1
|
Rotta G, Puca E, Cazzamalli S, Neri D, Dakhel Plaza S. Cytokine Biopharmaceuticals with "Activity-on-Demand" for Cancer Therapy. Bioconjug Chem 2024; 35:1075-1088. [PMID: 38885090 DOI: 10.1021/acs.bioconjchem.4c00187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Cytokines are small proteins that modulate the activity of the immune system. Because of their potent immunomodulatory properties, some recombinant cytokines have undergone clinical development and have gained marketing authorization for the therapy of certain forms of cancer. Recombinant cytokines are typically administered at ultralow doses, as many of them can cause substantial toxicity even at submilligram quantities. In an attempt to increase the therapeutic index, fusion proteins based on tumor-homing antibodies (also called "immunocytokines") have been considered, and some products in this class have reached late-stage clinical trials. While antibody-cytokine fusions, which preferentially localize in the neoplastic mass, can activate tumor-resident leukocytes and may be more efficacious than their nontargeted counterparts, such products typically conserve an intact cytokine activity, which may prevent escalation to curative doses. To further improve tolerability, several strategies have been conceived for the development of antibody-cytokine fusions with "activity-on-demand", acting on tumors but helping spare normal tissues from undesired toxicity. In this article, we have reviewed some of the most promising strategies, outlining their potential as well as possible limitations.
Collapse
Affiliation(s)
- Giulia Rotta
- Philochem AG, CH-8112 Otelfingen, Switzerland
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | | | | | - Dario Neri
- Philogen S.p.A, 53100 Siena, Italy
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | | |
Collapse
|
2
|
Di Nitto C, Ravazza D, Gilardoni E, Look T, Sun M, Prodi E, Moisoiu V, Pellegrino C, Manz MG, Puca E, Weller M, Weiss T, Neri D, De Luca R. An IL-7 fusion protein targeting EDA fibronectin upregulates TCF1 on CD8+ T-cells, preferentially accumulates to neoplastic lesions, and boosts PD-1 blockade. J Immunother Cancer 2024; 12:e008504. [PMID: 39142716 PMCID: PMC11332014 DOI: 10.1136/jitc-2023-008504] [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] [Accepted: 07/27/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Anti-PD-1 antibodies have revolutionized cancer immunotherapy due to their ability to induce long-lasting complete remissions in a proportion of patients. Current research efforts are attempting to identify biomarkers and suitable combination partners to predict or further improve the activity of immune checkpoint inhibitors. Antibody-cytokine fusions are a class of pharmaceuticals that showed the potential to boost the anticancer properties of other immunotherapies. Extradomain A-fibronectin (EDA-FN), which is expressed in most solid and hematological tumors but is virtually undetectable in healthy adult tissues, is an attractive target for the delivery of cytokine at the site of the disease. METHODS In this work, we describe the generation and characterization of a novel interleukin-7-based fusion protein targeting EDA-FN termed F8(scDb)-IL7. The product consists of the F8 antibody specific to the alternatively spliced EDA of FN in the single-chain diabody (scDb) format fused to human IL-7. RESULTS F8(scDb)-IL7 efficiently stimulates human peripheral blood mononuclear cells in vitro. Moreover, the product significantly increases the expression of T Cell Factor 1 (TCF-1) on CD8+T cells compared with an IL2-fusion protein. TCF-1 has emerged as a pivotal transcription factor that influences the durability and potency of immune responses against tumors. In preclinical cancer models, F8(scDb)-IL7 demonstrates potent single-agent activity and eradicates sarcoma lesions when combined with anti-PD-1. CONCLUSIONS Our results provide the rationale to explore the combination of F8(scDb)-IL7 with anti-PD-1 antibodies for the treatment of patients with cancer.
Collapse
Affiliation(s)
| | | | | | - Thomas Look
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
| | - Miaomiao Sun
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
| | | | - Vlad Moisoiu
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
| | - Christian Pellegrino
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Markus G. Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | | | - Michael Weller
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Dario Neri
- Philogen SpA, Siena, Italy
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
3
|
Santos L, Moreira JN, Abrunhosa A, Gomes C. Brain metastasis: An insight into novel molecular targets for theranostic approaches. Crit Rev Oncol Hematol 2024; 198:104377. [PMID: 38710296 DOI: 10.1016/j.critrevonc.2024.104377] [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: 12/05/2023] [Revised: 04/11/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024] Open
Abstract
Brain metastases (BrM) are common malignant lesions in the central nervous system, and pose a significant threat in advanced-stage malignancies due to delayed diagnosis and limited therapeutic options. Their distinct genomic profiles underscore the need for molecular profiling to tailor effective treatments. Recent advances in cancer biology have uncovered molecular drivers underlying tumor initiation, progression, and metastasis. This, coupled with the advances in molecular imaging technology and radiotracer synthesis, has paved the way for the development of innovative radiopharmaceuticals with enhanced specificity and affinity for BrM specific targets. Despite the challenges posed by the blood-brain barrier to effective drug delivery, several radiolabeled compounds have shown promise in detecting and targeting BrM. This manuscript provides an overview of the recent advances in molecular biomarkers used in nuclear imaging and targeted radionuclide therapy in both clinical and preclinical settings. Additionally, it explores potential theranostic applications addressing the unique challenges posed by BrM.
Collapse
Affiliation(s)
- Liliana Santos
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra 3000-548, Portugal; Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra 3000-548, Portugal
| | - João Nuno Moreira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra 3000-548, Portugal
| | - Antero Abrunhosa
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra 3000-548, Portugal
| | - Célia Gomes
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra 3000-548, Portugal; Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra 3000-548, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra 3000-075, Portugal.
| |
Collapse
|
4
|
Mai Z, Lin Y, Lin P, Zhao X, Cui L. Modulating extracellular matrix stiffness: a strategic approach to boost cancer immunotherapy. Cell Death Dis 2024; 15:307. [PMID: 38693104 PMCID: PMC11063215 DOI: 10.1038/s41419-024-06697-4] [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: 11/22/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024]
Abstract
The interplay between extracellular matrix (ECM) stiffness and the tumor microenvironment is increasingly recognized as a critical factor in cancer progression and the efficacy of immunotherapy. This review comprehensively discusses the key factors regulating ECM remodeling, including the activation of cancer-associated fibroblasts and the accumulation and crosslinking of ECM proteins. Furthermore, it provides a detailed exploration of how ECM stiffness influences the behaviors of both tumor and immune cells. Significantly, the impact of ECM stiffness on the response to various immunotherapy strategies, such as immune checkpoint blockade, adoptive cell therapy, oncolytic virus therapy, and therapeutic cancer vaccines, is thoroughly examined. The review also addresses the challenges in translating research findings into clinical practice, highlighting the need for more precise biomaterials that accurately mimic the ECM and the development of novel therapeutic strategies. The insights offered aim to guide future research, with the potential to enhance the effectiveness of cancer immunotherapy modalities.
Collapse
Affiliation(s)
- Zizhao Mai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
5
|
Rotta G, Gilardoni E, Ravazza D, Mock J, Seehusen F, Elsayed A, Puca E, De Luca R, Pellegrino C, Look T, Weiss T, Manz MG, Halin C, Neri D, Dakhel Plaza S. A novel strategy to generate immunocytokines with activity-on-demand using small molecule inhibitors. EMBO Mol Med 2024; 16:904-926. [PMID: 38448543 PMCID: PMC11018789 DOI: 10.1038/s44321-024-00034-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 03/08/2024] Open
Abstract
Cytokine-based therapeutics have been shown to mediate objective responses in certain tumor entities but suffer from insufficient selectivity, causing limiting toxicity which prevents dose escalation to therapeutically active regimens. The antibody-based delivery of cytokines significantly increases the therapeutic index of the corresponding payload but still suffers from side effects associated with peak concentrations of the product in blood upon intravenous administration. Here we devise a general strategy (named "Intra-Cork") to mask systemic cytokine activity without impacting anti-cancer efficacy. Our technology features the use of antibody-cytokine fusions, capable of selective localization at the neoplastic site, in combination with pathway-selective inhibitors of the cytokine signaling, which rapidly clear from the body. This strategy, exemplified with a tumor-targeted IL12 in combination with a JAK2 inhibitor, allowed to abrogate cytokine-driven toxicity without affecting therapeutic activity in a preclinical model of cancer. This approach is readily applicable in clinical practice.
Collapse
Affiliation(s)
- Giulia Rotta
- Philochem AG, CH-8112, Otelfingen, Switzerland
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | | | | | | | - Frauke Seehusen
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, University of Zurich, CH-8057, Zurich, Switzerland
| | - Abdullah Elsayed
- Philochem AG, CH-8112, Otelfingen, Switzerland
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093, Zurich, Switzerland
| | - Emanuele Puca
- Philochem AG, CH-8112, Otelfingen, Switzerland
- Philogen S.p.A, 53100, Siena, Italy
| | | | - Christian Pellegrino
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, CH-8091, Zurich, Switzerland
| | - Thomas Look
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, CH-8091, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, CH-8091, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, CH-8091, Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093, Zurich, Switzerland
| | - Dario Neri
- Philochem AG, CH-8112, Otelfingen, Switzerland.
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093, Zurich, Switzerland.
- Philogen S.p.A, 53100, Siena, Italy.
| | | |
Collapse
|
6
|
Giraudet AL. [Combination of internal and external beam radiotherapy]. Cancer Radiother 2023; 27:754-758. [PMID: 37953187 DOI: 10.1016/j.canrad.2023.08.005] [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: 05/30/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 11/14/2023]
Abstract
External beam radiation therapy and internal vectorized radiation therapy are two types of radiotherapy that can be used to treat cancer. They differ in the way they are administered, and the type of radiation used. Although they can be effective in treating cancer, they each have their own advantages and disadvantages, and their combination could be synergistic. Preclinical studies on combined internal and external beam radiation therapy have mainly used radiolabelled antibodies, whose bone marrow toxicity remains the limiting factor in increasing the administered activities. The use of small radioligands in clinical trials has shown to be better tolerated and more effective, which explains their rapid development. The results of preclinical studies on combined internal and external beam radiation therapy appear heterogeneous, making it impossible to determine an ideal therapeutic sequencing scheme, and complicating the transposition to clinical studies. The few clinical studies on combined internal and external beam radiation therapy available to date have demonstrated feasibility and tolerability. More work remains to be done in the fields of dosimetry and radiobiology, as well as in the sequencing of these two irradiation modalities to optimize their combination.
Collapse
Affiliation(s)
- A-L Giraudet
- Centre Léon-Bérard, 15, rue Gabriel-Sarrazin, 69008 Lyon, France.
| |
Collapse
|
7
|
Look T, Puca E, Bühler M, Kirschenbaum D, De Luca R, Stucchi R, Ravazza D, Di Nitto C, Roth P, Katzenelenbogen Y, Weiner A, Rindlisbacher L, Becher B, Amit I, Weller M, Neri D, Hemmerle T, Weiss T. Targeted delivery of tumor necrosis factor in combination with CCNU induces a T cell-dependent regression of glioblastoma. Sci Transl Med 2023; 15:eadf2281. [PMID: 37224228 DOI: 10.1126/scitranslmed.adf2281] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 05/01/2023] [Indexed: 05/26/2023]
Abstract
Glioblastoma is the most aggressive primary brain tumor with an unmet need for more effective therapies. Here, we investigated combination therapies based on L19TNF, an antibody-cytokine fusion protein based on tumor necrosis factor that selectively localizes to cancer neovasculature. Using immunocompetent orthotopic glioma mouse models, we identified strong anti-glioma activity of L19TNF in combination with the alkylating agent CCNU, which cured the majority of tumor-bearing mice, whereas monotherapies only had limited efficacy. In situ and ex vivo immunophenotypic and molecular profiling in the mouse models revealed that L19TNF and CCNU induced tumor DNA damage and treatment-associated tumor necrosis. In addition, this combination also up-regulated tumor endothelial cell adhesion molecules, promoted the infiltration of immune cells into the tumor, induced immunostimulatory pathways, and decreased immunosuppression pathways. MHC immunopeptidomics demonstrated that L19TNF and CCNU increased antigen presentation on MHC class I molecules. The antitumor activity was T cell dependent and completely abrogated in immunodeficient mouse models. On the basis of these encouraging results, we translated this treatment combination to patients with glioblastoma. The clinical translation is ongoing but already shows objective responses in three of five patients in the first recurrent glioblastoma patient cohort treated with L19TNF in combination with CCNU (NCT04573192).
Collapse
Affiliation(s)
- Thomas Look
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich 8091, Switzerland
| | | | - Marcel Bühler
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich 8091, Switzerland
| | - Daniel Kirschenbaum
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | - Patrick Roth
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich 8091, Switzerland
| | | | - Assaf Weiner
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Lukas Rindlisbacher
- Institute of Experimental Immunology, University of Zurich, Zurich 8057, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich 8057, Switzerland
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich 8091, Switzerland
| | | | | | - Tobias Weiss
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich 8091, Switzerland
| |
Collapse
|
8
|
Plüss L, Peissert F, Elsayed A, Rotta G, Römer J, Dakhel Plaza S, Villa A, Puca E, De Luca R, Oxenius A, Neri D. Generation and in vivo characterization of a novel high-affinity human antibody targeting carcinoembryonic antigen. MAbs 2023; 15:2217964. [PMID: 37243574 DOI: 10.1080/19420862.2023.2217964] [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: 02/20/2023] [Revised: 05/09/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023] Open
Abstract
There are no effective treatment options for most patients with metastatic colorectal cancer (mCRC). mCRC remains a leading cause of tumor-related death, with a five-year survival rate of only 15%, highlighting the urgent need for novel pharmacological products. Current standard drugs are based on cytotoxic chemotherapy, VEGF inhibitors, EGFR antibodies, and multikinase inhibitors. The antibody-based delivery of pro-inflammatory cytokines provides a promising and differentiated strategy to improve the treatment outcome for mCRC patients. Here, we describe the generation of a novel fully human monoclonal antibody (termed F4) targeting the carcinoembryonic antigen (CEA), a tumor-associated antigen overexpressed in colorectal cancer and other malignancies. The F4 antibody was selected by antibody phage display technology after two rounds of affinity maturation. F4 in single-chain variable fragment format bound to CEA in surface plasmon resonance with an affinity of 7.7 nM. Flow cytometry and immunofluorescence on human cancer specimens confirmed binding to CEA-expressing cells. F4 selectively accumulated in CEA-positive tumors, as evidenced by two orthogonal in vivo biodistribution studies. Encouraged by these results, we genetically fused murine interleukin (IL) 12 to F4 in the single-chain diabody format. F4-IL12 exhibited potent antitumor activity in two murine models of colon cancer. Treatment with F4-IL12 led to an increased density of tumor-infiltrating lymphocytes and an upregulation of interferon γ expression by tumor-homing lymphocytes. These data suggest that the F4 antibody is an attractive delivery vehicle for targeted cancer therapy.
Collapse
Affiliation(s)
- Louis Plüss
- Philochem AG, Libernstrasse 3, Otelfingen, Switzerland
- Department of Biology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | | - Abdullah Elsayed
- Philochem AG, Libernstrasse 3, Otelfingen, Switzerland
- Department of Biology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Giulia Rotta
- Philochem AG, Libernstrasse 3, Otelfingen, Switzerland
| | - Jonas Römer
- Department of Biology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | | | | - Emanuele Puca
- Philochem AG, Libernstrasse 3, Otelfingen, Switzerland
| | | | - Annette Oxenius
- Department of Biology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Dario Neri
- Philochem AG, Libernstrasse 3, Otelfingen, Switzerland
- Department of Biology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
- Philogen SpA, Località Bellaria, Sovicille, Italy
| |
Collapse
|
9
|
van der Heide CD, Dalm SU. Radionuclide imaging and therapy directed towards the tumor microenvironment: a multi-cancer approach for personalized medicine. Eur J Nucl Med Mol Imaging 2022; 49:4616-4641. [PMID: 35788730 PMCID: PMC9606105 DOI: 10.1007/s00259-022-05870-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022]
Abstract
Targeted radionuclide theranostics is becoming more and more prominent in clinical oncology. Currently, most nuclear medicine compounds researched for cancer theranostics are directed towards targets expressed in only a small subset of cancer types, limiting clinical applicability. The identification of cancer-specific targets that are (more) universally expressed will allow more cancer patients to benefit from these personalized nuclear medicine–based interventions. A tumor is not merely a collection of cancer cells, it also comprises supporting stromal cells embedded in an altered extracellular matrix (ECM), together forming the tumor microenvironment (TME). Since the TME is less genetically unstable than cancer cells, and TME phenotypes can be shared between cancer types, it offers targets that are more universally expressed. The TME is characterized by the presence of altered processes such as hypoxia, acidity, and increased metabolism. Next to the ECM, the TME consists of cancer-associated fibroblasts (CAFs), macrophages, endothelial cells forming the neo-vasculature, immune cells, and cancer-associated adipocytes (CAAs). Radioligands directed at the altered processes, the ECM, and the cellular components of the TME have been developed and evaluated in preclinical and clinical studies for targeted radionuclide imaging and/or therapy. In this review, we provide an overview of the TME targets and their corresponding radioligands. In addition, we discuss what developments are needed to further explore the TME as a target for radionuclide theranostics, with the hopes of stimulating the development of novel TME radioligands with multi-cancer, or in some cases even pan-cancer, application.
Collapse
Affiliation(s)
| | - Simone U Dalm
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
10
|
Wu AM. Imaging the host response to cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00114-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
11
|
Diagnosis of Glioblastoma by Immuno-Positron Emission Tomography. Cancers (Basel) 2021; 14:cancers14010074. [PMID: 35008238 PMCID: PMC8750680 DOI: 10.3390/cancers14010074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Neuroimaging has transformed the way brain tumors are diagnosed and treated. Although different non-invasive modalities provide very helpful information, in some situations, they present a limited value. By merging the specificity of antibodies with the resolution, sensitivity, and quantitative capabilities of positron emission tomography (PET), “Immuno-PET” allows us to conduct the non-invasive diagnosis and monitoring of patients over time using antibody-based probes as an in vivo, integrated, quantifiable, 3D, full-body “immunohistochemistry”, like a “virtual biopsy”. This review provides and focuses on immuno-PET applications and future perspectives of this promising imaging approach for glioblastoma. Abstract Neuroimaging has transformed neuro-oncology and the way that glioblastoma is diagnosed and treated. Magnetic Resonance Imaging (MRI) is the most widely used non-invasive technique in the primary diagnosis of glioblastoma. Although MRI provides very powerful anatomical information, it has proven to be of limited value for diagnosing glioblastomas in some situations. The final diagnosis requires a brain biopsy that may not depict the high intratumoral heterogeneity present in this tumor type. The revolution in “cancer-omics” is transforming the molecular classification of gliomas. However, many of the clinically relevant alterations revealed by these studies have not yet been integrated into the clinical management of patients, in part due to the lack of non-invasive biomarker-based imaging tools. An innovative option for biomarker identification in vivo is termed “immunotargeted imaging”. By merging the high target specificity of antibodies with the high spatial resolution, sensitivity, and quantitative capabilities of positron emission tomography (PET), “Immuno-PET” allows us to conduct the non-invasive diagnosis and monitoring of patients over time using antibody-based probes as an in vivo, integrated, quantifiable, 3D, full-body “immunohistochemistry” in patients. This review provides the state of the art of immuno-PET applications and future perspectives on this imaging approach for glioblastoma.
Collapse
|
12
|
Martín-Sabroso C, Lozza I, Torres-Suárez AI, Fraguas-Sánchez AI. Antibody-Antineoplastic Conjugates in Gynecological Malignancies: Current Status and Future Perspectives. Pharmaceutics 2021; 13:1705. [PMID: 34683998 PMCID: PMC8541375 DOI: 10.3390/pharmaceutics13101705] [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: 09/15/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 12/18/2022] Open
Abstract
In the last decade, antibody-drug conjugates (ADCs), normally formed by a humanized antibody and a small drug via a chemical cleavable or non-cleavable linker, have emerged as a potential treatment strategy in cancer disease. They allow to get a selective delivery of the chemotherapeutic agents at the tumor level, and, consequently, to improve the antitumor efficacy and, especially to decrease chemotherapy-related toxicity. Currently, nine antibody-drug conjugate-based formulations have been already approved and more than 80 are under clinical trials for the treatment of several tumors, especially breast cancer, lymphomas, and multiple myeloma. To date, no ADCs have been approved for the treatment of gynecological formulations, but many formulations have been developed and have reached the clinical stage, especially for the treatment of ovarian cancer, an aggressive disease with a low five-year survival rate. This manuscript analyzes the ADCs formulations that are under clinical research in the treatment of gynecological carcinomas, specifically ovarian, endometrial, and cervical tumors.
Collapse
Affiliation(s)
- Cristina Martín-Sabroso
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (C.M.-S.); (I.L.); (A.I.T.-S.)
- Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| | - Irene Lozza
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (C.M.-S.); (I.L.); (A.I.T.-S.)
| | - Ana Isabel Torres-Suárez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (C.M.-S.); (I.L.); (A.I.T.-S.)
- Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| | - Ana Isabel Fraguas-Sánchez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (C.M.-S.); (I.L.); (A.I.T.-S.)
- Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
13
|
Zhao X, Yang X, Wang X, Zhao X, Zhang Y, Liu S, Anderson GJ, Kim SJ, Li Y, Nie G. Penetration Cascade of Size Switchable Nanosystem in Desmoplastic Stroma for Improved Pancreatic Cancer Therapy. ACS NANO 2021; 15:14149-14161. [PMID: 34478262 DOI: 10.1021/acsnano.0c08860] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) cells are surrounded by a dense extracellular matrix (ECM), which greatly restricts the access of therapeutic agents, resulting in poor clinical response to chemotherapy. Transforming growth factor-β1 (TGF-β1) signaling plays a crucial role in construction of the desmoplastic stroma and provides potential targets for PDAC therapy. To surmount the pathological obstacle, we developed a size switchable nanosystem based on PEG-PLGA nanospheres encapsulated within liposomes for the combined delivery of vactosertib (VAC), a TGF-β1 receptor kinase inhibitor, and the cytotoxic drug paclitaxel (TAX). By surface modification of the liposomes with a peptide, APTEDB, the nanosystem can be anchored to abundant tumor-associated fibronectin in PDAC stroma and decreases its size by releasing encapsulated TAX-loaded nanospheres, as well as VAC after collapse of the liposomes. The inhibition of ECM hyperplasia by VAC allows TAX more ready access to the cancer cells in addition to its small size, thereby shrinking pancreatic tumor xenografts more effectively than a combination of the free drugs. This size switchable nanosystem enables sequential delivery of drugs at a fixed dose combination with simplified administration and provides an encouraging cascade approach of drug penetration for enhanced chemotherapy in cancers with a dense desmoplastic stroma.
Collapse
Affiliation(s)
- Xiaozheng Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xiao Yang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Xudong Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Shaoli Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Gregory J Anderson
- QIMR Berghofer Medical Research Institute, Royal Brisbane Hospital, Brisbane, Queensland 4029, Australia
| | - Seong-Jin Kim
- GILO Institute, GILO Foundation, Seoul 06668, Republic of Korea
- Medpacto Inc., 92 Myeongdal-ro, Seocho-gu, Seoul 06668, Republic of Korea
| | - Yiye Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- GBA Research Innovation Institute for Nanotechnology, Guangdong 510700, China
| |
Collapse
|
14
|
Zaccagna F, Grist JT, Quartuccio N, Riemer F, Fraioli F, Caracò C, Halsey R, Aldalilah Y, Cunningham CH, Massoud TF, Aloj L, Gallagher FA. Imaging and treatment of brain tumors through molecular targeting: Recent clinical advances. Eur J Radiol 2021; 142:109842. [PMID: 34274843 DOI: 10.1016/j.ejrad.2021.109842] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Molecular imaging techniques have rapidly progressed over recent decades providing unprecedented in vivo characterization of metabolic pathways and molecular biomarkers. Many of these new techniques have been successfully applied in the field of neuro-oncological imaging to probe tumor biology. Targeting specific signaling or metabolic pathways could help to address several unmet clinical needs that hamper the management of patients with brain tumors. This review aims to provide an overview of the recent advances in brain tumor imaging using molecular targeting with positron emission tomography and magnetic resonance imaging, as well as the role in patient management and possible therapeutic implications.
Collapse
Affiliation(s)
- Fulvio Zaccagna
- Division of Neuroimaging, Department of Medical Imaging, University of Toronto, Toronto, Canada.
| | - James T Grist
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom; Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom; Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom; Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Natale Quartuccio
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico Di Cristina Benfratelli, Palermo, Italy
| | - Frank Riemer
- Mohn Medical Imaging and Visualization Centre, University of Bergen, Bergen, Norway; Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Francesco Fraioli
- Institute of Nuclear Medicine, University College London, London, United Kingdom; NIHR University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Corradina Caracò
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Richard Halsey
- Institute of Nuclear Medicine, University College London, London, United Kingdom; NIHR University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Yazeed Aldalilah
- Institute of Nuclear Medicine, University College London, London, United Kingdom; NIHR University College London Hospitals Biomedical Research Centre, London, United Kingdom; Department of Radiology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Charles H Cunningham
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Tarik F Massoud
- Division of Neuroimaging and Neurointervention, Department of Radiology, Stanford University School of Medicine, Stanford, USA
| | - Luigi Aloj
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
15
|
Corroyer-Dulmont A, Jaudet C, Frelin AM, Fantin J, Weyts K, Vallis KA, Falzone N, Sibson NR, Chérel M, Kraeber-Bodéré F, Batalla A, Bardet S, Bernaudin M, Valable S. Radioimmunotherapy for Brain Metastases: The Potential for Inflammation as a Target of Choice. Front Oncol 2021; 11:714514. [PMID: 34504791 PMCID: PMC8423367 DOI: 10.3389/fonc.2021.714514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/03/2021] [Indexed: 11/18/2022] Open
Abstract
Brain metastases (BM) are frequently detected during the follow-up of patients with malignant tumors, particularly in those with advanced disease. Despite a major progress in systemic anti-cancer treatments, the average overall survival of these patients remains limited (6 months from diagnosis). Also, cognitive decline is regularly reported especially in patients treated with whole brain external beam radiotherapy (WBRT), due to the absorbed radiation dose in healthy brain tissue. New targeted therapies, for an earlier and/or more specific treatment of the tumor and its microenvironment, are needed. Radioimmunotherapy (RIT), a combination of a radionuclide to a specific antibody, appears to be a promising tool. Inflammation, which is involved in multiple steps, including the early phase, of BM development is attractive as a relevant target for RIT. This review will focus on the (1) early biomarkers of inflammation in BM pertinent for RIT, (2) state of the art studies on RIT for BM, and (3) the importance of dosimetry to RIT in BM. These two last points will be addressed in comparison to the conventional EBRT treatment, particularly with respect to the balance between tumor control and healthy tissue complications. Finally, because new diagnostic imaging techniques show a potential for the detection of BM at an early stage of the disease, we focus particularly on this therapeutic window.
Collapse
Affiliation(s)
- Aurélien Corroyer-Dulmont
- Medical Physics Department, CLCC François Baclesse, Caen, France
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, Caen, France
| | - Cyril Jaudet
- Medical Physics Department, CLCC François Baclesse, Caen, France
| | - Anne-Marie Frelin
- Grand accélérateur National d’Ions Lourds (GANIL), CEA/DRF-CNRS/IN2P3, Caen, France
| | - Jade Fantin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, Caen, France
| | - Kathleen Weyts
- Nuclear Medicine Department, CLCC François Baclesse, Caen, France
| | - Katherine A. Vallis
- Medical Research Council, Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Nicola R. Sibson
- Medical Research Council, Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Michel Chérel
- Team 13-Nuclear Oncology, CRCINA, INSERM, CNRS, Nantes University, Nantes, France
| | - Françoise Kraeber-Bodéré
- Team 13-Nuclear Oncology, CRCINA, INSERM, CNRS, Nantes University, Nantes, France
- Nuclear Medicine Department, University Hospital, Nantes, France
| | - Alain Batalla
- Medical Physics Department, CLCC François Baclesse, Caen, France
| | - Stéphane Bardet
- Nuclear Medicine Department, CLCC François Baclesse, Caen, France
| | - Myriam Bernaudin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, Caen, France
| | - Samuel Valable
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, Caen, France
| |
Collapse
|
16
|
Shooli H, Nemati R, Ahmadzadehfar H, Aboian M, Jafari E, Jokar N, Nabipour I, Dadgar H, Gholamrezanezhad A, Larvie M, Assadi M. Theranostics in Brain Tumors. PET Clin 2021; 16:397-418. [PMID: 34053584 DOI: 10.1016/j.cpet.2021.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Theranostic nuclear oncology, mainly in neuro-oncology (neurotheranostics), aims to combine cancer imaging and therapy using the same targeting molecule. This approach tries to identify patients who are most likely to benefit from tumor molecular radionuclide therapy. The ability of radioneurotheranostic agents to interact with cancer cells at the molecular level with high specificity can significantly improve the effectiveness of cancer therapy. A variety of biologic targets are under investigation for treating brain tumors. PET-based precision imaging can substantially improve the therapeutic efficacy of radiotheranostic approach in brain tumors.
Collapse
Affiliation(s)
- Hossein Shooli
- Department of Molecular Imaging and Radionuclide Therapy (MIRT), The Persian Gulf Nuclear Medicine Research Center, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Moallem St, Bushehr, Iran
| | - Reza Nemati
- Department of Neurology, Bushehr Medical University Hospital, Bushehr University of Medical Sciences, School of Medicine, Bushehr, Iran
| | | | - Mariam Aboian
- Department of Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Esmail Jafari
- Department of Molecular Imaging and Radionuclide Therapy (MIRT), The Persian Gulf Nuclear Medicine Research Center, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Moallem St, Bushehr, Iran
| | - Narges Jokar
- Department of Molecular Imaging and Radionuclide Therapy (MIRT), The Persian Gulf Nuclear Medicine Research Center, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Moallem St, Bushehr, Iran
| | - Iraj Nabipour
- Department of Internal Medicine (Division of Endocrinology), Bushehr Medical University Hospital, The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Habibollah Dadgar
- Cancer Research Center, RAZAVI Hospital, Imam Reza International University, Mashhad, Iran
| | - Ali Gholamrezanezhad
- Department of Diagnostic Radiology, Keck School of Medicine, University of Southern California (USC), 1520 San Pablo Street, Suite L1600, Los Angeles, CA 90033, USA
| | - Mykol Larvie
- Department of Radiology, Cleveland Clinic, Cleveland, OH, USA
| | - Majid Assadi
- Department of Molecular Imaging and Radionuclide Therapy (MIRT), The Persian Gulf Nuclear Medicine Research Center, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Moallem St, Bushehr, Iran.
| |
Collapse
|
17
|
Suman SK, Subramanian S, Mukherjee A. Combination radionuclide therapy: A new paradigm. Nucl Med Biol 2021; 98-99:40-58. [PMID: 34029984 DOI: 10.1016/j.nucmedbio.2021.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/23/2021] [Accepted: 05/06/2021] [Indexed: 12/15/2022]
Abstract
Targeted molecular radionuclide therapy (MRT) has shown its potential for the treatment of cancers of multiple origins. A combination therapy strategy employing two or more distinct therapeutic approaches in cancer management is aimed at circumventing tumor resistance by simultaneously targeting compensatory signaling pathways or bypassing survival selection mutations acquired in response to individual monotherapies. Combination radionuclide therapy (CRT) is a newer application of the concept, utilizing a combination of radiolabeled molecular targeting agents with chemotherapy and beam radiation therapy for enhanced therapeutic index. Encouraging results are reported with chemotherapeutic agents in combination with radiolabeled targeting molecules for cancer therapy. With increasing awareness of the various survival and stress response pathways activated after radionuclide therapy, different holistic combinations of MRT agents with radiosensitizers targeting such pathways have also been explored. MRT has also been studied in combination with beam radiotherapy modalities such as external beam radiation therapy and carbon ion radiation therapy to enhance the anti-tumor response. Nanotechnology aids in CRT by bringing together multiple monotherapies on a single nanostructure platform for treating cancers in a more precise or personalized way. CRT will be a key player in managing cancers if correctly tailored to the individual patient profile. The success of CRT lies in an in-depth understanding of the radiobiological principles and pathways activated in response.
Collapse
Affiliation(s)
- Shishu Kant Suman
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre; Homi Bhabha National Institute, Mumbai 400094, India
| | - Suresh Subramanian
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre; Homi Bhabha National Institute, Mumbai 400094, India
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre; Homi Bhabha National Institute, Mumbai 400094, India.
| |
Collapse
|
18
|
González-Gómez R, Pazo-Cid RA, Sarría L, Morcillo MÁ, Schuhmacher AJ. Diagnosis of Pancreatic Ductal Adenocarcinoma by Immuno-Positron Emission Tomography. J Clin Med 2021; 10:1151. [PMID: 33801810 PMCID: PMC8000738 DOI: 10.3390/jcm10061151] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 12/15/2022] Open
Abstract
Diagnosis of pancreatic ductal adenocarcinoma (PDAC) by current imaging techniques is useful and widely used in the clinic but presents several limitations and challenges, especially in small lesions that frequently cause radiological tumors infra-staging, false-positive diagnosis of metastatic tumor recurrence, and common occult micro-metastatic disease. The revolution in cancer multi-"omics" and bioinformatics has uncovered clinically relevant alterations in PDAC that still need to be integrated into patients' clinical management, urging the development of non-invasive imaging techniques against principal biomarkers to assess and incorporate this information into the clinical practice. "Immuno-PET" merges the high target selectivity and specificity of antibodies and engineered fragments toward a given tumor cell surface marker with the high spatial resolution, sensitivity, and quantitative capabilities of positron emission tomography (PET) imaging techniques. In this review, we detail and provide examples of the clinical limitations of current imaging techniques for diagnosing PDAC. Furthermore, we define the different components of immuno-PET and summarize the existing applications of this technique in PDAC. The development of novel immuno-PET methods will make it possible to conduct the non-invasive diagnosis and monitoring of patients over time using in vivo, integrated, quantifiable, 3D, whole body immunohistochemistry working like a "virtual biopsy".
Collapse
Affiliation(s)
- Ruth González-Gómez
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Roberto A. Pazo-Cid
- Medical Oncology Unit, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain;
| | - Luis Sarría
- Digestive Radiology Unit, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain;
| | - Miguel Ángel Morcillo
- Biomedical Application of Radioisotopes and Pharmacokinetics Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain
| | - Alberto J. Schuhmacher
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Fundación Aragonesa para la Investigación y el Desarrollo (ARAID), 50018 Zaragoza, Spain
| |
Collapse
|
19
|
Kumar K, Ghosh A. Radiochemistry, Production Processes, Labeling Methods, and ImmunoPET Imaging Pharmaceuticals of Iodine-124. Molecules 2021; 26:E414. [PMID: 33466827 PMCID: PMC7830191 DOI: 10.3390/molecules26020414] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 01/01/2023] Open
Abstract
Target-specific biomolecules, monoclonal antibodies (mAb), proteins, and protein fragments are known to have high specificity and affinity for receptors associated with tumors and other pathological conditions. However, the large biomolecules have relatively intermediate to long circulation half-lives (>day) and tumor localization times. Combining superior target specificity of mAbs and high sensitivity and resolution of the PET (Positron Emission Tomography) imaging technique has created a paradigm-shifting imaging modality, ImmunoPET. In addition to metallic PET radionuclides, 124I is an attractive radionuclide for radiolabeling of mAbs as potential immunoPET imaging pharmaceuticals due to its physical properties (decay characteristics and half-life), easy and routine production by cyclotrons, and well-established methodologies for radioiodination. The objective of this report is to provide a comprehensive review of the physical properties of iodine and iodine radionuclides, production processes of 124I, various 124I-labeling methodologies for large biomolecules, mAbs, and the development of 124I-labeled immunoPET imaging pharmaceuticals for various cancer targets in preclinical and clinical environments. A summary of several production processes, including 123Te(d,n)124I, 124Te(d,2n)124I, 121Sb(α,n)124I, 123Sb(α,3n)124I, 123Sb(3He,2n)124I, natSb(α, xn)124I, natSb(3He,n)124I reactions, a detailed overview of the 124Te(p,n)124I reaction (including target selection, preparation, processing, and recovery of 124I), and a fully automated process that can be scaled up for GMP (Good Manufacturing Practices) production of large quantities of 124I is provided. Direct, using inorganic and organic oxidizing agents and enzyme catalysis, and indirect, using prosthetic groups, 124I-labeling techniques have been discussed. Significant research has been conducted, in more than the last two decades, in the development of 124I-labeled immunoPET imaging pharmaceuticals for target-specific cancer detection. Details of preclinical and clinical evaluations of the potential 124I-labeled immunoPET imaging pharmaceuticals are described here.
Collapse
Affiliation(s)
- Krishan Kumar
- Laboratory for Translational Research in Imaging Pharmaceuticals, The Wright Center of Innovation in Biomedical Imaging, Department of Radiology, The Ohio State University, Columbus, OH 43212, USA;
| | | |
Collapse
|
20
|
Coenen HH, Ermert J. Expanding PET-applications in life sciences with positron-emitters beyond fluorine-18. Nucl Med Biol 2021; 92:241-269. [PMID: 32900582 DOI: 10.1016/j.nucmedbio.2020.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 07/09/2020] [Indexed: 12/20/2022]
Abstract
Positron-emission-tomography (PET) has become an indispensable diagnostic tool in modern nuclear medicine. Its outstanding molecular imaging features allow repetitive studies on one individual and with high sensitivity, though no interference. Rather few positron-emitters with near favourable physical properties, i.e. carbon-11 and fluorine-18, furnished most studies in the beginning, preferably if covalently bound as isotopic label of small molecules. With the advancement of PET-devices the scope of in vivo research in life sciences and especially that of medical applications expanded, and other than "standard" PET-nuclides received increasing significance, like the radiometals copper-64 and gallium-68. Especially during the last decades, positron-emitters of other chemical elements have gotten into the focus of interest, concomitant with the technical advancements in imaging and radionuclide production. With known nuclear imaging properties and main production methods of emerging positron-emitters their usefulness for medical application is promising and even proven for several ones already. Unfortunate decay properties could be corrected for, and β+-emitters, especially with a longer half-life, provided new possibilities for application where slower processes are of importance. Further on, (bio)chemical features of positron-emitters of other elements, among there many metals, not only expanded the field of classical clinical investigations, but also opened up new fields of application. Appropriately labelled peptides, proteins and nanoparticles lend itself as newer probes for PET-imaging, e.g. in theragnostic or PET/MR hybrid imaging. Furthermore, the potential of non-destructive in-vivo imaging with positron-emission-tomography directs the view on further areas of life sciences. Thus, exploiting the excellent methodology for basic research on molecular biochemical functions and processes is increasingly encouraged as well in areas outside of health, such as plant and environmental sciences.
Collapse
Affiliation(s)
- Heinz H Coenen
- Institut für Neurowissenschaften und Medizin, INM-5, Nuklearchemie, Forschungszentrum Jülich GmbH, D-52425 Jülich, Germany.
| | - Johannes Ermert
- Institut für Neurowissenschaften und Medizin, INM-5, Nuklearchemie, Forschungszentrum Jülich GmbH, D-52425 Jülich, Germany.
| |
Collapse
|
21
|
A novel format for recombinant antibody-interleukin-2 fusion proteins exhibits superior tumor-targeting properties in vivo. Oncotarget 2020; 11:3698-3711. [PMID: 33110477 PMCID: PMC7566808 DOI: 10.18632/oncotarget.27726] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/17/2020] [Indexed: 01/13/2023] Open
Abstract
The targeted delivery of interleukin-2 to the tumor is gaining attention as an avenue to potentiate the action of T and NK cells at the site of disease. We have previously described the fusion of the L19 antibody, specific to the EDB domain of fibronectin, with human interleukin-2, using a non-covalent homodimeric diabody format. Here, we describe four novel formats for the L19-IL2 fusion, featuring different arrangements of antibody and IL2. A comparative quantitative biodistribution analysis in tumor-bearing mice using radioiodinated proteins revealed that the novel format (L19L19-IL2, with the antibody in single-chain diabody format) exhibited the best biodistribution results. In vitro assays on peripheral blood mononuclear cells showed a decrease activation of regulatory T cells when single IL2 domain was used. In vivo, both L19-IL2 and L19L19-IL2 inhibited tumor growth in immunocompetent mouse models of cancer. T-cell analysis revealed similar levels of CD4+ and FoxP3+ cells, with an expansion of the CD8+ T cell in mice treated with L19-IL2 and L19L19-IL2. The percentage of CD4+ regulatory T cells was markedly decreased with L19L19-IL2 combined with a mouse-specific PD-1 blocker. Collectively, these data indicate that the new L19L19-IL2 format exhibits favorable tumor-homing properties and mediates a potent anti-cancer activity in vivo.
Collapse
|
22
|
Weiss T, Puca E, Silginer M, Hemmerle T, Pazahr S, Bink A, Weller M, Neri D, Roth P. Immunocytokines are a promising immunotherapeutic approach against glioblastoma. Sci Transl Med 2020; 12:12/564/eabb2311. [DOI: 10.1126/scitranslmed.abb2311] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022]
Abstract
Glioblastoma is a poorly immunogenic cancer, and the successes with recent immunotherapies in extracranial malignancies have, so far, not been translated to this devastating disease. Therefore, there is an urgent need for new strategies to convert the immunologically cold glioma microenvironment into a hot one to enable effective antitumor immunity. Using the L19 antibody, which is specific to a tumor-associated epitope of extracellular fibronectin, we developed antibody-cytokine fusions—immunocytokines—with interleukin-2 (IL2), IL12, or tumor necrosis factor (TNF). We showed that L19 accumulated in the tumor microenvironment of two orthotopic immunocompetent mouse glioma models. Furthermore, intravenous administration of L19-mIL12 or L19-mTNF cured a proportion of tumor-bearing mice, whereas L19-IL2 did not. This therapeutic activity was abolished in RAG−/− mice or upon depletion of CD4 or CD8 T cells, suggesting adaptive immunity. Mechanistically, both immunocytokines promoted tumor-infiltrating lymphocytes and increased the amounts of proinflammatory cytokines within the tumor microenvironment. In addition, L19-mTNF induced tumor necrosis. Systemic administration of the fully human L19-TNF fusion protein to patients with glioblastoma (NCT03779230) was safe, decreased regional blood perfusion within the tumor, and was associated with increasing tumor necrosis and an increase in tumor-infiltrating CD4 and CD8 T cells. The extensive preclinical characterization and subsequent clinical translation provide a robust basis for future studies with immunocytokines to treat malignant brain tumors.
Collapse
Affiliation(s)
- Tobias Weiss
- Department of Neurology and Brain Tumor Center, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland
| | - Emanuele Puca
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zürich, Switzerland
| | - Manuela Silginer
- Department of Neurology and Brain Tumor Center, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland
| | | | - Shila Pazahr
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| | - Andrea Bink
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| | - Michael Weller
- Department of Neurology and Brain Tumor Center, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zürich, Switzerland
| | - Patrick Roth
- Department of Neurology and Brain Tumor Center, University Hospital Zurich and University of Zurich, CH-8091 Zurich, Switzerland
| |
Collapse
|
23
|
Alfaleh MA, Alsaab HO, Mahmoud AB, Alkayyal AA, Jones ML, Mahler SM, Hashem AM. Phage Display Derived Monoclonal Antibodies: From Bench to Bedside. Front Immunol 2020; 11:1986. [PMID: 32983137 PMCID: PMC7485114 DOI: 10.3389/fimmu.2020.01986] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Monoclonal antibodies (mAbs) have become one of the most important classes of biopharmaceutical products, and they continue to dominate the universe of biopharmaceutical markets in terms of approval and sales. They are the most profitable single product class, where they represent six of the top ten selling drugs. At the beginning of the 1990s, an in vitro antibody selection technology known as antibody phage display was developed by John McCafferty and Sir. Gregory Winter that enabled the discovery of human antibodies for diverse applications, particularly antibody-based drugs. They created combinatorial antibody libraries on filamentous phage to be utilized for generating antigen specific antibodies in a matter of weeks. Since then, more than 70 phage–derived antibodies entered clinical studies and 14 of them have been approved. These antibodies are indicated for cancer, and non-cancer medical conditions, such as inflammatory, optical, infectious, or immunological diseases. This review will illustrate the utility of phage display as a powerful platform for therapeutic antibodies discovery and describe in detail all the approved mAbs derived from phage display.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.,Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, University of Tabuk, Tabuk, Saudi Arabia
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Anwar M Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
24
|
Lieverse RIY, Marcus D, van der Wiel AMA, Van Limbergen EJ, Theys J, Yaromina A, Lambin P, Dubois LJ. Human fibronectin extra domain B as a biomarker for targeted therapy in cancer. Mol Oncol 2020; 14:1555-1568. [PMID: 32386436 PMCID: PMC7332215 DOI: 10.1002/1878-0261.12705] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/15/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix protein fibronectin contains a domain that is rarely found in healthy adults and is almost exclusively expressed by newly formed blood vessels in tumours, particularly in solid tumours, different types of lymphoma and some leukaemias. This domain, called the extra domain B (ED‐B), thus has broad therapeutic potential. The antibody L19 has been developed to specifically target ED‐B and has shown therapeutic potential when combined with cytokines, such as IL‐2. In this review article, we discuss the preclinical research and clinical trials that highlight the potential of ED‐B targeting for the imaging and treatment of various types of cancer. ED‐B‐centred studies also highlight how proper patient stratification is of utmost importance for the successful implementation of novel antibody‐based targeted therapies.
Collapse
Affiliation(s)
- Relinde I Y Lieverse
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, The Netherlands
| | - Damiënne Marcus
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, The Netherlands
| | - Alexander M A van der Wiel
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, The Netherlands
| | - Evert J Van Limbergen
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, The Netherlands
| | - Jan Theys
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, The Netherlands
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, The Netherlands
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, The Netherlands
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, The Netherlands
| |
Collapse
|
25
|
Neri D. Antibody-Cytokine Fusions: Versatile Products for the Modulation of Anticancer Immunity. Cancer Immunol Res 2020; 7:348-354. [PMID: 30824549 DOI: 10.1158/2326-6066.cir-18-0622] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The remarkable clinical success of immune-checkpoint inhibitors for the treatment of a growing number of cancer types has sparked interest in the discovery of novel forms of immunotherapy, which may be used alone or in combination. In this context, cytokine-based therapeutics are well poised to play a role in modern cancer therapy. This article focuses on antibody-cytokine fusion proteins (also called "immunocytokines") as one class of biopharmaceuticals that can substantially improve the therapeutic index and, thus, the applicability of cytokine products. In many preclinical settings, antibodies can be used to preferentially deliver many (but not all) types of cytokines to primary and metastatic tumor lesions. The antibody-based delivery of certain proinflammatory payloads (such as IL2, IL12, and TNF) to the tumor microenvironment can lead to a dramatic potentiation of their anticancer activity. However, although some fusion proteins have advanced to late-stage clinical trials, much work remains to be done in order to fully characterize the mechanism of action and the pharmaceutical potential of immunocytokines in the clinical setting. Various factors contribute to in vivo performance, including the target antigen, the antibody properties, the nature of the payload, the format of the fusion protein, the dose, and schedule, as well as their use in combination with other therapeutic modalities. Protein engineering opportunities and insights in cancer immunology are contributing to the development of next-generation immunocytokine products and of novel therapeutic concepts, with the goal to increase antitumor activity and reduce systemic toxicity (a common problem for cytokine-based biopharmaceuticals).
Collapse
Affiliation(s)
- Dario Neri
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
26
|
Corroyer-Dulmont A, Valable S, Falzone N, Frelin-Labalme AM, Tietz O, Toutain J, Soto MS, Divoux D, Chazalviel L, Pérès EA, Sibson NR, Vallis KA, Bernaudin M. VCAM-1 targeted alpha-particle therapy for early brain metastases. Neuro Oncol 2020; 22:357-368. [PMID: 31538194 PMCID: PMC7162423 DOI: 10.1093/neuonc/noz169] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Brain metastases (BM) develop frequently in patients with breast cancer. Despite the use of external beam radiotherapy (EBRT), the average overall survival is short (6 months from diagnosis). The therapeutic challenge is to deliver molecularly targeted therapy at an early stage when relatively few metastatic tumor cells have invaded the brain. Vascular cell adhesion molecule 1 (VCAM-1), overexpressed by nearby endothelial cells during the early stages of BM development, is a promising target. The aim of this study was to investigate the therapeutic value of targeted alpha-particle radiotherapy, combining lead-212 (212Pb) with an anti-VCAM-1 antibody (212Pb-αVCAM-1). METHODS Human breast carcinoma cells that metastasize to the brain, MDA-231-Br-GFP, were injected into the left cardiac ventricle of nude mice. Twenty-one days after injection, 212Pb-αVCAM-1 uptake in early BM was determined in a biodistribution study and systemic/brain toxicity was evaluated. Therapeutic efficacy was assessed using MR imaging and histology. Overall survival after 212Pb-αVCAM-1 treatment was compared with that observed after standard EBRT. RESULTS 212Pb-αVCAM-1 was taken up into early BM with a tumor/healthy brain dose deposition ratio of 6 (5.52e108 and 0.92e108) disintegrations per gram of BM and healthy tissue, respectively. MRI analyses showed a statistically significant reduction in metastatic burden after 212Pb-αVCAM-1 treatment compared with EBRT (P < 0.001), translating to an increase in overall survival of 29% at 40 days post prescription (P < 0.01). No major toxicity was observed. CONCLUSIONS The present investigation demonstrates that 212Pb-αVCAM-1 specifically accumulates at sites of early BM causing tumor growth inhibition.
Collapse
Affiliation(s)
- Aurélien Corroyer-Dulmont
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, Caen, France
- Cancer Research UK and Medical Research Council, Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Samuel Valable
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, Caen, France
| | | | | | - Ole Tietz
- Cancer Research UK and Medical Research Council, Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Jérôme Toutain
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, Caen, France
| | - Manuel Sarmiento Soto
- Cancer Research UK and Medical Research Council, Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Didier Divoux
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, Caen, France
| | - Laurent Chazalviel
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, Caen, France
| | - Elodie A Pérès
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, Caen, France
| | - Nicola R Sibson
- Cancer Research UK and Medical Research Council, Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Katherine A Vallis
- Cancer Research UK and Medical Research Council, Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Myriam Bernaudin
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, Caen, France
| |
Collapse
|
27
|
Pruis IJ, van Dongen GAMS, Veldhuijzen van Zanten SEM. The Added Value of Diagnostic and Theranostic PET Imaging for the Treatment of CNS Tumors. Int J Mol Sci 2020; 21:E1029. [PMID: 32033160 PMCID: PMC7037158 DOI: 10.3390/ijms21031029] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/27/2020] [Accepted: 02/01/2020] [Indexed: 12/11/2022] Open
Abstract
This review highlights the added value of PET imaging in Central Nervous System (CNS) tumors, which is a tool that has rapidly evolved from a merely diagnostic setting to multimodal molecular diagnostics and the guidance of targeted therapy. PET is the method of choice for studying target expression and target binding behind the assumedly intact blood-brain barrier. Today, a variety of diagnostic PET tracers can be used for the primary staging of CNS tumors and to determine the effect of therapy. Additionally, theranostic PET tracers are increasingly used in the context of pharmaceutical and radiopharmaceutical drug development and application. In this approach, a single targeted drug is used for PET diagnosis, upon the coupling of a PET radionuclide, as well as for targeted (nuclide) therapy. Theranostic PET tracers have the potential to serve as a non-invasive whole body navigator in the selection of the most effective drug candidates and their most optimal dose and administration route, together with the potential to serve as a predictive biomarker in the selection of patients who are most likely to benefit from treatment. PET imaging supports the transition from trial and error medicine to predictive, preventive, and personalized medicine, hopefully leading to improved quality of life for patients and more cost-effective care.
Collapse
Affiliation(s)
- Ilanah J. Pruis
- Department of Radiology & Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands;
| | - Guus A. M. S. van Dongen
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Sophie E. M. Veldhuijzen van Zanten
- Department of Radiology & Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands;
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Department of Pediatrics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
28
|
Alfaleh MA, Alsaab HO, Mahmoud AB, Alkayyal AA, Jones ML, Mahler SM, Hashem AM. Phage Display Derived Monoclonal Antibodies: From Bench to Bedside. Front Immunol 2020. [PMID: 32983137 DOI: 10.3389/fimmu.2020.01986/bibtex] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
Monoclonal antibodies (mAbs) have become one of the most important classes of biopharmaceutical products, and they continue to dominate the universe of biopharmaceutical markets in terms of approval and sales. They are the most profitable single product class, where they represent six of the top ten selling drugs. At the beginning of the 1990s, an in vitro antibody selection technology known as antibody phage display was developed by John McCafferty and Sir. Gregory Winter that enabled the discovery of human antibodies for diverse applications, particularly antibody-based drugs. They created combinatorial antibody libraries on filamentous phage to be utilized for generating antigen specific antibodies in a matter of weeks. Since then, more than 70 phage-derived antibodies entered clinical studies and 14 of them have been approved. These antibodies are indicated for cancer, and non-cancer medical conditions, such as inflammatory, optical, infectious, or immunological diseases. This review will illustrate the utility of phage display as a powerful platform for therapeutic antibodies discovery and describe in detail all the approved mAbs derived from phage display.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, University of Tabuk, Tabuk, Saudi Arabia
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Anwar M Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
29
|
De Luca R, Gouyou B, Ongaro T, Villa A, Ziffels B, Sannino A, Buttinoni G, Galeazzi S, Mazzacuva M, Neri D. A Novel Fully-Human Potency-Matched Dual Cytokine-Antibody Fusion Protein Targets Carbonic Anhydrase IX in Renal Cell Carcinomas. Front Oncol 2019; 9:1228. [PMID: 31799191 PMCID: PMC6863974 DOI: 10.3389/fonc.2019.01228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/28/2019] [Indexed: 12/12/2022] Open
Abstract
Certain cytokines synergize in activating anti-cancer immunity at the site of disease and it may be desirable to generate biopharmaceutical agents, capable of simultaneous delivery of cytokine pairs to the tumor. In this article, we have described the cloning, expression and characterization of IL2-XE114-TNFmut, a dual-cytokine biopharmaceutical featuring the sequential fusion of interleukin-2 (IL2) with the XE114 antibody in scFv format and a tumor necrosis factor mutant (TNFmut). The fusion protein recognized the cognate antigen (carbonic anhydrase IX, a marker of hypoxia and of renal cell carcinoma) with high affinity and specificity. IL2-XE114-TNFmut formed a stable non-covalent homotrimeric structure, displayed cytokine activity in in vitro tests and preferentially localized to solid tumors in vivo. The product exhibited a partial growth inhibition of murine CT26 tumors transfected for carbonic anhydrase IX. When administered to Cynomolgus monkey as intravenous injection, IL2-XE114-TNFmut showed the expected plasma concentration of ~1,500 ng/ml at early time points, indicating the absence of any in vivo trapping events, and a half-life of ~2 h. IL2-XE114-TNFmut may thus be considered as a promising biopharmaceutical for the treatment of metastatic clear-cell renal cell carcinoma, since these tumors are known to be sensitive to IL2 and to TNF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gianluca Buttinoni
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | | | | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| |
Collapse
|
30
|
Ziffels B, Pretto F, Neri D. Intratumoral administration of IL2- and TNF-based fusion proteins cures cancer without establishing protective immunity. Immunotherapy 2019; 10:177-188. [PMID: 29370721 DOI: 10.2217/imt-2017-0119] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
AIM The combination of tumor-targeting IL2- and TNF-based antibody-cytokine fusions has exhibited encouraging results in mouse and men. Here, we studied their combination to assess efficacy and mechanism of action in four different immunocompetent mouse models of cancer. METHODS Mice receiving a single intratumoral injection of F8-IL2, F8-TNF or the combination were investigated for tumor-infiltrating leukocytes and rechallenged when cured. RESULTS In three models, a proportion of treated animals could be cured, most probably by infiltrating NK and CD8+ T cells. Most of the cured mice did not acquire protective immunity when rechallenged with the same tumor cell line. CONCLUSION Immunocompetent mouse tumor models may not be adequate enough to predict the search for more efficacious therapy regimens.
Collapse
Affiliation(s)
- Barbara Ziffels
- Department of Chemistry & Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, (ETH Zürich), Vladimir-Prelog-Weg 1-5/10, CH-8093 Zürich, Switzerland
| | | | - Dario Neri
- Department of Chemistry & Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, (ETH Zürich), Vladimir-Prelog-Weg 1-5/10, CH-8093 Zürich, Switzerland
| |
Collapse
|
31
|
Puttemans J, Lahoutte T, D'Huyvetter M, Devoogdt N. Beyond the Barrier: Targeted Radionuclide Therapy in Brain Tumors and Metastases. Pharmaceutics 2019; 11:pharmaceutics11080376. [PMID: 31374991 PMCID: PMC6723032 DOI: 10.3390/pharmaceutics11080376] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/12/2019] [Accepted: 07/17/2019] [Indexed: 01/10/2023] Open
Abstract
Brain tumors are notoriously difficult to treat. The blood-brain barrier provides a sanctuary site where residual and metastatic cancer cells can evade most therapeutic modalities. The delicate nature of the brain further complicates the decision of eliminating as much tumorous tissue as possible while protecting healthy tissue. Despite recent advances in immunotherapy, radiotherapy and systemic treatments, prognosis of newly diagnosed patients remains dismal, and recurrence is still a universal problem. Several strategies are now under preclinical and clinical investigation to optimize delivery and maximize the cytotoxic potential of pharmaceuticals with regards to brain tumors. This review provides an overview of targeted radionuclide therapy approaches for the treatment of primary brain tumors and brain metastases, with an emphasis on biological targeting moieties that specifically target key biomarkers involved in cancer development.
Collapse
Affiliation(s)
- Janik Puttemans
- In Vivo Cellular and Molecular Imaging Lab, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Tony Lahoutte
- In Vivo Cellular and Molecular Imaging Lab, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Matthias D'Huyvetter
- In Vivo Cellular and Molecular Imaging Lab, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Lab, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| |
Collapse
|
32
|
Yaghoubi S, Karimi MH, Lotfinia M, Gharibi T, Mahi-Birjand M, Kavi E, Hosseini F, Sineh Sepehr K, Khatami M, Bagheri N, Abdollahpour-Alitappeh M. Potential drugs used in the antibody-drug conjugate (ADC) architecture for cancer therapy. J Cell Physiol 2019; 235:31-64. [PMID: 31215038 DOI: 10.1002/jcp.28967] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 05/20/2019] [Indexed: 01/04/2023]
Abstract
Cytotoxic small-molecule drugs have a major influence on the fate of antibody-drug conjugates (ADCs). An ideal cytotoxic agent should be highly potent, remain stable while linked to ADCs, kill the targeted tumor cell upon internalization and release from the ADCs, and maintain its activity in multidrug-resistant tumor cells. Lessons learned from successful and failed experiences in ADC development resulted in remarkable progress in the discovery and development of novel highly potent small molecules. A better understanding of such small-molecule drugs is important for development of effective ADCs. The present review discusses requirements making a payload appropriate for antitumor ADCs and focuses on the main characteristics of commonly-used cytotoxic payloads that showed acceptable results in clinical trials. In addition, the present study represents emerging trends and recent advances of payloads used in ADCs currently under clinical trials.
Collapse
Affiliation(s)
- Sajad Yaghoubi
- Department of Clinical Microbiology, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Majid Lotfinia
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran.,Core Research Lab, Kashan University of Medical Sciences, Kashan, Iran
| | - Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Motahare Mahi-Birjand
- Infectious Disease Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Esmaeil Kavi
- Department of Nursing, School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Fahimeh Hosseini
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Koushan Sineh Sepehr
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Khatami
- NanoBioelectrochemistry Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | |
Collapse
|
33
|
Antibody-cytokine fusion proteins: Biopharmaceuticals with immunomodulatory properties for cancer therapy. Adv Drug Deliv Rev 2019; 141:67-91. [PMID: 30201522 DOI: 10.1016/j.addr.2018.09.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/29/2018] [Accepted: 09/04/2018] [Indexed: 01/07/2023]
Abstract
Cytokines have long been used for therapeutic applications in cancer patients. Substantial side effects and unfavorable pharmacokinetics limit their application and may prevent dose escalation to therapeutically active regimens. Antibody-cytokine fusion proteins (often referred to as immunocytokines) may help localize immunomodulatory cytokine payloads to the tumor, thereby activating anticancer immune responses. A variety of formats (e.g., intact IgGs or antibody fragments), molecular targets (e.g., extracellular matrix components and cell membrane antigens) and cytokine payloads have been considered for the development of this novel class of biopharmaceuticals. This review presents the basic concepts on the design and engineering of immunocytokines, reviews their potential limitations, points out emerging opportunities and summarizes key features of preclinical and clinical-stage products.
Collapse
|
34
|
Richards DA. Exploring alternative antibody scaffolds: Antibody fragments and antibody mimics for targeted drug delivery. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 30:35-46. [PMID: 30553519 DOI: 10.1016/j.ddtec.2018.10.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 05/20/2023]
Abstract
The field of targeted therapeutics has benefitted immeasurably from the development of high-affinity antibodies. These important ligands have facilitated the development of effective therapies, particularly when conjugated to potent cytotoxic payloads i.e. in antibody-drug conjugates (ADCs). The success of ADCs is evidenced by rapid adoption within the pharmaceuticals community; many major companies have dedicated ADC research programmes. However, despite the advantages, the field of ADCs has failed to live up to its full potential. Studies have emerged suggesting that traditional IgG scaffolds may not be the optimal format for targeted payload delivery. In response, the protein engineering community has begun to explore alternative high-binding protein scaffolds as antibody mimics. In this short review I will summarise the generation, modification, and application of emerging antibody fragments and synthetic antibody mimics, with a focus on their use as drug carriers. The review aims to highlight the advantages of antibody mimics, and how they could be employed to overcome the issues and limitations of traditional ADCs.
Collapse
Affiliation(s)
- Daniel A Richards
- Department of Chemistry, University College London, 20 Gordon Street, London, WC1H 0AJ, UK.
| |
Collapse
|
35
|
Tsai WTK, Wu AM. Aligning physics and physiology: Engineering antibodies for radionuclide delivery. J Labelled Comp Radiopharm 2018; 61:693-714. [PMID: 29537104 PMCID: PMC6105424 DOI: 10.1002/jlcr.3622] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/21/2018] [Accepted: 03/05/2018] [Indexed: 12/12/2022]
Abstract
The exquisite specificity of antibodies and antibody fragments renders them excellent agents for targeted delivery of radionuclides. Radiolabeled antibodies and fragments have been successfully used for molecular imaging and radioimmunotherapy (RIT) of cell surface targets in oncology and immunology. Protein engineering has been used for antibody humanization essential for clinical applications, as well as optimization of important characteristics including pharmacokinetics, biodistribution, and clearance. Although intact antibodies have high potential as imaging and therapeutic agents, challenges include long circulation time in blood, which leads to later imaging time points post-injection and higher blood absorbed dose that may be disadvantageous for RIT. Using engineered fragments may address these challenges, as size reduction and removal of Fc function decreases serum half-life. Radiolabeled fragments and pretargeting strategies can result in high contrast images within hours to days, and a reduction of RIT toxicity in normal tissues. Additionally, fragments can be engineered to direct hepatic or renal clearance, which may be chosen based on the application and disease setting. This review discusses aligning the physical properties of radionuclides (positron, gamma, beta, alpha, and Auger emitters) with antibodies and fragments and highlights recent advances of engineered antibodies and fragments in preclinical and clinical development for imaging and therapy.
Collapse
Affiliation(s)
- Wen-Ting K Tsai
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Anna M Wu
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
36
|
Ventura E, Weller M, Macnair W, Eschbach K, Beisel C, Cordazzo C, Claassen M, Zardi L, Burghardt I. TGF-β induces oncofetal fibronectin that, in turn, modulates TGF-β superfamily signaling in endothelial cells. J Cell Sci 2018; 131:jcs.209619. [PMID: 29158223 DOI: 10.1242/jcs.209619] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 11/14/2017] [Indexed: 12/31/2022] Open
Abstract
Gene splicing profiles are frequently altered in cancer, and the splice variants of fibronectin (FN) that contain the extra-domains A (EDA) or B (EDB), referred to as EDA+FN or EDB+FN, are highly upregulated in tumor vasculature. Transforming growth factor β (TGF-β) signaling has been attributed a pivotal role in glioblastoma, with TGF-β promoting angiogenesis and vessel remodeling. By using immunohistochemistry staining, we observed that the oncofetal FN isoforms EDA+FN and EDB+FN are expressed in glioblastoma vasculature. Ex vivo single-cell gene expression profiling of tumors by using CD31 and α-smooth muscle actin (αSMA) as markers for endothelial cells, and pericytes and vascular smooth muscle cells (VSMCs), respectively, confirmed the predominant expression of FN, EDA+FN and EDB+FN in the vascular compartment of glioblastoma. Specifically, within the CD31-positive cell population, we identified a positive correlation between the expression of EDA+FN and EDB+FN, and of molecules associated with TGF-β signaling. Further, TGF-β induced EDA+FN and EDB+FN in human cerebral microvascular endothelial cells and glioblastoma-derived endothelial cells in a SMAD3- and SMAD4-dependent manner. In turn, we found that FN modulated TGF-β superfamily signaling in endothelial cells via the EDA and EDB, pointing towards a bidirectional influence of oncofetal FN and TGF-β superfamily signaling.
Collapse
Affiliation(s)
- Elisa Ventura
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Michael Weller
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Will Macnair
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Katja Eschbach
- Department of Biosystems Science and Engineering, ETH Zürich, 4058 Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zürich, 4058 Basel, Switzerland
| | - Cinzia Cordazzo
- Sirius-biotech, c/o Advanced Biotechnology Center, 16132 Genoa, Italy
| | - Manfred Claassen
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Luciano Zardi
- Sirius-biotech, c/o Advanced Biotechnology Center, 16132 Genoa, Italy
| | - Isabel Burghardt
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| |
Collapse
|
37
|
Combined Yttrium-90 microsphere selective internal radiation therapy and external beam radiotherapy in patients with hepatocellular carcinoma: From clinical aspects to dosimetry. PLoS One 2018; 13:e0190098. [PMID: 29293557 PMCID: PMC5749761 DOI: 10.1371/journal.pone.0190098] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 12/10/2017] [Indexed: 11/19/2022] Open
Abstract
Purpose Selective internal radiation therapy (SIRT) is an effective treatment strategy for unresectable hepatocellular carcinoma (HCC) patients. However, the prognoses of patients with portal vein thrombosis, extra-hepatic metastases, or residual tumors remain poor when treated with SIRT alone. In these patients, sequential external beam radiotherapy (EBRT) may offer a chance of salvage. Here, we reported the clinical outcomes and the detailed dosimetry analysis of 22 patients treated with combination therapy. Methods Between October 2011 and May 2015, 22 consecutive patients who underwent EBRT after yttrium-90 (90Y) SIRT were included in this study. The post-SIRT 90Y bremsstrahlung SPECT/CT of each patient was transferred to dose distribution by adopting the local deposition hypothesis. The patient-specific 3-dimensional biological effective dose distribution of combined SIRT and EBRT was generated. The overall survival and safety were evaluated. The relationship between dosimetric parameters and liver toxicity was analyzed. Results The mean administered activity of SIRT was 1.50 GBq (range: 0.5–2.8). The mean prescribed dose of EBRT was 42.3 Gy (range: 15–63) in 14 fractions (range: 5–15) and was targeted to the residual liver tumor in 12 patients (55%), portal vein thrombosis in 11 patients (50%), and perihilar lymphadenopathies in 4 patients (18%). The overall 1-, 2-, and 3-year survival rates were 59.8%, 47.9%, and 47.9%, respectively. Overall, 8 patients (36%) developed > grade 2 liver toxicities, and the Child-Pugh score prior to EBRT strongly affected the toxicity risk. A dosimetry analysis restricted to 18 Child-Pugh A/B patients showed that the V100 (The fraction of normal liver exposed to more than 100 Gy) to V140 significance differed between patients who did or did not experience hepatotoxicity. The V110 was the strongest predictor of hepatotoxicity (18.6±11.6% vs 29.5±5.8%; P = 0.030). Conclusion Combined therapy is feasible and safe if patients are carefully selected. Specifically, 3-dimensional dosimetry is crucial for the evaluation of efficacy and toxicity. The normal liver V100 to V140 values of the combined dose should be as low as possible to minimize the risk of liver toxicity.
Collapse
|
38
|
Falzone N, Ackerman NL, Rosales LDLF, Bernal MA, Liu X, Peeters SGJA, Soto MS, Corroyer-Dulmont A, Bernaudin M, Grimoin E, Touzani O, Sibson NR, Vallis KA. Dosimetric evaluation of radionuclides for VCAM-1-targeted radionuclide therapy of early brain metastases. Theranostics 2018; 8:292-303. [PMID: 29290808 PMCID: PMC5743475 DOI: 10.7150/thno.22217] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/02/2017] [Indexed: 11/13/2022] Open
Abstract
Brain metastases develop frequently in patients with breast cancer, and present a pressing therapeutic challenge. Expression of vascular cell adhesion molecule 1 (VCAM-1) is upregulated on brain endothelial cells during the early stages of metastasis and provides a target for the detection and treatment of early brain metastases. The aim of this study was to use a model of early brain metastasis to evaluate the efficacy of α-emitting radionuclides, 149Tb, 211At, 212Pb, 213Bi and 225Ac; β-emitting radionuclides, 90Y, 161Tb and 177Lu; and Auger electron (AE)-emitters 67Ga, 89Zr, 111In and 124I, for targeted radionuclide therapy (TRT). METHODS Histologic sections and two photon microscopy of mouse brain parenchyma were used to inform a cylindrical vessel geometry using the Geant4 general purpose Monte Carlo (MC) toolkit with the Geant4-DNA low energy physics models. Energy deposition was evaluated as a radial function and the resulting phase spaces were superimposed on a DNA model to estimate double-strand break (DSB) yields for representative β- and α-emitters, 177Lu and 212Pb. Relative biological effectiveness (RBE) values were determined by only evaluating DNA damage due to physical interactions. RESULTS 177Lu produced 2.69 ± 0.08 DSB per GbpGy, without significant variation from the lumen of the vessel to a radius of 100 µm. The DSB yield of 212Pb included two local maxima produced by the 6.1 MeV and 8.8 MeV α-emissions from decay products, 212Bi and 212Po, with yields of 7.64 ± 0.12 and 9.15 ± 0.24 per GbpGy, respectively. Given its higher DSB yield 212Pb may be more effective for short range targeting of early micrometastatic lesions than 177Lu. CONCLUSION MC simulation of a model of early brain metastases provides invaluable insight into the potential efficacy of α-, β- and AE-emitting radionuclides for TRT. 212Pb, which has the attributes of a theranostic radionuclide since it can be used for SPECT imaging, showed a favorable dose profile and RBE.
Collapse
Affiliation(s)
- Nadia Falzone
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Nicole L. Ackerman
- Department of Physics and Astronomy, Agnes Scott College, Decatur, GA, United States of America
| | | | - Mario A. Bernal
- Departamento de Física Aplicada, Instituto de Física "Gleb Wataghin", UNICAMP, Campinas, Brazil
| | - Xiaoxuan Liu
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Sarah GJA Peeters
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Manuel Sarmiento Soto
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Aurélien Corroyer-Dulmont
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, Caen, France
| | - Myriam Bernaudin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, Caen, France
| | - Elisa Grimoin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, Caen, France
| | - Omar Touzani
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, Caen, France
| | - Nicola R. Sibson
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Katherine A. Vallis
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
39
|
Gill MR, Falzone N, Du Y, Vallis KA. Targeted radionuclide therapy in combined-modality regimens. Lancet Oncol 2017; 18:e414-e423. [PMID: 28677577 DOI: 10.1016/s1470-2045(17)30379-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 03/27/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Abstract
Targeted radionuclide therapy (TRT) is a branch of cancer medicine concerned with the use of radioisotopes, radiolabelled molecules, nanoparticles, or microparticles that either naturally accumulate in or are designed to target tumours. TRT combines the specificity of molecular and sometimes physical targeting with the potent cytotoxicity of ionising radiation. Targeting vectors for TRT include antibodies, antibody fragments, proteins, peptides, and small molecules. The diversity of available carrier molecules, together with the large panel of suitable radioisotopes with unique physicochemical properties, allows vector-radionuclide pairings to be matched to the molecular, pathological, and physical characteristics of a tumour. Some pairings are designed for dual therapeutic and diagnostic applications. Use of TRT is increasing with the adoption into practice of radium-223 dichloride for the treatment of bone metastases and with the ongoing clinical development of, among others, 177Lu-dodecanetetraacetic acid tyrosine-3-octreotate (DOTATATE) for the treatment of neuroendocrine tumours and 90Y-microspheres for the treatment of hepatic tumours. The increasing use of TRT raises the question of how best to integrate TRT into multimodality protocols. Achievements in this area and the future prospects of TRT are evaluated in this Review.
Collapse
Affiliation(s)
- Martin R Gill
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Nadia Falzone
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Yong Du
- The Royal Marsden Hospital NHS Foundation Trust, Sutton, Surrey, UK
| | - Katherine A Vallis
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK.
| |
Collapse
|
40
|
Ye XX, Zhao YY, Wang Q, Xiao W, Zhao J, Peng YJ, Cao DH, Lin WJ, Si-Tu MY, Li MZ, Zhang X, Zhang WG, Xia YF, Yang X, Feng GK, Zeng MS. EDB Fibronectin-Specific SPECT Probe 99mTc-HYNIC-ZD2 for Breast Cancer Detection. ACS OMEGA 2017; 2:2459-2468. [PMID: 30023665 PMCID: PMC6044779 DOI: 10.1021/acsomega.7b00226] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 05/19/2017] [Indexed: 06/08/2023]
Abstract
Extradomain-B fibronectin (EDB-FN), an oncofetal isoform of FN, is a promising diagnostic and therapeutic target of tumors, including breast cancer. Many EDB-FN-targeted drugs have been developed and have shown therapeutic effects in clinical trials. Molecular imaging to visualize EDB-FN-positive cancers may help select the right patients who will be benefit from EDB-FN-targeted therapy. Although a few EDB-FN-targeted imaging probes have been developed, the complicated manufacturing procedure and expensive material and equipment required limit their application for large-scale screening of EDB-FN-positive cancer patients. Thus, more simple and economic EDB-FN-targeted imaging probes are still urgently needed. Previously, we have identified a breast cancer-targeted peptide, CTVRTSADC. Coincidently, it was later identified as an EDB-FN-targeted peptide and named ZD2. In this study, we found a positive correlation between the binding activity of the ZD2 phage and the expression level of EDB-FN in breast cancer cells. Moreover, we observed the colocalization of the ZD2 peptide with EDB-FN in breast cancer cells. Furthermore, in vivo tumor targeting of the ZD2 phage, near-infrared fluorescence imaging, and flow cytometry showed tumor-specific homing of the ZD2 peptide in mice bearing EDB-FN-positive breast cancers. Importantly, on the basis of this EDB-FN-targeted ZD2 peptide, we developed a kit-formulated probe, 99mTc-HYNIC-ZD2, for single-photon-emission computed tomography (SPECT) imaging of breast cancer. The high tumor uptake of 99mTc-HYNIC-ZD2 demonstrated its feasibility for use in visualizing EDB-FN-positive breast cancers in vivo. This kit-formulated EDB-FN-targeted SPECT probe has potential clinical applications for precision screening of EDB-FN-positive cancer patients who may benefit from EDB-FN-targeted therapy.
Collapse
Affiliation(s)
- Xiao-Xuan Ye
- State
Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine,
Sun Yat-sen University Cancer Center, and Zhongshan School of Medicine, Guangzhou 510060, China
- Key
Laboratory of Functional Molecules from Marine Microorganisms, Zhongshan
School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yi-Ying Zhao
- Department of Neurosurgery, Biological Therapeutic
Center, Department of Medical
Imaging, Medical
Experimental Animal Center, Nuclear Medicine Department, and Radiation Oncology Center, State Key Laboratory of Oncology in South China, Collaborative
Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer
Center, Guangzhou 510060, China
| | - Qian Wang
- State
Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine,
Sun Yat-sen University Cancer Center, and Zhongshan School of Medicine, Guangzhou 510060, China
| | - Wei Xiao
- Department of Neurosurgery, Biological Therapeutic
Center, Department of Medical
Imaging, Medical
Experimental Animal Center, Nuclear Medicine Department, and Radiation Oncology Center, State Key Laboratory of Oncology in South China, Collaborative
Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer
Center, Guangzhou 510060, China
| | - Jing Zhao
- Department of Neurosurgery, Biological Therapeutic
Center, Department of Medical
Imaging, Medical
Experimental Animal Center, Nuclear Medicine Department, and Radiation Oncology Center, State Key Laboratory of Oncology in South China, Collaborative
Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer
Center, Guangzhou 510060, China
| | - Yong-Jian Peng
- Department of Neurosurgery, Biological Therapeutic
Center, Department of Medical
Imaging, Medical
Experimental Animal Center, Nuclear Medicine Department, and Radiation Oncology Center, State Key Laboratory of Oncology in South China, Collaborative
Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer
Center, Guangzhou 510060, China
| | - De-Hai Cao
- Department of Neurosurgery, Biological Therapeutic
Center, Department of Medical
Imaging, Medical
Experimental Animal Center, Nuclear Medicine Department, and Radiation Oncology Center, State Key Laboratory of Oncology in South China, Collaborative
Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer
Center, Guangzhou 510060, China
| | - Wen-Jie Lin
- Department of Neurosurgery, Biological Therapeutic
Center, Department of Medical
Imaging, Medical
Experimental Animal Center, Nuclear Medicine Department, and Radiation Oncology Center, State Key Laboratory of Oncology in South China, Collaborative
Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer
Center, Guangzhou 510060, China
| | - Min-Yi Si-Tu
- Department of Neurosurgery, Biological Therapeutic
Center, Department of Medical
Imaging, Medical
Experimental Animal Center, Nuclear Medicine Department, and Radiation Oncology Center, State Key Laboratory of Oncology in South China, Collaborative
Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer
Center, Guangzhou 510060, China
| | - Man-Zhi Li
- State
Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine,
Sun Yat-sen University Cancer Center, and Zhongshan School of Medicine, Guangzhou 510060, China
| | - Xing Zhang
- Department of Neurosurgery, Biological Therapeutic
Center, Department of Medical
Imaging, Medical
Experimental Animal Center, Nuclear Medicine Department, and Radiation Oncology Center, State Key Laboratory of Oncology in South China, Collaborative
Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer
Center, Guangzhou 510060, China
| | - Wei-Guang Zhang
- Department of Neurosurgery, Biological Therapeutic
Center, Department of Medical
Imaging, Medical
Experimental Animal Center, Nuclear Medicine Department, and Radiation Oncology Center, State Key Laboratory of Oncology in South China, Collaborative
Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer
Center, Guangzhou 510060, China
| | - Yun-Fei Xia
- Department of Neurosurgery, Biological Therapeutic
Center, Department of Medical
Imaging, Medical
Experimental Animal Center, Nuclear Medicine Department, and Radiation Oncology Center, State Key Laboratory of Oncology in South China, Collaborative
Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer
Center, Guangzhou 510060, China
| | - Xia Yang
- Key
Laboratory of Functional Molecules from Marine Microorganisms, Zhongshan
School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Guo-Kai Feng
- State
Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine,
Sun Yat-sen University Cancer Center, and Zhongshan School of Medicine, Guangzhou 510060, China
| | - Mu-Sheng Zeng
- State
Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine,
Sun Yat-sen University Cancer Center, and Zhongshan School of Medicine, Guangzhou 510060, China
| |
Collapse
|
41
|
Abstract
During cancer progression, the extracellular matrix (ECM) undergoes dramatic changes, which promote cancer cell migration and invasion. In the remodeled tumor ECM, fibronectin (FN) level is upregulated to assist tumor growth, progression, and invasion. FN serves as a central organizer of ECM molecules and mediates the crosstalk between the tumor microenvironment and cancer cells. Its upregulation is correlated with angiogenesis, cancer progression, metastasis, and drug resistance. A number of FN-targeting ligands have been developed for cancer imaging and therapy. Thus far, FN-targeting imaging agents have been tested for nuclear imaging, MRI, and fluorescence imaging, for tumor detection and localization. FN-targeting therapeutics, including nuclear medicine, chemotherapy drugs, cytokines, and photothermal moieties, were also developed in cancer therapy. Because of the prevalence of FN overexpression in cancer, FN targeting imaging agents and therapeutics have the promise of broad applications in the diagnosis, treatment, and image-guided interventions of many types of cancers. This review will summarize current understanding on the role of FN in cancer, discuss the design and development of FN-targeting agents, and highlight the applications of these FN-targeting agents in cancer imaging and therapy.
Collapse
Affiliation(s)
- Zheng Han
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|
42
|
Laforest R, Dehdashti F, Liu Y, Frye J, Frye S, Luehmann H, Sultan D, Shan JS, Freimark BD, Siegel BA. First-in-Man Evaluation of 124I-PGN650: A PET Tracer for Detecting Phosphatidylserine as a Biomarker of the Solid Tumor Microenvironment. Mol Imaging 2017; 16:1536012117733349. [PMID: 29037107 PMCID: PMC5648081 DOI: 10.1177/1536012117733349] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/17/2017] [Accepted: 08/20/2017] [Indexed: 11/24/2022] Open
Abstract
PURPOSE PGN650 is a F(ab')2 antibody fragment that targets phosphatidylserine (PS), a marker normally absent that becomes exposed on tumor cells and tumor vasculature in response to oxidative stress and increases in response to therapy. PGN650 was labeled with 124I to create a positron emission tomography (PET) agent as an in vivo biomarker for tumor microenvironment and response to therapy. In this phase 0 study, we evaluated the pharmacokinetics, safety, radiation dosimetry, and tumor targeting of this tracer in a cohort of patients with cancer. METHODS Eleven patients with known solid tumors received approximately 140 MBq (3.8 mCi) 124I-PGN650 intravenously and underwent positron emission tomography-computed tomography (PET/CT) approximately 1 hour, 3 hours, and either 24 hours or 48 hours later to establish tracer kinetics for the purpose of calculating radiation dosimetry (from integration of the organ time-activity curves and OLINDA/EXM using the adult male and female models). RESULTS Known tumor foci demonstrated mildly increased uptake, with the highest activity at the latest imaging time. There were no unexpected adverse events. The liver was the organ receiving the highest radiation dose (0.77 mGy/MBq); the effective dose was 0.41 mSv/MBq. CONCLUSION Although 124I-PGN650 is safe for human PET imaging, the tumor targeting with this agent in patients was less than previously observed in animal studies.
Collapse
Affiliation(s)
- Richard Laforest
- Division of Radiological Sciences, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Farrokh Dehdashti
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Yongjian Liu
- Division of Radiological Sciences, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
| | - Jennifer Frye
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Sarah Frye
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Hannah Luehmann
- Division of Radiological Sciences, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Deborah Sultan
- Division of Radiological Sciences, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | | | | | - Barry A. Siegel
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
43
|
Cazzamalli S, Corso AD, Neri D. Linker stability influences the anti-tumor activity of acetazolamide-drug conjugates for the therapy of renal cell carcinoma. J Control Release 2016; 246:39-45. [PMID: 27890855 DOI: 10.1016/j.jconrel.2016.11.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/26/2016] [Accepted: 11/17/2016] [Indexed: 10/20/2022]
Abstract
Small molecule-drug conjugates (SMDCs) are increasingly being considered as an alternative to antibody-drug conjugates (ADCs) for the selective delivery of anticancer agents to the tumor site, sparing normal tissues. Carbonic anhydrase IX (CAIX) is a membrane-bound enzyme, which is over-expressed in the majority of renal cell carcinomas and which can be efficiently targeted in vivo, using charged derivatives of acetazolamide, a small heteroaromatic sulfonamide. Here, we show that SMDC products, obtained by the coupling of acetazolamide with monomethyl auristatin E (MMAE) using dipeptide linkers, display a potent anti-tumoral activity in mice bearing xenografted SKRC-52 renal cell carcinomas. A comparative evaluation of four dipeptides revealed that SMDCs featuring valine-citrulline and valine-alanine linkers exhibited greater serum stability and superior therapeutic activity, compared to the counterparts with valine-lysine or valine-arginine linkers. The most active products substantially inhibited tumor growth over a prolonged period of time, in a tumor model for which sunitinib and sorafenib do not display therapeutic activity. However, complete tumor eradication was not possible even after ten intravenous injection. Macroscopic near-infrared imaging procedures confirmed that ligands had not lost the ability to selectively localize at the tumor site at the end of therapy and that the neoplastic masses continued to express CAIX. The findings are of mechanistic and of therapeutic significance, since CAIX is a non-internalizing membrane-associated antigen, which can be considered for targeted drug delivery applications in kidney cancer patients.
Collapse
Affiliation(s)
- Samuele Cazzamalli
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zürich (Switzerland)
| | - Alberto Dal Corso
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zürich (Switzerland)
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zürich (Switzerland)
| |
Collapse
|
44
|
Abstract
The past decade has witnessed impressive advances in cancer treatment ushered in by targeted and immunotherapies. However, with significantly prolonged survival, upon recurrence, more patients become inflicted by brain metastasis, which is mostly refractory to all currently available therapeutic regimens. Historically, brain metastasis is an understudied area in cancer research, partly due to the dearth of appropriate experimental models that closely simulate the special biological features of metastasis in the unique brain environment and to the sophistication of techniques required to perform in-depth studies of the extremely complex and challenging brain metastasis. Yet, with increasing clinical demand for more effective treatment options, brain metastasis research has rapidly advanced in recent years. The present review spotlights the recent major progresses in basic and translational studies of brain metastasis with focuses on new animal models, novel imaging technologies, omics "big data" resources, and some new and exciting biological insights on brain metastasis.
Collapse
|
45
|
Antibody-drug conjugates: Current status and future perspectives. Pharmacol Ther 2016; 167:48-59. [DOI: 10.1016/j.pharmthera.2016.07.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2016] [Indexed: 02/02/2023]
|
46
|
Cazzamalli S, Dal Corso A, Neri D. Acetazolamide Serves as Selective Delivery Vehicle for Dipeptide-Linked Drugs to Renal Cell Carcinoma. Mol Cancer Ther 2016; 15:2926-2935. [PMID: 27609641 DOI: 10.1158/1535-7163.mct-16-0283] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/27/2016] [Accepted: 08/28/2016] [Indexed: 01/12/2023]
Abstract
In most cases, cytotoxic drugs do not preferentially accumulate at the tumor site, causing unwanted toxicities and preventing dose escalation to therapeutically active regimens. Here, we show that acetazolamide derivatives, which bind to carbonic anhydrase IX (CAIX) on the surface of kidney cancer cells, selectively deliver payloads at the site of disease, sparing normal organs. Biodistribution studies, performed in tumor-bearing mice with acetazolamide derivatives bearing a technetium-99m chelator complex or a red fluorophore as payload, revealed a preferential tumor accumulation of the compound at doses up to 560 nmol/kg. The percentage of injected dose per gram in the tumor was dose-dependent and revealed optimal tumor:organ ratios at 140 nmol/kg, with a tumor:blood ratio of 80:1 at 6 hours. Acetazolamide, coupled to potent cytotoxic drugs via a dipeptide linker, exhibited a potent antitumor activity in nude mice bearing SKRC-52 renal cell carcinomas, whereas drug derivatives devoid of the acetazolamide moiety did not exhibit any detectable anticancer activity at the same doses. The observation of tumor regression with a noninternalizing ligand and with different cytotoxic moieties (MMAE and PNU-159682) indicates a general mechanism of action, based on the selective accumulation of the product on tumor cells, followed by the extracellular proteolytic release of the cytotoxic payload at the neoplastic site and the subsequent drug internalization into tumor cells. Acetazolamide-based drug conjugates may represent a promising class of targeted agents for the treatment of metastatic kidney cancer, as the majority of human clear cell renal cell carcinomas are strongly positive for CAIX. Mol Cancer Ther; 15(12); 2926-35. ©2016 AACR.
Collapse
Affiliation(s)
- Samuele Cazzamalli
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich (Switzerland)
| | - Alberto Dal Corso
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich (Switzerland)
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich (Switzerland).
| |
Collapse
|
47
|
Wang X, Sun Q, Shen S, Xu Y, Huang L. Nanotrastuzumab in combination with radioimmunotherapy: Can it be a viable treatment option for patients with HER2-positive breast cancer with brain metastasis? Med Hypotheses 2016; 88:79-81. [DOI: 10.1016/j.mehy.2015.12.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 12/08/2015] [Accepted: 12/15/2015] [Indexed: 10/22/2022]
|
48
|
Fibronectin-targeted drug delivery in cancer. Adv Drug Deliv Rev 2016; 97:101-10. [PMID: 26639577 DOI: 10.1016/j.addr.2015.11.014] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/21/2015] [Accepted: 11/23/2015] [Indexed: 01/09/2023]
Abstract
Fibronectin is an extracellular matrix protein with pivotal physiological and pathological functions in development and adulthood. Alternative splicing of the precursor mRNA, produced from the single copy fibronectin gene, occurs at three sites coding for the EDA, EDB and IIICS domains. Fibronectin isoforms comprising the EDA or EDB domains are known as oncofetal forms due to their developmental importance and their re-expression in tumors, contrasting with restricted presence in normal adult tissues. These isoforms are also recognized as important markers of angiogenesis, a crucial physiological process in development and required by tumor cells in cancer progression. Attributed to this feature, EDA and EDB domains have been extensively used for the targeted delivery of cytokines, cytotoxic agents, chemotherapy drugs and radioisotopes to fibronectin-expressing tumors to exert therapeutic effects on primary cancers and metastatic lesions. In addition to drug delivery, the EDA and EDB domains of fibronectin have also been utilized to develop imaging strategies for tumor tissues. Furthermore, EDA and EDB based vaccines seem to be promising for the treatment and prevention of certain cancer types. In this review, we will summarize recent advances in fibronectin EDA and EDB-based therapeutic strategies developed to treat cancer.
Collapse
|
49
|
Freise AC, Wu AM. In vivo imaging with antibodies and engineered fragments. Mol Immunol 2015; 67:142-52. [PMID: 25934435 PMCID: PMC4529772 DOI: 10.1016/j.molimm.2015.04.001] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/31/2015] [Accepted: 04/02/2015] [Indexed: 12/20/2022]
Abstract
Antibodies have clearly demonstrated their utility as therapeutics, providing highly selective and effective drugs to treat diseases in oncology, hematology, cardiology, immunology and autoimmunity, and infectious diseases. More recently, a pressing need for equally specific and targeted imaging agents for assessing disease in vivo, in preclinical models and patients, has emerged. This review summarizes strategies for developing and optimizing antibodies as targeted probes for use in non-invasive imaging using radioactive, optical, magnetic resonance, and ultrasound approaches. Recent advances in engineered antibody fragments and scaffolds, conjugation and labeling methods, and multimodality probes are highlighted. Importantly, antibody-based imaging probes are seeing new applications in detection and quantitation of cell surface biomarkers, imaging specific responses to targeted therapies, and monitoring immune responses in oncology and other diseases. Antibody-based imaging will provide essential tools to facilitate the transition to truly precision medicine.
Collapse
Affiliation(s)
- Amanda C Freise
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, USA
| | - Anna M Wu
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, USA.
| |
Collapse
|
50
|
Casi G, Neri D. Antibody–Drug Conjugates and Small Molecule–Drug Conjugates: Opportunities and Challenges for the Development of Selective Anticancer Cytotoxic Agents. J Med Chem 2015; 58:8751-61. [DOI: 10.1021/acs.jmedchem.5b00457] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Giulio Casi
- Philochem AG, Libernstrasse 3, CH8112 Otelfingen, Switzerland
| | - Dario Neri
- Department
of Chemistry and Applied Biosciences, Institute of Pharmaceutical
Sciences, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| |
Collapse
|