1
|
Liu M, Zhou S, Cao Y, Yang K, Xiao Y, Wang W. Characterization of MAP c21873-1 as a new counter-selectable marker for unmarked genetic modification of Pichia pastoris. Microb Cell Fact 2024; 23:224. [PMID: 39118053 PMCID: PMC11312372 DOI: 10.1186/s12934-024-02496-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: 04/18/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Selection markers are useful in genetic modification of yeast Pichia pastoris. However, the leakage of the promoter caused undesired expression of selection markers especially those toxic proteins like MazF, halting the cell growth and hampering the genetic manipulation in procaryotic system. In this study, a new counter-selectable marker-based strategy has been established for seamless modification with high efficiency and low toxicity. RESULTS At first, the leaky expression of the enhanced green fluorescent protein (EGFP) as a reporter gene under the control of six inducible promoters of P. pastoris was investigated in two hosts Escherichia coli and P. pastoris, respectively. The results demonstrated that the DAS1 and FDH1 promoters (PDAS1 and PFDH1) had the highest leakage expression activities in procaryotes and eukaryotes, and the DAS2 promoter (PDAS2) was inducible with medium strength but low leakage expression activity, all of which were selected for further investigation. Next, Mirabilis antiviral proteins (MAPs) c21873-1, c21873-1T (truncated form of c21873-1) and c23467 were mined as the new counter-selectable markers, and hygromycin B (Hyg B) resistance gene was used as the positive-selectable marker, respectively. Then, modular plasmids with MAP-target gene-Hyg B cassettes were constructed and used to transform into P. pastoris cells after linearization, and the target genes were integrated into its genome at the BmT1 locus through single-crossover homologous recombination (HR). After counter-selection induced by methanol medium, the markers c21873-1 and c21873-1T were recycled efficiently. But c23467 failed to be recycled due to its toxic effect on the P. pastoris cells. At last, the counter-selectable marker c21873-1 under the tightly regulated PDAS2 enabled the encoding genes of reporter EGFP and tested proteins to be integrated into the target locus and expressed successfully. CONCLUSIONS We have developed MAP c21873-1 as a novel counter-selectable marker which could perform efficient gene knock-in by site-directed HR. Upon counter-selection, the marker could be recycled for repeated use, and no undesirable sequences were introduced except for the target gene. This unmarked genetic modification strategy may be extended to other genetic modification including but not limited to gene knock-out and site-directed mutagenesis in future.
Collapse
Affiliation(s)
- Minzhi Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Key Laboratory of Biosynthesis of Natural Products of National Health Commission of the Peoples Republic of China, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Sihan Zhou
- Key Laboratory of Biosynthesis of Natural Products of National Health Commission of the Peoples Republic of China, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yunsong Cao
- Key Laboratory of Biosynthesis of Natural Products of National Health Commission of the Peoples Republic of China, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Keqin Yang
- Key Laboratory of Biosynthesis of Natural Products of National Health Commission of the Peoples Republic of China, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yao Xiao
- Key Laboratory of Biosynthesis of Natural Products of National Health Commission of the Peoples Republic of China, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Wei Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
- Key Laboratory of Biosynthesis of Natural Products of National Health Commission of the Peoples Republic of China, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
2
|
Yoon H, Savoy EA, Mesbahi N, Hendricksen AT, March GL, Fulton MD, Backer BS, Berkman CE. A PSMA-targeted doxorubicin small-molecule drug conjugate. Bioorg Med Chem Lett 2024; 104:129712. [PMID: 38521177 PMCID: PMC11062396 DOI: 10.1016/j.bmcl.2024.129712] [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: 11/26/2023] [Revised: 02/22/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
We developed a model small-molecule drug conjugate (SMDC) that employed doxorubicin as a representative chemotherapeutic targeted to the cell membrane biomarker PSMA (prostate-specific membrane antigen) expressed on prostate cancer cells. The strategy capitalized on the clatherin-mediated internalization of PSMA to facilitate the selective uptake and release of doxorubicin in the target cells. The SMDC was prepared and assessed for binding kinetics, plasma stability, cell toxicity, and specificity towards PSMA expressing prostate cancer cell lines. We observed high affinity of the SMDC for PSMA (IC50 5 nM) with irreversible binding, as well as specific effectiveness against PSMA(+) cells. These findings validated the strategy for a small molecule-based approach in targeted cancer therapy.
Collapse
Affiliation(s)
- Hosog Yoon
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Emily A Savoy
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Nooshin Mesbahi
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Aaron T Hendricksen
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Gabrielle L March
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Melody D Fulton
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Brian S Backer
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States
| | - Clifford E Berkman
- Washington State University, Department of Chemistry Pullman, WA 99164-4630, United States.
| |
Collapse
|
3
|
Savoy EA, Olatunji FP, Mesbahi N, Ballard RK, Lovingier CL, Hendricksen AT, Fulton MD, Berkman CE. PSMA-targeted SMART molecules outfitted with SN38. Bioorg Med Chem Lett 2024; 101:129657. [PMID: 38360419 DOI: 10.1016/j.bmcl.2024.129657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/18/2024] [Accepted: 02/10/2024] [Indexed: 02/17/2024]
Abstract
Herein, we report the modular synthesis and evaluation of a prostate-specific membrane antigen (PSMA) targeted small molecule drug conjugate (SMDC) carrying the chemotherapeutic agent, SN38. Due to the fluorogenic properties of SN38, payload release kinetics from the platform was observed in buffers representing the pH conditions of systemic circulation and cellular internalization. It was found that this platform is stable with minimal payload release at physiological pH with most rapid payload release observed at pH values representing the endosome complex. We confirmed selective payload release and chemotherapeutic efficacy for PSMA(+) prostate cancer cells over PSMA(-) cells. These results demonstrate that chemotherapeutic agents with limited solubility can be conjugated to a water-soluble targeting and linker platform without attenuating efficacy.
Collapse
Affiliation(s)
- Emily A Savoy
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Feyisola P Olatunji
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Nooshin Mesbahi
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Ryanne K Ballard
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Christine L Lovingier
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Aaron T Hendricksen
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Melody D Fulton
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States
| | - Clifford E Berkman
- Washington State University, Department of Chemistry, Pullman, WA 99164-4630, United States.
| |
Collapse
|
4
|
Calzada-Fraile D, Iborra S, Ramírez-Huesca M, Jorge I, Dotta E, Hernández-García E, Martín-Cófreces N, Nistal-Villán E, Veiga E, Vázquez J, Pasqual G, Sánchez-Madrid F. Immune synapse formation promotes lipid peroxidation and MHC-I upregulation in licensed dendritic cells for efficient priming of CD8 + T cells. Nat Commun 2023; 14:6772. [PMID: 37880206 PMCID: PMC10600134 DOI: 10.1038/s41467-023-42480-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023] Open
Abstract
Antigen cognate dendritic cell (DC)-T cell synaptic interactions drive activation of T cells and instruct DCs. Upon receiving CD4+ T cell help, post-synaptic DCs (psDCs) are licensed to generate CD8+ T cell responses. However, the cellular and molecular mechanisms that enable psDCs licensing remain unclear. Here, we describe that antigen presentation induces an upregulation of MHC-I protein molecules and increased lipid peroxidation on psDCs in vitro and in vivo. We also show that these events mediate DC licensing. In addition, psDC adoptive transfer enhances pathogen-specific CD8+ T responses and protects mice from infection in a CD8+ T cell-dependent manner. Conversely, depletion of psDCs in vivo abrogates antigen-specific CD8+ T cell responses during immunization. Together, our data show that psDCs enable CD8+ T cell responses in vivo during vaccination and reveal crucial molecular events underlying psDC licensing.
Collapse
Affiliation(s)
| | - Salvador Iborra
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | | | - Inmaculada Jorge
- Centro Nacional de Investigaciones Cardiovasculares, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029, Madrid, Spain
| | - Enrico Dotta
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Elena Hernández-García
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Noa Martín-Cófreces
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029, Madrid, Spain
- Dynamic Video Microscopy Unit, Immunology Department, Instituto de Investigación Sanitaria Hospital Universitario La Princesa, Universidad Autónoma de Madrid, 28006, Madrid, Spain
| | - Estanislao Nistal-Villán
- Microbiology Section, Departamento CC, Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Boadilla del Monte, 28668, Madrid, Spain
| | - Esteban Veiga
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029, Madrid, Spain
| | - Giulia Pasqual
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Francisco Sánchez-Madrid
- Centro Nacional de Investigaciones Cardiovasculares, 28029, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029, Madrid, Spain.
- Immunology Department, Instituto de Investigación Sanitaria Hospital Universitario La Princesa, Universidad Autónoma de Madrid, 28006, Madrid, Spain.
| |
Collapse
|
5
|
Steinbach C, Merchant A, Zaharie AT, Horak P, Marhold M, Krainer M. Current Developments in Cellular Therapy for Castration Resistant Prostate Cancer: A Systematic Review of Clinical Studies. Cancers (Basel) 2022; 14:5719. [PMID: 36428811 PMCID: PMC9688882 DOI: 10.3390/cancers14225719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/21/2022] [Indexed: 11/23/2022] Open
Abstract
Recently, the development of immunotherapies such as cellular therapy, monoclonal antibodies, vaccines and immunomodulators has revolutionized the treatment of various cancer entities. In order to close the existing gaps in knowledge about cellular immunotherapy, specifically focusing on the chimeric antigen receptors (CAR) T-cells, their benefits and application in clinical settings, we conducted a comprehensive systematic review. Two co-authors independently searched the literature and characterized the results. Out of 183 records, 26 were considered eligible. This review provides an overview of the cellular immunotherapy landscape in treating prostate cancer, honing in on the challenges of employing CAR T-cell therapy. CAR T-cell therapy is a promising avenue for research due to the presence of an array of different tumor specific antigens. In prostate cancer, the complex microenvironment of the tumor vastly contributes to the success or failure of immunotherapies.
Collapse
Affiliation(s)
- Christina Steinbach
- Internal Medicine I, Department of Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Almas Merchant
- Internal Medicine I, Department of Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Peter Horak
- Nationales Centrum für Tumorerkrankungen (NCT), 69120 Heidelberg, Germany
| | - Maximilian Marhold
- Internal Medicine I, Department of Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Krainer
- Internal Medicine I, Department of Oncology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
6
|
Wolf P. Targeted Toxins for the Treatment of Prostate Cancer. Biomedicines 2021; 9:biomedicines9080986. [PMID: 34440190 PMCID: PMC8391386 DOI: 10.3390/biomedicines9080986] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/02/2021] [Accepted: 08/07/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer is the second most common cancer and the fifth leading cause of cancer deaths worldwide. Despite improvements in diagnosis and treatment, new treatment options are urgently needed for advanced stages of the disease. Targeted toxins are chemical conjugates or fully recombinant proteins consisting of a binding domain directed against a target antigen on the surface of cancer cells and a toxin domain, which is transported into the cell for the induction of apoptosis. In the last decades, targeted toxins against prostate cancer have been developed. Several challenges, however, became apparent that prevented their direct clinical use. They comprise immunogenicity, low target antigen binding, endosomal entrapment, and lysosomal/proteasomal degradation of the targeted toxins. Moreover, their efficacy is impaired by prostate tumors, which are marked by a dense microenvironment, low target antigen expression, and apoptosis resistance. In this review, current findings in the development of targeted toxins against prostate cancer in view of effective targeting, reduction of immunogenicity, improvement of intracellular trafficking, and overcoming apoptosis resistance are discussed. There are promising approaches that should lead to the clinical use of targeted toxins as therapeutic alternatives for advanced prostate cancer in the future.
Collapse
Affiliation(s)
- Philipp Wolf
- Department of Urology, Medical Center, University of Freiburg, 79106 Freiburg, Germany; ; Tel.: +49-761-270-28921
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
7
|
Cytotoxic potential of bioactive seed proteins from Mallotus philippensis against various cancer cell lines. APPLIED NANOSCIENCE 2021. [DOI: 10.1007/s13204-021-01974-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
8
|
Kato J, O'Donnell RT, Abuhay M, Tuscano JM. Efficacy and toxicity of a CD22-targeted antibody-saporin conjugate in a xenograft model of non-Hodgkin's lymphoma. Oncoimmunology 2021; 1:1469-1475. [PMID: 23264893 PMCID: PMC3525602 DOI: 10.4161/onci.21815] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Antibody drug conjugates (ADCs) can deliver potent drugs to cancer cells by employing the specificity of monoclonal antibodies (mAbs). ADCs have demonstrated significant anticancer activity and, in 2011, brentuximab vedotin has been approved by the FDA for the treatment of Hodgkin's and anaplastic large cell lymphomas. CD22 is an ideal target for ADC against B-cell malignancies because of its lineage-specific expression and rapid internalization upon antibody binding. In this study, we evaluated the anti-CD22 mAb HB22.7 as a vehicle for the targeted delivery of the potent toxin saporin (SAP). In vitro, HB22.7-SAP was cytotoxic against a panel of non-Hodgkin's lymphoma (NHL) cell lines representing the most common types of the disease. Moreover, in a xenograft model of NHL, HB22.7-SAP significantly inhibited the growth of established lesions and completely prevented tumor development when treatment was initiated within 24 h from tumor-cell inoculation. HB22.7-SAP had no significant in vivo toxicity. In conclusion, HB22.7 constitutes a potential platform for CD22-targeted ADCs.
Collapse
Affiliation(s)
- Jason Kato
- Division of Hematology and Oncology; Department of Internal Medicine; University of California, Davis Cancer Center; Sacramento, CA USA
| | | | | | | |
Collapse
|
9
|
Marquez J, Dong J, Dong C, Tian C, Serrero G. Identification of Prostaglandin F2 Receptor Negative Regulator (PTGFRN) as an internalizable target in cancer cells for antibody-drug conjugate development. PLoS One 2021; 16:e0246197. [PMID: 33503070 PMCID: PMC7840024 DOI: 10.1371/journal.pone.0246197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/14/2021] [Indexed: 11/19/2022] Open
Abstract
Antibody-drug conjugates (ADC) are effective antibody-based therapeutics for hematopoietic and lymphoid tumors. However, there is need to identify new targets for ADCs, particularly for solid tumors and cancers with unmet needs. From a hybridoma library developed against cancer cells, we selected the mouse monoclonal antibody 33B7, which was able to bind to, and internalize, cancer cell lines. This antibody was used for identification of the target by immunoprecipitation and mass spectrometric analysis, followed by target validation. After target validation, 33B7 binding and target positivity were tested by flow cytometry and western blot analysis in several cancer cell lines. The ability of 33B7 conjugated to saporin to inhibit in vitro proliferation of PTFRN positive cell lines was investigated, as well as the 33B7 ADC in vivo effect on tumor growth in athymic mice. All flow cytometry and in vitro internalization assays were analyzed for statistical significance using a Welsh's T-test. Animal studies were analyzed using Two-Way Analysis of Variance (ANOVA) utilizing post-hoc Bonferroni analysis, and/or Mixed Effects analysis. The 33B7 cell surface target was identified as Prostaglandin F2 Receptor Negative Regulator (PTGFRN), a transmembrane protein in the Tetraspanin family. This target was confirmed by showing that PTGFRN-expressing cells bound and internalized 33B7, compared to PTGFRN negative cells. Cells able to bind 33B7 were PTGFRN-positive by Western blot analysis. In vitro treatment PTGFRN-positive cancer cell lines with the 33B7-saporin ADC inhibited their proliferation in a dose-dependent fashion. 33B7 conjugated to saporin was also able to block tumor growth in vivo in mouse xenografts when compared to a control ADC. These findings show that screening antibody libraries for internalizing antibodies in cancer cell lines is a good approach to identify new cancer targets for ADC development. These results suggest PTGFRN is a possible therapeutic target via antibody-based approach for certain cancers.
Collapse
Affiliation(s)
- Jorge Marquez
- Target Discovery Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore School of Pharmacy, Baltimore, Maryland, United States of America
| | - Jianping Dong
- Target Discovery Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
| | - Chun Dong
- Precision Antibody Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
| | - Changsheng Tian
- Target Discovery Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
| | - Ginette Serrero
- Target Discovery Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
- Precision Antibody Division, A&G Pharmaceutical, Inc., Columbia, Maryland, United States of America
| |
Collapse
|
10
|
Chimeric Antigen Receptor-Engineered T Cell Therapy for the Management of Patients with Metastatic Prostate Cancer: A Comprehensive Review. Int J Mol Sci 2021; 22:ijms22020640. [PMID: 33440664 PMCID: PMC7826945 DOI: 10.3390/ijms22020640] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer (PCa) has a vast clinical spectrum from the hormone-sensitive setting to castration-resistant metastatic disease. Thus, chemotherapy regimens and the administration of androgen receptor axis-targeted (ARAT) agents for advanced PCa have shown limited therapeutic efficacy. Scientific advances in the field of molecular medicine and technological developments over the last decade have paved the path for immunotherapy to become an essential clinical modality for the treatment of patients with metastatic PCa. However, several immunotherapeutic agents have shown poor outcomes in patients with advanced disease, possibly due to the low PCa mutational burden. Adoptive cellular approaches utilizing chimeric antigen receptor T cells (CAR-T) targeting cancer-specific antigens would be a solution for circumventing the immune tolerance mechanisms. The immunotherapeutic regimen of CAR-T cell therapy has shown potential in the eradication of hematologic malignancies, and current clinical objectives maintain the equivalent efficacy in the treatment of solid tumors, including PCa. This review will explore the current modalities of CAR-T therapy in the disease spectrum of PCa while describing key limitations of this immunotherapeutic approach and discuss future directions in the application of immunotherapy for the treatment of metastatic PCa and patients with advanced disease.
Collapse
|
11
|
Enzyme-responsive polymeric micelles of cabazitaxel for prostate cancer targeted therapy. Acta Biomater 2020; 113:501-511. [PMID: 32562805 DOI: 10.1016/j.actbio.2020.06.019] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/31/2022]
Abstract
Cabazitaxel, a novel tubulin inhibitor with poor affinity for P-glycoprotein, is a second-generation taxane holding great promise for the treatment of metastatic castration-resistant prostate cancer. However, its poor solubility and lack of target-ability limit its therapeutic applications. Herein, we develop a biodegradable, enzyme-responsive, and targeted polymeric micelle for cabazitaxel. The micelle is formed from two amphiphilic block copolymers. The first block copolymer consists of PEG, an enzyme-responsive peptide, and cholesterol; whereas the second block copolymer consists of a targeting ligand, PEG and cholesterol. The enzyme-responsive peptide is cleavable in the presence of matrixmetaloproteinase-2 (MMP-2), which is overexpressed in the tumor microenvironment of prostate cancer. The micelle showed a very low critical micelle concentration (CMC), high drug loading, and high entrapment efficiency. Release of cabazitaxel from the micelle is dependent on the cleavage of the enzyme-responsive peptide. Moreover, the micelle showed dramatically higher cellular uptake in prostate cancer cells compared to free cabazitaxel. Importantly, the ligand-coupled polymeric micelle demonstrated better inhibition of tumor growth in mice bearing prostate cancer xenografts compared to unmodified micelle and free cabazitaxel. Taken together, these findings suggest that the enzyme-responsive cabazitaxel micelle is a potent and promising drug delivery system for advanced prostate cancer therapy. STATEMENT OF SIGNIFICANCE: Herein, we develop a biodegradable, enzyme-responsive, and actively targeted polymer micelle for cabazitaxel, which is a novel tubulin inhibitor with poor affinity for P-glycoprotein. Despite cabazitaxel's great promise for metastatic castration-resistant prostate cancer, its poor solubility, lack of target-ability, and high systemic toxicity limit its therapeutic applications, and therefore a targeted delivery system is highly needed for cabazitaxel. Our results demonstrate the importance of active targeting in targeted prostate cancer therapy. Encapsulating cabazitaxel in the micelle increases its activity and is expected to reduce its systemic toxicity, which is a major hurdle in its clinical applications. Moreover, the polymeric micelle may servers as a promising nanoscale platform for the targeted delivery of other chemotherapeutic agents to prostate cancer.
Collapse
|
12
|
Schepisi G, Cursano MC, Casadei C, Menna C, Altavilla A, Lolli C, Cerchione C, Paganelli G, Santini D, Tonini G, Martinelli G, De Giorgi U. CAR-T cell therapy: a potential new strategy against prostate cancer. J Immunother Cancer 2019; 7:258. [PMID: 31619289 PMCID: PMC6794851 DOI: 10.1186/s40425-019-0741-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/13/2019] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer (PCa) is one of the main causes of cancer-related death in men. In the present immunotherapy era, several immunotherapeutic agents have been evaluated in PCa with poor results, possibly due to its low mutational burden. The recent development of chimeric antigen receptor (CAR)-T cell therapy redirected against cancer-specific antigens would seem to provide the means for bypassing immune tolerance mechanisms. CAR-T cell therapy has proven effective in eradicating hematologic malignancies and the challenge now is to obtain the same degree of in solid tumors, including PCa. In this study we review the principles that have guided the engineering of CAR-T cells and the specific prostatic antigens identified as possible targets for immunological and non-immunological therapies. We also provide a state-of-the-art overview of CAR-T cell therapy in PCa, defining the key obstacles to its development and underlining the mechanisms used to overcome these barriers. At present, although there are still many unanswered questions regarding CAR-T cell therapy, there is no doubt that it has the potential to become an important treatment option for urological malignancies.
Collapse
Affiliation(s)
- Giuseppe Schepisi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy.
| | | | - Chiara Casadei
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Cecilia Menna
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Amelia Altavilla
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Cristian Lolli
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Claudio Cerchione
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Giovanni Paganelli
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | | | | | - Giovanni Martinelli
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| |
Collapse
|
13
|
Kuan SL, Bergamini FRG, Weil T. Functional protein nanostructures: a chemical toolbox. Chem Soc Rev 2018; 47:9069-9105. [PMID: 30452046 PMCID: PMC6289173 DOI: 10.1039/c8cs00590g] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Indexed: 01/08/2023]
Abstract
Nature has evolved an optimal synthetic factory in the form of translational and posttranslational processes by which millions of proteins with defined primary sequences and 3D structures can be built. Nature's toolkit gives rise to protein building blocks, which dictates their spatial arrangement to form functional protein nanostructures that serve a myriad of functions in cells, ranging from biocatalysis, formation of structural networks, and regulation of biochemical processes, to sensing. With the advent of chemical tools for site-selective protein modifications and recombinant engineering, there is a rapid development to develop and apply synthetic methods for creating structurally defined, functional protein nanostructures for a broad range of applications in the fields of catalysis, materials and biomedical sciences. In this review, design principles and structural features for achieving and characterizing functional protein nanostructures by synthetic approaches are summarized. The synthetic customization of protein building blocks, the design and introduction of recognition units and linkers and subsequent assembly into structurally defined protein architectures are discussed herein. Key examples of these supramolecular protein nanostructures, their unique functions and resultant impact for biomedical applications are highlighted.
Collapse
Affiliation(s)
- Seah Ling Kuan
- Max-Planck Institute for Polymer Research
,
Ackermannweg 10
, 55128 Mainz
, Germany
.
;
- Institute of Inorganic Chemistry I – Ulm University
,
Albert-Einstein-Allee 11
, 89081 Ulm
, Germany
| | - Fernando R. G. Bergamini
- Institute of Chemistry
, Federal University of Uberlândia – UFU
,
38400-902 Uberlândia
, MG
, Brazil
| | - Tanja Weil
- Max-Planck Institute for Polymer Research
,
Ackermannweg 10
, 55128 Mainz
, Germany
.
;
- Institute of Inorganic Chemistry I – Ulm University
,
Albert-Einstein-Allee 11
, 89081 Ulm
, Germany
| |
Collapse
|
14
|
Lelieveldt LPWM, Kristyanto H, Pruijn GJM, Scherer HU, Toes REM, Bonger KM. Sequential Prodrug Strategy To Target and Eliminate ACPA-Selective Autoreactive B Cells. Mol Pharm 2018; 15:5565-5573. [PMID: 30289723 PMCID: PMC6282105 DOI: 10.1021/acs.molpharmaceut.8b00741] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
![]()
Autoreactive B cells are thought
to play a pivotal role in many
autoimmune diseases. Rheumatoid arthritis (RA) is an autoimmune disease
affecting ∼1% of the Western population and is hallmarked by
the presence of anticitrullinated proteins antibodies (ACPA) produced
by autoreactive B cells. We intend to develop a method to target and
selectively eliminate these autoreactive B cells using a sequential
antigen prodrug targeting strategy. As ACPA-expressing B cells are
thought to play essential roles in RA-disease pathogenesis, we used
this B cell response as a prototype to analyze the feasibility to
generate a construct consisting of a biologically silenced, that is,
blocked, antigen connected to a cytotoxic prodrug. Blocking of the
antigen is considered relevant as it is anticipated that circulating
autoantibodies will otherwise clear the antigen-prodrug before it
can reach the target cell. The antigen-prodrug can only bind to the
autoantigen-specific B cell receptor (BCR) upon enzymatic removal
of the blocking group in close proximity of the B cell surface. BCR
binding ultimately induces antigen-specific cytotoxicity after internalization
of the antigen. We have synthesized a cyclic citrullinated peptide
(CCP) antigen suitable for BCR binding and demonstrated that binding
by ACPA was impaired upon introduction of a carboxy-p-nitrobenzyl (CNBz) blocking group at the side chain of the citrulline
residue. Enzymatic removal of the CNBz moiety by nitroreductase fully
restored citrulline-specific recognition by both ACPA and ACPA-expressing
B cells and showed targeted cell death of CCP-recognizing B cells
only. These results mark an important step toward antigen-specific
B cell targeting in general and more specifically in RA, as successful
blocking and activation of citrullinated antigens forms the basis
for subsequent use of such construct as a prodrug in the context of
autoimmune diseases.
Collapse
Affiliation(s)
- Lianne P W M Lelieveldt
- Department of Biomolecular Chemistry, Institute for Molecules and Materials , Radboud University Nijmegen 6525 AJ , The Netherlands
| | - Hendy Kristyanto
- Department of Rheumatology , Leiden University Medical Center , Leiden , The Netherlands
| | - Ger J M Pruijn
- Department of Biomolecular Chemistry, Institute for Molecules and Materials , Radboud University Nijmegen 6525 AJ , The Netherlands
| | - Hans Ulrich Scherer
- Department of Rheumatology , Leiden University Medical Center , Leiden , The Netherlands
| | - René E M Toes
- Department of Rheumatology , Leiden University Medical Center , Leiden , The Netherlands
| | - Kimberly M Bonger
- Department of Biomolecular Chemistry, Institute for Molecules and Materials , Radboud University Nijmegen 6525 AJ , The Netherlands
| |
Collapse
|
15
|
PSMA-targeted polyinosine/polycytosine vector induces prostate tumor regression and invokes an antitumor immune response in mice. Proc Natl Acad Sci U S A 2017; 114:13655-13660. [PMID: 29229829 DOI: 10.1073/pnas.1714587115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
There is an urgent need for an effective treatment for metastatic prostate cancer (PC). Prostate tumors invariably overexpress prostate surface membrane antigen (PSMA). We designed a nonviral vector, PEI-PEG-DUPA (PPD), comprising polyethylenimine-polyethyleneglycol (PEI-PEG) tethered to the PSMA ligand, 2-[3-(1, 3-dicarboxy propyl)ureido] pentanedioic acid (DUPA), to treat PC. The purpose of PEI is to bind polyinosinic/polycytosinic acid (polyIC) and allow endosomal release, while DUPA targets PC cells. PolyIC activates multiple pathways that lead to tumor cell death and to the activation of bystander effects that harness the immune system against the tumor, attacking nontargeted neighboring tumor cells and reducing the probability of acquired resistance and disease recurrence. Targeting polyIC directly to tumor cells avoids the toxicity associated with systemic delivery. PPD selectively delivered polyIC into PSMA-overexpressing PC cells, inducing apoptosis, cytokine secretion, and the recruitment of human peripheral blood mononuclear cells (PBMCs). PSMA-overexpressing tumors in nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice with partially reconstituted immune systems were significantly shrunken following PPD/polyIC treatment, in all cases. Half of the tumors showed complete regression. PPD/polyIC invokes antitumor immunity, but unlike many immunotherapies does not need to be personalized for each patient. The potent antitumor effects of PPD/polyIC should spur its development for clinical use.
Collapse
|
16
|
Kessler C, Pardo A, Tur MK, Gattenlöhner S, Fischer R, Kolberg K, Barth S. Novel PSCA targeting scFv-fusion proteins for diagnosis and immunotherapy of prostate cancer. J Cancer Res Clin Oncol 2017; 143:2025-2038. [PMID: 28667390 DOI: 10.1007/s00432-017-2472-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/28/2017] [Indexed: 12/22/2022]
Abstract
PURPOSE Despite great progress in the diagnosis and treatment of localized prostate cancer (PCa), there remains a need for new diagnostic markers that can accurately distinguish indolent and aggressive variants. One promising approach is the antibody-based targeting of prostate stem cell antigen (PSCA), which is frequently overexpressed in PCa. Here, we show the construction of a molecular imaging probe comprising a humanized scFv fragment recognizing PSCA genetically fused to an engineered version of the human DNA repair enzyme O6-alkylguanine-DNA alkyltransferase (AGT), the SNAP-tag, enabling specific covalent coupling to various fluorophores for diagnosis of PCa. Furthermore, the recombinant immunotoxin (IT) PSCA(scFv)-ETA' comprising the PSCA(scFv) and a truncated version of Pseudomonas exotoxin A (PE, ETA') was generated. METHODS We analyzed the specific binding and internalization behavior of the molecular imaging probe PSCA(scFv)-SNAP in vitro by flow cytometry and live cell imaging, compared to the corresponding IT PSCA(scFv)-ETA'. The cytotoxic activity of PSCA(scFv)-ETA' was tested using cell viability assays. Specific binding was confirmed on formalin-fixed paraffin-embedded tissue specimen of early and advanced PCa. RESULTS Alexa Fluor® 647 labeling of PSCA(scFv)-SNAP confirmed selective binding to PSCA, leading to rapid internalization into the target cells. The recombinant IT PSCA(scFv)-ETA' showed selective binding leading to internalization and efficient elimination of target cells. CONCLUSIONS Our data demonstrate, for the first time, the specific binding, internalization, and cytotoxicity of a scFv-based fusion protein targeting PSCA. Immunohistochemical staining confirmed the specific ex vivo binding to primary PCa material.
Collapse
Affiliation(s)
- Claudia Kessler
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany
- Institute of Applied Medical Engineering, University Hospital RWTH Aachen, Aachen, Germany
| | - Alessa Pardo
- Institute of Applied Medical Engineering, University Hospital RWTH Aachen, Aachen, Germany
| | - Mehmet K Tur
- Institute for Pathology, Justus-Liebig University, Giessen, Germany
| | | | - Rainer Fischer
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany
- Institute of Molecular Biotechnology (Biology VII), RWTH Aachen University, Aachen, Germany
| | - Katharina Kolberg
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany
- Institute of Applied Medical Engineering, University Hospital RWTH Aachen, Aachen, Germany
| | - Stefan Barth
- South African Research Chair in Cancer Biotechnology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Anzio Road, Observatory, 7925, South Africa.
| |
Collapse
|
17
|
Abuhay M, Kato J, Tuscano E, Barisone GA, Sidhu RS, O'Donnell RT, Tuscano JM. The HB22.7-vcMMAE antibody-drug conjugate has efficacy against non-Hodgkin lymphoma mouse xenografts with minimal systemic toxicity. Cancer Immunol Immunother 2016; 65:1169-75. [PMID: 27506529 PMCID: PMC7643839 DOI: 10.1007/s00262-016-1873-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 07/19/2016] [Indexed: 10/21/2022]
Abstract
In this study, HB22.7, an anti-CD22 monoclonal antibody, was used for specific, targeted delivery of monomethyl auristatin E (MMAE) to non-Hodgkin lymphoma (NHL). MMAE was covalently coupled to HB22.7 through a valine-citrulline peptide linker (vc). Maleimide-functionalized vcMMAE (mal-vcMMAE) was reacted with thiols of the partially reduced mAb. Approximately 4 molecules of MMAE were conjugated to HB22.7 as determined by residual thiol measurement and hydrophobic interaction chromatography-HPLC (HIC-HPLC). HB22.7-vcMMAE antibody-drug conjugate (ADC) retained its binding to Ramos NHL cells and also exhibited potent and specific in vitro cytotoxicity on a panel of B cell NHL cell lines with IC50s of 20-284 ng/ml. HB22.7-vcMMAE also showed potent efficacy in vivo against established NHL xenografts using the DoHH2 and Granta 519 cell lines. One dose of the ADC induced complete and persistent response in all DoHH2 xenografts and 90 % of Granta xenografts. Minimal toxicity was observed. In summary, HB22.7-vcMMAE is an effective ADC that should be evaluated for clinical translation.
Collapse
Affiliation(s)
- Mastewal Abuhay
- Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis, UCDHS 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA
| | - Jason Kato
- Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis, UCDHS 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA
| | - Emily Tuscano
- Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis, UCDHS 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA
| | - Gustavo A Barisone
- Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis, UCDHS 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA
| | - Ranjit S Sidhu
- Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis, UCDHS 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA
| | - Robert T O'Donnell
- Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis, UCDHS 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA
- Department of Veterans' Affairs, Northern California Healthcare System, Mather, CA, USA
| | - Joseph M Tuscano
- Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis, UCDHS 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA.
- Department of Veterans' Affairs, Northern California Healthcare System, Mather, CA, USA.
| |
Collapse
|
18
|
Lee HJ, Yoon YI, Bae YJ. Theragnostic ultrasound using microbubbles in the treatment of prostate cancer. Ultrasonography 2016; 35:309-17. [PMID: 27197842 PMCID: PMC5040139 DOI: 10.14366/usg.16006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/14/2016] [Accepted: 04/14/2016] [Indexed: 12/19/2022] Open
Abstract
The use of gas-filled microbubbles in perfusion monitoring as intravascular ultrasound contrast agents has recently become more common. Additionally, microbubbles are employed as carriers of pharmaceutical substances or genes. Microbubbles have great potential to improve the delivery of therapeutic materials into cells and to modify vascular permeability, causing increased extravasation of drugs and drug carriers. Prostate cancer is the most common neoplasm in Europe and America, with an incidence twice to three times that of lung and colorectal cancer. Its incidence is still rising in Asian countries, including Japan and Korea. In this review, we present current strategies regarding the synthesis of microbubbles with targeted ligands on their surfaces, with a focus on prostate cancer.
Collapse
Affiliation(s)
- Hak Jong Lee
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.,Program in Nanoscience and Technology, Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Young Il Yoon
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea.,Program in Nanoscience and Technology, Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Yun Jung Bae
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
19
|
Zeng M, Zheng M, Lu D, Wang J, Jiang W, Sha O. Anti-tumor activities and apoptotic mechanism of ribosome-inactivating proteins. CHINESE JOURNAL OF CANCER 2015; 34:325-34. [PMID: 26184404 PMCID: PMC4593346 DOI: 10.1186/s40880-015-0030-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/14/2015] [Indexed: 01/22/2023]
Abstract
Ribosome-inactivating proteins (RIPs) belong to a family of enzymes that attack eukaryotic ribosomes and potently inhibit cellular protein synthesis. RIPs possess several biomedical properties, including anti-viral and anti-tumor activities. Multiple RIPs are known to inhibit tumor cell proliferation through inducing apoptosis in a variety of cancers, such as breast cancer, leukemia/lymphoma, and hepatoma. This review focuses on the anti-tumor activities of RIPs and their apoptotic effects through three closely related pathways: mitochondrial, death receptor, and endoplasmic reticulum pathways.
Collapse
Affiliation(s)
- Meiqi Zeng
- School of Medicine, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
| | - Manyin Zheng
- School of Medicine, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
| | - Desheng Lu
- School of Medicine, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
| | - Jun Wang
- School of Medicine, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
| | - Wenqi Jiang
- School of Medicine, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
- School of Medicine, Shenzhen University, Shenzhen, 518060, Guangdong, People's Republic of China.
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, 510060, People's Republic of China.
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Ou Sha
- School of Medicine, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
| |
Collapse
|
20
|
Barve A, Jin W, Cheng K. Prostate cancer relevant antigens and enzymes for targeted drug delivery. J Control Release 2014; 187:118-32. [PMID: 24878184 DOI: 10.1016/j.jconrel.2014.05.035] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 05/13/2014] [Accepted: 05/17/2014] [Indexed: 12/26/2022]
Abstract
Chemotherapy is one of the most widely used approaches in combating advanced prostate cancer, but its therapeutic efficacy is usually insufficient due to poor specificity and associated toxicity. Lack of targeted delivery to prostate cancer cells is also the primary obstacles in achieving feasible therapeutic effect of other promising agents including peptide, protein, and nucleic acid. Consequently, there remains a critical need for strategies to increase the selectivity of anti-prostate cancer agents. This review will focus on various prostate cancer-relevant antigens and enzymes that could be exploited for prostate cancer targeted drug delivery. Among various targeting strategies, active targeting is the most advanced approach to specifically deliver drugs to their designated cancer cells. In this approach, drug carriers are modified with targeting ligands that can specifically bind to prostate cancer-specific antigens. Moreover, there are several specific enzymes in the tumor microenvironment of prostate cancer that can be exploited for stimulus-responsive drug delivery systems. These systems can specifically release the active drug in the tumor microenvironment of prostate cancer, leading to enhanced tumor penetration efficiency.
Collapse
Affiliation(s)
- Ashutosh Barve
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City 64108, USA
| | - Wei Jin
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City 64108, USA
| | - Kun Cheng
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City 64108, USA.
| |
Collapse
|
21
|
Bispecific small molecule-antibody conjugate targeting prostate cancer. Proc Natl Acad Sci U S A 2013; 110:17796-801. [PMID: 24127589 DOI: 10.1073/pnas.1316026110] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Bispecific antibodies, which simultaneously target CD3 on T cells and tumor-associated antigens to recruit cytotoxic T cells to cancer cells, are a promising new approach to the treatment of hormone-refractory prostate cancer. Here we report a site-specific, semisynthetic method for the production of bispecific antibody-like therapeutics in which a derivative of the prostate-specific membrane antigen-binding small molecule DUPA was selectively conjugated to a mutant αCD3 Fab containing the unnatural amino acid, p-acetylphenylalanine, at a defined site. Homogeneous conjugates were generated in excellent yields and had good solubility. The efficacy of the conjugate was optimized by modifying the linker structure, relative binding orientation, and stoichiometry of the ligand. The optimized conjugate showed potent and selective in vitro activity (EC50 ~ 100 pM), good serum half-life, and potent in vivo activity in prophylactic and treatment xenograft mouse models. This semisynthetic approach is likely to be applicable to the generation of additional bispecific agents using drug-like ligands selective for other cell-surface receptors.
Collapse
|
22
|
Saporin-S6: a useful tool in cancer therapy. Toxins (Basel) 2013; 5:1698-722. [PMID: 24105401 PMCID: PMC3813907 DOI: 10.3390/toxins5101698] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/17/2013] [Accepted: 09/22/2013] [Indexed: 01/24/2023] Open
Abstract
Thirty years ago, the type 1 ribosome-inactivating protein (RIP) saporin-S6 (also known as saporin) was isolated from Saponaria officinalis L. seeds. Since then, the properties and mechanisms of action of saporin-S6 have been well characterized, and it has been widely employed in the construction of conjugates and immunotoxins for different purposes. These immunotoxins have shown many interesting results when used in cancer therapy, particularly in hematological tumors. The high enzymatic activity, stability and resistance to conjugation procedures and blood proteases make saporin-S6 a very useful tool in cancer therapy. High efficacy has been reported in clinical trials with saporin-S6-containing immunotoxins, at dosages that induced only mild and transient side effects, which were mainly fever, myalgias, hepatotoxicity, thrombocytopenia and vascular leak syndrome. Moreover, saporin-S6 triggers multiple cell death pathways, rendering impossible the selection of RIP-resistant mutants. In this review, some aspects of saporin-S6, such as the chemico-physical characteristics, the structural properties, its endocytosis, its intracellular routing and the pathogenetic mechanisms of the cell damage, are reported. In addition, the recent progress and developments of saporin-S6-containing immunotoxins in cancer immunotherapy are summarized, including in vitro and in vivo pre-clinical studies and clinical trials.
Collapse
|
23
|
Baiz D, Hassan S, Choi YA, Flores A, Karpova Y, Yancey D, Pullikuth A, Sui G, Sadelain M, Debinski W, Kulik G. Combination of the PI3K inhibitor ZSTK474 with a PSMA-targeted immunotoxin accelerates apoptosis and regression of prostate cancer. Neoplasia 2013; 15:1172-83. [PMID: 24204196 PMCID: PMC3819633 DOI: 10.1593/neo.13986] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 08/07/2013] [Accepted: 08/09/2013] [Indexed: 12/12/2022]
Abstract
The phosphoinositide 3-kinase (PI3K) pathway is activated in most advanced prostate cancers, yet so far treatments with PI3K inhibitors have been at best tumorostatic in preclinical cancer models and do not show significant antitumor efficacy in clinical trials. Results from tissue culture experiments in prostate cancer cells suggest that PI3K inhibitors should be combined with other cytotoxic agents; however, the general toxicity of such combinations prevents translating these experimental data into preclinical and clinical models. We investigated the emerging concept of tumor-targeted synthetic lethality in prostate cancer cells by using the pan-PI3K inhibitor ZSTK474 and the immunotoxin J591PE, a protein chimera between the single-chain variable fragment of the monoclonal antibody J591 against the prostate-specific membrane antigen (PSMA) and the truncated form of the Pseudomonas aeruginosa exotoxin A (PE38QQR). The combination of ZSTK474 and J591PE increased apoptosis within 6 hours and cell death (monitored at 24-48 hours) in the PSMA-expressing cells LNCaP, C4-2, and C4-2Luc but not in control cells that do not express PSMA (PC3 and BT549 cells). Mechanistic analysis suggested that induction of apoptosis requires Bcl-2-associated death promoter (BAD) dephosphorylation and decreased expression of myeloid leukemia cell differentiation protein 1 (MCL-1). A single injection of ZSTK474 and J591PE into engrafted prostate cancer C4-2Luc cells led to consistent and stable reduction of luminescence within 6 days. These results suggest that the combination of a PI3K inhibitor and a PSMA-targeted protein synthesis inhibitor toxin represents a promising novel strategy for advanced prostate cancer therapy that should be further investigated.
Collapse
Affiliation(s)
- Daniele Baiz
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC
| | - Sazzad Hassan
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC
| | - Young A Choi
- Department of Neurosurgery and Brain Tumor Center of Excellence, Wake Forest School of Medicine, Winston-Salem, NC
| | - Anabel Flores
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC
| | - Yelena Karpova
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC
| | - Dana Yancey
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC
| | - Ashok Pullikuth
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC
| | - Guangchao Sui
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC
| | - Michel Sadelain
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Waldemar Debinski
- Department of Neurosurgery and Brain Tumor Center of Excellence, Wake Forest School of Medicine, Winston-Salem, NC
| | - George Kulik
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC
| |
Collapse
|
24
|
Abstract
Ribosome-inactivating proteins (RIPs) were first isolated over a century ago and have been shown to be catalytic toxins that irreversibly inactivate protein synthesis. Elucidation of atomic structures and molecular mechanism has revealed these proteins to be a diverse group subdivided into two classes. RIPs have been shown to exhibit RNA N-glycosidase activity and depurinate the 28S rRNA of the eukaryotic 60S ribosomal subunit. In this review, we compare archetypal RIP family members with other potent toxins that abolish protein synthesis: the fungal ribotoxins which directly cleave the 28S rRNA and the newly discovered Burkholderia lethal factor 1 (BLF1). BLF1 presents additional challenges to the current classification system since, like the ribotoxins, it does not possess RNA N-glycosidase activity but does irreversibly inactivate ribosomes. We further discuss whether the RIP classification should be broadened to include toxins achieving irreversible ribosome inactivation with similar turnovers to RIPs, but through different enzymatic mechanisms.
Collapse
Affiliation(s)
- Matthew J Walsh
- RNA Biology Laboratory; Sheffield Institute for Translational Neuroscience (SITraN); Department of Neuroscience; University of Sheffield; Sheffield, UK
| | - Jennifer E Dodd
- RNA Biology Laboratory; Sheffield Institute for Translational Neuroscience (SITraN); Department of Neuroscience; University of Sheffield; Sheffield, UK
| | - Guillaume M Hautbergue
- RNA Biology Laboratory; Sheffield Institute for Translational Neuroscience (SITraN); Department of Neuroscience; University of Sheffield; Sheffield, UK
| |
Collapse
|
25
|
Zhang F, Shan L, Liu Y, Neville D, Woo JH, Chen Y, Korotcov A, Lin S, Huang S, Sridhar R, Liang W, Wang PC. An anti-PSMA bivalent immunotoxin exhibits specificity and efficacy for prostate cancer imaging and therapy. Adv Healthc Mater 2013; 2:736-44. [PMID: 23184611 PMCID: PMC3741670 DOI: 10.1002/adhm.201200254] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Revised: 08/22/2012] [Indexed: 11/10/2022]
Abstract
Prostate specific membrane antigen (PSMA) is overexpressed on prostate tumor cells and the neovascular endothelia various solid tumors. A bivalent immunotoxin generated by fusing a fold-back single-chain diabody derived from the Fv fragments of an anti-PSMA monoclonal antibody with a truncated diphtheria toxin (DT) containing the activity and translocation domains [A-dmDT390-scfbDb(PSMA)] might be suitable for targeted therapy of tumors that overexpress PSMA. In this study, a PSMA-positive and a PSMA-negative prostate cancer cell lines were treated with immunotoxin A-dmDT390-scfbDb(PSMA) in order to study the tumor targeting specificity and therapeutic potential of the immunotoxin. The cellular uptake and selective toxicity of the immunotoxin were evident in monolayer cultures of PSMA-positive LNCaP prostate cancer cells but not in cultures of PSMA-negative PC-3 prostate cancer cells. Cellular accumulation of A-dmDT390-scfbDb(PSMA) increased with increasing incubation times and concentrations in LNCaP cells. The proportion of apoptotic LNCaP cells increased upon incubation with increasing doses of the fold-back immunotoxin. Optical imaging and MRI with the Alexa Fluor 680-labeled A-dmDT390-scfbDb(PSMA) confirmed the specific targeting and therapeutic efficacy of this immunotoxin towards PSMA-positive LNCaP solid tumor xenografts in athymic nude mice.
Collapse
Affiliation(s)
- Fayun Zhang
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, 20060, USA
- Protein & Peptide Pharmaceutical Laboratory, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Liang Shan
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, 20060, USA
- National Center for Biotechnology Information, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | - Jung-Hee Woo
- Cancer Research Institute of Scott and White Healthcare, Texas A&M Health Science Center, Temple, TX 76502, USA
| | - Yue Chen
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, 20060, USA
| | - Alexandru Korotcov
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, 20060, USA
| | - Stephen Lin
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, 20060, USA
| | - Sophia Huang
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, 20060, USA
| | - Rajagopalan Sridhar
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, 20060, USA
| | - Wei Liang
- Protein & Peptide Pharmaceutical Laboratory, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Paul C. Wang
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, 20060, USA
| |
Collapse
|
26
|
Pan S, Wang F, Huang P, Xu T, Zhang L, Xu J, Li Q, Xia W, Sun R, Huang L, Peng Y, Qin X, Shu Y, Hu Z, Shen H. The study on newly developed McAb NJ001 specific to non-small cell lung cancer and its biological characteristics. PLoS One 2012; 7:e33009. [PMID: 22479355 PMCID: PMC3316548 DOI: 10.1371/journal.pone.0033009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2011] [Accepted: 02/02/2012] [Indexed: 01/05/2023] Open
Abstract
Monoclonal antibody (McAb) is the key tool for cancer immunodiagnosis and immunotherapy. McAb-based immunotherapy that targets tumor antigens has had great achivement. In this study, a cell clone which kept secreting high-titer IgG1-type McAb named NJ001 against human non-small cell lung cancer (NSCLC) cells was obtained. The titer of purified NJ001 was 2×106. The antigen named SP70 of NSCLC specifically identified by NJ001 was proved to be a protein with the relative molecular mass (Mr) of 70 kDa. The results of immunohistochemical staining indicated that NJ001 could positively react to NSCLC, but weak positively or negatively react to human small-cell lung cancer (SCLC), pulmonary pseudotumor and other epithelial tumors. In soft agar assay, the colony formation efficiency in NJ001 groups decreased in a dose-dependent manner. For the concentration of 100 µg/ml, 200 µg/ml and 400 µg/ml, the inhibition ratio of colony formation was 23.4%, 62.5% and 100% respectively. Meanwhile, NJ001 caused significant reduction in tumor volume and tumor weight compared to control mice in lung cancer xenograft model. The tumor growth inhibition ratio in 200 µg, 400 µg and 800 µg NJ001 groups was 10.44%, 37.29% and 44.04%, respectively. NJ001 also led to cytomorphological changes and induced the apoptosis of human lung adenocarcinoma cell line SPC-A1 significantly. The newly developed NJ001 selectively reacted to NSCLC and exhibited anti-tumor activity both in vitro and in vivo. NJ001 is of great value concerning immunodiagnostics and immunotherapy for NSCLC and holds promise for further research regarding the mechanism underlying tumor progression of NSCLC.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/immunology
- Adenocarcinoma/pathology
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigens, Neoplasm/immunology
- Apoptosis/drug effects
- Blotting, Western
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Female
- Humans
- Hybridomas
- Immunohistochemistry
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Molecular Weight
- Time Factors
- Tumor Burden/drug effects
- Tumor Burden/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Shiyang Pan
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Tumor-associated antigens for specific immunotherapy of prostate cancer. Cancers (Basel) 2012; 4:193-217. [PMID: 24213236 PMCID: PMC3712678 DOI: 10.3390/cancers4010193] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 02/14/2012] [Accepted: 02/16/2012] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PCa) is the most common noncutaneous cancer diagnosis and the second leading cause of cancer-related deaths among men in the United States. Effective treatment modalities for advanced metastatic PCa are limited. Immunotherapeutic strategies based on T cells and antibodies represent interesting approaches to prevent progression from localized to advanced PCa and to improve survival outcomes for patients with advanced disease. CD8+ cytotoxic T lymphocytes (CTLs) efficiently recognize and destroy tumor cells. CD4+ T cells augment the antigen-presenting capacity of dendritic cells and promote the expansion of tumor-reactive CTLs. Antibodies mediate their antitumor effects via antibody-dependent cellular cytotoxicity, activation of the complement system, improving the uptake of coated tumor cells by phagocytes, and the functional interference of biological pathways essential for tumor growth. Consequently, several tumor-associated antigens (TAAs) have been identified that represent promising targets for T cell- or antibody-based immunotherapy. These TAAs comprise proteins preferentially expressed in normal and malignant prostate tissues and molecules which are not predominantly restricted to the prostate, but are overexpressed in various tumor entities including PCa. Clinical trials provide evidence that specific immunotherapeutic strategies using such TAAs represent safe and feasible concepts for the induction of immunological and clinical responses in PCa patients. However, further improvement of the current approaches is required which may be achieved by combining T cell- and/or antibody-based strategies with radio-, hormone-, chemo- or antiangiogenic therapy.
Collapse
|
28
|
Fu W, Madan E, Yee M, Zhang H. Progress of molecular targeted therapies for prostate cancers. Biochim Biophys Acta Rev Cancer 2011; 1825:140-52. [PMID: 22146293 DOI: 10.1016/j.bbcan.2011.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 11/18/2011] [Accepted: 11/19/2011] [Indexed: 01/16/2023]
Abstract
Prostate cancer remains the most commonly diagnosed malignancy and the second leading cause of cancer-related deaths in men in the United States. The current standard of care consists of prostatectomy and radiation therapy, which may often be supplemented with hormonal therapies. Recurrence is common, and many develop metastatic prostate cancer for which chemotherapy is only moderately effective. It is clear that novel therapies are needed for the treatment of the malignant forms of prostate cancer that recur after initial therapies, such as hormone refractory (HRPC) or castration resistant prostate cancer (CRPC). With advances in understanding of the molecular mechanisms of cancer, we have witnessed unprecedented progress in developing new forms of targeted therapy. Several targeted therapeutic agents have been developed and clinically used for the treatment of solid tumors such as breast cancer, non-small cell lung cancer, and renal cancer. Some of these reagents modulate growth factors and/or their receptors, which are abundant in cancer cells. Other reagents target the downstream signal transduction, survival pathways, and angiogenesis pathways that are abnormally activated in transformed cells or metastatic tumors. We will review current developments in this field, focusing specifically on treatments that can be applied to prostate cancers. Finally we will describe aspects of the future direction of the field with respect to discovering biomarkers to aid in identifying responsive prostate cancer patients.
Collapse
Affiliation(s)
- Weihua Fu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, USA
| | | | | | | |
Collapse
|
29
|
Taylor RM, Huber DL, Monson TC, Ali AMS, Bisoffi M, Sillerud LO. Multifunctional iron platinum stealth immunomicelles: targeted detection of human prostate cancer cells using both fluorescence and magnetic resonance imaging. JOURNAL OF NANOPARTICLE RESEARCH : AN INTERDISCIPLINARY FORUM FOR NANOSCALE SCIENCE AND TECHNOLOGY 2011; 13:4717-4729. [PMID: 22121333 PMCID: PMC3223933 DOI: 10.1007/s11051-011-0439-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) are the most common type of contrast agents used in contrast agent-enhanced magnetic resonance imaging (MRI). Still, there is a great deal of room for improvement, and nanoparticles with increased MRI relaxivities are needed to increase the contrast enhancement in MRI applied to various medical conditions including cancer. We report the synthesis of superparamagnetic iron platinum nanoparticles (SIPPs) and subsequent encapsulation using PEGylated phospholipids to create stealth immunomicelles (DSPE-SIPPs) that can be specifically targeted to human prostate cancer cell lines and detected using both MRI and fluorescence imaging. SIPP cores and DSPE-SIPPs were 8.5 ± 1.6 nm and 42.9 ± 8.2 nm in diameter, respectively, and the SIPPs had a magnetic moment of 120 A m(2)/kg iron. J591, a monoclonal antibody against prostate specific membrane antigen (PSMA), was conjugated to the DSPE-SIPPs (J591-DSPE-SIPPs), and specific targeting of J591-DSPE-SIPPs to PSMA-expressing human prostate cancer cell lines was demonstrated using fluorescence confocal microscopy. The transverse relaxivity of the DSPE-SIPPs, measured at 4.7 Tesla, was 300.6 ± 8.5 s(-1) mM(-1), which is 13-fold better than commercially available SPIONs (23.8 ± 6.9 s(-1) mM(-1)) and ~3-fold better than reported relaxivities for Feridex(®) and Resovist(®). Our data suggest that J591-DSPE-SIPPs specifically target human prostate cancer cells in vitro, are superior contrast agents in T(2)-weighted MRI, and can be detected using fluorescence imaging. To our knowledge, this is the first report on the synthesis of multifunctional SIPP micelles and using SIPPs for the specific detection of prostate cancer.
Collapse
Affiliation(s)
- Robert M. Taylor
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Dale L. Huber
- Center for Integrated Nanotechnologies, Sandia National Laboratories, Albuquerque, NM 87111, USA
| | - Todd C. Monson
- Nanomaterials Sciences Department, Sandia National Laboratories, Albuquerque, NM 87111, USA
| | - Abdul-Mehdi S. Ali
- Department of Earth and Planetary Sciences, University of New Mexico, Albuquerque, NM 87131, USA
| | - Marco Bisoffi
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Laurel O. Sillerud
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| |
Collapse
|
30
|
Polito L, Bortolotti M, Pedrazzi M, Bolognesi A. Immunotoxins and other conjugates containing saporin-s6 for cancer therapy. Toxins (Basel) 2011; 3:697-720. [PMID: 22069735 PMCID: PMC3202841 DOI: 10.3390/toxins3060697] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 05/27/2011] [Accepted: 06/03/2011] [Indexed: 11/18/2022] Open
Abstract
Ribosome-inactivating proteins (RIPs) are a family of plant toxins that permanently damage ribosomes and possibly other cellular substrates, thus causing cell death. RIPs are mostly divided in two types: Type 1 RIPs that are single-chain enzymatic proteins, and type 2 RIPs that consist of an active A chain (similar to a type 1 RIP) linked to a B chain with lectin properties. RIP-containing conjugates have been used in many experimental strategies against cancer cells, often showing great efficacy in clinical trials. Saporin-S6, a type 1 RIP extracted from Saponaria officinalis L. seeds, has been extensively utilized to construct anti-cancer conjugates because of its high enzymatic activity, stability and resistance to conjugation procedures, resulting in the efficient killing of target cells. This review summarizes saporin-S6-containing conjugates and their application in cancer therapy, considering in-vitro and in-vivo studies both in animal models and in clinical trials. The review is structured on the basis of the targeting of hematological versus solid tumors and on the antigen recognized on the cell surface.
Collapse
Affiliation(s)
- Letizia Polito
- Department of Experimental Pathology, "Alma Mater Studiorum" University of Bologna, via San Giacomo 14, 40126-Bologna, Italy.
| | | | | | | |
Collapse
|
31
|
Thie H, Toleikis L, Li J, von Wasielewski R, Bastert G, Schirrmann T, Esteves IT, Behrens CK, Fournes B, Fournier N, de Romeuf C, Hust M, Dübel S. Rise and fall of an anti-MUC1 specific antibody. PLoS One 2011; 6:e15921. [PMID: 21264246 PMCID: PMC3021526 DOI: 10.1371/journal.pone.0015921] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 12/07/2010] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND So far, human antibodies with good affinity and specificity for MUC1, a transmembrane protein overexpressed on breast cancers and ovarian carcinomas, and thus a promising target for therapy, were very difficult to generate. RESULTS A human scFv antibody was isolated from an immune library derived from breast cancer patients immunised with MUC1. The anti-MUC1 scFv reacted with tumour cells in more than 80% of 228 tissue sections of mamma carcinoma samples, while showing very low reactivity with a large panel of non-tumour tissues. By mutagenesis and phage display, affinity of scFvs was increased up to 500fold to 5,7×10(-10) M. Half-life in serum was improved from below 1 day to more than 4 weeks and was correlated with the dimerisation tendency of the individual scFvs. The scFv bound to T47D and MCF-7 mammalian cancer cell lines were recloned into the scFv-Fc and IgG format resulting in decrease of affinity of one binder. The IgG variants with the highest affinity were tested in mouse xenograft models using MCF-7 and OVCAR tumour cells. However, the experiments showed no significant decrease in tumour growth or increase in the survival rates. To study the reasons for the failure of the xenograft experiments, ADCC was analysed in vitro using MCF-7 and OVCAR3 target cells, revealing a low ADCC, possibly due to internalisation, as detected for MCF-7 cells. CONCLUSIONS Antibody phage display starting with immune libraries and followed by affinity maturation is a powerful strategy to generate high affinity human antibodies to difficult targets, in this case shown by the creation of a highly specific antibody with subnanomolar affinity to a very small epitope consisting of four amino acids. Despite these "best in class" binding parameters, the therapeutic success of this antibody was prevented by the target biology.
Collapse
Affiliation(s)
- Holger Thie
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | - Lars Toleikis
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | - Jiandong Li
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | | | | | - Thomas Schirrmann
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | | | | | | | | | | | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | - Stefan Dübel
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
- * E-mail:
| |
Collapse
|
32
|
de Virgilio M, Lombardi A, Caliandro R, Fabbrini MS. Ribosome-inactivating proteins: from plant defense to tumor attack. Toxins (Basel) 2010; 2:2699-737. [PMID: 22069572 PMCID: PMC3153179 DOI: 10.3390/toxins2112699] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 10/29/2010] [Accepted: 11/04/2010] [Indexed: 12/02/2022] Open
Abstract
Ribosome-inactivating proteins (RIPs) are EC3.2.32.22 N-glycosidases that recognize a universally conserved stem-loop structure in 23S/25S/28S rRNA, depurinating a single adenine (A4324 in rat) and irreversibly blocking protein translation, leading finally to cell death of intoxicated mammalian cells. Ricin, the plant RIP prototype that comprises a catalytic A subunit linked to a galactose-binding lectin B subunit to allow cell surface binding and toxin entry in most mammalian cells, shows a potency in the picomolar range. The most promising way to exploit plant RIPs as weapons against cancer cells is either by designing molecules in which the toxic domains are linked to selective tumor targeting domains or directly delivered as suicide genes for cancer gene therapy. Here, we will provide a comprehensive picture of plant RIPs and discuss successful designs and features of chimeric molecules having therapeutic potential.
Collapse
Affiliation(s)
| | - Alessio Lombardi
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, Milan, Italy;
| | - Rocco Caliandro
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Bari, Italy;
| | - Maria Serena Fabbrini
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, Milan, Italy;
| |
Collapse
|