1
|
Chen X, Habib S, Alexandru M, Chauhan J, Evan T, Troka JM, Rahimi A, Esapa B, Tull TJ, Ng WZ, Fitzpatrick A, Wu Y, Geh JLC, Lloyd-Hughes H, Palhares LCGF, Adams R, Bax HJ, Whittaker S, Jacków-Malinowska J, Karagiannis SN. Chondroitin Sulfate Proteoglycan 4 (CSPG4) as an Emerging Target for Immunotherapy to Treat Melanoma. Cancers (Basel) 2024; 16:3260. [PMID: 39409881 PMCID: PMC11476251 DOI: 10.3390/cancers16193260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
Immunotherapies, including checkpoint inhibitor antibodies, have precipitated significant improvements in clinical outcomes for melanoma. However, approximately half of patients do not benefit from approved treatments. Additionally, apart from Tebentafusp, which is approved for the treatment of uveal melanoma, there is a lack of immunotherapies directly focused on melanoma cells. This is partly due to few available targets, especially those expressed on the cancer cell surface. Chondroitin sulfate proteoglycan 4 (CSPG4) is a cell surface molecule overexpressed in human melanoma, with restricted distribution and low expression in non-malignant tissues and involved in several cancer-promoting and dissemination pathways. Here, we summarize the current understanding of the expression and functional significance of CSPG4 in health and melanoma, and we outline immunotherapeutic strategies. These include monoclonal antibodies, antibody-drug conjugates (ADCs), chimeric-antigen receptor (CAR) T cells, and other strategies such as anti-idiotypic and mimotope vaccines to raise immune responses against CSPG4-expressing melanomas. Several showed promising functions in preclinical models of melanoma, yet few have reached clinical testing, and none are approved for therapeutic use. Obstacles preventing that progress include limited knowledge of CSPG4 function in human cancer and a lack of in vivo models that adequately represent patient immune responses and human melanoma biology. Despite several challenges, immunotherapy directed to CSPG4-expressing melanoma harbors significant potential to transform the treatment landscape.
Collapse
Affiliation(s)
- Xinyi Chen
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Shabana Habib
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Madalina Alexandru
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Jitesh Chauhan
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Theodore Evan
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Joanna M. Troka
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Avigail Rahimi
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Benjamina Esapa
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Thomas J. Tull
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Wen Zhe Ng
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Amanda Fitzpatrick
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
- Oncology Department, Guy’s and St Thomas’ Hospitals, London SE1 9RT, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Innovation Hub, Guy’s Hospital, London SE1 9RT, UK
| | - Yin Wu
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Innovation Hub, Guy’s Hospital, London SE1 9RT, UK
- Peter Gorer Department of Immunobiology, Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Jenny L. C. Geh
- St John’s Institute of Dermatology, Guy’s, King’s and St. Thomas’ Hospitals NHS Foundation Trust, London SE1 9RT, UK
- Department of Plastic Surgery, Guy’s, King’s and St. Thomas’ Hospitals, London SE1 9RT, UK
| | - Hawys Lloyd-Hughes
- Department of Plastic Surgery, Guy’s, King’s and St. Thomas’ Hospitals, London SE1 9RT, UK
| | - Lais C. G. F. Palhares
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Rebecca Adams
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Heather J. Bax
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Sean Whittaker
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Joanna Jacków-Malinowska
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
| | - Sophia N. Karagiannis
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, London SE1 9RT, UK (J.M.T.); (A.R.); (H.J.B.)
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Innovation Hub, Guy’s Hospital, London SE1 9RT, UK
| |
Collapse
|
2
|
Houerbi N, Kim J, Overbey EG, Batra R, Schweickart A, Patras L, Lucotti S, Ryon KA, Najjar D, Meydan C, Damle N, Chin C, Narayanan SA, Guarnieri JW, Widjaja G, Beheshti A, Tobias G, Vatter F, Hirschberg JW, Kleinman A, Afshin EE, MacKay M, Chen Q, Miller D, Gajadhar AS, Williamson L, Tandel P, Yang Q, Chu J, Benz R, Siddiqui A, Hornburg D, Gross S, Shirah B, Krumsiek J, Mateus J, Mao X, Matei I, Mason CE. Secretome profiling reveals acute changes in oxidative stress, brain homeostasis, and coagulation following short-duration spaceflight. Nat Commun 2024; 15:4862. [PMID: 38862464 PMCID: PMC11166969 DOI: 10.1038/s41467-024-48841-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/15/2024] [Indexed: 06/13/2024] Open
Abstract
As spaceflight becomes more common with commercial crews, blood-based measures of crew health can guide both astronaut biomedicine and countermeasures. By profiling plasma proteins, metabolites, and extracellular vesicles/particles (EVPs) from the SpaceX Inspiration4 crew, we generated "spaceflight secretome profiles," which showed significant differences in coagulation, oxidative stress, and brain-enriched proteins. While >93% of differentially abundant proteins (DAPs) in vesicles and metabolites recovered within six months, the majority (73%) of plasma DAPs were still perturbed post-flight. Moreover, these proteomic alterations correlated better with peripheral blood mononuclear cells than whole blood, suggesting that immune cells contribute more DAPs than erythrocytes. Finally, to discern possible mechanisms leading to brain-enriched protein detection and blood-brain barrier (BBB) disruption, we examined protein changes in dissected brains of spaceflight mice, which showed increases in PECAM-1, a marker of BBB integrity. These data highlight how even short-duration spaceflight can disrupt human and murine physiology and identify spaceflight biomarkers that can guide countermeasure development.
Collapse
Affiliation(s)
- Nadia Houerbi
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Eliah G Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Richa Batra
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Annalise Schweickart
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional Biology and Medicine program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Laura Patras
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Serena Lucotti
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Krista A Ryon
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Deena Najjar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Namita Damle
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Christopher Chin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - S Anand Narayanan
- Department of Nutrition & Integrative Physiology, Florida State University, Tallahassee, FL, USA
| | - Joseph W Guarnieri
- Center of Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Gabrielle Widjaja
- Center of Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Afshin Beheshti
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Gabriel Tobias
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Seer, Inc., Redwood City, CA, 94065, USA
| | - Fanny Vatter
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Seer, Inc., Redwood City, CA, 94065, USA
| | | | - Ashley Kleinman
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Evan E Afshin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Matthew MacKay
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Qiuying Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Dawson Miller
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | | | | | | | - Qiu Yang
- Seer, Inc., Redwood City, CA, 94065, USA
| | | | - Ryan Benz
- Seer, Inc., Redwood City, CA, 94065, USA
| | | | | | - Steven Gross
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Bader Shirah
- Department of Neuroscience, King Faisal Specialist Hospital & Research Centre, Jeddah, Saudi Arabia
| | - Jan Krumsiek
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional Biology and Medicine program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jaime Mateus
- Space Exploration Technologies Corporation (SpaceX), Hawthorne, CA, USA
| | - Xiao Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA, 92350, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA.
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
- Tri-Institutional Biology and Medicine program, Weill Cornell Medicine, New York, NY, 10021, USA.
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
- WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
3
|
KUROKAWA T, IMAI K. Chondroitin sulfate proteoglycan 4: An attractive target for antibody-based immunotherapy. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2024; 100:293-308. [PMID: 38735753 PMCID: PMC11260911 DOI: 10.2183/pjab.100.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/11/2024] [Indexed: 05/14/2024]
Abstract
Multifunctional molecules involved in tumor progression and metastasis have been identified as valuable targets for immunotherapy. Among these, chondroitin sulfate proteoglycan 4 (CSPG4), a significant tumor cell membrane-bound proteoglycan, has emerged as a promising target, especially in light of advances in chimeric antigen receptor (CAR) T-cell therapy. The profound bioactivity of CSPG4 and its role in pivotal processes such as tumor proliferation, migration, and neoangiogenesis underline its therapeutic potential. We reviewed the molecular intricacies of CSPG4, its functional attributes within tumor cells, and the latest clinical-translational advances targeting it. Strategies such as blocking monoclonal antibodies, conjugate therapies, bispecific antibodies, small-molecule inhibitors, CAR T-cell therapies, trispecific killer engagers, and ribonucleic acid vaccines against CSPG4 were assessed. CSPG4 overexpression in diverse tumors and its correlation with adverse prognostic outcomes emphasize its significance in cancer biology. These findings suggest that targeting CSPG4 offers a promising avenue for future cancer therapy, with potential synergistic effects when combined with existing treatments.
Collapse
Affiliation(s)
- Tomohiro KUROKAWA
- Department of Medical Epigenomics Research, Fukushima Medical University, Fukushima, Japan
- Department of Surgery, Jyoban Hospital of Tokiwa Foundation, Fukushima, Japan
| | - Kohzoh IMAI
- Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
4
|
Grossauer A, Uranowska K, Kitzwögerer M, Mostegel M, Breiteneder H, Hafner C. Immunohistochemical detection of the chondroitin sulfate proteoglycan 4 protein in primary and metastatic melanoma. Oncol Lett 2023; 26:382. [PMID: 37559576 PMCID: PMC10407859 DOI: 10.3892/ol.2023.13968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/22/2023] [Indexed: 08/11/2023] Open
Abstract
Treatment of malignant melanoma, the most aggressive form of skin cancer, continues to be a major challenge for clinicians. New targeted therapies with kinase inhibitors or drugs which modify the immune response are often accompanied by the development of resistance or severe side effects. In this context, chondroitin sulfate proteoglycan 4 (CSPG4), a highly immunogenic melanoma tumor antigen, could be a potential target for alternative therapeutic approaches. The aim of the present study was to identify differences in the levels of CSPG4 protein expression in primary and metastatic melanomas as well as to analyze correlations between CSPG4 expression and histopathological data and patient characteristics. A total of 189 melanoma tissue samples from Lower Austria, including primary melanomas and melanoma metastases, were immunohistochemically stained for the expression of CSPG4 and statistical analyses were performed. A total of 65.6% of melanoma tissue samples stained positive for the expression of CSPG4. Primary nodular and primary superficial spreading melanomas demonstrated a significantly higher number of positively stained tissue samples for CSPG4 compared with primary lentigo maligna melanomas. No significant differences in the expression of CSPG4 were demonstrated between primary melanomas and melanoma metastases. The present study supports the advancement of the understanding of CSPG4 tissue expression patterns in melanoma patients and provides additional information for further investigation of CSPG4 as a potential therapeutic target.
Collapse
Affiliation(s)
- Anna Grossauer
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, A-3100 St. Poelten, Austria
- Department of Pathology, University Hospital Krems, Karl Landsteiner University of Health Sciences, A-3500 Krems an der Donau, Austria
| | - Karolina Uranowska
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, A-3100 St. Poelten, Austria
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Melitta Kitzwögerer
- Department of Pathology, University Hospital St. Poelten, A-3100 St. Poelten, Austria
| | - Margit Mostegel
- Department of Pathology, University Hospital Krems, Karl Landsteiner University of Health Sciences, A-3500 Krems an der Donau, Austria
| | - Heimo Breiteneder
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Christine Hafner
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, A-3100 St. Poelten, Austria
- Karl Landsteiner Institute of Dermatological Research, Karl Landsteiner Gesellschaft, A-3100 St. Poelten, Austria
| |
Collapse
|
5
|
Isakova A, Artykov A, Vorontsova Y, Dolgikh D, Kirpichnikov M, Gasparian M, Yagolovich A. Application of an Autoinduction Strategy to Optimize the Heterologous Production of an Antitumor Bispecific Fusion Protein Based on the TRAIL Receptor-Selective Mutant Variant in Escherichia coli. Mol Biotechnol 2023; 65:581-589. [PMID: 36094644 DOI: 10.1007/s12033-022-00561-6] [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: 07/15/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
Autoinduction is a simple approach for heterologous protein expression that helps to achieve the high-level production of recombinant proteins in soluble form. In this work, we investigated if the application of an autoinduction strategy could help to optimize the production of bifunctional protein SRH-DR5-B, the DR5-specific TRAIL variant DR5-B fused to a VEGFR2-specific peptide SRHTKQRHTALH for dual antitumor and antiangiogenic activity. The protein was expressed in Escherichia coli SHuffle B T7, BL21(DE3), and BL21(DE3)pLysS strains. By IPTG induction, the highest expression level was in SHuffle B T7, while by autoinduction, the similar expression level was achieved in BL21(DE3)pLysS. However, in SHuffle B T7, only 45% of IPTG-induced SRH-DR5-B was expressed in soluble form, in contrast to 75% autoinduced in BL21(DE3)pLysS. The yield of purified SRH-DR5-B protein expressed by autoinduction in BL21(DE3)pLysS was 28 ± 4.5 mg per 200 ml of cell culture, which was 1.4 times higher than the yield from IPTG-induced SHuffle B T7. Regardless of the production method, SRH-DR5-B was equally cytotoxic to BxPC-3 human tumor cells expressing DR5 and VEGFR2 receptors. Thus, the production of SRH-DR5-B by autoinduction in the E. coli BL21(DE3)pLysS strain is an efficient, technologically simple, and economical technique that allows to obtain a large amount of active protein from the cytoplasmic cell fraction. Our work demonstrates that the strategy of induction of protein expression is no less important than the strain selection.
Collapse
Affiliation(s)
- Alina Isakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia
| | - Artem Artykov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia
| | - Yekaterina Vorontsova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia
| | - Dmitry Dolgikh
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia.,Faculty of Biology, Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Mikhail Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia.,Faculty of Biology, Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Marine Gasparian
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia
| | - Anne Yagolovich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia. .,Faculty of Biology, Lomonosov Moscow State University, 119991, Moscow, Russia.
| |
Collapse
|
6
|
Li J, Wuethrich A, Zhang Z, Wang J, Lin LL, Behren A, Wang Y, Trau M. SERS Multiplex Profiling of Melanoma Circulating Tumor Cells for Predicting the Response to Immune Checkpoint Blockade Therapy. Anal Chem 2022; 94:14573-14582. [PMID: 36222247 DOI: 10.1021/acs.analchem.2c02398] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Immune checkpoint blockade (ICB) therapy has achieved remarkable success in many cancers including melanoma. However, ICB therapy benefits only a small proportion of patients and produces severe side effects for some patients. Thus, there is an urgent need to identify patients who are more likely to respond to ICB therapy to improve outcomes and minimize side effects. To predict ICB therapy responses, we design a surface-enhanced Raman scattering (SERS) assay for multiplex profiling of circulating tumor cells (CTCs) under basal and interferon-γ (IFN-γ) stimulation. Through simultaneous ensemble and single-cell measurements of CTCs, the SERS assay can reveal tumor heterogeneity and offer a comprehensive CTC phenotype for decision-making. Anisotropic gold-silver alloy nanoboxes are utilized as SERS plasmonic substrates for improved signal readouts of CTC surface biomarkers. By generating a unique CTC signature with four surface biomarkers, the developed assay enables the differentiation of CTCs from three different patient-derived melanoma cell lines. Significantly, in a cohort of 14 melanoma patients who received programmed cell death-1 blockade therapy, the changes of CTC signature induced by IFN-γ stimulation to CTCs show the potential to predict responders. We expect that the SERS assay can help select patients for receiving ICB therapy in other cancers.
Collapse
Affiliation(s)
- Junrong Li
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan430079, P. R. China.,Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD4072, Australia
| | - Alain Wuethrich
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD4072, Australia
| | - Zhen Zhang
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD4072, Australia
| | - Jing Wang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou350007, P. R. China
| | - Lynlee L Lin
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD4102, Australia
| | - Andreas Behren
- Oliva Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, VIC3086, Australia.,Department of Medicine, University of Melbourne, Heidelberg, VIC3010, Australia
| | - Yuling Wang
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW2109, Australia
| | - Matt Trau
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD4072, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD4072, Australia
| |
Collapse
|
7
|
Las Heras K, Royo F, Garcia-Vallicrosa C, Igartua M, Santos-Vizcaino E, Falcon-Perez JM, Hernandez RM. Extracellular vesicles from hair follicle-derived mesenchymal stromal cells: isolation, characterization and therapeutic potential for chronic wound healing. Stem Cell Res Ther 2022; 13:147. [PMID: 35395929 PMCID: PMC8994406 DOI: 10.1186/s13287-022-02824-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/23/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) and their extracellular vesicles (MSC-EVs) have demonstrated to elicit immunomodulatory and pro-regenerative properties that are beneficial for the treatment of chronic wounds. Thanks to different mediators, MSC-EVs have shown to play an important role in the proliferation, migration and cell survival of different skin cell populations. However, there is still a big bid to achieve the most effective, suitable and available source of MSC-EVs. METHODS We isolated, characterized and compared medium-large EVs (m-lEVs) and small EVs (sEVs) obtained from hair follicle-derived MSCs (HF-MSCs) against the gold standard in regenerative medicine, EVs isolated from adipose tissue-derived MSCs (AT-MSCs). RESULTS We demonstrated that HF-EVs, as well as AT-EVs, expressed typical MSC-EVs markers (CD9, CD44, CD63, CD81 and CD105) among other different functional markers. We showed that both cell types were able to increase human dermal fibroblasts (HDFs) proliferation and migration. Moreover, both MSC-EVs were able to increase angiogenesis in human umbilical vein endothelial cells (HUVECs) and protect HDFs exposed to a hyperglycemic environment from oxidative stress and cytotoxicity. CONCLUSIONS Taken together, HF-EVs demonstrated to exhibit comparable potential to that of AT-EVs as promising candidates in the treatment of chronic wounds.
Collapse
Affiliation(s)
- Kevin Las Heras
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Félix Royo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Exosomes Laboratory, 48160, Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), 28029, Madrid, Spain
| | - Clara Garcia-Vallicrosa
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Exosomes Laboratory, 48160, Derio, Spain
| | - Manoli Igartua
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Juan M Falcon-Perez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Exosomes Laboratory, 48160, Derio, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), 28029, Madrid, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013, Bilbao, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU), 01006, Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain.
| |
Collapse
|
8
|
Uranowska K, Samadaei M, Kalic T, Pinter M, Breiteneder H, Hafner C. A chondroitin sulfate proteoglycan 4‑specific monoclonal antibody inhibits melanoma cell invasion in a spheroid model. Int J Oncol 2021; 59:70. [PMID: 34318902 PMCID: PMC8357264 DOI: 10.3892/ijo.2021.5250] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022] Open
Abstract
The overexpression of chondroitin sulfate proteoglycan 4 (CSPG4) is associated with several tumor types, including malignant melanoma, squamous cell carcinoma, triple-negative breast carcinoma, oligodendrocytomas or gliomas. Due to its restricted distribution in normal tissues, CSPG4 has been considered a potential target for several antitumor approaches, including monoclonal antibody (mAb) therapies. The aim of the present study was to characterize the impact of the CSPG4-specific mAb clone 9.2.27 on its own or in combination with the commonly used BRAF-selective inhibitor, PLX4032, on different functions of melanoma cells to assess the potential synergistic effects. The BRAF V600-mutant human melanoma cell lines, M14 (CSPG4-negative) and WM164 (CSPG4-positive), were exposed to the CSPG4-specific 9.2.27 mAb and/or PLX4032. Cell viability and colony formation capacity were evaluated. A 3D-cell culture spheroid model was used to assess the invasive properties of the treated cells. In addition, flow cytometric analysis of apoptosis and cell cycle analyses were performed. Incubation of the WM164 cell line with CSPG4-specific 9.2.27 mAb decreased viability, colony formation ability and the invasive capacity of CSPG4-positive tumor cells, which was not the case for the CSPG4-negative M14 cell line. Combined treatment of the WM164 cells with 9.2.27 mAb plus PLX4032 did not exert any significant additional effect in comparison to treatment with PLX4032 alone in the clonogenic and invasion assays. M14 cell cycle distribution was not influenced by the CSPG4-specific 9.2.27 mAb. By contrast, the exposure of WM164 cells to the mAb resulted in an arrest of the cells in the S phase. Moreover, combined treatment of the WM164 cells led to a significantly increased accumulation of cells in the subG1 phase, combined with a decrease of cells in the G2/M phase. On the whole, findings of the present study indicate that the CSPG4-specific 9.2.27 mAb exerts an anti-invasive effect on CSPG4-positive melanoma spheroids, which is not enhanced by BRAF inhibition. These findings provide the basis for further investigations on the effects of anti-CSPG4-based treatments of CSPG4-positive tumors.
Collapse
Affiliation(s)
- Karolina Uranowska
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, A-3100 St. Poelten, Austria
| | - Mahzeiar Samadaei
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Tanja Kalic
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, A-3100 St. Poelten, Austria
| | - Matthias Pinter
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Heimo Breiteneder
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Christine Hafner
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, A-3100 St. Poelten, Austria
| |
Collapse
|
9
|
Safarzadeh Kozani P, Safarzadeh Kozani P, O'Connor RS. In Like a Lamb; Out Like a Lion: Marching CAR T Cells Toward Enhanced Efficacy in B-ALL. Mol Cancer Ther 2021; 20:1223-1233. [PMID: 33903140 PMCID: PMC8285067 DOI: 10.1158/1535-7163.mct-20-1089] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/26/2021] [Accepted: 04/19/2021] [Indexed: 11/16/2022]
Abstract
Combining synthetic biology with adoptive T-cell transfer has led to promising advances in the treatment of relapsed/refractory B-cell acute lymphoblastic leukemia (R/R B-ALL), diffuse large B-cell lymphoma (DLBCL), and mantle cell lymphoma (MCL). Chimeric antigen receptors (CARs) are synthetic receptors that redirect T-cell specificity against cancer. CARs include "built-in" signaling domains that reprogram T-cell metabolism, enhance effector function, and support long-term persistence. Despite their success in blood-based malignancies, relapse can occur in CD19-redirected CAR T-cell therapies for several reasons, including poor engraftment, impaired in vivo proliferation, and T-cell senescence. Herein, we explain how subtle alterations in CAR design may overcome barriers to effective adoptive immunotherapy. We also discuss how the physiochemical properties of the single-chain variable fragment (scFv) affect differentiation and persistence. Moreover, we describe innovative advances in CAR engineering and provide insight into the development of humanized scFvs whose proposed benefits include increased persistence and improved clinical outcomes. Tumor cells can evade CAR T-cell-mediated detection and elimination due to the emergence or presence of CD19-negative leukemic cell subpopulations. We also discuss the opportunities and challenges in targeting other B-ALL-associated antigens. Identifying alternate targets is fundamentally necessary to restore the success of CAR T-cell therapies in CD19-negative patients with B-ALL.
Collapse
MESH Headings
- Animals
- Antigens, CD19/immunology
- Antigens, Neoplasm/immunology
- Disease Management
- Genetic Engineering
- Humans
- Immunity
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/etiology
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Research Design
- T-Cell Antigen Receptor Specificity/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Treatment Outcome
- Tumor Escape/immunology
Collapse
Affiliation(s)
- Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
- Student Research Committee, Medical Biotechnology Research Center, School of Nursing, Midwifery, and Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Roddy S O'Connor
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Madheswaran S, Mungra N, Biteghe FAN, De la Croix Ndong J, Arowolo AT, Adeola HA, Ramamurthy D, Naran K, Khumalo NP, Barth S. Antibody-Based Targeted Interventions for the Diagnosis and Treatment of Skin Cancers. Anticancer Agents Med Chem 2021; 21:162-186. [PMID: 32723261 DOI: 10.2174/1871520620666200728123006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/19/2020] [Accepted: 04/30/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cutaneous malignancies most commonly arise from skin epidermal cells. These cancers may rapidly progress from benign to a metastatic phase. Surgical resection represents the gold standard therapeutic treatment of non-metastatic skin cancer while chemo- and/or radiotherapy are often used against metastatic tumors. However, these therapeutic treatments are limited by the development of resistance and toxic side effects, resulting from the passive accumulation of cytotoxic drugs within healthy cells. OBJECTIVE This review aims to elucidate how the use of monoclonal Antibodies (mAbs) targeting specific Tumor Associated Antigens (TAAs) is paving the way to improved treatment. These mAbs are used as therapeutic or diagnostic carriers that can specifically deliver cytotoxic molecules, fluorophores or radiolabels to cancer cells that overexpress specific target antigens. RESULTS mAbs raised against TAAs are widely in use for e.g. differential diagnosis, prognosis and therapy of skin cancers. Antibody-Drug Conjugates (ADCs) particularly show remarkable potential. The safest ADCs reported to date use non-toxic photo-activatable Photosensitizers (PSs), allowing targeted Photodynamic Therapy (PDT) resulting in targeted delivery of PS into cancer cells and selective killing after light activation without harming the normal cell population. The use of near-infrared-emitting PSs enables both diagnostic and therapeutic applications upon light activation at the specific wavelengths. CONCLUSION Antibody-based approaches are presenting an array of opportunities to complement and improve current methods employed for skin cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Suresh Madheswaran
- Medical Biotechnology & Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Neelakshi Mungra
- Medical Biotechnology & Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Fleury A N Biteghe
- Department of Radiation Oncology and Biomedical Sciences, Cedars-Sinai Medical, 8700 Beverly Blvd, Los Angeles, CA, United States
| | - Jean De la Croix Ndong
- Department of Orthopedic Surgery, New York University Langone Orthopedic Hospital, 301 East 17th Street, New York, NY, United States
| | - Afolake T Arowolo
- The Hair and Skin Research Lab, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | - Henry A Adeola
- The Hair and Skin Research Lab, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | - Dharanidharan Ramamurthy
- Medical Biotechnology & Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Krupa Naran
- Medical Biotechnology & Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Nonhlanhla P Khumalo
- The Hair and Skin Research Lab, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | - Stefan Barth
- Medical Biotechnology & Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
11
|
Apoptosis-Inducing TNF Superfamily Ligands for Cancer Therapy. Cancers (Basel) 2021; 13:cancers13071543. [PMID: 33801589 PMCID: PMC8036978 DOI: 10.3390/cancers13071543] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is a complex disease with apoptosis evasion as one of its hallmarks; therefore, apoptosis induction in transformed cells seems a promising approach as a cancer treatment. TNF apoptosis-inducing ligands, which are naturally present in the body and possess tumoricidal activity, are attractive candidates. The most studied proteins are TNF-α, FasL, and TNF-related apoptosis-inducing ligand (TRAIL). Over the years, different recombinant TNF family-derived apoptosis-inducing ligands and agonists have been designed. Their stability, specificity, and half-life have been improved because most of the TNF ligands have the disadvantages of having a short half-life and affinity to more than one receptor. Here, we review the outlook on apoptosis-inducing ligands as cancer treatments in diverse preclinical and clinical stages and summarize strategies of overcoming their natural limitations to improve their effectiveness.
Collapse
|
12
|
Evaluating the Combination of Radioimmunotherapy and Immunotherapy in a Melanoma Mouse Model. Int J Mol Sci 2020; 21:ijms21030773. [PMID: 31991626 PMCID: PMC7037880 DOI: 10.3390/ijms21030773] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 01/02/2023] Open
Abstract
Immunotherapy has changed the oncology landscape during the last decade and become standard of care for several cancers. The combinations of immunotherapy with other treatment modalities are also being investigated. One of the challenges to investigate such combinations is to identify suitable mouse models for the pre-clinical experiments. In the past, we and other researchers showed that murine B16-F10 melanoma in C57Bl6 mice is refractory to treatment with immune checkpoint inhibitors. In this work we studied the suitability of an alternative syngeneic model, Cloudman S91 murine melanoma in DBA/2 mouse (DBA/2NCrl), to study the combination of immunotherapy targeting PD-1 and radioimmunotherapy targeting melanin. DBA/2 male and female mice were injected subcutaneously with 3-6 million Cloudman S91 cells. When the tumors reached ~150 mm3 volume, the animals were treated intraperitoneally with PBS (sham), h8C3 unlabeled (cold) antibody to melanin, immunotherapy with anti-PD-1 antibody, radioimmunotherapy with 213Bismuth (213Bi)-labeled h8C3 antibody, or several combinations of immunotherapy and radioimmunotherapy. Treatments with immunotherapy alone produced very modest effect on the tumor size, while combination therapy resulted in significant slowing down of the tumor growth, increased animal survival, and no decrease in animal body weight. We conclude that Cloudman S91 murine melanoma in DBA/2 mouse is a suitable model to evaluate combination of immunotherapy of melanoma with tangentially targeted treatments.
Collapse
|
13
|
Rozga P, Kloska D, Pawlak S, Teska-Kaminska M, Galazka M, Bukato K, Pieczykolan A, Jaworski A, Molga-Kaczmarska A, Kopacz A, Badyra B, Kachamakova-Trojanowska N, Zolnierkiewicz O, Targosz-Korecka M, Poleszak K, Szymanik M, Zerek B, Pieczykolan J, Jozkowicz A, Grochot-Przeczek A. Novel engineered TRAIL-based chimeric protein strongly inhibits tumor growth and bypasses TRAIL resistance. Int J Cancer 2020; 147:1117-1130. [PMID: 31863596 DOI: 10.1002/ijc.32845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 09/23/2019] [Accepted: 09/26/2019] [Indexed: 01/07/2023]
Abstract
Targeting of the TRAIL-DR4/5 pathway was proposed as a promising approach for specific induction of apoptosis in cancer cells. Clinical trials, however, showed inadequate efficiency of TRAIL as a monotherapy. It is a widely held view that the application of multifunctional molecules or combination therapy may lead to substantial improvement. Here, we demonstrate the effectiveness and safety of a novel chimeric protein, AD-O51.4, which is a TRAIL equipped with positively charged VEGFA-derived effector peptides. The study was performed in multiple cancer cell line- and patient-derived xenografts. A pharmacokinetic profile was established in monkeys. AD-O51.4 strongly inhibits tumor growth, even leading to complete long-term tumor remission. Neither mice nor monkeys treated with AD-O51.4 demonstrate symptoms of drug toxicity. AD-O51.4 exhibits a satisfactory half-life in plasma and accumulates preferentially in tumors. The cellular mechanism of AD-O51.4 activity involves both cytotoxic effects in tumor cells and antiangiogenic effects on the endothelium. The presence of DRs in cancer cells is crucial for AD-O51.4-driven apoptosis execution. The TRAIL component of the fusion molecule serves as an apoptosis inducer and a cellular anchor for the effector peptides in TRAIL-sensitive and TRAIL-resistant cancer cells, respectively. The FADD-dependent pathway, however, seems to be not indispensable in death signal transduction; thus, AD-O51.4 is capable of bypassing the refractoriness of TRAIL. AD-O51.4-driven cell death, which exceeds TRAIL activity, is achieved due to the N-terminally fused polypeptide, containing VEGFA-derived effector peptides. The high anticancer efficiency of AD-O51.4 combined with its safety has led to the entry of AD-O51.4 into toxicological studies.
Collapse
Affiliation(s)
- Piotr Rozga
- Department of Drug Discovery, Adamed Pharma S.A. Pienkow, Czosnow, Poland
| | - Damian Kloska
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Sebastian Pawlak
- Department of Drug Discovery, Adamed Pharma S.A. Pienkow, Czosnow, Poland
| | | | - Marlena Galazka
- Department of Drug Discovery, Adamed Pharma S.A. Pienkow, Czosnow, Poland
| | - Katarzyna Bukato
- Department of Drug Discovery, Adamed Pharma S.A. Pienkow, Czosnow, Poland
| | - Anna Pieczykolan
- Department of Drug Discovery, Adamed Pharma S.A. Pienkow, Czosnow, Poland
| | - Albert Jaworski
- Department of Drug Discovery, Adamed Pharma S.A. Pienkow, Czosnow, Poland
| | | | - Aleksandra Kopacz
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Bogna Badyra
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Neli Kachamakova-Trojanowska
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Olga Zolnierkiewicz
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Marta Targosz-Korecka
- Department of Physics of Nanostructures and Nanotechnology, Institute of Physics, Jagiellonian University, Krakow, Poland
| | - Katarzyna Poleszak
- Department of Drug Discovery, Adamed Pharma S.A. Pienkow, Czosnow, Poland
| | - Michal Szymanik
- Department of Drug Discovery, Adamed Pharma S.A. Pienkow, Czosnow, Poland
| | - Bartlomiej Zerek
- Department of Drug Discovery, Adamed Pharma S.A. Pienkow, Czosnow, Poland
| | - Jerzy Pieczykolan
- Department of Drug Discovery, Adamed Pharma S.A. Pienkow, Czosnow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
14
|
Double-Edged Lipid Nanoparticles Combining Liposome-Bound TRAIL and Encapsulated Doxorubicin Showing an Extraordinary Synergistic Pro-Apoptotic Potential. Cancers (Basel) 2019; 11:cancers11121948. [PMID: 31817469 PMCID: PMC6966652 DOI: 10.3390/cancers11121948] [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: 11/01/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/29/2022] Open
Abstract
Although TRAIL (TNF-related apoptosis-inducing ligand, also known as Apo2L) was described as capable of inducing apoptosis in transformed cells while sparing normal cells, limited results obtained in clinical trials has limited its use as an anti-tumor agent. Consequently, novel TRAIL formulations with enhanced bioactivity are necessary for overcoming resistance to conventional soluble TRAIL (sTRAIL) exhibited by many primary tumors. Our group has generated artificial liposomes with sTRAIL anchored on their surface (large unilamellar vesicle (LUV)-TRAIL), which have shown a greater cytotoxic activity both in vitro and in vivo when compared to sTRAIL against distinct hematologic and epithelial carcinoma cells. In this study, we have improved LUV-TRAIL by loading doxorubicin (DOX) in its liposomal lumen (LUVDOX-TRAIL) in order to improve their cytotoxic potential. LUVDOX-TRAIL killed not only to a higher extent, but also with a much faster kinetic than LUV-TRAIL. In addition, the concerted action of the liposomal DOX and TRAIL was specific of the liposomal DOX and was not observed when with soluble DOX. The cytotoxicity induced by LUVDOX-TRAIL was proven to rely on two processes due to different molecular mechanisms: a dynamin-mediated internalization of the doxorubicin-loaded particle, and the strong activation of caspase-8 exerted by the liposomal TRAIL. Finally, greater cytotoxic activity of LUVDOX-TRAIL was also observed in vivo in a tumor xenograft model. Therefore, we developed a novel double-edged nanoparticle combining the cytotoxic potential of DOX and TRAIL, showing an exceptional and remarkable synergistic effect between both agents.
Collapse
|
15
|
Payandeh Z, Yarahmadi M, Nariman-Saleh-Fam Z, Tarhriz V, Islami M, Aghdam AM, Eyvazi S. Immune therapy of melanoma: Overview of therapeutic vaccines. J Cell Physiol 2019; 234:14612-14621. [PMID: 30706472 DOI: 10.1002/jcp.28181] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Melanoma is the most serious type of skin cancer which develops from the occurrence of genetic mutations in the melanocytes. Based on the features of melanoma tumors such as location, genetic profile and stage, there are several therapeutic strategies including surgery, chemotherapy, and radiotherapy. However, because of the appearance resistance mechanisms, the efficiency of these treatments strategies may be reduced. It has been demonstrated that therapeutic monoclonal antibodies can improve the efficiency of melanoma therapies. Recently, several mAbs, such as nivolumab, pembrolizumab, and ipilimumab, were approved for the immunotherapy of melanoma. The antibodies inhibit immune checkpoint receptors such as CTL4 and pd-1. Another therapeutic strategy for the treatment of melanoma is cancer vaccines, which improve clinical outcomes in patients. The combination therapy using antibodies and gene vaccine give us a new perspective in the treatment of melanoma patients. Herein, we present the recent progressions in the melanoma immunotherapy, especially dendritic cells mRNA vaccines by reviewing recent literature.
Collapse
Affiliation(s)
- Zahra Payandeh
- Immunology Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maral Yarahmadi
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ziba Nariman-Saleh-Fam
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Tarhriz
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Islami
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Shirin Eyvazi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Biotechnology Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Wiesinger M, März J, Kummer M, Schuler G, Dörrie J, Schuler-Thurner B, Schaft N. Clinical-Scale Production of CAR-T Cells for the Treatment of Melanoma Patients by mRNA Transfection of a CSPG4-Specific CAR under Full GMP Compliance. Cancers (Basel) 2019; 11:cancers11081198. [PMID: 31426437 PMCID: PMC6721485 DOI: 10.3390/cancers11081198] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/14/2019] [Accepted: 08/14/2019] [Indexed: 02/06/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cells already showed impressive clinical regressions in leukemia and lymphoma. However, the development of CAR-T cells against solid tumors lags behind. Here we present the clinical-scale production of CAR-T cells for the treatment of melanoma under full GMP compliance. In this approach a CAR, specific for chondroitin sulfate proteoglycan 4 (CSPG4) is intentionally transiently expressed by mRNA electroporation for safety reasons. The clinical-scale protocol was optimized for: (i) expansion of T cells, (ii) electroporation efficiency, (iii) viability, (iv) cryopreservation, and (v) potency. Four consistency runs resulted in CAR-T cells in clinically sufficient numbers, i.e., 2.4 × 109 CAR-expressing T cells, starting from 1.77x108 PBMCs, with an average expansion of 13.6x, an electroporation efficiency of 88.0% CAR-positive cells, a survival of 74.1% after electroporation, and a viability of 84% after cryopreservation. Purity was 98.7% CD3+ cells, with 78.1% CD3+/CD8+ T cells and with minor contaminations of 1.2% NK cells and 0.6% B cells. The resulting CAR-T cells were tested for cytolytic activity after cryopreservation and showed antigen-specific and very efficient lysis of tumor cells. Although our work is descriptive rather than investigative in nature, we expect that providing this clinically applicable protocol to generate sufficient numbers of mRNA-transfected CAR-T cells will help in moving the field of adoptive cell therapy of cancer forward.
Collapse
Affiliation(s)
- Manuel Wiesinger
- Department of Dermatology, Universtitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany
| | - Johannes März
- Department of Dermatology, Universtitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany
| | - Mirko Kummer
- Department of Dermatology, Universtitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany
| | - Gerold Schuler
- Department of Dermatology, Universtitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universtitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany
| | - Beatrice Schuler-Thurner
- Department of Dermatology, Universtitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universtitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany.
| |
Collapse
|
17
|
Role of cell surface proteoglycans in cancer immunotherapy. Semin Cancer Biol 2019; 62:48-67. [PMID: 31336150 DOI: 10.1016/j.semcancer.2019.07.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/05/2019] [Accepted: 07/17/2019] [Indexed: 12/23/2022]
Abstract
Over the past few decades, understanding how tumor cells evade the immune system and their communication with their tumor microenvironment, has been the subject of intense investigation, with the aim of developing new cancer immunotherapies. The current therapies against cancer such as monoclonal antibodies against checkpoint inhibitors, adoptive T-cell transfer, cytokines, vaccines, and oncolytic viruses have managed to improve the clinical outcome of the patients. However, in some tumor entities, the response is limited and could benefit from the identification of novel therapeutic targets. It is known that tumor-extracellular matrix interplay and matrix remodeling are necessary for anti-tumor and pro-tumoral immune responses. Proteoglycans are dominant components of the extracellular matrix and are a highly heterogeneous group of proteins characterized by the covalent attachment of a specific linear carbohydrate chain of the glycosaminoglycan type. At cell surfaces, these molecules modulate the expression and activity of cytokines, chemokines, growth factors, adhesion molecules, and function as signaling co-receptors. By these mechanisms, proteoglycans influence the behavior of cancer cells and their microenvironment during the progression of solid tumors and hematopoietic malignancies. In this review, we discuss why cell surface proteoglycans are attractive pharmacological targets in cancer, and we present current and recent developments in cancer immunology and immunotherapy utilizing proteoglycan-targeted strategies.
Collapse
|
18
|
Molecular Mode of Action of TRAIL Receptor Agonists-Common Principles and Their Translational Exploitation. Cancers (Basel) 2019; 11:cancers11070954. [PMID: 31284696 PMCID: PMC6678900 DOI: 10.3390/cancers11070954] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its death receptors TRAILR1/death receptor 4 (DR4) and TRAILR2/DR5 trigger cell death in many cancer cells but rarely exert cytotoxic activity on non-transformed cells. Against this background, a variety of recombinant TRAIL variants and anti-TRAIL death receptor antibodies have been developed and tested in preclinical and clinical studies. Despite promising results from mice tumor models, TRAIL death receptor targeting has failed so far in clinical studies to show satisfying anti-tumor efficacy. These disappointing results can largely be explained by two issues: First, tumor cells can acquire TRAIL resistance by several mechanisms defining a need for combination therapies with appropriate sensitizing drugs. Second, there is now growing preclinical evidence that soluble TRAIL variants but also bivalent anti-TRAIL death receptor antibodies typically require oligomerization or plasma membrane anchoring to achieve maximum activity. This review discusses the need for oligomerization and plasma membrane attachment for the activity of TRAIL death receptor agonists in view of what is known about the molecular mechanisms of how TRAIL death receptors trigger intracellular cell death signaling. In particular, it will be highlighted which consequences this has for the development of next generation TRAIL death receptor agonists and their potential clinical application.
Collapse
|
19
|
CSPG4-Specific CAR T Cells for High-Risk Childhood B Cell Precursor Leukemia. Int J Mol Sci 2019; 20:ijms20112764. [PMID: 31195686 PMCID: PMC6600602 DOI: 10.3390/ijms20112764] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 02/06/2023] Open
Abstract
The advent of CD19-specific chimeric antigen receptor (CAR) T cells has proven to be a powerful asset in the arsenal of cancer immunotherapy of acute lymphoblastic leukemia and certain B cell lymphomas. However, a sizable portion of patients treated with CD19-CAR T cells relapse with CD19-negative cancer cells, necessitating the quest for back-up antigens. Chondroitin sulfate proteoglycan 4 (CSPG4) expression has been reported on leukemic blasts bearing the ill-fated MLL 11q23 rearrangement. We aimed at exploring the use of CSPG4-specific CAR T cells against mixed-lineage leukemia (MLL)-rearranged leukemic blasts, using the precursor B cell leukemia cell line KOPN8 (MLL–MLLT1 translocation) as a model. First, we confirmed CSPG4 expression on KOPN8 cells. Bulk T cells electroporated with mRNA encoding a CSPG4-specific CAR upregulated activation markers and secreted the Th1 cytokines TNF and IFNγ in an antigen-specific manner upon co-culture with KOPN8 cells. More importantly, CSPG4-specific CAR T cells evinced specific degranulation towards KOPN8 cells and specifically lysed KOPN8 target cells in chromium lysis experiments. CSPG4 is a well-established CAR target in cutaneous melanoma. Here, we provide proof-of-principle data for the use of CSPG4-specific CAR T cells against MLL-translocated leukemias.
Collapse
|
20
|
Aya-Bonilla C, Gray ES, Manikandan J, Freeman JB, Zaenker P, Reid AL, Khattak MA, Frank MH, Millward M, Ziman M. Immunomagnetic-Enriched Subpopulations of Melanoma Circulating Tumour Cells (CTCs) Exhibit Distinct Transcriptome Profiles. Cancers (Basel) 2019; 11:cancers11020157. [PMID: 30769764 PMCID: PMC6406574 DOI: 10.3390/cancers11020157] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 02/06/2023] Open
Abstract
Cutaneous melanoma circulating tumour cells (CTCs) are phenotypically and molecularly heterogeneous. We profiled the gene expression of CTC subpopulations immunomagnetic-captured by targeting either the melanoma-associated marker, MCSP, or the melanoma-initiating marker, ABCB5. Firstly, the expression of a subset of melanoma genes was investigated by RT-PCR in MCSP-enriched and ABCB5-enriched CTCs isolated from a total of 59 blood draws from 39 melanoma cases. Of these, 6 MCSP- and 6 ABCB5-enriched CTC fractions were further analysed using a genome-wide gene expression microarray. The transcriptional programs of both CTC subtypes included cell survival maintenance, cell proliferation, and migration pathways. ABCB5-enriched CTCs were specifically characterised by up-regulation of genes involved in epithelial to mesenchymal transition (EMT), suggesting an invasive phenotype. These findings underscore the presence of at least two distinct melanoma CTC subpopulations with distinct transcriptional programs, which may have distinct roles in disease progression and response to therapy.
Collapse
Affiliation(s)
- Carlos Aya-Bonilla
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia.
| | - Elin S Gray
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia.
| | | | - James B Freeman
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia.
| | - Pauline Zaenker
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia.
| | - Anna L Reid
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia.
| | - Muhammad A Khattak
- School of Medicine, University of Western Australia, Crawley, WA 6009, Australia.
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia.
| | - Markus H Frank
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia.
- Transplantation Research Program, Boston Children's Hospital and Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| | - Michael Millward
- School of Medicine, University of Western Australia, Crawley, WA 6009, Australia.
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia.
| | - Mel Ziman
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia.
- School of Biomedical Science, University of Western Australia, Crawley, WA 6009, Australia.
| |
Collapse
|
21
|
Karamanos NK, Piperigkou Z, Theocharis AD, Watanabe H, Franchi M, Baud S, Brézillon S, Götte M, Passi A, Vigetti D, Ricard-Blum S, Sanderson RD, Neill T, Iozzo RV. Proteoglycan Chemical Diversity Drives Multifunctional Cell Regulation and Therapeutics. Chem Rev 2018; 118:9152-9232. [DOI: 10.1021/acs.chemrev.8b00354] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Aichi 480-1195, Japan
| | - Marco Franchi
- Department for Life Quality Studies, University of Bologna, Rimini 47100, Italy
| | - Stéphanie Baud
- Université de Reims Champagne-Ardenne, Laboratoire SiRMa, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Stéphane Brézillon
- Université de Reims Champagne-Ardenne, Laboratoire de Biochimie Médicale et Biologie Moléculaire, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster 48149, Germany
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Sylvie Ricard-Blum
- University Claude Bernard Lyon 1, CNRS, UMR 5246, Institute of Molecular and Supramolecular Chemistry and Biochemistry, Villeurbanne 69622, France
| | - Ralph D. Sanderson
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| |
Collapse
|
22
|
Marsavela G, Aya-Bonilla CA, Warkiani ME, Gray ES, Ziman M. Melanoma circulating tumor cells: Benefits and challenges required for clinical application. Cancer Lett 2018; 424:1-8. [PMID: 29548820 DOI: 10.1016/j.canlet.2018.03.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/01/2018] [Accepted: 03/09/2018] [Indexed: 02/07/2023]
Abstract
The implementation of novel therapeutic interventions has improved the survival rates of melanoma patients with metastatic disease. Nonetheless, only 33% of treated cases exhibit long term responses. Circulating tumor cell (CTC) measurements are currently of clinical value in breast, prostate and colorectal cancers. However, the clinical utility of melanoma CTCs (MelCTCs) is still unclear due to challenges that appear intrinsic to MelCTCs (i.e. rarity, heterogeneity) and a lack of standardization in their isolation, across research laboratories. Here, we review the latest developments, pinpoint the challenges in MelCTC isolation and address their potential role in melanoma management.
Collapse
Affiliation(s)
- G Marsavela
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - C A Aya-Bonilla
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia.
| | - M E Warkiani
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia; School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia; Institute of Molecular Medicine, Sechenov First Moscow State University, Moscow, Russia
| | - E S Gray
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - M Ziman
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia; School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Australia
| |
Collapse
|
23
|
Ilieva KM, Cheung A, Mele S, Chiaruttini G, Crescioli S, Griffin M, Nakamura M, Spicer JF, Tsoka S, Lacy KE, Tutt ANJ, Karagiannis SN. Chondroitin Sulfate Proteoglycan 4 and Its Potential As an Antibody Immunotherapy Target across Different Tumor Types. Front Immunol 2018; 8:1911. [PMID: 29375561 PMCID: PMC5767725 DOI: 10.3389/fimmu.2017.01911] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 12/14/2017] [Indexed: 12/18/2022] Open
Abstract
Overexpression of the chondroitin sulfate proteoglycan 4 (CSPG4) has been associated with the pathology of multiple types of such as melanoma, breast cancer, squamous cell carcinoma, mesothelioma, neuroblastoma, adult and pediatric sarcomas, and some hematological cancers. CSPG4 has been reported to exhibit a role in the growth and survival as well as in the spreading and metastasis of tumor cells. CSPG4 is overexpressed in several malignant diseases, while it is thought to have restricted and low expression in normal tissues. Thus, CSPG4 has become the target of numerous anticancer treatment approaches, including monoclonal antibody-based therapies. This study reviews key potential anti-CSPG4 antibody and immune-based therapies and examines their direct antiproliferative/metastatic and immune activating mechanisms of action.
Collapse
Affiliation(s)
- Kristina M Ilieva
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Anthony Cheung
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Silvia Mele
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Giulia Chiaruttini
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Silvia Crescioli
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Merope Griffin
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Mano Nakamura
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom.,Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London, United Kingdom
| | - James F Spicer
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London, United Kingdom
| | - Katie E Lacy
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Andrew N J Tutt
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom.,Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| |
Collapse
|
24
|
Ishii K, Matsuoka I, Kajiwara S, Sasaki T, Miki M, Kato M, Kanda H, Arima K, Shiraishi T, Sugimura Y. Additive naftopidil treatment synergizes docetaxel-induced apoptosis in human prostate cancer cells. J Cancer Res Clin Oncol 2017; 144:89-98. [PMID: 29098395 DOI: 10.1007/s00432-017-2536-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 10/24/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE Docetaxel (DTX) is a standard chemotherapeutic drug for castration-resistant prostate cancer (CRPC), although adverse events are common. To overcome this problem, researchers have evaluated the efficacy of DTX treatment in combination with other drugs. Naftopidil is a tubulin-binding drug with fewer adverse events, implying the usefulness of this drug in clinical applications when combined with DTX. Here, we investigated the efficacy of additive naftopidil treatment in combination with DTX on prostate cancer (PCa) cells. METHODS The effects of combination treatment with DTX plus naftopidil were analyzed using two animal models of LNCaP cells plus PrSC xenografts (sub-renal capsule grafting) and PC-3 xenografts (intratibial injection). RESULTS Combination treatment with DTX plus naftopidil significantly inhibited cell growth in LNCaP cells compared with DTX alone. Analysis of the cooperativity index (CI) showed that combination treatment exhibited additive effects on DTX-induced growth inhibition in LNCaP cells. In contrast, combination treatment showed more than an additive (synergistic) effect on DTX-induced apoptosis in LNCaP and PC-3 cells. In LNCaP cells plus PrSC xenografts, combination treatment showed synergistic effects on DTX-induced apoptosis. The synergistic effects of naftopidil on DTX-induced apoptosis were also observed in PC-3 xenografts. CONCLUSIONS Our results demonstrated that additive naftopidil treatment in combination with DTX increased the efficacy of DTX for the treatment of LNCaP and PC-3 tumors in vivo. Thus, additive naftopidil treatment showed a synergistic effect on DTX-induced apoptosis in PCa cells in vitro and in vivo, suggesting that this treatment approach may yield improved clinical benefits compared with DTX alone.
Collapse
Affiliation(s)
- Kenichiro Ishii
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.,Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Izumi Matsuoka
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Shinya Kajiwara
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Takeshi Sasaki
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Manabu Miki
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Manabu Kato
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Hideki Kanda
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Kiminobu Arima
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Taizo Shiraishi
- Department of Diagnostic Pathology, Kuwana City Medical Center, Kuwana, Mie, Japan
| | - Yoshiki Sugimura
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| |
Collapse
|
25
|
Dubuisson A, Micheau O. Antibodies and Derivatives Targeting DR4 and DR5 for Cancer Therapy. Antibodies (Basel) 2017; 6:E16. [PMID: 31548531 PMCID: PMC6698863 DOI: 10.3390/antib6040016] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/16/2017] [Accepted: 10/19/2017] [Indexed: 02/07/2023] Open
Abstract
Developing therapeutics that induce apoptosis in cancer cells has become an increasingly attractive approach for the past 30 years. The discovery of tumor necrosis factor (TNF) superfamily members and more specifically TNF-related apoptosis-inducing ligand (TRAIL), the only cytokine of the family capable of eradicating selectively cancer cells, led to the development of numerous TRAIL derivatives targeting death receptor 4 (DR4) and death receptor 5 (DR5) for cancer therapy. With a few exceptions, preliminary attempts to use recombinant TRAIL, agonistic antibodies, or derivatives to target TRAIL agonist receptors in the clinic have been fairly disappointing. Nonetheless, a tremendous effort, worldwide, is being put into the development of novel strategic options to target TRAIL receptors. Antibodies and derivatives allow for the design of novel and efficient agonists. We summarize and discuss here the advantages and drawbacks of the soar of TRAIL therapeutics, from the first developments to the next generation of agonistic products, with a particular insight on new concepts.
Collapse
Affiliation(s)
- Agathe Dubuisson
- University Bourgogne Franche-Comté, INSERM, LNC UMR1231, F-21079 Dijon, France.
- CovalAb, Research Department, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France.
- INSERM, UMR1231, Laboratoire d'Excellence LipSTIC, F-21079 Dijon, France.
| | - Olivier Micheau
- University Bourgogne Franche-Comté, INSERM, LNC UMR1231, F-21079 Dijon, France.
- CovalAb, Research Department, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France.
- INSERM, UMR1231, Laboratoire d'Excellence LipSTIC, F-21079 Dijon, France.
| |
Collapse
|
26
|
A versatile pretargeting approach for tumour-selective delivery and activation of TNF superfamily members. Sci Rep 2017; 7:13301. [PMID: 29038485 PMCID: PMC5643434 DOI: 10.1038/s41598-017-13530-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/25/2017] [Indexed: 11/08/2022] Open
Abstract
TNFR superfamily (TNFRSF) members have important immunoregulatory functions and are of clear interest for cancer immunotherapy. Various TNFRSF agonists have been clinically evaluated, but have met with limited efficacy and/or toxicity. Recent insights indicate that 'first-generation' TNFRSF agonists lack efficacy as they do not effectively cross-link their corresponding receptor. Reversely, ubiquitous TNFRSF receptor(s) cross-linking by CD40 and Fas agonistic antibodies resulted in dose-limiting liver toxicity. To overcome these issues, we developed a novel pretargeting strategy exploiting recombinant fusion proteins in which a soluble form of TRAIL, FasL or CD40L is genetically fused to a high-affinity anti-fluorescein scFv antibody fragment (scFvFITC). Fusion proteins scFvFITC:sTRAIL and scFvFITC:sFasL induced potent target antigen-restricted apoptosis in a panel of cancer lines and in primary patient-derived cancer cells, but only when pretargeted with a relevant FITC-labelled antitumour antibody. In a similar pretargeting setting, fusion protein scFvFITC:sCD40L promoted tumour-directed maturation of immature monocyte-derived dendritic cells (iDCs). This novel tumour-selective pretargeting approach may be used to improve efficacy and/or reduce possible off-target toxicity of TNFSF ligands for cancer immunotherapy.
Collapse
|
27
|
Gamie Z, Kapriniotis K, Papanikolaou D, Haagensen E, Da Conceicao Ribeiro R, Dalgarno K, Krippner-Heidenreich A, Gerrand C, Tsiridis E, Rankin KS. TNF-related apoptosis-inducing ligand (TRAIL) for bone sarcoma treatment: Pre-clinical and clinical data. Cancer Lett 2017; 409:66-80. [PMID: 28888998 DOI: 10.1016/j.canlet.2017.08.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 01/25/2023]
Abstract
Bone sarcomas are rare, highly malignant mesenchymal tumours that affect teenagers and young adults, as well as older patients. Despite intensive, multimodal therapy, patients with bone sarcomas have poor 5-year survival, close to 50%, with lack of improvement over recent decades. TNF-related apoptosis-inducing ligand (TRAIL), a member of the tumour necrosis factor (TNF) ligand superfamily (TNFLSF), has been found to induce apoptosis in cancer cells while sparing nontransformed cells, and may therefore offer a promising new approach to treatment. We cover the existing preclinical and clinical evidence about the use of TRAIL and other death receptor agonists in bone sarcoma treatment. In vitro studies indicate that TRAIL and other death receptor agonists are generally potent against bone sarcoma cell lines. Ewing's sarcoma cell lines present the highest sensitivity, whereas osteosarcoma and chondrosarcoma cell lines are considered less sensitive. In vivo studies also demonstrate satisfactory results, especially in Ewing's sarcoma xenograft models. However, the few clinical trials in the literature show only low or moderate efficacy of TRAIL in treating bone sarcoma. Potential strategies to overcome the in vivo resistance reported include co-administration with other drugs and the potential to deliver TRAIL on the surface of primed mesenchymal or immune cells and the use of targeted single chain antibodies such as scFv-scTRAIL.
Collapse
Affiliation(s)
- Zakareya Gamie
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Konstantinos Kapriniotis
- Academic Orthopedic Department, "PapaGeorgiou" General Hospital, Thessaloniki, Greece; CORE-Center for Orthopedic Research at CIRI-AUTh, Aristotle University Medical School, Thessaloniki, Hellas, Greece.
| | - Dimitra Papanikolaou
- Academic Orthopedic Department, "PapaGeorgiou" General Hospital, Thessaloniki, Greece; CORE-Center for Orthopedic Research at CIRI-AUTh, Aristotle University Medical School, Thessaloniki, Hellas, Greece.
| | - Emma Haagensen
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Ricardo Da Conceicao Ribeiro
- School of Mechanical and Systems Engineering, Stephenson Building, Claremont Road, Newcastle Upon Tyne, NE1 7RU, UK.
| | - Kenneth Dalgarno
- School of Mechanical and Systems Engineering, Stephenson Building, Claremont Road, Newcastle Upon Tyne, NE1 7RU, UK.
| | - Anja Krippner-Heidenreich
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Craig Gerrand
- North of England Bone and Soft Tissue Tumour Service, Freeman Hospital, Freeman Road, Newcastle Upon Tyne, NE7 7DN, UK.
| | - Eleftherios Tsiridis
- Academic Orthopedic Department, "PapaGeorgiou" General Hospital, Thessaloniki, Greece; CORE-Center for Orthopedic Research at CIRI-AUTh, Aristotle University Medical School, Thessaloniki, Hellas, Greece; Secretary General European Hip Society, Austria.
| | - Kenneth Samora Rankin
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| |
Collapse
|
28
|
Rolih V, Barutello G, Iussich S, De Maria R, Quaglino E, Buracco P, Cavallo F, Riccardo F. CSPG4: a prototype oncoantigen for translational immunotherapy studies. J Transl Med 2017; 15:151. [PMID: 28668095 PMCID: PMC5494135 DOI: 10.1186/s12967-017-1250-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/21/2017] [Indexed: 12/21/2022] Open
Abstract
Thanks to striking progress in both the understanding of anti-tumor immune response and the characterization of several tumor associated antigens (TAA), a more rational design and more sophisticated strategies for anti-tumor vaccination have been possible. However, the effectiveness of cancer vaccines in clinical trial is still partial, indicating that additional studies are needed to optimize their design and their pre-clinical testing. Indeed, anti-tumor vaccination success relies on the choice of the best TAA to be targeted and on the translational power of the pre-clinical model used to assess its efficacy. The chondroitin sulfate proteoglycan-4 (CSPG4) is a cell surface proteoglycan overexpressed in a huge range of human and canine neoplastic lesions by tumor cells, tumor microenvironment and cancer initiating cells. CSPG4 plays a central role in the oncogenic pathways required for malignant progression and metastatization. Thanks to these features and to its poor expression in adult healthy tissues, CSPG4 represents an ideal oncoantigen and thus an attractive target for anti-tumor immunotherapy. In this review we explore the potential of CSPG4 immune-targeting. Moreover, since it has been clearly demonstrated that spontaneous canine tumors mimic the progression of human malignancies better than any other pre-clinical model available so far, we reported also our results indicating that CSPG4 DNA vaccination is safe and effective in significantly increasing the survival of canine melanoma patients. Therefore, anti-CSPG4 vaccination strategy could have a substantial impact for the treatment of the wider population of spontaneous CSPG4-positive tumor affected dogs with a priceless translational value and a revolutionary implication for human oncological patients.
Collapse
Affiliation(s)
- Valeria Rolih
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| | - Selina Iussich
- Department of Veterinary Sciences, University of Torino, 10095 Grugliasco, Italy
| | - Raffaella De Maria
- Department of Veterinary Sciences, University of Torino, 10095 Grugliasco, Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Torino, 10095 Grugliasco, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy
| |
Collapse
|
29
|
CSPG4: A Target for Selective Delivery of Human Cytolytic Fusion Proteins and TRAIL. Biomedicines 2017; 5:biomedicines5030037. [PMID: 28657611 PMCID: PMC5618295 DOI: 10.3390/biomedicines5030037] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/06/2017] [Accepted: 06/09/2017] [Indexed: 01/08/2023] Open
Abstract
Chondroitin-sulfate proteoglycan 4 (CSPG4) is a transmembrane glycoprotein overexpressed on malignant cells in several cancer types with only limited expression on normal cells. CSPG4 is implicated in several signaling pathways believed to drive cancer progression, particularly proliferation, motility and metastatic spread. Expression may serve as a prognostic marker for survival and risk of relapse in treatment-resistant malignancies including melanoma, triple negative breast cancer, rhabdomyosarcoma and acute lymphoblastic leukemia. This tumor-associated overexpression of CSPG4 points towards a highly promising therapeutic target for antibody-guided cancer therapy. Monoclonal αCSPG4 antibodies have been shown to inhibit cancer progression by blocking ligand access to the CSPG4 extracellular binding sites. Moreover, CSPG4-directed antibody conjugates have been shown to be selectively internalized by CSPG4-expressing cancer cells via endocytosis. CSPG4-directed immunotherapy may be approached in several ways, including: (1) antibody-based fusion proteins for the selective delivery of a pro-apoptotic factors such as tumor necrosis factor-related apoptosis-inducing ligand to agonistic death receptors 4 and 5 on the cell surface; and (2) CSPG4-specific immunotoxins which bind selectively to diseased cells expressing CSPG4, are internalized by them and induce arrest of biosynthesis, closely followed by initiation of apoptotic signaling. Here we review various methods of exploiting tumor-associated CSPG4 expression to improve targeted cancer therapy.
Collapse
|
30
|
Hendriks D, Choi G, de Bruyn M, Wiersma VR, Bremer E. Antibody-Based Cancer Therapy: Successful Agents and Novel Approaches. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 331:289-383. [PMID: 28325214 DOI: 10.1016/bs.ircmb.2016.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since their discovery, antibodies have been viewed as ideal candidates or "magic bullets" for use in targeted therapy in the fields of cancer, autoimmunity, and chronic inflammatory disorders. A wave of antibody-dedicated research followed, which resulted in the clinical approval of a first generation of monoclonal antibodies for cancer therapy such as rituximab (1997) and cetuximab (2004), and infliximab (2002) for the treatment of autoimmune diseases. More recently, the development of antibodies that prevent checkpoint-mediated inhibition of T cell responses invigorated the field of cancer immunotherapy. Such antibodies induced unprecedented long-term remissions in patients with advanced stage malignancies, most notably melanoma and lung cancer, that do not respond to conventional therapies. In this review, we will recapitulate the development of antibody-based therapy, and detail recent advances and new functions, particularly in the field of cancer immunotherapy. With the advent of recombinant DNA engineering, a number of rationally designed molecular formats of antibodies and antibody-derived agents have become available, and we will discuss various molecular formats including antibodies with improved effector functions, bispecific antibodies, antibody-drug conjugates, antibody-cytokine fusion proteins, and T cells genetically modified with chimeric antigen receptors. With these exciting advances, new antibody-based treatment options will likely enter clinical practice and pave the way toward more successful control of malignant diseases.
Collapse
Affiliation(s)
- D Hendriks
- Department of Surgery, Translational Surgical Oncology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - G Choi
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - M de Bruyn
- Department of Obstetrics & Gynecology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - V R Wiersma
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.
| | - E Bremer
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands; University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
31
|
Abstract
Sigma1 (also known as sigma-1 receptor, Sig1R, σ1 receptor) is a unique pharmacologically regulated integral membrane chaperone or scaffolding protein. The majority of publications on the subject have focused on the neuropharmacology of Sigma1. However, a number of publications have also suggested a role for Sigma1 in cancer. Although there is currently no clinically used anti-cancer drug that targets Sigma1, a growing body of evidence supports the potential of Sigma1 ligands as therapeutic agents to treat cancer. In preclinical models, compounds with affinity for Sigma1 have been reported to inhibit cancer cell proliferation and survival, cell adhesion and migration, tumor growth, to alleviate cancer-associated pain, and to have immunomodulatory properties. This review will highlight that although the literature supports a role for Sigma1 in cancer, several fundamental questions regarding drug mechanism of action and the physiological relevance of aberrant SIGMAR1 transcript and Sigma1 protein expression in certain cancers remain unanswered or only partially answered. However, emerging lines of evidence suggest that Sigma1 is a component of the cancer cell support machinery, that it facilitates protein interaction networks, that it allosterically modulates the activity of its associated proteins, and that Sigma1 is a selectively multifunctional drug target.
Collapse
Affiliation(s)
- Felix J Kim
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Philadelphia, PA, USA.
| | - Christina M Maher
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, USA
| |
Collapse
|
32
|
Hendriks D, He Y, Koopmans I, Wiersma VR, van Ginkel RJ, Samplonius DF, Helfrich W, Bremer E. Programmed Death Ligand 1 (PD-L1)-targeted TRAIL combines PD-L1-mediated checkpoint inhibition with TRAIL-mediated apoptosis induction. Oncoimmunology 2016; 5:e1202390. [PMID: 27622071 PMCID: PMC5007955 DOI: 10.1080/2162402x.2016.1202390] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/07/2016] [Accepted: 06/12/2016] [Indexed: 01/18/2023] Open
Abstract
Antibodies that block PD-L1/PD-1 immune checkpoints restore the activity of functionally-impaired antitumor T cells. These antibodies show unprecedented clinical benefit in various advanced cancers, particularly in melanoma. However, only a subset of cancer patients responds to current PD-L1/PD-1-blocking strategies, highlighting the need for further advancements in PD-L1/PD-1-based immunotherapy. Here, we report on a novel approach designed to combine PD-L1 checkpoint inhibition with the tumor-selective induction of apoptosis by TNF-related Apoptosis Inducing Ligand (TRAIL). In brief, a new bi-functional fusion protein, designated anti-PD-L1:TRAIL, was constructed comprising a PD-L1-blocking antibody fragment genetically fused to the extracellular domain of the pro-apoptotic tumoricidal protein TRAIL. Treatment of PD-L1-expressing cancer cells with anti-PD-L1:TRAIL induced PD-L1-directed TRAIL-mediated cancer cell death. Treatment of T cells with anti-PD-L1:TRAIL augmented T cell activation, as evidenced by increased proliferation, secretion of IFNγ and enhanced killing of cancer cell lines and primary patient-derived cancer cells in mixed T cell/cancer cell culture experiments. Of note, elevated levels of IFNγ further upregulated PD-L1 on cancer cells and simultaneously sensitized cancer cells to TRAIL-mediated apoptosis by anti-PD-L1:TRAIL. Additionally, anti-PD-L1:TRAIL converted immunosuppressive PD-L1-expressing myeloid cells into pro-apoptotic effector cells that triggered TRAIL-mediated cancer cell death. In conclusion, combining PD-L1 checkpoint inhibition with TRAIL-mediated induction of apoptosis using anti-PD-L1:TRAIL yields promising multi-fold and mutually reinforcing anticancer activity that may be exploited to enhance the efficacy of therapeutic PD-L1/PD-1 checkpoint inhibition.
Collapse
Affiliation(s)
- Djoke Hendriks
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Yuan He
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Iris Koopmans
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Valerie R Wiersma
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Robert J van Ginkel
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Douwe F Samplonius
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Wijnand Helfrich
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Edwin Bremer
- University of Exeter Medical School, St Luke's Campus, Exeter, Devon, UK; University of Groningen, University Medical Center Groningen, Department of Experimental Hematology, Section Immunohematology, Cancer Research Center Groningen (CRCG), Groningen, the Netherlands
| |
Collapse
|
33
|
de Miguel D, Lemke J, Anel A, Walczak H, Martinez-Lostao L. Onto better TRAILs for cancer treatment. Cell Death Differ 2016; 23:733-47. [PMID: 26943322 PMCID: PMC4832109 DOI: 10.1038/cdd.2015.174] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/11/2015] [Accepted: 12/17/2015] [Indexed: 01/01/2023] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), also known as Apo-2 ligand (Apo2L), is a member of the TNF cytokine superfamily. By cross-linking TRAIL-Receptor (TRAIL-R) 1 or TRAIL-R2, also known as death receptors 4 and 5 (DR4 and DR5), TRAIL has the capability to induce apoptosis in a wide variety of tumor cells while sparing vital normal cells. The discovery of this unique property among TNF superfamily members laid the foundation for testing the clinical potential of TRAIL-R-targeting therapies in the cancer clinic. To date, two of these therapeutic strategies have been tested clinically: (i) recombinant human TRAIL and (ii) antibodies directed against TRAIL-R1 or TRAIL-R2. Unfortunately, however, these TRAIL-R agonists have basically failed as most human tumors are resistant to apoptosis induction by them. It recently emerged that this is largely due to the poor agonistic activity of these agents. Consequently, novel TRAIL-R-targeting agents with increased bioactivity are currently being developed with the aim of rendering TRAIL-based therapies more active. This review summarizes these second-generation novel formulations of TRAIL and other TRAIL-R agonists, which exhibit enhanced cytotoxic capacity toward cancer cells, thereby providing the potential of being more effective when applied clinically than first-generation TRAIL-R agonists.
Collapse
Affiliation(s)
- D de Miguel
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
| | - J Lemke
- UCL Cancer Institute, Faculty of Medical Sciences, University College London, London, UK
| | - A Anel
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
| | - H Walczak
- UCL Cancer Institute, Faculty of Medical Sciences, University College London, London, UK
| | - L Martinez-Lostao
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
- Instituto de Nanociencia de Aragón, Zaragoza, Spain
| |
Collapse
|
34
|
Amoury M, Mladenov R, Nachreiner T, Pham AT, Hristodorov D, Di Fiore S, Helfrich W, Pardo A, Fey G, Schwenkert M, Thepen T, Kiessling F, Hussain AF, Fischer R, Kolberg K, Barth S. A novel approach for targeted elimination of CSPG4-positive triple-negative breast cancer cells using a MAP tau-based fusion protein. Int J Cancer 2016; 139:916-27. [DOI: 10.1002/ijc.30119] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 02/24/2016] [Accepted: 03/03/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Manal Amoury
- Department of Experimental Medicine and Immunotherapy; Institute for Applied Medical Engineering, University Hospital RWTH Aachen; Germany
| | - Radoslav Mladenov
- Department of Experimental Medicine and Immunotherapy; Institute for Applied Medical Engineering, University Hospital RWTH Aachen; Germany
| | - Thomas Nachreiner
- Department of Experimental Medicine and Immunotherapy; Institute for Applied Medical Engineering, University Hospital RWTH Aachen; Germany
| | - Anh-Tuan Pham
- Department of Experimental Medicine and Immunotherapy; Institute for Applied Medical Engineering, University Hospital RWTH Aachen; Germany
| | - Dmitrij Hristodorov
- Department of Experimental Medicine and Immunotherapy; Institute for Applied Medical Engineering, University Hospital RWTH Aachen; Germany
| | - Stefano Di Fiore
- Department of Pharmaceutical Product Development; Fraunhofer Institute for Molecular Biology and Applied Ecology; Aachen Germany
| | - Wijnand Helfrich
- Department of Surgery, Laboratory for Translational Surgical Oncology; University of Groningen, University Medical Center Groningen; The Netherlands
| | - Alessa Pardo
- Department of Experimental Medicine and Immunotherapy; Institute for Applied Medical Engineering, University Hospital RWTH Aachen; Germany
| | - Georg Fey
- Department of Biology; Friedrich Alexander University Erlangen-Nuremberg; Germany
| | | | - Theophilus Thepen
- Department of Pharmaceutical Product Development; Fraunhofer Institute for Molecular Biology and Applied Ecology; Aachen Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen; Germany
| | - Ahmad F. Hussain
- Department of Gynecology and Obstetrics; University Hospital RWTH Aachen; Germany
| | - Rainer Fischer
- Department of Pharmaceutical Product Development; Fraunhofer Institute for Molecular Biology and Applied Ecology; Aachen Germany
| | - Katharina Kolberg
- Department of Experimental Medicine and Immunotherapy; Institute for Applied Medical Engineering, University Hospital RWTH Aachen; Germany
- Department of Pharmaceutical Product Development; Fraunhofer Institute for Molecular Biology and Applied Ecology; Aachen Germany
| | - Stefan Barth
- South African Research Chair in Cancer Biotechnology, Institute of Infectious Disease and Molecular Medicine (IDM), Department of Integrative Biomedical Sciences, Faculty of Health Sciences; University of Cape Town; Observatory South Africa
| |
Collapse
|
35
|
Brünker P, Wartha K, Friess T, Grau-Richards S, Waldhauer I, Koller CF, Weiser B, Majety M, Runza V, Niu H, Packman K, Feng N, Daouti S, Hosse RJ, Mössner E, Weber TG, Herting F, Scheuer W, Sade H, Shao C, Liu B, Wang P, Xu G, Vega-Harring S, Klein C, Bosslet K, Umaña P. RG7386, a Novel Tetravalent FAP-DR5 Antibody, Effectively Triggers FAP-Dependent, Avidity-Driven DR5 Hyperclustering and Tumor Cell Apoptosis. Mol Cancer Ther 2016; 15:946-57. [PMID: 27037412 DOI: 10.1158/1535-7163.mct-15-0647] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 01/18/2016] [Indexed: 11/16/2022]
Abstract
Dysregulated cellular apoptosis and resistance to cell death are hallmarks of neoplastic initiation and disease progression. Therefore, the development of agents that overcome apoptosis dysregulation in tumor cells is an attractive therapeutic approach. Activation of the extrinsic apoptotic pathway is strongly dependent on death receptor (DR) hyperclustering on the cell surface. However, strategies to activate DR5 or DR4 through agonistic antibodies have had only limited clinical success. To pursue an alternative approach for tumor-targeted induction of apoptosis, we engineered a bispecific antibody (BsAb), which simultaneously targets fibroblast-activation protein (FAP) on cancer-associated fibroblasts in tumor stroma and DR5 on tumor cells. We hypothesized that bivalent binding to both FAP and DR5 leads to avidity-driven hyperclustering of DR5 and subsequently strong induction of apoptosis in tumor cells but not in normal cells. Here, we show that RG7386, an optimized FAP-DR5 BsAb, triggers potent tumor cell apoptosis in vitro and in vivo in preclinical tumor models with FAP-positive stroma. RG7386 antitumor efficacy was strictly FAP dependent, was independent of FcR cross-linking, and was superior to conventional DR5 antibodies. In combination with irinotecan or doxorubicin, FAP-DR5 treatment resulted in substantial tumor regression in patient-derived xenograft models. FAP-DR5 also demonstrated single-agent activity against FAP-expressing malignant cells, due to cross-binding of FAP and DR5 across tumor cells. Taken together, these data demonstrate that RG7386, a novel and potent antitumor agent in both mono- and combination therapies, overcomes limitations of previous DR5 antibodies and represents a promising approach to conquer tumor-associated resistance to apoptosis. Mol Cancer Ther; 15(5); 946-57. ©2016 AACR.
Collapse
Affiliation(s)
- Peter Brünker
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Katharina Wartha
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Thomas Friess
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Sandra Grau-Richards
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Inja Waldhauer
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Claudia Ferrara Koller
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Barbara Weiser
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Meher Majety
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Valeria Runza
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Huifeng Niu
- Roche Pharma Research and Early Development, Roche Innovation Center New York, New York, New York
| | - Kathryn Packman
- Roche Pharma Research and Early Development, Roche Innovation Center New York, New York, New York
| | - Ningping Feng
- Roche Pharma Research and Early Development, Roche Innovation Center New York, New York, New York
| | - Sherif Daouti
- Roche Pharma Research and Early Development, Roche Innovation Center New York, New York, New York
| | - Ralf J Hosse
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Ekkehard Mössner
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Thomas G Weber
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Frank Herting
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Werner Scheuer
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Hadassah Sade
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Cuiying Shao
- Roche Pharma Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Bin Liu
- Roche Pharma Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Peng Wang
- Roche Pharma Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Gary Xu
- Roche Pharma Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Suzana Vega-Harring
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Klaus Bosslet
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Pablo Umaña
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland.
| |
Collapse
|
36
|
van der Steen SC, van Tilborg AA, Vallen MJ, Bulten J, van Kuppevelt TH, Massuger LF. Prognostic significance of highly sulfated chondroitin sulfates in ovarian cancer defined by the single chain antibody GD3A11. Gynecol Oncol 2016; 140:527-36. [DOI: 10.1016/j.ygyno.2015.12.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/14/2015] [Accepted: 12/23/2015] [Indexed: 10/22/2022]
|
37
|
Melanoma-Directed Activation of Apoptosis Using a Bispecific Antibody Directed at MCSP and TRAIL Receptor-2/Death Receptor-5. J Invest Dermatol 2015; 136:541-544. [PMID: 26967487 DOI: 10.1016/j.jid.2015.11.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
38
|
Krug C, Birkholz K, Paulus A, Schwenkert M, Schmidt P, Hoffmann N, Hombach A, Fey G, Abken H, Schuler G, Schuler-Thurner B, Dörrie J, Schaft N. Stability and activity of MCSP-specific chimeric antigen receptors (CARs) depend on the scFv antigen-binding domain and the protein backbone. Cancer Immunol Immunother 2015; 64:1623-35. [PMID: 26515978 PMCID: PMC11028909 DOI: 10.1007/s00262-015-1767-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 10/16/2015] [Indexed: 12/26/2022]
Abstract
Chimeric antigen receptor (CAR)-modified T cells emerged as effective tools in the immunotherapy of cancer but can produce severe on-target off-tissue toxicities. This risk can conceivably be overcome, at least partially, by transient transfection. The design of CARs, however, has so far not been optimized for use in non-permanent T cell modification. Here we compared the performance of T cells modified with three different first- and second-generation CARs, each specific for MCSP (HMW-MAA) which is commonly expressed by melanoma cells. Upon RNA transfer, the expression of all receptors was limited in time. The second-generation CARs, which combined CD28-CD3ζ signaling, were expressed at higher levels and more prolonged than first-generation CARs with CD3ζ only. The CD28 domain increased the cytokine production, but had only an indirect effect on the lytic capacity, by prolonging the CAR expression. Especially for the second-generation CARs, the scFv clearly impacted the level and duration of CAR expression and the T cell performance. Thus, we identified a CAR high in both expression and anti-tumor cell reactivity. T cells transfected with this CAR increased the mean survival time of mice after challenge with melanoma cells. To facilitate clinical application, this CAR was used to redirect T cells from late-stage melanoma patients by RNA transfection. These T cells mediated effective antigen-specific tumor cell lysis and release of pro-inflammatory cytokines, even after cryoconservation of the transfected T cells. Taken together, the analysis identified a CAR with superior anti-melanoma performance after RNA transfer which is a promising candidate for clinical exploration.
Collapse
MESH Headings
- Animals
- Carrier Proteins/immunology
- Cell Line, Tumor
- Disease Models, Animal
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Melanoma/immunology
- Melanoma/physiopathology
- Mice
- Mitochondrial Proteins/genetics
- Mitochondrial Proteins/immunology
- Protein Stability
- Protein Structure, Tertiary
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Single-Chain Antibodies/metabolism
Collapse
Affiliation(s)
- Christian Krug
- Department of Dermatology, Universitätsklinikum Erlangen, Hartmannstraße 14, 91052, Erlangen, Germany
- Department of Biology, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Katrin Birkholz
- Department of Dermatology, Universitätsklinikum Erlangen, Hartmannstraße 14, 91052, Erlangen, Germany
| | - Alexander Paulus
- Department of Dermatology, Universitätsklinikum Erlangen, Hartmannstraße 14, 91052, Erlangen, Germany
| | - Michael Schwenkert
- Department of Biology, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Patrick Schmidt
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Department I Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Nicole Hoffmann
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Department I Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Andreas Hombach
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Department I Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Georg Fey
- Department of Biology, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Hinrich Abken
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Department I Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Gerold Schuler
- Department of Dermatology, Universitätsklinikum Erlangen, Hartmannstraße 14, 91052, Erlangen, Germany
| | - Beatrice Schuler-Thurner
- Department of Dermatology, Universitätsklinikum Erlangen, Hartmannstraße 14, 91052, Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen, Hartmannstraße 14, 91052, Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen, Hartmannstraße 14, 91052, Erlangen, Germany.
| |
Collapse
|
39
|
Wiersma VR, de Bruyn M, Shi C, Gooden MJM, Wouters MCA, Samplonius DF, Hendriks D, Nijman HW, Wei Y, Zhou J, Helfrich W, Bremer E. C-type lectin-like molecule-1 (CLL1)-targeted TRAIL augments the tumoricidal activity of granulocytes and potentiates therapeutic antibody-dependent cell-mediated cytotoxicity. MAbs 2015; 7:321-30. [PMID: 25760768 DOI: 10.1080/19420862.2015.1007811] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
The therapeutic effect of anti-cancer monoclonal antibodies stems from their capacity to opsonize targeted cancer cells with subsequent phagocytic removal, induction of antibody-dependent cell-mediated cytotoxicity (ADCC) or induction of complement-mediated cytotoxicity (CDC). The major immune effector cells involved in these processes are natural killer (NK) cells and granulocytes. The latter and most prevalent blood cell population contributes to phagocytosis, but is not effective in inducing ADCC. Here, we report that targeted delivery of the tumoricidal protein tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to granulocyte marker C-type lectin-like molecule-1 (CLL1), using fusion protein CLL1:TRAIL, equips granulocytes with high levels of TRAIL. Upon CLL1-selective binding of this fusion protein, granulocytes acquire additional TRAIL-mediated cytotoxic activity that, importantly, potentiates antibody-mediated cytotoxicity of clinically used therapeutic antibodies (e.g., rituximab, cetuximab). Thus, CLL1:TRAIL could be used as an adjuvant to optimize the clinical potential of anticancer antibody therapy by augmenting tumoricidal activity of granulocytes.
Collapse
Affiliation(s)
- Valerie R Wiersma
- a University of Groningen; University Medical Center Groningen (UMCG) ; Department of Surgery; Translational Surgical Oncology ; Groningen , The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
van Waarde A, Rybczynska AA, Ramakrishnan NK, Ishiwata K, Elsinga PH, Dierckx RAJO. Potential applications for sigma receptor ligands in cancer diagnosis and therapy. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1848:2703-14. [PMID: 25173780 DOI: 10.1016/j.bbamem.2014.08.022] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/04/2014] [Accepted: 08/19/2014] [Indexed: 01/03/2023]
Abstract
Sigma receptors (sigma-1 and sigma-2) represent two independent classes of proteins. Their endogenous ligands may include the hallucinogen N,N-dimethyltryptamine (DMT) and sphingolipid-derived amines which interact with sigma-1 receptors, besides steroid hormones (e.g., progesterone) which bind to both sigma receptor subpopulations. The sigma-1 receptor is a ligand-regulated molecular chaperone with various ion channels and G-protein-coupled membrane receptors as clients. The sigma-2 receptor was identified as the progesterone receptor membrane component 1 (PGRMC1). Although sigma receptors are over-expressed in tumors and up-regulated in rapidly dividing normal tissue, their ligands induce significant cell death only in tumor tissue. Sigma ligands may therefore be used to selectively eradicate tumors. Multiple mechanisms appear to underlie cell killing after administration of sigma ligands, and the signaling pathways are dependent both on the type of ligand and the type of tumor cell. Recent evidence suggests that the sigma-2 receptor is a potential tumor and serum biomarker for human lung cancer and an important target for inhibiting tumor invasion and cancer progression. Current radiochemical efforts are focused on the development of subtype-selective radioligands for positron emission tomography (PET) imaging. Right now, the mostpromising tracers are [18F]fluspidine and [18F]FTC-146 for sigma-1 receptors and [11C]RHM-1 and [18F]ISO-1 for the sigma-2 subtype. Nanoparticles coupled to sigma ligands have shown considerable potential for targeted delivery of antitumor drugs in animal models of cancer, but clinical studies exploring this strategy in cancer patients have not yet been reported. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Aren van Waarde
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Anna A Rybczynska
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Nisha K Ramakrishnan
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Kiichi Ishiwata
- Tokyo Metropolitan Institute of Gerontology, Research Team for Neuroimaging, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo 173-0015, Japan
| | - Philip H Elsinga
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; University of Ghent, University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| |
Collapse
|
41
|
de Bruyn M, Wiersma VR, Helfrich W, Eggleton P, Bremer E. The ever-expanding immunomodulatory role of calreticulin in cancer immunity. Front Oncol 2015; 5:35. [PMID: 25750898 PMCID: PMC4335099 DOI: 10.3389/fonc.2015.00035] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 01/31/2015] [Indexed: 01/03/2023] Open
Abstract
Calreticulin is a pleiotropic molecule that normally resides in the lumen of the endoplasmic reticulum (ER). Here, it has various functions, ranging from regulation of calcium homeostasis to ensuring proper protein folding. More recently, calreticulin gained special interest for its extracellular functions, where it has direct immunomodulatory activity. In this respect, calreticulin activates dendritic cells and macrophages. In addition, certain anti-cancer therapies induce the translocation of calreticulin from the ER to the cell surface of dying cancer cells, where calreticulin dictates the immunogenicity of these cells. Interestingly, treatment with tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) also induces membrane calreticulin exposure on cancer cells. As shown here, calreticulin directly interacts with TRAIL and its receptor-signaling complex, as well as with other TNF family members. Of note, TRAIL is a well known immunomodulatory molecule, and is expressed on the surface of natural killer T-cells. Therefore, calreticulin may have an as yet unrecognized wide(r) impact on immunity, with the TNF-ligand family modulating virtually all aspects of the immune response.
Collapse
Affiliation(s)
- Marco de Bruyn
- Department of Gynecologic Oncology, University Medical Center Groningen (UMCG), University of Groningen , Groningen , Netherlands
| | - Valerie R Wiersma
- Department of Surgery, Translational Surgical Oncology, University Medical Center Groningen (UMCG), University of Groningen , Groningen , Netherlands
| | - Wijnand Helfrich
- Department of Surgery, Translational Surgical Oncology, University Medical Center Groningen (UMCG), University of Groningen , Groningen , Netherlands
| | | | - Edwin Bremer
- Department of Surgery, Translational Surgical Oncology, University Medical Center Groningen (UMCG), University of Groningen , Groningen , Netherlands ; University of Exeter Medical School , Exeter , UK
| |
Collapse
|
42
|
Nicolosi PA, Dallatomasina A, Perris R. Theranostic impact of NG2/CSPG4 proteoglycan in cancer. Theranostics 2015; 5:530-44. [PMID: 25767619 PMCID: PMC4350014 DOI: 10.7150/thno.10824] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 12/03/2014] [Indexed: 12/27/2022] Open
Abstract
NG2/CSPG4 is an unusual cell-membrane integral proteoglycan widely recognized to be a prognostic factor, a valuable tool for ex vivo and non-invasive molecular diagnostics and, by virtue of its tight association with malignancy, a tantalizing therapeutic target in several tumour types. Although the biology behind its involvement in cancer progression needs to be better understood, implementation of NG2/CSPG4 in the routine clinical practice is attainable and has the potential to contribute to an improved individualized management of cancer patients. In this context, its polymorphic nature seems to be particularly valuable in the effort to standardize informative diagnostic procedures and consolidate forcible immunotherapeutic treatment strategies. We discuss here the underpinnings for this potential and highlight the benefits of taking advantage of the intra-tumour and inter-patient variability in the regulation of NG2/CSPG4 expression. We envision that NG2/CSPG4 may effectively be exploited in therapeutic interventions aimed at averting resistance to target therapy agents and at interfering with secondary lesion formation and/or tumour recurrence.
Collapse
|
43
|
Vallen MJ, van der Steen SC, van Tilborg AA, Massuger LF, van Kuppevelt TH. Sulfated sugars in the extracellular matrix orchestrate ovarian cancer development: ‘When sweet turns sour’. Gynecol Oncol 2014; 135:371-81. [DOI: 10.1016/j.ygyno.2014.08.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 08/15/2014] [Accepted: 08/16/2014] [Indexed: 01/14/2023]
|
44
|
Yamaguchi M, Nishii Y, Nakamura K, Aoki H, Hirai S, Uchida H, Sakuma Y, Hamada H. Development of a sensitive screening method for selecting monoclonal antibodies to be internalized by cells. Biochem Biophys Res Commun 2014; 454:600-3. [PMID: 25450699 DOI: 10.1016/j.bbrc.2014.10.133] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 10/28/2014] [Indexed: 11/16/2022]
Abstract
Antibody-drug conjugates (ADCs), drugs developed by conjugation of an anticancer agent to a monoclonal antibody (mAb), have lately attracted attention in cancer therapy because ADCs can directly bind cancer cells and kill them. Although mAbs for ADCs must be internalized by the target cells, few methods are available for screening mAbs for their ability to be internalized by cells. We have developed a recombinant protein, termed DT3C, which consists of diphtheria toxin (DT) lacking the receptor-binding domain but containing the C1, C2, and C3 domains of Streptococcus protein G (3C). When a mAb-DT3C conjugate, which functions in vitro like an ADC, reduces the viability of cancer cells, the mAb being tested must have been internalized by the target cells. DT3C can thus be a tool to identify efficiently and easily mAbs that can be internalized by cells, thereby enhancing the development of promising ADCs.
Collapse
Affiliation(s)
- Miki Yamaguchi
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan.
| | - Yukari Nishii
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Kiminori Nakamura
- Department of Cell Biological Science, Faculty of Advanced Life Science, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido 001-0021, Japan
| | - Haruka Aoki
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Sachie Hirai
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Hiroaki Uchida
- Laboratory of Oncology, School of Life Science, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo 192-0392, Japan
| | - Yuji Sakuma
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Hirofumi Hamada
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan; Laboratory of Oncology, School of Life Science, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo 192-0392, Japan
| |
Collapse
|
45
|
Brehm H, Niesen J, Mladenov R, Stein C, Pardo A, Fey G, Helfrich W, Fischer R, Gattenlöhner S, Barth S. A CSPG4-specific immunotoxin kills rhabdomyosarcoma cells and binds to primary tumor tissues. Cancer Lett 2014; 352:228-35. [PMID: 25016058 DOI: 10.1016/j.canlet.2014.07.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/01/2014] [Accepted: 07/03/2014] [Indexed: 11/15/2022]
Abstract
The treatment of rhabdomyosarcoma (RMS) remains challenging, with metastatic and alveolar RMS offering a particularly poor prognosis. Therefore, the identification and evaluation of novel antigens, which are suitable targets for immunotherapy, is one attractive possibility to improve the treatment of this disease. Here we show that chondroitin sulfate proteoglycan 4 (CSPG4) is expressed on RMS cell lines and RMS patient material. We evaluated the immunotoxin (IT) αMCSP-ETA', which specifically recognizes CSPG4 on the RMS cell lines RD, FL-OH1, TE-671 and Rh30. It is internalized rapidly, induces apoptosis and thus kills RMS cells selectively. We also demonstrate the specific binding of this IT to RMS primary tumor material from three different patients.
Collapse
Affiliation(s)
- Hannes Brehm
- Department of Experimental Medicine and Immunotherapy, Institute for Applied Medical Engineering, University Hospital RWTH Aachen, Aachen, Germany
| | - Judith Niesen
- Department of Pharmaceutical Product Development, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Radoslav Mladenov
- Department of Experimental Medicine and Immunotherapy, Institute for Applied Medical Engineering, University Hospital RWTH Aachen, Aachen, Germany; Department of Pharmaceutical Product Development, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Christoph Stein
- Department of Experimental Medicine and Immunotherapy, Institute for Applied Medical Engineering, University Hospital RWTH Aachen, Aachen, Germany; Department of Pharmaceutical Product Development, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Alessa Pardo
- Department of Experimental Medicine and Immunotherapy, Institute for Applied Medical Engineering, University Hospital RWTH Aachen, Aachen, Germany
| | - Georg Fey
- Department of Biology, Friedrich Alexander Universität Erlangen-Nürnberg, Germany
| | - Wijnand Helfrich
- Laboratory for Translational Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Rainer Fischer
- Department of Pharmaceutical Product Development, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany; Institute of Molecular Biotechnology (Biology VII), RWTH Aachen University, Aachen, Germany
| | | | - Stefan Barth
- Department of Experimental Medicine and Immunotherapy, Institute for Applied Medical Engineering, University Hospital RWTH Aachen, Aachen, Germany; Department of Pharmaceutical Product Development, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
| |
Collapse
|
46
|
Malas S, Harrasser M, Lacy KE, Karagiannis SN. Antibody therapies for melanoma: new and emerging opportunities to activate immunity (Review). Oncol Rep 2014; 32:875-86. [PMID: 24969320 PMCID: PMC4121424 DOI: 10.3892/or.2014.3275] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/06/2014] [Indexed: 12/21/2022] Open
Abstract
The interface between malignant melanoma and patient immunity has long been recognised and efforts to treat this most lethal form of skin cancer by activating immune responses with cytokine, vaccine and also antibody immunotherapies have demonstrated promise in limited subsets of patients. In the present study, we discuss different antibody immunotherapy approaches evaluated in the context of melanoma, each designed to act on distinct targets and to employ different mechanisms to restrict tumour growth and spread. Monoclonal antibodies recognising melanoma-associated antigens such as CSPG4/MCSP and targeting elements of tumour-associated vasculature (VEGF) have constituted long-standing translational approaches aimed at reducing melanoma growth and metastasis. Recent insights into mechanisms of immune regulation and tumour-immune cell interactions have helped to identify checkpoint molecules on immune (CTLA4, PD-1) and tumour (PD-L1) cells as promising therapeutic targets. Checkpoint blockade with antibodies to activate immune responses and perhaps to counteract melanoma-associated immunomodulatory mechanisms led to the first clinical breakthrough in the form of an anti-CTLA4 monoclonal antibody. Novel modalities to target key mechanisms of immune suppression and to redirect potent effector cell subsets against tumours are expected to improve clinical outcomes and to provide previously unexplored avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Sadek Malas
- St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine and NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals, King's College London, London SE1 9RT, UK
| | - Micaela Harrasser
- St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine and NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals, King's College London, London SE1 9RT, UK
| | - Katie E Lacy
- St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine and NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals, King's College London, London SE1 9RT, UK
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine and NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals, King's College London, London SE1 9RT, UK
| |
Collapse
|
47
|
Warta R, Herold-Mende C, Chaisaingmongkol J, Popanda O, Mock A, Mogler C, Osswald F, Herpel E, Küstner S, Eckstein V, Plass C, Plinkert P, Schmezer P, Dyckhoff G. Reduced promoter methylation and increased expression of CSPG4 negatively influences survival of HNSCC patients. Int J Cancer 2014; 135:2727-34. [DOI: 10.1002/ijc.28906] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 03/12/2014] [Accepted: 03/28/2014] [Indexed: 11/07/2022]
Affiliation(s)
- Rolf Warta
- Department of Otorhinolaryngology; Head and Neck Surgery, University of Heidelberg; Heidelberg Germany
- Division of Experimental Neurosurgery, Department of Neurosurgery; University of Heidelberg; Heidelberg Germany
| | - Christel Herold-Mende
- Department of Otorhinolaryngology; Head and Neck Surgery, University of Heidelberg; Heidelberg Germany
- Division of Experimental Neurosurgery, Department of Neurosurgery; University of Heidelberg; Heidelberg Germany
| | - Jittiporn Chaisaingmongkol
- Division of Epigenomics and Cancer Risk Factors; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Odilia Popanda
- Division of Epigenomics and Cancer Risk Factors; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Andreas Mock
- Department of Otorhinolaryngology; Head and Neck Surgery, University of Heidelberg; Heidelberg Germany
| | - Carolin Mogler
- NCT Tissue Bank, National Center for Tumor Diseases; Heidelberg Germany
- Institute of Pathology, University of Heidelberg; Heidelberg Germany
| | - Florian Osswald
- Department of Otorhinolaryngology; Head and Neck Surgery, University of Heidelberg; Heidelberg Germany
| | - Esther Herpel
- NCT Tissue Bank, National Center for Tumor Diseases; Heidelberg Germany
- Institute of Pathology, University of Heidelberg; Heidelberg Germany
| | - Sabine Küstner
- Department of Otorhinolaryngology; Head and Neck Surgery, University of Heidelberg; Heidelberg Germany
| | - Volker Eckstein
- Department of Medicine V; University of Heidelberg; Heidelberg Germany
| | - Christoph Plass
- Division of Epigenomics and Cancer Risk Factors; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Peter Plinkert
- Department of Otorhinolaryngology; Head and Neck Surgery, University of Heidelberg; Heidelberg Germany
| | - Peter Schmezer
- Division of Epigenomics and Cancer Risk Factors; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Gerhard Dyckhoff
- Department of Otorhinolaryngology; Head and Neck Surgery, University of Heidelberg; Heidelberg Germany
| |
Collapse
|
48
|
CD70-restricted specific activation of TRAILR1 or TRAILR2 using scFv-targeted TRAIL mutants. Cell Death Dis 2014; 5:e1035. [PMID: 24481449 PMCID: PMC4040681 DOI: 10.1038/cddis.2013.555] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 12/10/2013] [Accepted: 12/13/2013] [Indexed: 12/22/2022]
Abstract
To combine the CD27 stimulation inhibitory effect of blocking CD70 antibodies with an antibody-dependent cellular cytotoxicity (ADCC)-independent, cell death-inducing activity for targeting of CD70-expressing tumors, we evaluated here fusion proteins of the apoptosis-inducing TNF family member TRAIL and a single-chain variable fragment (scFv) derived from a high-affinity llama-derived anti-human CD70 antibody (lαhCD70). A fusion protein of scFv:lαhCD70 with TNC-TRAIL, a stabilized form of TRAIL, showed strongly enhanced apoptosis induction upon CD70 binding and furthermore efficiently interfered with CD70-CD27 interaction. Noteworthy, introduction of recently identified mutations that discriminate between TRAILR1 and TRAILR2 binding into the TRAIL part of scFv:lαhCD70-TNC-TRAIL resulted in TRAIL death receptor-specific fusion proteins with CD70-restricted activity.
Collapse
|
49
|
Tumor targeting properties of antibody fusion proteins based on different members of the murine tumor necrosis superfamily. J Biotechnol 2013; 172:73-6. [PMID: 24384233 DOI: 10.1016/j.jbiotec.2013.12.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 12/14/2013] [Indexed: 11/24/2022]
Abstract
The tumor necrosis factor superfamily (TNFSF) consists of more than 20 members that can modulate cellular and immunological functions, including cell survival and the stimulation of an inflammatory response. Many TNF superfamily members display potent anticancer activity when used as recombinant proteins in vitro and in vivo. While TNF, TRAIL and FasL have already been used as payloads in antibody-based pharmacodelivery strategies, most TNF superfamily members have not yet been investigated as antibody payloads. Here, we report the cloning, production and characterization of eight novel antibody fusion proteins based on CD40L, FasL, TRAIL, LiGHT, VEGI, lymphotoxin alpha, lymphotoxin beta and lymphotoxin alpha1/beta2. The monoclonal antibody F8 was chosen as fusion partner of proven tumor targeting performance, which recognizes the alternatively-spliced EDA domain of fibronectin, a marker of angiogenesis. A quantitative biodistribution analysis performed with radioiodinated protein preparations in tumor-bearing mice revealed that TRAIL and lymphotoxin alpha1/beta2 were able to selectively accumulate at the tumor site, while all other members of the TNF superfamily abrogated the selective tumor targeting performance of the parental antibody or accumulated also in healthy tissues. The study indicates that even cytokines, which are closely related in terms of structure and function, may have a substantially different impact on the biodistribution and functional properties of the corresponding fusions with disease-homing antibodies.
Collapse
|
50
|
Wang LH, Ni CW, Lin YZ, Yin L, Jiang CB, Lv CT, Le Y, Lang Y, Zhao CY, Yang K, Jiao BH, Yin J. Targeted induction of apoptosis in glioblastoma multiforme cells by an MRP3-specific TRAIL fusion protein in vitro. Tumour Biol 2013; 35:1157-68. [PMID: 24272336 DOI: 10.1007/s13277-013-1155-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 08/28/2013] [Indexed: 10/26/2022] Open
Abstract
Single-chain Fv fragments (scFvs) consist of the variable heavy-chain (VH) and variable light-chain (VL) domains, which are the smallest immunoglobulin fragments containing the whole antigen-binding site. Human soluble tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) proves to acquire a potent pro-apoptotic activity only after selective binding to a predefined tumor cell surface antigen and has no off-target effects towards normal cells. Glioblastoma multiforme (GBM) is the most frequent and aggressive type of brain tumor and overexpresses human multidrug resistance protein 3 (MRP3). In this study, we designed a novel fusion protein, termed scFvM58-sTRAIL, in which the MRP3-specific scFv antibody M58 was genetically fused to the N-terminus of human soluble TRAIL (sTRAIL). The recombinant scFvM58-sTRAIL fusion protein, expressed in Escherichia coli, was purified by chromatography and tested for cytotoxicity. scFvM58-sTRAIL showed a significant apoptosis-inducing activity towards MRP3-positive GBM cells in vitro. The pro-apoptotic activity of scFvM58-sTRAIL towards GBM cells was strongly inhibited in the presence of the parental scFvM58 antibody, suggesting that cytotoxic activity is MRP3-restricted. In a control experiment with MRP3-negative Jurkat cells, scFvM58-sTRAIL did not induce apparent apoptosis. In addition, through target antigen-restricted binding, scFvM58-sTRAIL was capable of activating not only TRAIL-R1 but also TRAIL-R2. In conclusion, our results suggest that fusion protein scFvM58-sTRAIL with specificity for MRP3 is a highly selective therapeutic agent and may provide an alternative therapy for human GBM.
Collapse
Affiliation(s)
- Liang-Hua Wang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Xiangyin Road No. 800, Shanghai, 200433, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|