in osteosarcoma progression. SPC24 knockdown in 143B and U2OS osteosarcoma cells decreased cell growth, survival and invasiveness. The SPC24 knockdown cells also exhibited low EGFR, Ras and phospho-ERK levels and high E-cadherin levels, suggesting inhibition of EGFR/Ras/ERK signaling and epithelial-to-mesenchymal transitioning. Xenografted SPC24 knockdown osteosarcoma cells showed reduced tumor growth in nude mice with decreased EGFR and phospho-ERK levels and increased E-cadherin levels. By contrast, human osteosarcoma tissue samples showed high SPC24 and phospho-ERK levels and low E-cadherin levels. These results suggest SPC24 promotes osteosarcoma progression by increasing EGFR/Ras/ERK signaling.
Collapse
Affiliation(s)
- Jun Sheng
- Department of Orthopedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Mengchen Yin
- Department of Orthopedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhengwang Sun
- Department of Orthopedics, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Xia Kang
- Department of Orthopedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Da Liu
- Department of Orthopedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Kai Jiang
- Department of Orthopedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Jia Xu
- Department of Personnel Office, Traditional Chinese Medical Hospital of Zhuji, Zhuji, Zhejiang, China
| | - Feixing Zhao
- Department of Pathology, Zhuji People's Hospital of Zhejiang Province, Zhuji, Zhejiang, China
| | - Qunfeng Guo
- Department of Orthopedics, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Wei Zheng
- Department of Orthopedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| |
Collapse
|
22
|
Targeting pediatric sarcoma with a bispecific ligand immunotoxin targeting urokinase and epidermal growth factor receptors. Oncotarget 2017; 9:11938-11947. [PMID: 29552283 PMCID: PMC5844719 DOI: 10.18632/oncotarget.21187] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/07/2017] [Indexed: 12/11/2022] Open
Abstract
Children with high risk sarcoma have a poor prognosis despite surgical resection, irradiation and chemotherapy. Alternative therapies are urgently needed. Urokinase-type plasminogen activator receptor (uPAR) and epidermal growth factor receptor (EGFR) are surface proteins expressed by some pediatric sarcomas. We show for the first time that a de-immunized bispecific ligand toxin, EGFATFKDEL, directed against EGFR and uPAR, successfully targets pediatric sarcoma. Using flow cytometry, we identified a rhabdomyosarcoma (RMS) cell line, RH30, that expresses both uPAR and EGFR, and a Ewing sarcoma (EWS) cell line, TC-71, that expresses only uPAR. We tested the differential sensitivity of these two sarcoma cell lines to toxin-induced killing, using both in vitro assays and an in vivo murine model. We show that pediatric sarcomas are highly sensitive to EGFATFKDEL (at subnanomolar concentrations) in vitro. In vivo, tumor growth was significantly attenuated after treatment with EGFTFKDEL, compared to untreated controls, in both RH30 and TC-71 tumor bearing mice. In addition, we found that simultaneously targeting both receptors in a dual positive cell line was more effective than targeting a single receptor or antigen, resulting in a greater tumor response, including complete tumor regression in an animal model of bulky disease. Our findings provide support for further exploration of bispecific targeting of pediatric sarcomas with bispecific ligand toxins, such as EGFATFKDEL.
Collapse
|
23
|
Amphiregulin enhances intercellular adhesion molecule-1 expression and promotes tumor metastasis in human osteosarcoma. Oncotarget 2016; 6:40880-95. [PMID: 26503469 PMCID: PMC4747375 DOI: 10.18632/oncotarget.5679] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 09/13/2015] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is a common, high malignant, and metastatic bone cancer. Amphiregulin (AREG) has been associated with cancer cellular activities. However, the effect of AREG on metastasis activity in human osteosarcoma cells has yet to be determined. We determined that AREG increases the expression of intercellular adhesion molecule-1 (ICAM-1) through PI3K/Akt signaling pathway via its interaction with the epidermal growth factor receptor, thus resulting in the enhanced cell migration of osteosarcoma. Furthermore, AREG stimulation increased the association of NF-κB to ICAM-1 promoter which then up-regulated ICAM-1 expression. Finally, we observed that shRNA silencing of AREG decreased osteosarcoma metastasis in vivo. Our findings revealed a relationship between osteosarcoma metastatic potential and AREG expression and the modulating effect of AREG on ICAM-1 expression.
Collapse
|
24
|
Mantovani FB, Morrison JA, Mutsaers AJ. Effects of epidermal growth factor receptor kinase inhibition on radiation response in canine osteosarcoma cells. BMC Vet Res 2016; 12:82. [PMID: 27245053 PMCID: PMC4888507 DOI: 10.1186/s12917-016-0707-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 05/24/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Radiation therapy is a palliative treatment modality for canine osteosarcoma, with transient improvement in analgesia observed in many cases. However there is room for improvement in outcome for these patients. It is possible that the addition of sensitizing agents may increase tumor response to radiation therapy and prolong quality of life. Epidermal growth factor receptor (EGFR) expression has been documented in canine osteosarcoma and higher EGFR levels have been correlated to a worse prognosis. However, effects of EGFR inhibition on radiation responsiveness in canine osteosarcoma have not been previously characterized. This study examined the effects of the small molecule EGFR inhibitor erlotinib on canine osteosarcoma radiation responses, target and downstream protein expression in vitro. Additionally, to assess the potential impact of treatment on tumor angiogenesis, vascular endothelial growth factor (VEGF) levels in conditioned media were measured. RESULTS Erlotinib as a single agent reduced clonogenic survival in two canine osteosarcoma cell lines and enhanced the impact of radiation in one out of three cell lines investigated. In cell viability assays, erlotinib enhanced radiation effects and demonstrated single agent effects. Erlotinib did not alter total levels of EGFR, nor inhibit downstream protein kinase B (PKB/Akt) activation. On the contrary, erlotinib treatment increased phosphorylated Akt in these osteosarcoma cell lines. VEGF levels in conditioned media increased after erlotinib treatment as a single agent and in combination with radiation in two out of three cell lines investigated. However, VEGF levels decreased with erlotinib treatment in the third cell line. CONCLUSIONS Erlotinib treatment promoted modest enhancement of radiation effects in canine osteosarcoma cells, and possessed activity as a single agent in some cell lines, indicating a potential role for EGFR inhibition in the treatment of a subset of osteosarcoma patients. The relative radioresistance of osteosarcoma cells does not appear to be related to EGFR signalling exclusively. Angiogenic responses to radiation and kinase inhibitors are similarly likely to be multifactorial and require further investigation.
Collapse
Affiliation(s)
- Fernanda B Mantovani
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Jodi A Morrison
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Anthony J Mutsaers
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada.
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
25
|
Baranski Z, Booij TH, Kuijjer ML, de Jong Y, Cleton-Jansen AM, Price LS, van de Water B, Bovée JVMG, Hogendoorn PCW, Danen EHJ. MEK inhibition induces apoptosis in osteosarcoma cells with constitutive ERK1/2 phosphorylation. Genes Cancer 2016; 6:503-12. [PMID: 26807203 PMCID: PMC4701229 DOI: 10.18632/genesandcancer.91] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Conventional high-grade osteosarcoma is the most common primary bone cancer with relatively high incidence in young people. Recurrent and metastatic tumors are difficult to treat. We performed a kinase inhibitor screen in two osteosarcoma cell lines, which identified MEK1/2 inhibitors. These inhibitors were further validated in a panel of six osteosarcoma cell lines. Western blot analysis was performed to assess ERK activity and efficacy of MEK inhibition. A 3D culture system was used to validate results from 2D monolayer cultures. Gene expression analysis was performed to identify differentially expressed gene signatures in sensitive and resistant cell lines. Activation of the AKT signaling network was explored using Western blot and pharmacological inhibition. In the screen, Trametinib, AZD8330 and TAK-733 decreased cell viability by more than 50%. Validation in six osteosarcoma cell lines identified three cell lines as resistant and three as sensitive to the inhibitors. Western blot analysis of ERK activity revealed that sensitive lines had high constitutive ERK activity. Treatment with the three MEK inhibitors in a 3D culture system validated efficacy in inhibition of osteosarcoma viability. MEK1/2 inhibition represents a candidate treatment strategy for osteosarcomas displaying high MEK activity as determined by ERK phosphorylation status.
Collapse
Affiliation(s)
- Zuzanna Baranski
- Division of Toxicology, Leiden/Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Tijmen H Booij
- Division of Toxicology, Leiden/Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Marieke L Kuijjer
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yvonne de Jong
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Leo S Price
- Division of Toxicology, Leiden/Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Bob van de Water
- Division of Toxicology, Leiden/Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Judith V M G Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Erik H J Danen
- Division of Toxicology, Leiden/Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
26
|
Sevelda F, Mayr L, Kubista B, Lötsch D, van Schoonhoven S, Windhager R, Pirker C, Micksche M, Berger W. EGFR is not a major driver for osteosarcoma cell growth in vitro but contributes to starvation and chemotherapy resistance. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:134. [PMID: 26526352 PMCID: PMC4630894 DOI: 10.1186/s13046-015-0251-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/23/2015] [Indexed: 11/28/2022]
Abstract
Background Enhanced signalling via the epidermal growth factor receptor (EGFR) is a hallmark of multiple human carcinomas. However, in recent years data have accumulated that EGFR might also be hyperactivated in human sarcomas. Aim of this study was to investigate the influence of EGFR inhibition on cell viability and its interaction with chemotherapy response in osteosarcoma cell lines. Methods We have investigated a panel of human osteosarcoma cell lines regarding EGFR expression and downstream signalling. To test its potential applicability as therapeutic target, inhibition of EGFR by gefitinib was combined with osteosarcoma chemotherapeutics and cell viability, migration, and cell death assays were performed. Results Osteosarcoma cells expressed distinctly differing levels of functional EGFR reaching in some cases high amounts. Functionality of EGFR in osteosarcoma cells was proven by EGF-mediated activation of both MAPK and PI3K/AKT pathway (determined by phosphorylation of ERK1/2, AKT, S6, and GSK3β). The EGFR-specific inhibitor gefitinib blocked EGF-mediated downstream signal activation. At standard in vitro culture conditions, clinically achievable gefitinib doses demonstrated only limited cytotoxic activity, however, significantly reduced long-term colony formation and cell migration. In contrast, under serum-starvation conditions active gefitinib doses were distinctly reduced while EGF promoted starvation survival. Importantly, gefitinib significantly supported the anti-osteosarcoma activities of doxorubicin and methotrexate regarding cell survival and migratory potential. Conclusion Our data suggest that EGFR is not a major driver for osteosarcoma cell growth but contributes to starvation- and chemotherapy-induced stress survival. Consequently, combination approaches including EGFR inhibitors should be evaluated for treatment of high-grade osteosarcoma patients. Electronic supplementary material The online version of this article (doi:10.1186/s13046-015-0251-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Florian Sevelda
- Department of Orthopaedics, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria. .,Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, Borschkegasse 8a, 1090, Vienna, Austria.
| | - Lisa Mayr
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, Borschkegasse 8a, 1090, Vienna, Austria.
| | - Bernd Kubista
- Department of Orthopaedics, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| | - Daniela Lötsch
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, Borschkegasse 8a, 1090, Vienna, Austria.
| | - Sushilla van Schoonhoven
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, Borschkegasse 8a, 1090, Vienna, Austria.
| | - Reinhard Windhager
- Department of Orthopaedics, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, Borschkegasse 8a, 1090, Vienna, Austria.
| | - Michael Micksche
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, Borschkegasse 8a, 1090, Vienna, Austria.
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, Borschkegasse 8a, 1090, Vienna, Austria.
| |
Collapse
|
27
|
Wang H, Yang Q, Fu Z, Zuo D, Hua Y, Cai Z. ErbB receptors as prognostic and therapeutic drug targets in bone and soft tissue sarcomas. Cancer Invest 2014; 32:533-42. [PMID: 25347730 DOI: 10.3109/07357907.2014.964409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
ErbB receptors have been intensely studied to understand their importance in cancer biology and as therapeutic targets, and many ErbB inhibitors are now used in the clinical setting. A large number of studies have been conducted to examine the expression of ErbB family members in bone and soft tissue sarcomas, including osteosarcomas, synovial sarcomas, Ewing sarcomas, rhabdomyosarcomas, and so on. Nevertheless, the clinical implications of ErbB receptors remain elusive. To illustrate the potential of ErbB family members as prognostic and therapeutic drug targets in bone and soft tissue sarcomas, we summarized the molecular evidence and observations from clinical and basic trials.
Collapse
Affiliation(s)
- Hongsheng Wang
- 1Department of Orthopedics, Shanghai 1st People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
28
|
Kasza A. IL-1 and EGF regulate expression of genes important in inflammation and cancer. Cytokine 2013; 62:22-33. [PMID: 23481102 DOI: 10.1016/j.cyto.2013.02.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/09/2013] [Accepted: 02/05/2013] [Indexed: 02/08/2023]
Abstract
This review focuses on the mechanisms by which the expression of specific genes is regulated by two proteins that are important in inflammation and cancer, namely the pro-inflammatory cytokine interleukin (IL)-1β and epidermal growth factor (EGF). In the review the receptors that recognize factors that cause inflammation are described with main focus on the receptors associated with activation of IL-1β. The function of IL-1β and pathways leading to activation of transcription factors, particularly NFκB and Elk-1 are analyzed. Then the mechanisms of EGF action, with particular emphasis of the activation of Elk-1 are illustrated. The link between aberrant signaling of EGF receptor family members and cancer development is explained. The relationship between inflammation and tumorigenesis is discussed.
Collapse
Affiliation(s)
- Aneta Kasza
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland.
| |
Collapse
|
29
|
Lee JA, Ko Y, Kim DH, Lim JS, Kong CB, Cho WH, Jeon DG, Lee SY, Koh JS. Epidermal growth factor receptor: is it a feasible target for the treatment of osteosarcoma? Cancer Res Treat 2012; 44:202-9. [PMID: 23091447 PMCID: PMC3467424 DOI: 10.4143/crt.2012.44.3.202] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 06/27/2012] [Indexed: 11/23/2022] Open
Abstract
Purpose Features of epidermal growth factor receptor (EGFR) expression in osteosarcoma and in vitro efficacies of EGFR inhibitors against osteosarcoma cells were evaluated. Materials and Methods Thirty biopsy samples of osteosarcoma patients were retrospectively analyzed for EGFR protein expression by immunohistochemistry. Relationships between EGFR expression and clinicopathologic characteristics and treatment outcomes were evaluated. Four osteosarcoma cell lines were analyzed for EGFR and p-EGFR expression by western blotting. Efficacies of gefitinib and BIBW2992 on osteosarcoma cells were evaluated using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Tyrosine kinase domains in exons 18 to 21 were sequenced and gene expression analyses of EGFR and PTEN were performed in four osteosarcoma cell lines. Results EGFR protein was expressed in 27 (90%) samples (6 low, 12 intermediate, 9 high) and in three cell lines. Intermediate or high staining for EGFR was related to a tumor volume<150 mL (p<0.001) and histologic subtype other than osteoblastic type (p=0.03). However, EGFR expression was not associated with histologic response to preoperative chemotherapy or survival. Gefitinib and BIBW 2992 did not have any significant inhibitory effect on cell viabilities. DNA sequencing analysis revealed three osteosarcoma cell lines have single base changes at codon 2361 of exon 20 (G to A), without affecting translation results. Furthermore, no mutation was found to be associated with constitutive EGFR activation. Conclusion In the present study, gefitinib and BIBW2992 were not effective against osteosarcoma cells. However, as osteosarcoma cells express EGFR, further studies are necessary to explore the potential of other therapeutic agents targeting EGFR.
Collapse
Affiliation(s)
- Jun Ah Lee
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Expression of epidermal growth factor receptor in canine osteosarcoma: association with clinicopathological parameters and prognosis. Vet J 2012; 193:412-9. [PMID: 22436430 DOI: 10.1016/j.tvjl.2012.02.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 02/13/2012] [Accepted: 02/15/2012] [Indexed: 11/23/2022]
Abstract
Expression of epidermal growth factor receptor (EGFR) is associated with aggressive growth and metastasis of a range of tumours, including osteosarcomas (OS), although some studies have reported no relevance to clinicopathological events or prognosis. The present study evaluated EGFR mRNA and protein expression in a panel of OS cell lines, normal bones, frozen primary OS and tissue microarrays. EGFR expression was significantly elevated in primary OS compared to normal bones and in metastases of OS to the lungs in comparison with extrapulmonary sites. However, there were no clinical or pathological associations with mRNA expression levels in frozen tumours. Tissue microarray analysis demonstrated that a subset of canine OS with high EGFR expression was associated with significantly shorter survival times and disease-free intervals. Cytoplasmic expression of EGFR was present in 75% of metastases and was similar to expression in primary tumours. EGFR expression alone is not a reliable predictor of outcome and other markers are necessary for further prognostic stratification of dogs with OS. However, these findings suggest that a subset of dogs may benefit from anti-EGFR adjuvant therapies.
Collapse
|
31
|
Balla P, Moskovszky L, Sapi Z, Forsyth R, Knowles H, Athanasou NA, Szendroi M, Kopper L, Rajnai H, Pinter F, Petak I, Benassi MS, Picci P, Conti A, Krenacs T. Epidermal growth factor receptor signalling contributes to osteoblastic stromal cell proliferation, osteoclastogenesis and disease progression in giant cell tumour of bone. Histopathology 2012; 59:376-89. [PMID: 22034878 DOI: 10.1111/j.1365-2559.2011.03948.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIMS Epidermal growth factor receptor (EGFR) is implicated in bone remodelling. The aim was to determine whether EGFR protein expression contributes to the aggressiveness and recurrence potential of giant cell tumour of bone (GCTB), an osteolytic primary bone tumour that can exhibit markedly variable clinical behaviour. METHODS AND RESULTS Immunohistochemical analysis on tissue microarrays (TMA) of 231 primary, 97 recurrent, 17 metastatic and 26 malignant GCTBs was performed using TMA analysis software and whole digital slides allowing validated scoring. EGFR expression was restricted to neoplastic stromal cells and was significantly more frequent in recurrent (71 of 92; 77%) than in non-recurrent GCTBs (86 of 162; 53%) (P = 0.002); and in clinicoradiologically aggressive (31 of 43; 72%) than latent (27 of 54; 50%) cases (P = 0.034). Detecting phosphotyrosine epitopes pY1068 and -pY1173 indicated active EGFR signalling, and finding EGFR ligands EGF and transforming growth factor-α restricted to cells of the monocytic lineage suggested paracrine EGFR activation in stromal cells. In functional studies EGF supported proliferation of GCTB stromal cells, and the addition of EGF and macrophage-colony stimulating factor promoted osteoclastogenesis. CONCLUSION In GCTB, EGFR signalling in neoplastic stromal cells may contribute to disease progression through promoting stromal cell proliferation and osteoclastogenesis.
Collapse
Affiliation(s)
- Peter Balla
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Immunotherapy targeting HER2 with genetically modified T cells eliminates tumor-initiating cells in osteosarcoma. Cancer Gene Ther 2011; 19:212-7. [PMID: 22173710 DOI: 10.1038/cgt.2011.83] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Despite radical surgery and multi-agent chemotherapy, less than one third of patients with recurrent or metastatic osteosarcoma (OS) survive. The limited efficacy of current therapeutic approaches to target tumor-initiating cells (TICs) may explain this dismal outcome. The purpose of this study was to assess the impact of modified T cells expressing a human epidermal growth factor receptor (HER2)-specific chimeric antigen receptor in the OS TIC compartment of human established cell lines. Using the sarcosphere formation assay, we found that OS TICs were resistant to increasing methotrexate concentrations. In contrast, HER2-specific T cells decreased markedly sarcosphere formation capacity and the ability to generate bone tumors in immunodeficient mice after orthotopic transplantation. In vivo, administration of HER2-specific T cells significantly reduced TICs in bulky tumors as judged by decreased sarcosphere forming efficiency in OS cells isolated from explanted tumors. We demonstrate that HER2-specific T cells target drug resistant TICs in established OS cell lines, suggesting that incorporating immunotherapy into current treatment strategies for OS has the potential to improve outcomes.
Collapse
|
33
|
Erlotinib prevents experimental metastases of human small cell lung cancer cells with no epidermal growth factor receptor expression. Clin Exp Metastasis 2011; 29:207-16. [PMID: 22170031 DOI: 10.1007/s10585-011-9443-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 12/06/2011] [Indexed: 02/07/2023]
Abstract
Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) show dramatic antitumor activity in a subset of patients with non-small cell lung cancer who have an active mutation in the epidermal growth factor receptor (EGFR) gene. On the other hand, some lung cancer patients with wild type EGFR also respond to EGFR-TKIs, suggesting that EGFR-TKIs have an effect on host cells as well as tumor cells. However, the effect of EGFR-TKIs on host microenvironments is largely unknown. A multiple organ metastasis model was previously established in natural killer cell-depleted severe combined immunodeficient mice using human lung cancer cells. This model was used to investigate the therapeutic efficacy of erlotinib, an EGFR-TKI, on multiple organ metastases induced by human small cell lung cancer cells (SBC-5 cells) that did not express EGFR. Although erlotinib did not have any effect on the proliferation of SBC-5 cells in vitro, it significantly suppressed bone and lung metastases in vivo, but not liver metastases. An immunohistochemical analysis revealed that, erlotinib significantly suppressed the number of osteoclasts in bone metastases, whereas no difference was seen in microvessel density. Moreover, erlotinib inhibited EGF-induced receptor activator of nuclear factor kappa-B expression in an osteoblastic cell line (MC3T3-E1 cells). These results strongly suggested that erlotinib prevented bone metastases by affecting host microenvironments irrespective of its direct effect on tumor cells.
Collapse
|
34
|
McCleese JK, Bear MD, Kulp SK, Mazcko C, Khanna C, London CA. Met interacts with EGFR and Ron in canine osteosarcoma. Vet Comp Oncol 2011; 11:124-39. [PMID: 22235915 DOI: 10.1111/j.1476-5829.2011.00309.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 10/26/2011] [Accepted: 11/05/2011] [Indexed: 12/24/2022]
Abstract
The receptor tyrosine kinase (RTK) Met is known to be over-expressed in canine osteosarcoma (OSA). In human cancers, the RTKs Met, epidermal growth factor receptor (EGFR) and Ron are frequently co-expressed and engage in heterodimerization, altering signal transduction and promoting resistance to targeted therapeutics. We found that EGFR and Ron are expressed in canine OSA cell lines and primary tissues, EGFR and Ron are frequently phosphorylated in OSA tumour samples, and Met is co-associated with EGFR and Ron in canine OSA cell lines. Transforming growth factor alpha (TGFα) and hepatocyte growth factor (HGF) stimulation induced amplification of ERK1/2 and STAT3 phosphorylation in OSA cells and Met was phosphorylated following TGFα stimulation providing evidence for receptor cross-talk. Lastly, treatment of OSA cells with combined gefitinib and crizotinib inhibited cell proliferation in an additive manner. Together, these data support the notion that Met, EGFR and Ron interact in OSA cells and as such, may represent viable targets for therapeutic intervention.
Collapse
Affiliation(s)
- J K McCleese
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
35
|
Pahl JH, Ruslan SEN, Buddingh EP, Santos SJ, Szuhai K, Serra M, Gelderblom H, Hogendoorn PC, Egeler RM, Schilham MW, Lankester AC. Anti-EGFR Antibody Cetuximab Enhances the Cytolytic Activity of Natural Killer Cells toward Osteosarcoma. Clin Cancer Res 2011; 18:432-41. [DOI: 10.1158/1078-0432.ccr-11-2277] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
36
|
Choy E, Hornicek F, MacConaill L, Harmon D, Tariq Z, Garraway L, Duan Z. High-throughput genotyping in osteosarcoma identifies multiple mutations in phosphoinositide-3-kinase and other oncogenes. Cancer 2011; 118:2905-14. [PMID: 22006429 DOI: 10.1002/cncr.26617] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 09/09/2011] [Accepted: 09/12/2011] [Indexed: 12/12/2022]
Abstract
BACKGROUND The identification of new genes that are mutated in osteosarcomas is critical to developing a better understanding of the molecular pathogenesis of this disease and discovering new targets for therapeutic development. METHODS The authors identified somatic nonsynonymous coding mutations in oncogenes associated with human cancers and hotspot mutations from tumor suppressor genes that were either well described in the literature or observed multiple times in human cancer sequencing efforts. Then, 961 mutations in 89 genes were systematically characterized across 98 osteosarcoma tumor samples and cell lines. All identified mutations were replicated on an independent platform using homogeneous mass extend matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. RESULTS In total, 14 mutations were identified in at least 1 osteosarcoma tumor sample or cell line. Some of the genetic changes identified were in tumor suppressor genes previously identified as altered in osteosarcoma: p53 (arginine→histidine at codon 273 [R273H], R→cysteine at codon 723 [R273C], and tyrosine→C at codon 163 [Y163C]) and retinoblastoma 1 (RB1) (glutamic acid→* at codon 137 [E137*]). Notably, multiple mutations were identified in phosphoinositide-3-kinase (PI3K), catalytic, alpha polypeptide (PIK3CA) (H1047R, E→lysine at codon 545 [E545K], and H→proline at codon 701 [H701P]) that were not observed previously in osteosarcoma. In addition, mutations in v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) (glycine→serine at codon 12 [G12S]); cubilin (CUBN) (isolucine→valine at codon 3189 [I3189V]; observed in 2 separate tumor samples); cadherin 1, type 1, epithelial (CDH1) (alanine→threonine at codon 617 [A617T]; observed in 2 separate tumor samples); catenin (cadherin-associated protein), beta 1, 88 kDa (CTNNB1) (asparagine→S at codon 287 [N287S]); and fibrous sheath CABYR binding protein (FSCB) (S→leucine at codon 775 [S775L]) were observed. CONCLUSIONS In this largest mutational profiling of osteosarcoma to date, the authors identified for the first time several mutations involving the PI3K pathway, adding osteosarcoma to the growing list of malignancies with PI3K mutations. In addition, they initiated a mutational map detailing DNA sequence changes across a variety of osteosarcoma subtypes and offered new candidates for therapeutic targeting.
Collapse
Affiliation(s)
- Edwin Choy
- Division of Hematology Oncology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Thurber GM, Figueiredo JL, Weissleder R. Detection limits of intraoperative near infrared imaging for tumor resection. J Surg Oncol 2011; 102:758-64. [PMID: 20872807 DOI: 10.1002/jso.21735] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND AND OBJECTIVES The application of fluorescent molecular imaging to surgical oncology is a developing field with the potential to reduce morbidity and mortality. However, the detection thresholds and other requirements for successful intervention remain poorly understood. Here we modeled and experimentally validated depth and size of detection of tumor deposits, trade-offs in coverage and resolution of areas of interest, and required pharmacokinetics of probes based on differing levels of tumor target presentation. METHODS Three orthotopic tumor models were imaged by widefield epifluorescence and confocal microscopes, and the experimental results were compared with pharmacokinetic models and light scattering simulations to determine detection thresholds. RESULTS Widefield epifluorescence imaging can provide sufficient contrast to visualize tumor margins and detect tumor deposits 3-5 mm deep based on labeled monoclonal antibodies at low objective magnification. At higher magnification, surface tumor deposits at cellular resolution are detectable at TBR ratios achieved with highly expressed antigens. CONCLUSIONS A widefield illumination system with the capability for macroscopic surveying and microscopic imaging provides the greatest utility for varying surgical goals. These results have implications for system and agent designs, which ultimately should aid complete resection in most surgical beds and provide real-time feedback to obtain clean margins.
Collapse
Affiliation(s)
- Greg M Thurber
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
38
|
Susa M, Choy E, Liu X, Schwab J, Hornicek FJ, Mankin H, Duan Z. Cyclin G-associated kinase is necessary for osteosarcoma cell proliferation and receptor trafficking. Mol Cancer Ther 2010; 9:3342-50. [PMID: 20881269 DOI: 10.1158/1535-7163.mct-10-0637] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Osteosarcoma is the most frequent primary malignant bone tumor among the children. The advent of neoadjuvant chemotherapy significantly improved the prognosis of patients with osteosarcoma in the 1980s, but it has since plateaued in the past decades. Recently, one of the most researched areas in sarcoma treatment is tyrosine kinases. Here, we describe research on a serine/threonine kinase, cyclin G-associated kinase (GAK), which has not been reported in osteosarcoma previously. In this study, a lentiviral based human shRNA library was utilized to screen for kinases in KHOS and U-2OS osteosarcoma cells. The expression of GAK was examined in osteosarcoma and the effect on cell proliferation was analyzed by GAK siRNA knockdown. The level of GAK expression and its correlation to prognosis was analyzed in osteosarcoma tissue microarray. The effect of GAK depletion on insulin-like growth factor and epidermal growth factor receptor-mediated signal transduction was analyzed by Western blot. We observed that GAK was overexpressed in both osteosarcoma cell lines and tissue samples when compared with human osteoblasts. GAK knockdown by siRNA decreased cell proliferation in both drug-sensitive and multidrug-resistant osteosarcoma cell lines. Immunohistochemistry of osteosarcoma tissue microarray revealed that overexpression of GAK was associated with poor prognosis. Finally, knockdown of GAK resulted in alterations of receptor trafficking and several downstream proteins. In conclusion, our results suggest that osteosarcoma cell proliferation and survival are dependent on GAK. These findings may lead to the development of new therapeutic options for osteosarcoma.
Collapse
Affiliation(s)
- Michiro Susa
- Department of Orthopaedic Surgery, and SarcomaBiology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Zhu C, Liu J, Wang X. [Detection of EGFR and COX-2 expression by immunohistochemical method on a tissue microarray section in lung cancer and biological significance]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2010; 13:107-11. [PMID: 20673501 PMCID: PMC6000521 DOI: 10.3779/j.issn.1009-3419.2010.02.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2009] [Revised: 12/18/2009] [Indexed: 11/05/2022]
Abstract
背景与目的 研究表明表皮生长因子受体(epidermal growth factor receptor, EGFR)和环氧合酶-2 (cyclooxygenase-2, COX-2)在多种实体瘤中存在高表达, 并且可以通过相应的信号通路调节肿瘤的生长、侵袭和转移。本研究旨在探讨EGFR和COX-2在人类肺癌组织中表达的生物学意义及相互之间的关系。 方法 应用组织芯片技术结合免疫组织化学SP法检测89例原发肺癌、12例淋巴结转移性肺癌、12例癌前病变(不典型腺瘤样增生)和10例正常肺组织中EGFR、COX-2蛋白的表达情况。 结果 EGFR在肺癌组、癌前病变组、淋巴结转移性肺癌组中的阳性表达率分别为59.6%(53/89)、41.7%(5/12)和66.7%(8/12), COX-2在上述三组中的阳性表达率分别为52.8%(47/89)、41.7%(5/12)和66.7%(8/12), 均较正常组明显升高(P < 0.05)。EGFR和COX-2的表达与肺癌的组织学类型、临床分期和淋巴结转移有关(P < 0.05), 而与组织学分级、性别、年龄无关(P > 0.05)。COX-2的表达还与肺癌的大体类型有关(P < 0.05)。EGFR和COX-2的表达呈正相关(P < 0.01)。
结论 EGFR和COX-2在肺癌中的异常表达与肺癌的发生、发展和恶性程度有关, 两者存在一定的协同作用。检测EGFR和COX-2的表达有助于肺癌的临床诊断和预后评估。
Collapse
Affiliation(s)
- Congzhong Zhu
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China
| | | | | |
Collapse
|
40
|
Hughes DPM. Strategies for the targeted delivery of therapeutics for osteosarcoma. Expert Opin Drug Deliv 2010; 6:1311-21. [PMID: 19761419 DOI: 10.1517/17425240903280422] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Conventional therapy for osteosarcoma has reached a plateau of 60 - 70%, a 5-year survival rate that has changed little in two decades, highlighting the need for new approaches. OBJECTIVE To review the alternative means of delivering effective therapy for osteosarcoma that reach beyond the central venous catheter. METHODS Drawing on the author's own experiences providing care to high-risk osteosarcoma patients and reviewing the last two decades of literature describing sarcoma therapy, available information is summarized about potential osteosarcoma treatments that deliver therapy by a less conventional route. RESULTS/CONCLUSIONS Intra-arterial chemotherapy has a limited impact on survival, but may help to achieve a better limb salvage. Intrapleural chemotherapy is important for managing malignant effusions. The development of inhalation therapies, treatments that target new bone formation such as bisphosphonates, chemically targeted radiation and antibody-based therapies all have potential to improve osteosarcoma therapy.
Collapse
Affiliation(s)
- Dennis P M Hughes
- The Children's Cancer Hospital at MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 853, Houston, TX 77030, USA.
| |
Collapse
|
41
|
Geryk-Hall M, Hughes DPM. Critical signaling pathways in bone sarcoma: candidates for therapeutic interventions. Curr Oncol Rep 2009; 11:446-53. [PMID: 19840522 DOI: 10.1007/s11912-009-0061-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Bone sarcomas cause disproportionate morbidity and mortality and desperately need new therapies as there has been little improvement in outcomes in 20 years. Identification of critical signaling pathways, including type 1 insulin-like growth factor receptor (IGF-1R) for Ewing sarcoma and possibly osteosarcoma, and the ERBB and the Wnt signaling pathways for osteosarcoma, have emerged as receptors mediating vital signals for bone sarcoma. Akt, mammalian target of rapamycin (mTOR), phosphoinositide 3-kinases, mitogen-activated protein kinase kinase, extracellular signal-regulated kinases, and Ras pathway play key roles in at least some tumors, and inhibition of mTOR in particular will likely lead to improved survival, although clinical trials are still underway. The Notch pathway and ezrin are essential for osteosarcoma metastasis, and Fas downregulation is necessary for survival of metastases in lungs. As little is known about chondrosarcoma signaling, more preclinical work is needed. By defining vital signaling pathways in bone sarcomas, small molecule inhibitors can be applied rationally, leading to longer survival and reducing morbidity and late effects from intensive chemotherapy.
Collapse
Affiliation(s)
- Mandy Geryk-Hall
- Department of Pediatrics Research, Unit 853, Children's Cancer Hospital, The University of Texas, M. D. Anderson Cancer Center, PO Box 301402, Houston, TX 77030, USA
| | | |
Collapse
|
42
|
Schneider MR, Sibilia M, Erben RG. The EGFR network in bone biology and pathology. Trends Endocrinol Metab 2009; 20:517-24. [PMID: 19819718 DOI: 10.1016/j.tem.2009.06.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Revised: 06/18/2009] [Accepted: 06/23/2009] [Indexed: 10/20/2022]
Abstract
The resorption, formation and maintenance of bone are coordinated by the action of several hormones, growth factors and transcription factors. Recent experiments based on genetically modified mouse models, gene microarrays and pharmacological intervention indicate that the epidermal growth factor receptor (EGFR) system plays important roles in skeletal biology and pathology. This network, including a family of seven growth factors - the EGFR ligands - and the related tyrosine kinase receptors EGFR (ERBB1), ERBB2, ERBB3 and ERBB4, regulates aspects such as proliferation and differentiation of osteoblasts, chondrocytes and osteoclasts, parathyroid hormone-mediated bone formation and cancer metastases in bone. Here, we summarize and discuss the role of the EGFR and its ligands in skeletal biology and pathology.
Collapse
|
43
|
Freeman SS, Allen SW, Ganti R, Wu J, Ma J, Su X, Neale G, Dome JS, Daw NC, Khoury JD. Copy number gains in EGFR and copy number losses in PTEN are common events in osteosarcoma tumors. Cancer 2008; 113:1453-61. [PMID: 18704985 DOI: 10.1002/cncr.23782] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Osteosarcoma cell lines and tumors have been shown to express epidermal growth factor receptor (EGFR) and harbor amplifications at the EGFR locus. In this study, the authors further analyzed the genomic features of EGFR in osteosarcoma tumors and investigated whether they correlate with phosphatase and tensin homolog (PTEN) expression and copy number status. METHODS EGFR and PTEN expression was assessed by immunohistochemistry (n = 28), and copy number alterations at the EGFR and PTEN loci were surveyed using Affymetrix (Santa Clara, Calif) 50K single nucleotide polymorphism (SNP) arrays (n = 31) and fluorescence in situ hybridization (FISH) (n = 27). The EGFR tyrosine kinase domain was sequenced to survey for activating mutations (n = 34). In addition, EGFRvIII expression was assessed using reverse transcriptase polymerase chain reaction (n = 24). Results were correlated with available clinical information on 59 patients, with a median age of 14.1 years (range, 5-23 years) and median follow-up of 4.4 years. RESULTS EGFR expression was detected in the majority of osteosarcoma tumors surveyed (23 of 28; 82%). SNP arrays revealed evidence of frequent copy number gains at 7p11.2 and losses at 10q23.21. A sizeable subset (16 of 27 cases; 59%) showed gains at the EGFR locus using FISH (amplification in 4 of 27 [15%] and balanced chromosome 7 polysomy in 12 of 27 [44%]), and 12 cases showed deletions at the PTEN locus (biallelic deletions in 4 of 27 [15%] and monoallelic deletion in 9 of 27 [33%]). No activating mutations in the EGFR tyrosine kinase domain, EGFRvIII expression, or association with clinical findings were detected. CONCLUSIONS EGFR expression and genomic gains at the EGFR locus are prevalent in osteosarcoma tumors, which also commonly harbor deletions at the PTEN locus.
Collapse
Affiliation(s)
- Serena S Freeman
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Messerschmitt PJ, Rettew AN, Brookover RE, Garcia RM, Getty PJ, Greenfield EM. Specific tyrosine kinase inhibitors regulate human osteosarcoma cells in vitro. Clin Orthop Relat Res 2008; 466:2168-75. [PMID: 18607665 PMCID: PMC2493014 DOI: 10.1007/s11999-008-0338-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Accepted: 05/21/2008] [Indexed: 01/31/2023]
Abstract
Inhibitors of specific tyrosine kinases are attractive lead compounds for development of targeted chemotherapies for many tumors, including osteosarcoma. We asked whether inhibition of specific tyrosine kinases would decrease the motility, colony formation, and/or invasiveness by human osteosarcoma cell lines (TE85, MNNG, 143B, SAOS-2, LM-7). An EGF-R inhibitor reduced motility of all five cell lines by 50% to 80%. In contrast, an IGF-1R inhibitor preferentially reduced motility by 42% in LM-7 cells and a met inhibitor preferentially reduced motility by 80% in MNNG cells. The inhibitors of EGF-R, IGF-1R, and met reduced colony formation by more than 80% in all tested cell lines (TE85, MNNG, 143B). The EGF-R inhibitor reduced invasiveness by 62% in 143B cells. The JAK inhibitor increased motility of SAOS-2 and LM7 cells without affecting colony formation or invasiveness. Inhibitors of HER-2, NGF-R, and PDGF-Rs did not affect motility, invasiveness, or colony formation. These results support the hypothesis that specific tyrosine kinases regulate tumorigenesis and/or metastasis in osteosarcoma.
Collapse
Affiliation(s)
- Patrick J Messerschmitt
- Department of Orthopaedic Surgery, University Hospitals Case Medical Center, Case Western Reserve University, 11100 Euclid Avenue, 6th Floor Hanna House, Cleveland, OH 44118, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Sibilia M, Kroismayr R, Lichtenberger BM, Natarajan A, Hecking M, Holcmann M. The epidermal growth factor receptor: from development to tumorigenesis. Differentiation 2007; 75:770-87. [DOI: 10.1111/j.1432-0436.2007.00238.x] [Citation(s) in RCA: 246] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|