1
|
Pezzella M, Quintarelli C, Quadraccia MC, Sarcinelli A, Manni S, Iaffaldano L, Ottaviani A, Ciccone R, Camera A, D'Amore ML, Di Cecca S, Sinibaldi M, Guercio M, Aurigemma M, De Falco P, Fustaino V, Rota R, Pomella S, Cassandri M, Di Giannatale A, Agrati C, Bordoni V, Guarracino F, Massa M, Del Baldo G, Becilli M, Milano GM, Del Bufalo F, Locatelli F, De Angelis B. Tumor-derived G-CSF induces an immunosuppressive microenvironment in an osteosarcoma model, reducing response to CAR.GD2 T-cells. J Hematol Oncol 2024; 17:127. [PMID: 39695851 DOI: 10.1186/s13045-024-01641-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
Sarcomas are rare, mesenchymal tumors, representing about 10-15% of all childhood cancers. GD2 is a suitable target for chimeric antigen receptor (CAR) T-cell therapy due to its overexpression in several solid tumors. In this preclinical study, we investigated the potential use of iCasp9.2A.GD2.CAR-CD28.4-1BBζ (CAR.GD2) T-cells as a treatment option for patients who have GD2-positive sarcomas and we sought to identify factors shaping hostile tumor microenvironment in this setting. GD2 expression was evaluated by flow-cytometry on primary tumor biopsies of pediatric sarcoma patients. GD2 expression in sarcoma cells was also evaluated in response to an enhancer of zeste homolog 2 (EZH2) inhibitor (Tazemetostat). The antitumor activity of CAR.GD2 T-cells was evaluated both in vitro and in vivo preclinical models of orthotopic and/or metastatic soft-tissue and bone sarcomas. GD2 expression was detected in 55% of the primary tumors. Notably, the Osteosarcoma and Alveolar Rhabdomyosarcomas subtypes exhibited the highest GD2 expression levels, while Ewing sarcoma showed the lowest. CAR.GD2 T-cells show a significant tumor control both in vitro and in vivo models of GD2-expressing tumors. Pretreatment with an EZH2 inhibitor (Tazemetostat) upregulating GD2 expression, sensitizes GD2dim sarcoma cells to CAR.GD2 T-cells cytotoxic activity. Moreover, in mouse models of disseminated Rhabdomyosarcomas and orthotopic Osteosarcoma, CAR.GD2 T-cells showed both a vigorous anti-tumor activity and long-term persistence as compared to un-transduced T-cells. The presence of immunosuppressive murine myeloid-derived suppressor (MDSC) cells significantly reduces long-term anti-tumour activity of infused CAR.GD2 T-cells. Tumor-derived G-CSF was found to be one of the key factors driving expansion of immunosuppressive murine and human MDSC, thus indirectly limiting the efficacy of CAR.GD2 T-cells. Our preclinical data strongly suggest that CAR.GD2 T-cells hold promise as a potential therapeutic option for the treatment of patients with GD2-positive sarcomas. Strategies to tackle hostile immunosuppressive MDSC are desirable to optimize CAR.GD2 T-cell activity.
Collapse
Affiliation(s)
- Michele Pezzella
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Concetta Quintarelli
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
- Department of Clinical Medicine and Surgery, Federico II University of Naples, 80131, Naples, Italy
| | - Maria C Quadraccia
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, 00185, Rome, Italy
| | - Andrea Sarcinelli
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
- PhD Program in Immunology, Molecular Medicine and Applied Biotechnologies, Faculty of Medicine, University of Rome Tor Vergata, 00173, Rome, Italy
| | - Simona Manni
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Laura Iaffaldano
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Alessio Ottaviani
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Roselia Ciccone
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Antonio Camera
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Maria L D'Amore
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Stefano Di Cecca
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Matilde Sinibaldi
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Marika Guercio
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Mariasole Aurigemma
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, 00185, Rome, Italy
| | - Pamela De Falco
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Valentina Fustaino
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Rossella Rota
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Silvia Pomella
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Matteo Cassandri
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Angela Di Giannatale
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Chiara Agrati
- Unit of Pathogen Specific Immunity, Bambino Gesù Children's Hospital, IRCCS, 00145, Rome, Italy
| | - Veronica Bordoni
- Unit of Pathogen Specific Immunity, Bambino Gesù Children's Hospital, IRCCS, 00145, Rome, Italy
| | - Federica Guarracino
- Unit of Pathogen Specific Immunity, Bambino Gesù Children's Hospital, IRCCS, 00145, Rome, Italy
| | - Michele Massa
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Giada Del Baldo
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Marco Becilli
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Giuseppe M Milano
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Francesca Del Bufalo
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy
| | - Franco Locatelli
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy.
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, S. Onofrio Square, 00165, Rome, Italy.
| | - Biagio De Angelis
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, San Paolo N°15 Street, 00146, Rome, Italy.
| |
Collapse
|
2
|
Zhao K, Wu C, Li X, Niu M, Wu D, Cui X, Zhao H. From mechanism to therapy: the journey of CD24 in cancer. Front Immunol 2024; 15:1401528. [PMID: 38881902 PMCID: PMC11176514 DOI: 10.3389/fimmu.2024.1401528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 04/25/2024] [Indexed: 06/18/2024] Open
Abstract
CD24 is a glycosylphosphatidylinositol-anchored protein that is expressed in a wide range of tissues and cell types. It is involved in a variety of physiological and pathological processes, including cell adhesion, migration, differentiation, and apoptosis. Additionally, CD24 has been studied extensively in the context of cancer, where it has been found to play a role in tumor growth, invasion, and metastasis. In recent years, there has been growing interest in CD24 as a potential therapeutic target for cancer treatment. This review summarizes the current knowledge of CD24, including its structure, function, and its role in cancer. Finally, we provide insights into potential clinical application of CD24 and discuss possible approaches for the development of targeted cancer therapies.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Caifeng Wu
- Department of Hand and Foot, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangjun Li
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mengchao Niu
- Department of Operation Room, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dan Wu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaofeng Cui
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
3
|
Liatsou I, Fu Y, Li Z, Hasan M, Guo X, Yu J, Piccolo J, Cartee A, Wang H, Du Y, Bryan J, Gabrielson K, Kraitchman DL, Sgouros G. Therapeutic efficacy of an alpha-particle emitter labeled anti-GD2 humanized antibody against osteosarcoma-a proof of concept study. Eur J Nucl Med Mol Imaging 2024; 51:1409-1420. [PMID: 38108831 DOI: 10.1007/s00259-023-06528-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
PURPOSE Current treatments for osteosarcoma (OS) have a poor prognosis, particularly for patients with metastasis and recurrence, underscoring an urgent need for new targeted therapies to improve survival. Targeted alpha-particle therapy selectively delivers cytotoxic payloads to tumors with radiolabeled molecules that recognize tumor-associated antigens. We have recently demonstrated the potential of an FDA approved, humanized anti-GD2 antibody, hu3F8, as a targeted delivery vector for radiopharmaceutical imaging of OS. The current study aims to advance this system for alpha-particle therapy of OS. METHODS The hu3F8 antibody was radiolabeled with actinium-225, and the safety and therapeutic efficacy of the [225Ac]Ac-DOTA-hu3F8 were evaluated in both orthotopic murine xenografts of OS and spontaneously occurring OS in canines. RESULTS Significant antitumor activity was proven in both cases, leading to improved overall survival. In the murine xenograft's case, tumor growth was delayed by 16-18 days compared to the untreated cohort as demonstrated by bioluminescence imaging. The results were further validated with magnetic resonance imaging at 33 days after treatment, and microcomputed tomography and planar microradiography post-mortem. Histological evaluations revealed radiation-induced renal toxicity, manifested as epithelial cell karyomegaly and suggestive polyploidy in the kidneys, suggesting rapid recovery of renal function after radiation damage. Treatment of the two canine patients delayed the progression of metastatic spread, with an overall survival time of 211 and 437 days and survival beyond documented metastasis of 111 and 84 days, respectively. CONCLUSION This study highlights the potential of hu3F8-based alpha-particle therapy as a promising treatment strategy for OS.
Collapse
Affiliation(s)
- Ioanna Liatsou
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Yingli Fu
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhi Li
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mahmud Hasan
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xin Guo
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jing Yu
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joseph Piccolo
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Allison Cartee
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hao Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yong Du
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeffrey Bryan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
| | - Kathleen Gabrielson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dara L Kraitchman
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - George Sgouros
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Trautwein NF, Schwenck J, Seitz C, Seith F, Calderón E, von Beschwitz S, Singer S, Reischl G, Handgretinger R, Schäfer J, Lang P, Pichler BJ, Schulte JH, la Fougère C, Dittmann H. A novel approach to guide GD2-targeted therapy in pediatric tumors by PET and [ 64Cu]Cu-NOTA-ch14.18/CHO. Theranostics 2024; 14:1212-1223. [PMID: 38323317 PMCID: PMC10845206 DOI: 10.7150/thno.92481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 12/23/2023] [Indexed: 02/08/2024] Open
Abstract
Background: The tumor-associated disialoganglioside GD2 is a bona fide immunotherapy target in neuroblastoma and other childhood tumors, including Ewing sarcoma and osteosarcoma. GD2-targeting antibodies proved to be effective in neuroblastoma and GD2-targeting chimeric antigen receptors (CAR)- expressing T cells as well as natural killer T cells (NKTs) are emerging. However, assessment of intra- and intertumoral heterogeneity has been complicated by ineffective immunohistochemistry as well as sampling bias in disseminated disease. Therefore, a non-invasive approach for the assessment and visualization of GD2 expression in-vivo is of upmost interest and might enable a more appropriate treatment stratification. Methods: Recently, [64Cu]Cu-NOTA-ch14.18/CHO (64Cu-GD2), a radiolabeled GD2-antibody for imaging with Positron-Emission-Tomography (PET) was developed. We here report our first clinical patients' series (n = 11) in different pediatric tumors assessed with 64Cu-GD2 PET/MRI. GD2-expression in tumors and tissue uptake in organs was evaluated by semiquantitative measurements of standardized uptake values (SUV) with PET/MRI on day 1 p.i. (n = 11) as well as on day 2 p.i. (n = 6). Results: In 8 of 9 patients with suspicious tumor lesions on PET/MRI at least one metastasis showed an increased 64Cu-GD2 uptake and a high tracer uptake (SUVmax > 10) was measured in 4 of those 8 patients. Of note, sufficient image quality with high tumor to background contrast was readily achieved on day 1. In case of 64Cu-GD2-positive lesions, an excellent tumor to background ratio (at least 6:1) was observed in bones, muscles or lungs, while lower tumor to background contrast was seen in the spleen, liver and kidneys. Furthermore, we demonstrated extensive tumor heterogeneity between patients as well as among different metastatic sites in individual patients. Dosimetry assessment revealed a whole-body dose of only 0.03 mGy/MBq (range 0.02-0.04). Conclusion: 64Cu-GD2 PET/MRI enables the non-invasive assessment of individual heterogeneity of GD2 expression, which challenges our current clinical practice of patient selection, stratification and immunotherapy application scheme for treatment with anti-GD2 directed therapies.
Collapse
Affiliation(s)
- Nils Florian Trautwein
- Department of Nuclear Medicine and Clinical Molecular Imaging, University of Tübingen
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen
| | - Johannes Schwenck
- Department of Nuclear Medicine and Clinical Molecular Imaging, University of Tübingen
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen
| | - Christian Seitz
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen
- Department of Pediatric Hematology and Oncology, University of Tübingen
| | - Ferdinand Seith
- Department of Diagnostic and Interventional Radiology, University of Tübingen
| | - Eduardo Calderón
- Department of Nuclear Medicine and Clinical Molecular Imaging, University of Tübingen
| | | | - Stephan Singer
- Department of Pathology, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Gerald Reischl
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen
| | | | - Jürgen Schäfer
- Department of Diagnostic and Interventional Radiology, University of Tübingen
| | - Peter Lang
- Department of Pediatric Hematology and Oncology, University of Tübingen
| | - Bernd J. Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen
- German Cancer Consortium (DKTK), Partner Site Tübingen, Germany
| | | | - Christian la Fougère
- Department of Nuclear Medicine and Clinical Molecular Imaging, University of Tübingen
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen
- German Cancer Consortium (DKTK), Partner Site Tübingen, Germany
| | - Helmut Dittmann
- Department of Nuclear Medicine and Clinical Molecular Imaging, University of Tübingen
| |
Collapse
|
5
|
Eichholz T, Heubach F, Arendt AM, Seitz C, Brecht IB, Ebinger M, Flaadt T, Süsskind D, Richter L, Hülsenbeck I, Zerweck L, Göricke S, Paulsen F, Dombrowski F, Flotho C, Schönberger S, Ketteler P, Schulte J, Lang P. Targeted therapies in retinoblastoma: GD2-directed immunotherapy following autologous stem cell transplantation and evaluation of alternative target B7-H3. Cancer Immunol Immunother 2024; 73:19. [PMID: 38240863 PMCID: PMC10798927 DOI: 10.1007/s00262-023-03587-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/10/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND GD2-directed immunotherapy is highly effective in the treatment of high-risk neuroblastoma (NB), and might be an interesting target also in other high-risk tumors. METHODS The German-Austrian Retinoblastoma Registry, Essen, was searched for patients, who were treated with anti-GD2 monoclonal antibody (mAb) dinutuximab beta (Db) in order to evaluate toxicity, response and outcome in these patients. Additionally, we evaluated anti-GD2 antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) in retinoblastoma cell lines in vitro. Furthermore, in vitro cytotoxicity assays directed against B7-H3 (CD276), a new identified potential target in RB, were performed. RESULTS We identified four patients with relapsed stage IV retinoblastoma, who were treated with Db following autologous stem cell transplantation (ASCT). Two out of two evaluable patients with detectable tumors responded to immunotherapy. One of these and another patient who received immunotherapy without residual disease relapsed 10 and 12 months after start of Db. The other patients remained in remission until last follow-up 26 and 45 months, respectively. In vitro, significant lysis of RB cell lines by ADCC and CDC with samples from patients and healthy donors and anti-GD2 and anti-CD276-mAbs were demonstrated. CONCLUSION Anti-GD2-directed immunotherapy represents an additional therapeutic option in high-risk metastasized RB. Moreover, CD276 is another target of interest.
Collapse
Affiliation(s)
- Thomas Eichholz
- University Children's Hospital, Eberhard Karls University, Abteilung I, Hoppe-Seyler-Str. 1, 72076, Tuebingen, Germany.
| | - Florian Heubach
- University Children's Hospital, Eberhard Karls University, Abteilung I, Hoppe-Seyler-Str. 1, 72076, Tuebingen, Germany
| | - Anne-Marie Arendt
- University Children's Hospital, Eberhard Karls University, Abteilung I, Hoppe-Seyler-Str. 1, 72076, Tuebingen, Germany
| | - Christian Seitz
- University Children's Hospital, Eberhard Karls University, Abteilung I, Hoppe-Seyler-Str. 1, 72076, Tuebingen, Germany
| | - Ines B Brecht
- University Children's Hospital, Eberhard Karls University, Abteilung I, Hoppe-Seyler-Str. 1, 72076, Tuebingen, Germany
| | - Martin Ebinger
- University Children's Hospital, Eberhard Karls University, Abteilung I, Hoppe-Seyler-Str. 1, 72076, Tuebingen, Germany
| | - Tim Flaadt
- University Children's Hospital, Eberhard Karls University, Abteilung I, Hoppe-Seyler-Str. 1, 72076, Tuebingen, Germany
| | - Daniela Süsskind
- Department of Ophthalmology, Eberhard Karls University, Tuebingen, Germany
| | - Lisa Richter
- Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Isabel Hülsenbeck
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Leonie Zerweck
- Department of Radiology, Diagnostic and Interventional Neuroradiology, Eberhard Karls University, Tuebingen, Germany
| | - Sophia Göricke
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Frank Paulsen
- Department of Radiation Oncology, Eberhard Karls University, Tuebingen, Germany
| | - Frank Dombrowski
- Institute of Pathology, University Medicine of Greifswald, Greifswald, Germany
| | - Christian Flotho
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Schönberger
- Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
- RB-Registry, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Petra Ketteler
- Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
- RB-Registry, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Johannes Schulte
- University Children's Hospital, Eberhard Karls University, Abteilung I, Hoppe-Seyler-Str. 1, 72076, Tuebingen, Germany
| | - Peter Lang
- University Children's Hospital, Eberhard Karls University, Abteilung I, Hoppe-Seyler-Str. 1, 72076, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tuebingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
6
|
Jeremiasse B, Rijs Z, Angoelal KR, Hiemcke-Jiwa LS, de Boed EA, Kuppen PJK, Sier CFM, van Driel PBAA, van de Sande MAJ, Wijnen MHWA, Rios AC, van der Steeg AFW. Evaluation of Potential Targets for Fluorescence-Guided Surgery in Pediatric Ewing Sarcoma: A Preclinical Proof-of-Concept Study. Cancers (Basel) 2023; 15:3896. [PMID: 37568714 PMCID: PMC10417270 DOI: 10.3390/cancers15153896] [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: 06/28/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Fluorescence-guided surgery (FGS), based on fluorescent tracers binding to tumor-specific biomarkers, could assist surgeons to achieve complete tumor resections. This study evaluated potential biomarkers for FGS in pediatric Ewing sarcoma (ES). Immunohistochemistry (IHC) was performed to assess CD99, CXCR4, CD117, NPY-R-Y1, and IGF-1R expression in ES biopsies and resection specimens. LINGO-1 and GD2 evaluation did not work on the acquired tissue. Based on the immunoreactive scores, anti-CD99 and anti-CD117 were evaluated for binding specificity using flow cytometry and immunofluorescence microscopy. Anti-GD2, a tracer in the developmental phase, was also tested. These three tracers were topically applied to a freshly resected ES tumor and adjacent healthy tissue. IHC demonstrated moderate/strong CD99 and CD117 expression in ES tumor samples, while adjacent healthy tissue had limited expression. Flow cytometry and immunofluorescence microscopy confirmed high CD99 expression, along with low/moderate CD117 and low GD2 expression, in ES cell lines. Topical anti-CD99 and anti-GD2 application on ES tumor showed fluorescence, while anti-CD117 did not show fluorescence for this patient. In conclusion, CD99-targeting tracers hold promise for FGS of ES. CD117 and GD2 tracers could be potential alternatives. The next step towards development of ES-specific FGS tracers could be ex vivo topical application experiments on a large cohort of ES patients.
Collapse
Affiliation(s)
- Bernadette Jeremiasse
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (B.J.); (K.R.A.); (M.A.J.v.d.S.); (M.H.W.A.W.); (A.F.W.v.d.S.)
| | - Zeger Rijs
- Department of Orthopedic Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Karieshma R. Angoelal
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (B.J.); (K.R.A.); (M.A.J.v.d.S.); (M.H.W.A.W.); (A.F.W.v.d.S.)
| | - Laura S. Hiemcke-Jiwa
- Department of Pathology, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (L.S.H.-J.); (E.A.d.B.)
- Department of Pathology, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Ella A. de Boed
- Department of Pathology, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (L.S.H.-J.); (E.A.d.B.)
| | - Peter J. K. Kuppen
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (P.J.K.K.); (C.F.M.S.)
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (P.J.K.K.); (C.F.M.S.)
| | | | - Michiel A. J. van de Sande
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (B.J.); (K.R.A.); (M.A.J.v.d.S.); (M.H.W.A.W.); (A.F.W.v.d.S.)
- Department of Orthopedic Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marc H. W. A. Wijnen
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (B.J.); (K.R.A.); (M.A.J.v.d.S.); (M.H.W.A.W.); (A.F.W.v.d.S.)
| | - Anne C. Rios
- Research Department, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
- Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, The Netherlands
| | - Alida F. W. van der Steeg
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (B.J.); (K.R.A.); (M.A.J.v.d.S.); (M.H.W.A.W.); (A.F.W.v.d.S.)
| |
Collapse
|
7
|
Bareke H, Ibáñez-Navarro A, Guerra-García P, González Pérez C, Rubio-Aparicio P, Plaza López de Sabando D, Sastre-Urgelles A, Ortiz-Cruz EJ, Pérez-Martínez A. Prospects and Advances in Adoptive Natural Killer Cell Therapy for Unmet Therapeutic Needs in Pediatric Bone Sarcomas. Int J Mol Sci 2023; 24:ijms24098324. [PMID: 37176035 PMCID: PMC10178897 DOI: 10.3390/ijms24098324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/25/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Malignant bone tumors are aggressive tumors, with a high tendency to metastasize, that are observed most frequently in adolescents during rapid growth spurts. Pediatric patients with malignant bone sarcomas, Ewing sarcoma and osteosarcoma, who present with progressive disease have dire survival rates despite aggressive therapy. These therapies can have long-term effects on bone growth, such as decreased bone mineral density and reduced longitudinal growth. New therapeutic approaches are therefore urgently needed for targeting pediatric malignant bone tumors. Harnessing the power of the immune system against cancer has improved the survival rates dramatically in certain cancer types. Natural killer (NK) cells are a heterogeneous group of innate effector cells that possess numerous antitumor effects, such as cytolysis and cytokine production. Pediatric sarcoma cells have been shown to be especially susceptible to NK-cell-mediated killing. NK-cell adoptive therapy confers numerous advantages over T-cell adoptive therapy, including a good safety profile and a lack of major histocompatibility complex restriction. NK-cell immunotherapy has the potential to be a new therapy for pediatric malignant bone tumors. In this manuscript, we review the general characteristics of osteosarcoma and Ewing sarcoma, discuss the long-term effects of sarcoma treatment on bones, and the barriers to effective immunotherapy in bone sarcomas. We then present the laboratory and clinical studies on NK-cell immunotherapy for pediatric malignant bone tumors. We discuss the various donor sources and NK-cell types, the engineering of NK cells and combinatorial treatment approaches that are being studied to overcome the current challenges in adoptive NK-cell therapy, while suggesting approaches for future studies on NK-cell immunotherapy in pediatric bone tumors.
Collapse
Affiliation(s)
- Halin Bareke
- Translational Research Group in Pediatric Oncology, Haematopoietic Transplantation and Cell Therapy, Hospital La Paz Institute for Health Research, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Adrián Ibáñez-Navarro
- Translational Research Group in Pediatric Oncology, Haematopoietic Transplantation and Cell Therapy, Hospital La Paz Institute for Health Research, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Pilar Guerra-García
- Department of Pediatric Hemato-Oncology, La Paz University Hospital, 28046 Madrid, Spain
| | - Carlos González Pérez
- Department of Pediatric Hemato-Oncology, La Paz University Hospital, 28046 Madrid, Spain
| | - Pedro Rubio-Aparicio
- Department of Pediatric Hemato-Oncology, La Paz University Hospital, 28046 Madrid, Spain
| | | | - Ana Sastre-Urgelles
- Department of Pediatric Hemato-Oncology, La Paz University Hospital, 28046 Madrid, Spain
| | - Eduardo José Ortiz-Cruz
- Department of Orthopedic Surgery and Traumatology, La Paz University Hospital, 28046 Madrid, Spain
| | - Antonio Pérez-Martínez
- Translational Research Group in Pediatric Oncology, Haematopoietic Transplantation and Cell Therapy, Hospital La Paz Institute for Health Research, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
- Department of Pediatric Hemato-Oncology, La Paz University Hospital, 28046 Madrid, Spain
- School of Medicine, Autonomous University of Madrid, 28046 Madrid, Spain
| |
Collapse
|