1
|
Mairaville C, Broyon M, Maurel M, Chentouf M, Chiavarina B, Turtoi A, Pirot N, Martineau P. Identification of monoclonal antibodies from naive antibody phage-display libraries for protein detection in formalin-fixed paraffin-embedded tissues. J Immunol Methods 2024; 532:113730. [PMID: 39059744 DOI: 10.1016/j.jim.2024.113730] [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: 04/08/2024] [Revised: 07/05/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Most antibodies used in immunohistochemistry (IHC) have been developed by animal immunization. We wanted to explore naive antibody repertoires displayed on filamentous phages as a source of full-length antibodies for IHC on Formalin-Fixed and Paraffin-Embedded (FFPE) tissues. We used two isogenic mouse fibroblast cell lines that express or not human HER2 to generate positive and negative FFPE pseudo-tissue respectively. Using these pseudo-tissues and previously described approaches based on differential panning, we isolated very efficient antibody clones, but not against HER2. To optimize HER2 targeting and tissue specificity, we first performed 3-4 rounds of in vitro panning using recombinant HER2 extracellular domain (ECD) to enrich the phage library in HER2 binders, followed by one panning round using the two FFPE pseudo-tissues to retain binders for IHC conditions. We then analyzed the bound phages using next-generation sequencing to identify antibody sequences specifically associated with the HER2-positive pseudo-tissue. Using this approach, the top-ranked clone identified by sequencing was specific to the HER2-positive pseudo-tissue and showed a staining pattern similar to that of the antibody used for the clinical diagnosis of HER2-positive breast cancer. However, we could not optimize staining on other tissues, showing that specificity was restricted to the tissue used for selection and screening. Therefore, future optimized protocols must consider tissue diversity early during the selection by panning using a wide collection of tissue types.
Collapse
Affiliation(s)
| | - Morgane Broyon
- BCM, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Margaux Maurel
- IRCM, Univ. Montpellier, ICM, INSERM, Montpellier, France
| | | | | | - Andrei Turtoi
- IRCM, Univ. Montpellier, ICM, INSERM, Montpellier, France
| | - Nelly Pirot
- IRCM, Univ. Montpellier, ICM, INSERM, Montpellier, France; BCM, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | | |
Collapse
|
2
|
Bidkar AP, Wang S, Bobba KN, Chan E, Bidlingmaier S, Egusa EA, Peter R, Ali U, Meher N, Wadhwa A, Dhrona S, Dasari C, Beckford-Vera D, Su Y, Tang R, Zhang L, He J, Wilson DM, Aggarwal R, VanBrocklin HF, Seo Y, Chou J, Liu B, Flavell RR. Treatment of Prostate Cancer with CD46-targeted 225Ac Alpha Particle Radioimmunotherapy. Clin Cancer Res 2023; 29:1916-1928. [PMID: 36917693 PMCID: PMC10183825 DOI: 10.1158/1078-0432.ccr-22-3291] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/19/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023]
Abstract
PURPOSE Radiopharmaceutical therapy is changing the standard of care in prostate cancer and other malignancies. We previously reported high CD46 expression in prostate cancer and developed an antibody-drug conjugate and immunoPET agent based on the YS5 antibody, which targets a tumor-selective CD46 epitope. Here, we present the preparation, preclinical efficacy, and toxicity evaluation of [225Ac]DOTA-YS5, a radioimmunotherapy agent based on the YS5 antibody. EXPERIMENTAL DESIGN [225Ac]DOTA-YS5 was developed, and its therapeutic efficiency was tested on cell-derived (22Rv1, DU145), and patient-derived (LTL-545, LTL484) prostate cancer xenograft models. Biodistribution studies were carried out on 22Rv1 tumor xenograft models to confirm the targeting efficacy. Toxicity analysis of the [225Ac]DOTA-YS5 was carried out on nu/nu mice to study short-term (acute) and long-term (chronic) toxicity. RESULTS Biodistribution study shows that [225Ac]DOTA-YS5 agent delivers high levels of radiation to the tumor tissue (11.64% ± 1.37%ID/g, 28.58% ± 10.88%ID/g, 29.35% ± 7.76%ID/g, and 31.78% ± 5.89%ID/g at 24, 96, 168, and 408 hours, respectively), compared with the healthy organs. [225Ac]DOTA-YS5 suppressed tumor size and prolonged survival in cell line-derived and patient-derived xenograft models. Toxicity analysis revealed that the 0.5 μCi activity levels showed toxicity to the kidneys, likely due to redistribution of daughter isotope 213Bi. CONCLUSIONS [225Ac]DOTA-YS5 suppressed the growth of cell-derived and patient-derived xenografts, including prostate-specific membrane antigen-positive and prostate-specific membrane antigen-deficient models. Overall, this preclinical study confirms that [225Ac]DOTA-YS5 is a highly effective treatment and suggests feasibility for clinical translation of CD46-targeted radioligand therapy in prostate cancer.
Collapse
Affiliation(s)
- Anil P. Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Sinan Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Kondapa Naidu Bobba
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Emily Chan
- Department of Pathology, University of California, San Francisco, California
| | - Scott Bidlingmaier
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Emily A. Egusa
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Robin Peter
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- Department of Nuclear Engineering, University of California, Berkeley, California
| | - Umama Ali
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Niranjan Meher
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Anju Wadhwa
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Suchi Dhrona
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Chandrashekhar Dasari
- Department of Surgery, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California
| | - Denis Beckford-Vera
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Ryan Tang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Li Zhang
- Department of Medicine and the Department of Epidemiology and Biostatistics, University of California, Berkeley, California
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia
| | - David M. Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Rahul Aggarwal
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Henry F. VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Jonathan Chou
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Robert R. Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California
| |
Collapse
|
3
|
Li J, Huang T, Hua J, Wang Q, Su Y, Chen P, Bidlingmaier S, Li A, Xie Z, Bidkar AP, Shen S, Shi W, Seo Y, Flavell RR, Gioeli D, Dreicer R, Li H, Liu B, He J. CD46 targeted 212Pb alpha particle radioimmunotherapy for prostate cancer treatment. J Exp Clin Cancer Res 2023; 42:61. [PMID: 36906664 PMCID: PMC10007843 DOI: 10.1186/s13046-023-02636-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/01/2023] [Indexed: 03/13/2023] Open
Abstract
We recently identified CD46 as a novel prostate cancer cell surface antigen that shows lineage independent expression in both adenocarcinoma and small cell neuroendocrine subtypes of metastatic castration resistant prostate cancer (mCRPC), discovered an internalizing human monoclonal antibody YS5 that binds to a tumor selective CD46 epitope, and developed a microtubule inhibitor-based antibody drug conjugate that is in a multi-center phase I trial for mCRPC (NCT03575819). Here we report the development of a novel CD46-targeted alpha therapy based on YS5. We conjugated 212Pb, an in vivo generator of alpha-emitting 212Bi and 212Po, to YS5 through the chelator TCMC to create the radioimmunoconjugate, 212Pb-TCMC-YS5. We characterized 212Pb-TCMC-YS5 in vitro and established a safe dose in vivo. We next studied therapeutic efficacy of a single dose of 212Pb-TCMC-YS5 using three prostate cancer small animal models: a subcutaneous mCRPC cell line-derived xenograft (CDX) model (subcu-CDX), an orthotopically grafted mCRPC CDX model (ortho-CDX), and a prostate cancer patient-derived xenograft model (PDX). In all three models, a single dose of 0.74 MBq (20 µCi) 212Pb-TCMC-YS5 was well tolerated and caused potent and sustained inhibition of established tumors, with significant increases of survival in treated animals. A lower dose (0.37 MBq or 10 µCi 212Pb-TCMC-YS5) was also studied on the PDX model, which also showed a significant effect on tumor growth inhibition and prolongation of animal survival. These results demonstrate that 212Pb-TCMC-YS5 has an excellent therapeutic window in preclinical models including PDXs, opening a direct path for clinical translation of this novel CD46-targeted alpha radioimmunotherapy for mCRPC treatment.
Collapse
Affiliation(s)
- Jun Li
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Guangdong, 518036, China
| | - Tao Huang
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
| | - Jun Hua
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Nuclear Medicine, Chongqing Cancer Hospital, Chongqing University, Chongqing, China
| | - Qiong Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, University of Virginia, Charlottesville, VA, 22903, USA
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, CA, 94110, USA
| | - Ping Chen
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Guangdong, 518036, China
| | - Scott Bidlingmaier
- Department of Anesthesia, University of California, San Francisco, CA, 94110, USA
| | - Allan Li
- Department of Anesthesia, University of California, San Francisco, CA, 94110, USA
| | - Zhongqiu Xie
- Department of Pathology, University of Virginia, Charlottesville, VA, 22903, USA
| | - Anil P Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94110, USA
| | - Sui Shen
- Department of Radiation Oncology, University of Alabama, Birmingham, AL, 35233, USA
| | - Weibin Shi
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94110, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94110, USA
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94110, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94110, USA
| | - Daniel Gioeli
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22903, USA
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22903, USA
| | - Robert Dreicer
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Medicine, University of Virginia, Charlottesville, VA, 22903, USA
| | - Hui Li
- Department of Pathology, University of Virginia, Charlottesville, VA, 22903, USA
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22903, USA
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, CA, 94110, USA.
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94110, USA.
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA.
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22903, USA.
| |
Collapse
|
4
|
Condelipes PGM, Fontes PM, Godinho-Santos A, Brás EJS, Marques V, Afonso MB, Rodrigues CMP, Chu V, Gonçalves J, Conde JP. Towards personalized antibody cancer therapy: development of a microfluidic cell culture device for antibody selection. LAB ON A CHIP 2022; 22:4717-4728. [PMID: 36349999 DOI: 10.1039/d2lc00918h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Antibody therapy has been one of the most successful therapies for a wide range of diseases, including cancer. One way of expediting antibody therapy development is through phage display technology. Here, by screening thousands of randomly assembled peptide sequences, it is possible to identify potential therapeutic candidates. Conventional screening technologies do not accommodate perfusion through the system, as is the case of standard plate-based cultures. This leads to a poor translation of the experimental results obtained in vitro when moving to a more physiologically relevant setting, such as the case of preclinical animal models or clinical trials. Microfluidics is a technology that can improve screening efficacy by replicating more physiologically relevant conditions such as shear stress. In this work, a polydimethylsiloxane/polystyrene-based microfluidic system for a continuously perfused culture of cancer cells is reported. Human colorectal adenocarcinoma cells (HCT116) expressing CXCR4 were used as a cell target. Fluorescently labeled M13 phages anti-CXCR4 were used to study the efficiency of the microfluidic system as a tool to study the binding kinetics of the engineered bacteriophages. Using our microfluidic platform, we estimated a dissociation constant of 0.45 pM for the engineered phage. Additionally, a receptor internalization assay was developed using SDF-1α to verify phage specificity to the CXCR4 receptor. Upon receptor internalization there was a signal reduction, proving that the anti-CXCR4 fluorescently labelled M13 phages bound specifically to the CXCR4 receptor. The simplicity and ease of use of the microfluidic device design presented in this work can form the basis of a generic platform that facilitates the study and optimization of therapies based on interaction with biological entities such as mammalian cells.
Collapse
Affiliation(s)
- Pedro G M Condelipes
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| | - Pedro Mendes Fontes
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Godinho-Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Eduardo J S Brás
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
- IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Vanda Marques
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Marta B Afonso
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Virginia Chu
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
| | - João Gonçalves
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - João Pedro Conde
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
5
|
Hyrenius-Wittsten A, Su Y, Park M, Garcia JM, Alavi J, Perry N, Montgomery G, Liu B, Roybal KT. SynNotch CAR circuits enhance solid tumor recognition and promote persistent antitumor activity in mouse models. Sci Transl Med 2021; 13:eabd8836. [PMID: 33910981 PMCID: PMC8594452 DOI: 10.1126/scitranslmed.abd8836] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 01/11/2021] [Accepted: 03/16/2021] [Indexed: 12/21/2022]
Abstract
The first clinically approved engineered chimeric antigen receptor (CAR) T cell therapies are remarkably effective in a subset of hematological malignancies with few therapeutic options. Although these clinical successes have been exciting, CAR T cells have hit roadblocks in solid tumors that include the lack of highly tumor-specific antigens to target, opening up the possibility of life-threatening "on-target/off-tumor" toxicities, and problems with T cell entry into solid tumor and persistent activity in suppressive tumor microenvironments. Here, we improve the specificity and persistent antitumor activity of therapeutic T cells with synthetic Notch (synNotch) CAR circuits. We identify alkaline phosphatase placental-like 2 (ALPPL2) as a tumor-specific antigen expressed in a spectrum of solid tumors, including mesothelioma and ovarian cancer. ALPPL2 can act as a sole target for CAR therapy or be combined with tumor-associated antigens such as melanoma cell adhesion molecule (MCAM), mesothelin, or human epidermal growth factor receptor 2 (HER2) in synNotch CAR combinatorial antigen circuits. SynNotch CAR T cells display superior control of tumor burden when compared to T cells constitutively expressing a CAR targeting the same antigens in mouse models of human mesothelioma and ovarian cancer. This was achieved by preventing CAR-mediated tonic signaling through synNotch-controlled expression, allowing T cells to maintain a long-lived memory and non-exhausted phenotype. Collectively, we establish ALPPL2 as a clinically viable cell therapy target for multiple solid tumors and demonstrate the multifaceted therapeutic benefits of synNotch CAR T cells.
Collapse
Affiliation(s)
- Axel Hyrenius-Wittsten
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Minhee Park
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Julie M Garcia
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Josef Alavi
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nathaniel Perry
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Garrett Montgomery
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bin Liu
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA.
- Department of Anesthesia, University of California, San Francisco, San Francisco, CA 94110, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kole T Roybal
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Gladstone-UCSF Institute for Genomic Immunology, San Francisco, CA 94158, USA
- UCSF Cell Design Institute, San Francisco, CA 94158, USA
| |
Collapse
|
6
|
Antibody Identification for Antigen Detection in Formalin-Fixed Paraffin-Embedded Tissue Using Phage Display and Naïve Libraries. Antibodies (Basel) 2021; 10:antib10010004. [PMID: 33466676 PMCID: PMC7839037 DOI: 10.3390/antib10010004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/15/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Immunohistochemistry is a widely used technique for research and diagnostic purposes that relies on the recognition by antibodies of antigens expressed in tissues. However, tissue processing and particularly formalin fixation affect the conformation of these antigens through the formation of methylene bridges. Although antigen retrieval techniques can partially restore antigen immunoreactivity, it is difficult to identify antibodies that can recognize their target especially in formalin-fixed paraffin-embedded tissues. Most of the antibodies currently used in immunohistochemistry have been obtained by animal immunization; however, in vitro display techniques represent alternative strategies that have not been fully explored yet. This review provides an overview of phage display-based antibody selections using naïve antibody libraries on various supports (fixed cells, dissociated tissues, tissue fragments, and tissue sections) that have led to the identification of antibodies suitable for immunohistochemistry.
Collapse
|
7
|
Zhou T, Wang J. Laser Capture Microdissection of Vascular Endothelial Cells from Frozen Heart Tissues. Methods Mol Biol 2021; 2319:105-110. [PMID: 34331248 DOI: 10.1007/978-1-0716-1480-8_12] [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] [Indexed: 06/13/2023]
Abstract
Laser capture microdissection (LCM) enables researchers to selectively evaluate gene expression profiling of a specific cell type within a tissue. Vascular endothelial cells (EC) line the inside of vessel lumen and play an essential role in new blood vessel formation. It remains a challenge to determine vascular ECs-specific genes expression in vivo. Here, we described a method to dissect vascular ECs from the frozen heart tissue by LCM. The total RNA or proteins are then extracted from the ECs for further analysis.
Collapse
Affiliation(s)
- Tianhao Zhou
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA.
| | - Jian Wang
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| |
Collapse
|
8
|
Wang S, Li J, Hua J, Su Y, Beckford-Vera DR, Zhao W, Jayaraman M, Huynh TL, Zhao N, Wang YH, Huang Y, Qin F, Shen S, Gioeli D, Dreicer R, Sriram R, Egusa EA, Chou J, Feng FY, Aggarwal R, Evans MJ, Seo Y, Liu B, Flavell RR, He J. Molecular Imaging of Prostate Cancer Targeting CD46 Using ImmunoPET. Clin Cancer Res 2020; 27:1305-1315. [PMID: 33293372 DOI: 10.1158/1078-0432.ccr-20-3310] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/19/2020] [Accepted: 12/03/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE We recently identified CD46 as a novel therapeutic target in prostate cancer. In this study, we developed a CD46-targeted PET radiopharmaceutical, [89Zr]DFO-YS5, and evaluated its performance for immunoPET imaging in murine prostate cancer models. EXPERIMENTAL DESIGN [89Zr]DFO-YS5 was prepared and its in vitro binding affinity for CD46 was measured. ImmunoPET imaging was conducted in male athymic nu/nu mice bearing DU145 [AR-, CD46+, prostate-specific membrane antigen-negative (PSMA-)] or 22Rv1 (AR+, CD46+, PSMA+) tumors, and in NOD/SCID gamma mice bearing patient-derived adenocarcinoma xenograft, LTL-331, and neuroendocrine prostate cancers, LTL-331R and LTL-545. RESULTS [89Zr]DFO-YS5 binds specifically to the CD46-positive human prostate cancer DU145 and 22Rv1 xenografts. In biodistribution studies, the tumor uptake of [89Zr]DFO-YS5 was 13.3 ± 3.9 and 11.2 ± 2.5 %ID/g, respectively, in DU145 and 22Rv1 xenografts, 4 days postinjection. Notably, [89Zr]DFO-YS5 demonstrated specific uptake in the PSMA- and AR-negative DU145 model. [89Zr]DFO-YS5 also showed uptake in the patient-derived LTL-331 and -331R models, with particularly high uptake in the LTL-545 neuroendocrine prostate cancer tumors (18.8 ± 5.3, 12.5 ± 1.8, and 32 ± 5.3 %ID/g in LTL-331, LTL-331R, and LTL-545, respectively, at 4 days postinjection). CONCLUSIONS [89Zr]DFO-YS5 is an excellent PET imaging agent across a panel of prostate cancer models, including in both adenocarcinoma and neuroendocrine prostate cancer, both cell line- and patient-derived xenografts, and both PSMA-positive and -negative tumors. It demonstrates potential for clinical translation as an imaging agent, theranostic platform, and companion biomarker in prostate cancer.
Collapse
Affiliation(s)
- Sinan Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Jun Li
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia.,Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Jun Hua
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia.,Department of Nuclear Medicine, Chongqing University Cancer Hospital, Chongqing, P.R. China
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Denis R Beckford-Vera
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Walter Zhao
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Mayuri Jayaraman
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Tony L Huynh
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Ning Zhao
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Yung-Hua Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Yangjie Huang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Fujun Qin
- Department of Pathology, University of Virginia, Charlottesville, Virginia
| | - Sui Shen
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daniel Gioeli
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia.,University of Virginia Cancer Center, Charlottesville, Virginia
| | - Robert Dreicer
- University of Virginia Cancer Center, Charlottesville, Virginia.,Departments of Medicine and Urology, University of Virginia, Charlottesville, Virginia
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Emily A Egusa
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Jonathan Chou
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Felix Y Feng
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Rahul Aggarwal
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Division of Hematology and Oncology, University of California, San Francisco, San Francisco, California
| | - Michael J Evans
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California.,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California.,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, San Francisco, California. .,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California. .,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia. .,University of Virginia Cancer Center, Charlottesville, Virginia
| |
Collapse
|
9
|
Sherbenou DW, Su Y, Behrens CR, Aftab BT, Perez de Acha O, Murnane M, Bearrows SC, Hann BC, Wolf JL, Martin TG, Liu B. Potent Activity of an Anti-ICAM1 Antibody-Drug Conjugate against Multiple Myeloma. Clin Cancer Res 2020; 26:6028-6038. [PMID: 32917735 PMCID: PMC7669584 DOI: 10.1158/1078-0432.ccr-20-0400] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/15/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE New therapies have changed the outlook for patients with multiple myeloma, but novel agents are needed for patients who are refractory or relapsed on currently approved drug classes. Novel targets other than CD38 and BCMA are needed for new immunotherapy development, as resistance to daratumumab and emerging anti-BCMA approaches appears inevitable. One potential target of interest in myeloma is ICAM1. Naked anti-ICAM1 antibodies were active in preclinical models of myeloma and safe in patients, but showed limited clinical efficacy. Here, we sought to achieve improved targeting of multiple myeloma with an anti-ICAM1 antibody-drug conjugate (ADC). EXPERIMENTAL DESIGN Our anti-ICAM1 human mAb was conjugated to an auristatin derivative, and tested against multiple myeloma cell lines in vitro, orthotopic xenografts in vivo, and patient samples ex vivo. The expression of ICAM1 was also measured by quantitative flow cytometry in patients spanning from diagnosis to the daratumumab-refractory state. RESULTS The anti-ICAM1 ADC displayed potent anti-myeloma cytotoxicity in vitro and in vivo. In addition, we have verified that ICAM1 is highly expressed on myeloma cells and shown that its expression is further accentuated by the presence of bone marrow microenvironmental factors. In primary samples, ICAM1 is differentially overexpressed on multiple myeloma cells compared with normal cells, including daratumumab-refractory patients with decreased CD38. In addition, ICAM1-ADC showed selective cytotoxicity in multiple myeloma primary samples. CONCLUSIONS We propose that anti-ICAM1 ADC should be further studied for toxicity, and if safe, tested for clinical efficacy in patients with relapsed or refractory multiple myeloma.
Collapse
Affiliation(s)
- Daniel W Sherbenou
- Department of Medicine, University of California at San Francisco, California
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Yang Su
- Department of Anesthesia, University of California at San Francisco, California
| | | | - Blake T Aftab
- Department of Medicine, University of California at San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Olivia Perez de Acha
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Megan Murnane
- Department of Medicine, University of California at San Francisco, California
| | - Shelby C Bearrows
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Byron C Hann
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Jeffery L Wolf
- Department of Medicine, University of California at San Francisco, California
| | - Thomas G Martin
- Department of Medicine, University of California at San Francisco, California
| | - Bin Liu
- Department of Anesthesia, University of California at San Francisco, California.
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| |
Collapse
|
10
|
Su Y, Zhang X, Bidlingmaier S, Behrens CR, Lee NK, Liu B. ALPPL2 Is a Highly Specific and Targetable Tumor Cell Surface Antigen. Cancer Res 2020; 80:4552-4564. [PMID: 32868383 PMCID: PMC7572689 DOI: 10.1158/0008-5472.can-20-1418] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/29/2020] [Accepted: 08/19/2020] [Indexed: 11/16/2022]
Abstract
Identification of tumor-specific cell surface antigens has proven challenging, as the vast majority of tumor-associated antigens are also expressed in normal tissues. In mesothelioma, we identified a highly specific tumor cell surface antigen that can be targeted for therapy development. Mesothelioma is caused by malignant transformation of the mesothelium, is incurable, and can be categorized into three histologic subtypes: epithelioid, biphasic, and sarcomatoid. To identity novel mesothelioma cell surface antigens with broad subtype coverage and high tissue specificity, we have previously selected phage antibody display libraries on live mesothelioma cells and tissues following counterselection on normal cells and identified a panel of human antibodies that bind all subtypes of mesothelioma, but not normal mesothelium. One of the antibodies, M25, showed high specificity against an antigen we identify here as ALPPL2. IHC on normal human tissues found that ALPPL2 is expressed only on placental trophoblasts, but not on any other normal tissues. This significant tissue specificity and broad tumor type coverage suggest that ALPPL2 could be an excellent cell surface target for therapeutic development against mesothelioma. To evaluate therapeutic potential of ALPPL2 targeting, an ALPPL2-targeted antibody-drug conjugate was developed and demonstrated potent and specific tumor killing in vitro and in vivo against both epithelioid and sarcomatoid mesothelioma. Thus, ALPPL2 belongs to a rare class of cell surface antigens classified as truly tumor specific and is well suited for therapy development against ALPPL2-expressing tumors. SIGNIFICANCE: These findings identify ALPP2 as a true tumor-specific cell surface antigen whose tissue specificity enables the development of novel therapies.
Collapse
Affiliation(s)
- Yang Su
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Xin Zhang
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Scott Bidlingmaier
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Christopher R Behrens
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Nam-Kyung Lee
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, San Francisco, California.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| |
Collapse
|
11
|
He J, Feng J, Su Y, Seo Y, Liu B. Quantitative 99mTc Labeling Kit for HYNIC-Conjugated Single Chain Antibody Fragments Targeting Malignant Mesothelioma. Bioconjug Chem 2020; 31:1750-1755. [PMID: 32628464 DOI: 10.1021/acs.bioconjchem.0c00319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Single chain antibody fragment (scFv) is a promising agent for imaging and targeted therapy. The objective of the study is to evaluate a kit formulation for 99mTc labeling of scFv for tumor imaging. The scFv was engineered to contain a cysteine tag to accommodate the specific conjugation of HYNIC and subsequent 99mTc labeling. The labeling conditions were formulated to allow instantaneous one-pot quantitative labeling. The reproducibility of labeling was evaluated at various time points during kit storage at -20 °C. In vitro cell binding experiments and HPLC analysis were performed to assess binding affinity and radiolabel stability, respectively. In vivo tumor targeting study was performed in xenograft models with biodistribution studied at 1, 3, and 24 h post-injection. The optimized kit with 5 μg SnF2, pH 5.5, and 50 μg GH along with as low as 15 μg of HYNIC-cys-scFv provided high labeling yield (>95%), high specific activity (1.8 × 107 Ci/Mol), and robust reproducibility with shelf life up to 90 days when stored at -20 °C. The in vitro cell binding study showed the labeled scFv maintained the binding capability with an apparent KD of ∼27 nM. The animal study using tumor-bearing mice showed high tumor uptake at 16.9%ID/g 24 h post-injection along with rapid blood clearance (0.18%ID/g) and kidney excretion (44%ID/g), resulting in very high contrast (tumor/muscle >200:1). A kit formulation for 99mTc labeling of scFvs targeting mesothelioma was developed based on specific HYNIC conjugation and GH (Glucoheptonate) as a coligand, producing not only high specific activity, but also improved tumor uptake. This convenient one-pot labeling method has the potential for translation into clinical use and is applicable to other scFvs as well.
Collapse
Affiliation(s)
- Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia 22908, United States.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
| | - Jinjin Feng
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, California 94143, United States
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States.,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California 94143, United States
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, California 94143, United States.,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California 94143, United States
| |
Collapse
|
12
|
Xu H, Cao B, Li Y, Mao C. Phage nanofibers in nanomedicine: Biopanning for early diagnosis, targeted therapy, and proteomics analysis. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1623. [PMID: 32147974 DOI: 10.1002/wnan.1623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/02/2020] [Accepted: 02/04/2020] [Indexed: 12/16/2022]
Abstract
Display of a peptide or protein of interest on the filamentous phage (also known as bacteriophage), a biological nanofiber, has opened a new route for disease diagnosis and therapy as well as proteomics. Earlier phage display was widely used in protein-protein or antigen-antibody studies. In recent years, its application in nanomedicine is becoming increasingly popular and encouraging. We aim to review the current status in this research direction. For better understanding, we start with a brief introduction of basic biology and structure of the filamentous phage. We present the principle of phage display and library construction method on the basis of the filamentous phage. We summarize the use of the phage displayed peptide library for selecting peptides with high affinity against cells or tissues. We then review the recent applications of the selected cell or tissue targeting peptides in developing new targeting probes and therapeutics to advance the early diagnosis and targeted therapy of different diseases in nanomedicine. We also discuss the integration of antibody phage display and modern proteomics in discovering new biomarkers or target proteins for disease diagnosis and therapy. Finally, we propose an outlook for further advancing the potential impact of phage display on future nanomedicine. This article is categorized under: Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Hong Xu
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Binrui Cao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Yan Li
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
13
|
Wilson HD, Li X, Peng H, Rader C. A Sortase A Programmable Phage Display Format for Improved Panning of Fab Antibody Libraries. J Mol Biol 2018; 430:4387-4400. [PMID: 30213726 DOI: 10.1016/j.jmb.2018.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 09/01/2018] [Accepted: 09/05/2018] [Indexed: 11/26/2022]
Abstract
Phage display of combinatorial antibody libraries is a versatile tool in the field of antibody engineering, with diverse applications including monoclonal antibody (mAb) discovery, affinity maturation, and humanization. To improve the selection efficiency of antibody libraries, we developed a new phagemid display system that addresses the complication of bald phage propagation. The phagemid facilitates the biotinylation of fragment of antigen binding (Fab) antibody fragments displayed on phage via Sortase A catalysis and the subsequent enrichment of Fab-displaying phage during selections. In multiple contexts, this selection approach improved the enrichment of target-reactive mAbs by depleting background phage. Panels of cancer cell line-reactive mAbs with high diversity and specificity were isolated from a naïve chimeric rabbit/human Fab library using this approach, highlighting its potential to accelerate antibody engineering efforts and to empower concerted antibody drug and target discovery.
Collapse
Affiliation(s)
- Henry D Wilson
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Xiuling Li
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Haiyong Peng
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Christoph Rader
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
14
|
Su Y, Liu Y, Behrens CR, Bidlingmaier S, Lee NK, Aggarwal R, Sherbenou DW, Burlingame AL, Hann BC, Simko JP, Premasekharan G, Paris PL, Shuman MA, Seo Y, Small EJ, Liu B. Targeting CD46 for both adenocarcinoma and neuroendocrine prostate cancer. JCI Insight 2018; 3:121497. [PMID: 30185663 DOI: 10.1172/jci.insight.121497] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/24/2018] [Indexed: 12/25/2022] Open
Abstract
Although initially responsive to androgen signaling inhibitors (ASIs), metastatic castration-resistant prostate cancer (mCRPC) inevitably develops and is incurable. In addition to adenocarcinoma (adeno), neuroendocrine prostate cancer (NEPC) emerges to confer ASI resistance. We have previously combined laser capture microdissection and phage antibody display library selection on human cancer specimens and identified novel internalizing antibodies binding to tumor cells residing in their tissue microenvironment. We identified the target antigen for one of these antibodies as CD46, a multifunctional protein that is best known for negatively regulating the innate immune system. CD46 is overexpressed in primary tumor tissue and CRPC (localized and metastatic; adeno and NEPC), but expressed at low levels on normal tissues except for placental trophoblasts and prostate epithelium. Abiraterone- and enzalutamide-treated mCRPC cells upregulate cell surface CD46 expression. Genomic analysis showed that the CD46 gene is gained in 45% abiraterone-resistant mCRPC patients. We conjugated a tubulin inhibitor to our macropinocytosing anti-CD46 antibody and showed that the resulting antibody-drug conjugate (ADC) potently and selectively kills both adeno and NEPC cell lines in vitro (sub-nM EC50) but not normal cells. CD46 ADC regressed and eliminated an mCRPC cell line xenograft in vivo in both subcutaneous and intrafemoral models. Exploratory toxicology studies of the CD46 ADC in non-human primates demonstrated an acceptable safety profile. Thus, CD46 is an excellent target for antibody-based therapy development, which has potential to be applicable to both adenocarcinoma and neuroendocrine types of mCRPC that are resistant to current treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Rahul Aggarwal
- Department of Medicine.,Helen Diller Family Comprehensive Cancer Center
| | | | | | | | - Jeffry P Simko
- Helen Diller Family Comprehensive Cancer Center.,Department of Pathology
| | | | - Pamela L Paris
- Helen Diller Family Comprehensive Cancer Center.,Department of Urology, and
| | | | - Youngho Seo
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
| | - Eric J Small
- Department of Medicine.,Helen Diller Family Comprehensive Cancer Center.,Department of Urology, and
| | - Bin Liu
- Department of Anesthesia.,Helen Diller Family Comprehensive Cancer Center
| |
Collapse
|
15
|
Lee NK, Bidlingmaier S, Su Y, Liu B. Modular Construction of Large Non-Immune Human Antibody Phage-Display Libraries from Variable Heavy and Light Chain Gene Cassettes. Methods Mol Biol 2018; 1701:61-82. [PMID: 29116500 DOI: 10.1007/978-1-4939-7447-4_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Monoclonal antibodies and antibody-derived therapeutics have emerged as a rapidly growing class of biological drugs for the treatment of cancer, autoimmunity, infection, and neurological diseases. To support the development of human antibodies, various display techniques based on antibody gene repertoires have been constructed over the last two decades. In particular, scFv-antibody phage display has been extensively utilized to select lead antibodies against a variety of target antigens. To construct a scFv phage display that enables efficient antibody discovery, and optimization, it is desirable to develop a system that allows modular assembly of highly diverse variable heavy chain and light chain (Vκ and Vλ) repertoires. Here, we describe modular construction of large non-immune human antibody phage-display libraries built on variable gene cassettes from heavy chain and light chain repertoires (Vκ- and Vλ-light can be made into independent cassettes). We describe utility of such libraries in antibody discovery and optimization through chain shuffling.
Collapse
Affiliation(s)
- Nam-Kyung Lee
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, 1001 Potrero Avenue, 1305, San Francisco, CA, 94110, USA
| | - Scott Bidlingmaier
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, 1001 Potrero Avenue, 1305, San Francisco, CA, 94110, USA
| | - Yang Su
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, 1001 Potrero Avenue, 1305, San Francisco, CA, 94110, USA
| | - Bin Liu
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, 1001 Potrero Avenue, 1305, San Francisco, CA, 94110, USA.
| |
Collapse
|
16
|
Giansanti F, Flavell DJ, Angelucci F, Fabbrini MS, Ippoliti R. Strategies to Improve the Clinical Utility of Saporin-Based Targeted Toxins. Toxins (Basel) 2018; 10:toxins10020082. [PMID: 29438358 PMCID: PMC5848183 DOI: 10.3390/toxins10020082] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/29/2018] [Accepted: 02/11/2018] [Indexed: 02/06/2023] Open
Abstract
Plant Ribosome-inactivating proteins (RIPs) including the type I RIP Saporin have been used for the construction of Immunotoxins (ITxs) obtained via chemical conjugation of the toxic domain to whole antibodies or by generating genetic fusions to antibody fragments/targeting domains able to direct the chimeric toxin against a desired sub-population of cancer cells. The high enzymatic activity, stability and resistance to conjugation procedures and especially the possibility to express recombinant fusions in yeast, make Saporin a well-suited tool for anti-cancer therapy approaches. Previous clinical work on RIPs-based Immunotoxins (including Saporin) has shown that several critical issues must be taken into deeper consideration to fully exploit their therapeutic potential. This review focuses on possible combinatorial strategies (chemical and genetic) to augment Saporin-targeted toxin efficacy. Combinatorial approaches may facilitate RIP escape into the cytosolic compartment (where target ribosomes are), while genetic manipulations may minimize potential adverse effects such as vascular-leak syndrome or may identify T/B cell epitopes in order to decrease the immunogenicity following similar strategies as those used in the case of bacterial toxins such as Pseudomonas Exotoxin A or as for Type I RIP Bouganin. This review will further focus on strategies to improve recombinant production of Saporin-based chimeric toxins.
Collapse
Affiliation(s)
- Francesco Giansanti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, I-67100 L'Aquila, Italy.
| | - David J Flavell
- The Simon Flavell Leukaemia Research Laboratory (Leukaemia Busters), Southampton General Hospital, Southampton, SO16 8AT, UK.
| | - Francesco Angelucci
- Department of Life, Health and Environmental Sciences, University of L'Aquila, I-67100 L'Aquila, Italy.
| | | | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, I-67100 L'Aquila, Italy.
| |
Collapse
|
17
|
Lee NK, Zhang Y, Su Y, Bidlingmaier S, Sherbenou DW, Ha KD, Liu B. Cell-type specific potent Wnt signaling blockade by bispecific antibody. Sci Rep 2018; 8:766. [PMID: 29335534 PMCID: PMC5768681 DOI: 10.1038/s41598-017-17539-z] [Citation(s) in RCA: 9] [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: 08/14/2017] [Accepted: 11/13/2017] [Indexed: 01/07/2023] Open
Abstract
Cell signaling pathways are often shared between normal and diseased cells. How to achieve cell type-specific, potent inhibition of signaling pathways is a major challenge with implications for therapeutic development. Using the Wnt/β-catenin signaling pathway as a model system, we report here a novel and generally applicable method to achieve cell type-selective signaling blockade. We constructed a bispecific antibody targeting the Wnt co-receptor LRP6 (the effector antigen) and a cell type-associated antigen (the guide antigen) that provides the targeting specificity. We found that the bispecific antibody inhibits Wnt-induced reporter activities with over one hundred-fold enhancement in potency, and in a cell type-selective manner. Potency enhancement is dependent on the expression level of the guide antigen on the target cell surface and the apparent affinity of the anti-guide antibody. Both internalizing and non-internalizing guide antigens can be used, with internalizing bispecific antibody being able to block signaling by all ligands binding to the target receptor due to its removal from the cell surface. It is thus feasible to develop bispecific-based therapeutic strategies that potently and selectively inhibit signaling pathways in a cell type-selective manner, creating opportunity for therapeutic targeting.
Collapse
Affiliation(s)
- Nam-Kyung Lee
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, 1001 Potrero Ave., 1305, San Francisco, CA, 94110-1305, USA
| | - Yafeng Zhang
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, 1001 Potrero Ave., 1305, San Francisco, CA, 94110-1305, USA
| | - Yang Su
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, 1001 Potrero Ave., 1305, San Francisco, CA, 94110-1305, USA
| | - Scott Bidlingmaier
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, 1001 Potrero Ave., 1305, San Francisco, CA, 94110-1305, USA
| | - Daniel W Sherbenou
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, 1001 Potrero Ave., 1305, San Francisco, CA, 94110-1305, USA
| | - Kevin D Ha
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, 1001 Potrero Ave., 1305, San Francisco, CA, 94110-1305, USA
| | - Bin Liu
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, 1001 Potrero Ave., 1305, San Francisco, CA, 94110-1305, USA.
| |
Collapse
|
18
|
Combine Phage Antibody Display Library Selection on Patient Tissue Specimens with Laser Capture Microdissection to Identify Novel Human Antibodies Targeting Clinically Relevant Tumor Antigens. Methods Mol Biol 2018; 1701:331-347. [PMID: 29116514 DOI: 10.1007/978-1-4939-7447-4_18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A functional approach to generate tumor-targeting human monoclonal antibodies is through selection of phage antibody display libraries directly on tumor cells. Although technically convenient, the use of cancer cell lines for the selection has limitations as those cell lines often undergo genetic and epigenetic changes during prolonged in vitro culture and alter their cell surface antigen expression profile. The key is to develop a technology that allows selection of phage antibody display libraries on tumor cells in situ residing in their natural tissue microenvironment. Laser capture microdissection (LCM) permits the precise procurement of tumor cells from human cancer patient tissue sections. Here, we describe a LCM-based method for selecting phage antibodies against tumor cells in situ using both fresh frozen and paraffin-embedded tissues. To restrict the selection to antibodies that bind internalizing epitopes, the method utilizes a polyclonal phage population pre-enriched for internalizing phage antibodies. The ability to recognize tumor cells in situ residing in their natural tissue microenvironment and to deliver payload intracellularly makes these LCM-selected antibodies attractive candidates for the development of targeted cancer therapeutics.
Collapse
|
19
|
Lykkemark S, Mandrup OA, Jensen MB, Just J, Kristensen P. A novel excision selection method for isolation of antibodies binding antigens expressed specifically by rare cells in tissue sections. Nucleic Acids Res 2017; 45:e107. [PMID: 28369551 PMCID: PMC5499801 DOI: 10.1093/nar/gkx207] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 03/21/2017] [Indexed: 12/22/2022] Open
Abstract
There is a growing appreciation of single cell technologies to provide increased biological insight and allow development of improved therapeutics. The central dogma explains why single cell technologies is further advanced in studies targeting nucleic acids compared to proteins, as nucleic acid amplification makes experimental detection possible. Here we describe a novel method for single round phage display selection of antibody fragments from genetic libraries targeting antigens expressed by rare cells in tissue sections. We present and discuss the results of two selections of antibodies recognizing antigens expressed by perivascular cells surrounding capillaries located in a human brain section; with the aim of identifying biomarkers expressed by pericytes. The area targeted for selection was identified by a known biomarker and morphological appearance, however in situ hybridizations to nucleic acids can also be used for the identification of target cells. The antibody selections were performed directly on the tissue sections followed by excision of the target cells using a glass capillary attached to micromanipulation equipment. Antibodies bound to the target cells were characterized using ELISA, immunocytochemistry and immunohistochemistry. The described method will provide a valuable tool for the discovery of novel biomarkers on rare cells in all types of tissues.
Collapse
Affiliation(s)
- Simon Lykkemark
- Department of Clinical Medicine, Aarhus University, Nørrebrogade 44, 8000 Aarhus C, Denmark.,Sino-Danish Centre for Education and Research (SDC), Niels Jensens Vej 2, 8000 Aarhus C, Denmark
| | - Ole Aalund Mandrup
- Department of Engineering, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus C, Denmark
| | - Mads Bjørnkjær Jensen
- Department of Engineering, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus C, Denmark
| | - Jesper Just
- Department of Molecular Biology and Genetics, Gustav Wieds Vej 10, 8000 Aarhus C, Denmark
| | - Peter Kristensen
- Department of Engineering, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus C, Denmark
| |
Collapse
|
20
|
Petrenko V, Gillespie J. Paradigm shift in bacteriophage-mediated delivery of anticancer drugs: from targeted 'magic bullets' to self-navigated 'magic missiles'. Expert Opin Drug Deliv 2017; 14:373-384. [PMID: 27466706 PMCID: PMC5544533 DOI: 10.1080/17425247.2016.1218463] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION New phage-directed nanomedicines have emerged recently as a result of the in-depth study of the genetics and structure of filamentous phage and evolution of phage display and phage nanobiotechnology. This review focuses on the progress made in the development of the cancer-targeted nanomaterials and discusses the trends in using phage as a bioselectable molecular navigation system. Areas covered: The merging of phage display technologies with nanotechnology in recent years has proved promising in different areas of medicine and technology, such as medical diagnostics, molecular imaging, vaccine development and targeted drug/gene delivery, which is the focus of this review. The authors used data obtained from their research group and sourced using Science Citation Index (Web of Science) and NCBI PubMed search resources. Expert opinion: First attempts of adapting traditional concepts of direct targeting of tumor using phage-targeted nanomedicines has shown minimal improvements. With discovery and study of biological and technical barriers that prevent anti-tumor drug delivery, a paradigm shift from traditional drug targeting to nanomedicine navigation systems is required. The advanced bacteriophage-driven self-navigation systems are thought to overcome those barriers using more precise, localized phage selection methods, multi-targeting 'promiscuous' ligands and advanced multifunctional nanomedicine platforms.
Collapse
Affiliation(s)
- V.A. Petrenko
- Department of Pathobiology, Auburn University, AL 36849, USA
| | - J.W. Gillespie
- Department of Pathobiology, Auburn University, AL 36849, USA
| |
Collapse
|
21
|
Ha KD, Bidlingmaier SM, Su Y, Lee NK, Liu B. Identification of Novel Macropinocytosing Human Antibodies by Phage Display and High-Content Analysis. Methods Enzymol 2017; 585:91-110. [PMID: 28109445 PMCID: PMC8671048 DOI: 10.1016/bs.mie.2016.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Internalizing antibodies have great potential for the development of targeted therapeutics. Antibodies that internalize via the macropinocytosis pathway are particularly promising since macropinocytosis is capable of mediating rapid, bulk uptake and is selectively upregulated in many cancers. We hereby describe a method for identifying antibodies that internalize via macropinocytosis by screening phage-displayed single-chain antibody selection outputs with an automated fluorescent microscopy-based high-content analysis platform. Furthermore, this method can be similarly applied to other endocytic pathways if other fluorescent, pathway-specific, soluble markers are available.
Collapse
Affiliation(s)
| | | | | | | | - Bin Liu
- Corresponding author Department of Anesthesia, University of California at San Francisco, 1001 Potrero Ave., Box 1305, San Francisco, CA 94110-1305,
| |
Collapse
|
22
|
Sherbenou DW, Aftab BT, Su Y, Behrens CR, Wiita A, Logan AC, Acosta-Alvear D, Hann BC, Walter P, Shuman MA, Wu X, Atkinson JP, Wolf JL, Martin TG, Liu B. Antibody-drug conjugate targeting CD46 eliminates multiple myeloma cells. J Clin Invest 2016; 126:4640-4653. [PMID: 27841764 DOI: 10.1172/jci85856] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 10/06/2016] [Indexed: 12/21/2022] Open
Abstract
Multiple myeloma is incurable by standard approaches because of inevitable relapse and development of treatment resistance in all patients. In our prior work, we identified a panel of macropinocytosing human monoclonal antibodies against CD46, a negative regulator of the innate immune system, and constructed antibody-drug conjugates (ADCs). In this report, we show that an anti-CD46 ADC (CD46-ADC) potently inhibited proliferation in myeloma cell lines with little effect on normal cells. CD46-ADC also potently eliminated myeloma growth in orthometastatic xenograft models. In primary myeloma cells derived from bone marrow aspirates, CD46-ADC induced apoptosis and cell death, but did not affect the viability of nontumor mononuclear cells. It is of clinical interest that the CD46 gene resides on chromosome 1q, which undergoes genomic amplification in the majority of relapsed myeloma patients. We found that the cell surface expression level of CD46 was markedly higher in patient myeloma cells with 1q gain than in those with normal 1q copy number. Thus, genomic amplification of CD46 may serve as a surrogate for target amplification that could allow patient stratification for tailored CD46-targeted therapy. Overall, these findings indicate that CD46 is a promising target for antibody-based treatment of multiple myeloma, especially in patients with gain of chromosome 1q.
Collapse
|
23
|
Tonry CL, Leacy E, Raso C, Finn SP, Armstrong J, Pennington SR. The Role of Proteomics in Biomarker Development for Improved Patient Diagnosis and Clinical Decision Making in Prostate Cancer. Diagnostics (Basel) 2016; 6:E27. [PMID: 27438858 PMCID: PMC5039561 DOI: 10.3390/diagnostics6030027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/28/2016] [Accepted: 07/07/2016] [Indexed: 02/06/2023] Open
Abstract
Prostate Cancer (PCa) is the second most commonly diagnosed cancer in men worldwide. Although increased expression of prostate-specific antigen (PSA) is an effective indicator for the recurrence of PCa, its intended use as a screening marker for PCa is of considerable controversy. Recent research efforts in the field of PCa biomarkers have focused on the identification of tissue and fluid-based biomarkers that would be better able to stratify those individuals diagnosed with PCa who (i) might best receive no treatment (active surveillance of the disease); (ii) would benefit from existing treatments; or (iii) those who are likely to succumb to disease recurrence and/or have aggressive disease. The growing demand for better prostate cancer biomarkers has coincided with the development of improved discovery and evaluation technologies for multiplexed measurement of proteins in bio-fluids and tissues. This review aims to (i) provide an overview of these technologies as well as describe some of the candidate PCa protein biomarkers that have been discovered using them; (ii) address some of the general limitations in the clinical evaluation and validation of protein biomarkers; and (iii) make recommendations for strategies that could be adopted to improve the successful development of protein biomarkers to deliver improvements in personalized PCa patient decision making.
Collapse
Affiliation(s)
- Claire L Tonry
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland.
| | - Emma Leacy
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland.
| | - Cinzia Raso
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland.
| | - Stephen P Finn
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland.
| | | | - Stephen R Pennington
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
24
|
Xia X, Feng H, Li C, Qin C, Song Y, Zhang Y, Lan X. 99mTc-labeled estradiol as an estrogen receptor probe: Preparation and preclinical evaluation. Nucl Med Biol 2015; 43:89-96. [PMID: 26466867 DOI: 10.1016/j.nucmedbio.2015.09.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 08/22/2015] [Accepted: 09/08/2015] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Most breast cancers express estrogen receptors (ERs). Noninvasive imaging of ER expression may be helpful for planning therapy of ER+ tumors. We developed a new ER- binding probe, (99m)Tc-labeled estradiol, with diethylenetriaminepentaacetic acid (DTPA) as a chelating ligand, and assessed its targeting ability in vitro and in vivo. METHODS 3-Aminoethyl estradiol was synthesized in two steps from estrone, followed by (99m)Tc labeling. Western blotting and immunofluorescence staining were used to detect ER expression in MCF-7 and MDA-MB-231 breast cancer cells. Saturation binding and specific binding were performed by incubating MCF-7 cells with increasing concentrations of (99m)Tc-DTPA-estradiol. Cell uptake, efflux, and blocking assays were also performed. To test (99m)Tc-DTPA-estradiol in vivo, nude mice bearing either MCF-7- (high ER expression) or MDA-MB-231- derived tumors (low ER expression) were injected with (99m)Tc-DTPA-estradiol, and underwent single-photon emission-computed tomography (SPECT). Mice injected with excess unlabeled DTPA-estradiol were used as controls. Ex vivo gamma-counting of tissues from normal and tumor-bearing mice was used to evaluate (99m)Tc-DTPA-estradiol biodistribution. RESULTS The radiochemical purity of (99m)Tc-DTPA-estradiol was 98.3%±1.3% with a specific activity of 33.1±1.5 MBq/μmol (n=3). Western blotting and immunofluorescence staining confirmed extensive expression of ERs by the MCF-7 cells, and less extensive expression by MDA-MB-231 cells. There was high binding affinity of (99m)Tc-DTPA-estradiol to MCF-7 cells with a>45% specific rate of total cell uptake. SPECT images and the biodistribution study results showed significantly higher uptake by MCF-7 tumors (6.06±0.38 %ID/g) than by MDA-MB-231 tumors (1.57±0.28 %ID/g). Pre-injection of MCF-7 tumor-bearing nude mice with excess unlabeled DTPA-estradiol significantly reduced tumor uptake of (99m)Tc-DTPA-estradiol (2.24±0.28 %ID/g), suggesting that (99m)Tc-DTPA-estradiol specifically targets ERs in tumors. CONCLUSIONS (99m)Tc-DTPA-estradiol can be synthesized with satisfactory labeling efficiency and stability. (99m)Tc-DTPA-estradiol specifically targeted ERs in vitro and in vivo with favorable pharmacokinetics, allowing ER receptor expression assessment with SPECT imaging.
Collapse
Affiliation(s)
- Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Hongyan Feng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Chongjiao Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Chunxia Qin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yiling Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| |
Collapse
|
25
|
ten Haaf A, Pscherer S, Fries K, Barth S, Gattenlöhner S, Tur MK. Phage display-based on-slide selection of tumor-specific antibodies on formalin-fixed paraffin-embedded human tissue biopsies. Immunol Lett 2015; 166:65-78. [DOI: 10.1016/j.imlet.2015.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/22/2015] [Accepted: 05/25/2015] [Indexed: 10/24/2022]
|
26
|
Larsen SA, Meldgaard T, Lykkemark S, Mandrup OA, Kristensen P. Selection of cell-type specific antibodies on tissue-sections using phage display. J Cell Mol Med 2015; 19:1939-48. [PMID: 25808085 PMCID: PMC4549044 DOI: 10.1111/jcmm.12568] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/29/2015] [Indexed: 12/29/2022] Open
Abstract
With the advent of modern technologies enabling single cell analysis, it has become clear that small sub-populations of cells or even single cells can drive the phenotypic appearance of tissue, both diseased and normal. Nucleic acid based technologies allowing single cell analysis has been faster to mature, while technologies aimed at analysing the proteome at a single cell level is still lacking behind, especially technologies which allow single cell analysis in tissue. Introducing methods, that allows such analysis, will pave the way for discovering new biomarkers with more clinical relevance, as these may be unique for microenvironments only present in tissue and will avoid artifacts introduced by in vitro studies. Here, we introduce a technology enabling biomarker identification on small sub-populations of cells within a tissue section. Phage antibody libraries are applied to the tissue sections, followed by washing to remove non-bound phage particles. To eliminate phage antibodies binding to antigens ubiquitously expressed and retrieve phage antibodies binding specifically to antigens expressed by the sub-population of cells, the area of interest is protected by a ‘shadow stick’. The phage antibodies on the remaining areas on the slide are exposed to UV light, which introduces cross-links in the phage genome, thus rendering them non-replicable. In this work we applied the technology, guided by CD31 expressing endothelial cells, to isolate recombinant antibodies specifically binding biomarkers expressed either by the cell or in the microenvironment surrounding the endothelial cell.
Collapse
Affiliation(s)
- Simon Asbjørn Larsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Simon Lykkemark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Sino-Danish Centre for Education and Research (SDC), Aarhus, Denmark
| | | | | |
Collapse
|
27
|
Selection strategies for anticancer antibody discovery: searching off the beaten path. Trends Biotechnol 2015; 33:292-301. [PMID: 25819764 DOI: 10.1016/j.tibtech.2015.02.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/24/2015] [Accepted: 02/25/2015] [Indexed: 01/13/2023]
Abstract
Antibody-based drugs represent one of the most successful and promising therapeutic approaches in oncology. Large combinatorial phage antibody libraries are available for the identification of therapeutic antibodies and various technologies exist for their further conversion into multivalent and multispecific formats optimized for the desired pharmacokinetics and the pathological context. However, there is no technology for antigen profiling of intact tumors to identify tumor markers targetable with antibodies. Such constraints have led to a relative paucity of tumor-associated antigens for antibody targeting in oncology. Here we review novel approaches aimed at the identification of antibody-targetable, accessible antigens in intact tumors. We hope that such advanced selection approaches will be useful in the development of next-generation antibody therapies for cancer.
Collapse
|
28
|
Bidlingmaier S, Su Y, Liu B. Combining Phage and Yeast Cell Surface Antibody Display to Identify Novel Cell Type-Selective Internalizing Human Monoclonal Antibodies. Methods Mol Biol 2015; 1319:51-63. [PMID: 26060069 DOI: 10.1007/978-1-4939-2748-7_3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Using phage antibody display, large libraries can be generated and screened to identify monoclonal antibodies with affinity for target antigens. However, while library size and diversity is an advantage of the phage display method, there is limited ability to quantitatively enrich for specific binding properties such as affinity. One way of overcoming this limitation is to combine the scale of phage display selections with the flexibility and quantitativeness of FACS-based yeast surface display selections. In this chapter we describe protocols for generating yeast surface antibody display libraries using phage antibody display selection outputs as starting material and FACS-based enrichment of target antigen-binding clones from these libraries. These methods should be widely applicable for the identification of monoclonal antibodies with specific binding properties.
Collapse
Affiliation(s)
- Scott Bidlingmaier
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, 1001 Potrero Avenue, 1305, San Francisco, CA, 94110, USA
| | | | | |
Collapse
|
29
|
Selecting an Optimal Antibody for Antibody- Drug Conjugate Therapy. ANTIBODY-DRUG CONJUGATES 2015. [DOI: 10.1007/978-3-319-13081-1_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
30
|
Bidlingmaier S, Liu B. Identification of Posttranslational Modification-Dependent Protein Interactions Using Yeast Surface Displayed Human Proteome Libraries. Methods Mol Biol 2015; 1319:193-202. [PMID: 26060076 DOI: 10.1007/978-1-4939-2748-7_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The identification of proteins that interact specifically with posttranslational modifications such as phosphorylation is often necessary to understand cellular signaling pathways. Numerous methods for identifying proteins that interact with posttranslational modifications have been utilized, including affinity-based purification and analysis, protein microarrays, phage display, and tethered catalysis. Although these techniques have been used successfully, each has limitations. Recently, yeast surface-displayed human proteome libraries have been utilized to identify protein fragments with affinity for various target molecules, including phosphorylated peptides. When coupled with fluorescently activated cell sorting and high throughput methods for the analysis of selection outputs, yeast surface-displayed human proteome libraries can rapidly and efficiently identify protein fragments with affinity for any soluble ligand that can be fluorescently detected, including posttranslational modifications. In this review we compare the use of yeast surface display libraries to other methods for the identification of interactions between proteins and posttranslational modifications and discuss future applications of the technology.
Collapse
Affiliation(s)
- Scott Bidlingmaier
- Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 1001 Potrero Avenue, 1305, San Francisco, CA, 94110, USA
| | | |
Collapse
|
31
|
Ha KD, Bidlingmaier SM, Zhang Y, Su Y, Liu B. High-content analysis of antibody phage-display library selection outputs identifies tumor selective macropinocytosis-dependent rapidly internalizing antibodies. Mol Cell Proteomics 2014; 13:3320-31. [PMID: 25149096 PMCID: PMC4256486 DOI: 10.1074/mcp.m114.039768] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Many forms of antibody-based targeted therapeutics, including antibody drug conjugates, utilize the internalizing function of the targeting antibody to gain intracellular entry into tumor cells. Ideal antibodies for developing such therapeutics should be capable of both tumor-selective binding and efficient endocytosis. The macropinocytosis pathway is capable of both rapid and bulk endocytosis, and recent studies have demonstrated that it is selectively up-regulated by cancer cells. We hypothesize that receptor-dependent macropinocytosis can be achieved using tumor-targeting antibodies that internalize via the macropinocytosis pathway, improving potency and selectivity of the antibody-based targeted therapeutic. Although phage antibody display libraries have been utilized to find antibodies that bind and internalize to target cells, no methods have been described to screen for antibodies that internalize specifically via macropinocytosis. We hereby describe a novel screening strategy to identify phage antibodies that bind and rapidly enter tumor cells via macropinocytosis. We utilized an automated microscopic imaging-based, High Content Analysis platform to identify novel internalizing phage antibodies that colocalize with macropinocytic markers from antibody libraries that we have generated previously by laser capture microdissection-based selection, which are enriched for internalizing antibodies binding to tumor cells in situ residing in their tissue microenvironment (Ruan, W., Sassoon, A., An, F., Simko, J. P., and Liu, B. (2006) Identification of clinically significant tumor antigens by selecting phage antibody library on tumor cells in situ using laser capture microdissection. Mol. Cell. Proteomics. 5, 2364–2373). Full-length human IgG molecules derived from macropinocytosing phage antibodies retained the ability to internalize via macropinocytosis, validating our screening strategy. The target antigen for a cross-species binding antibody with a highly active macropinocytosis activity was identified as ephrin type-A receptor 2. Antibody-toxin conjugates created using this macropinocytosing IgG were capable of potent and receptor-dependent killing of a panel of EphA2-positive tumor cell lines in vitro. These studies identify novel methods to screen for and validate antibodies capable of receptor-dependent macropinocytosis, allowing further exploration of this highly efficient and tumor-selective internalization pathway for targeted therapy development.
Collapse
Affiliation(s)
- Kevin D Ha
- From the ‡Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110-1305
| | - Scott M Bidlingmaier
- From the ‡Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110-1305
| | - Yafeng Zhang
- From the ‡Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110-1305
| | - Yang Su
- From the ‡Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110-1305
| | - Bin Liu
- From the ‡Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110-1305
| |
Collapse
|
32
|
Zhang ZF, Shan X, Wang YX, Wang W, Feng SY, Cui YB. Screening and selection of peptides specific for esophageal cancer cells from a phage display peptide library. J Cardiothorac Surg 2014; 9:76. [PMID: 24779651 PMCID: PMC4018990 DOI: 10.1186/1749-8090-9-76] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 04/24/2014] [Indexed: 01/11/2023] Open
Abstract
Background Esophageal cancer is a common malignant tumor of the gastrointestinal tract and is typically diagnosed at an advanced stage due to the absence of early clinical symptoms. Although surgery, chemotherapy, and radiotherapy represent the major treatment methods employed for this cancer, the prognosis of esophageal cancer remains poor. Methods A Ph.D.-12TM Phage Display Peptide Library was screened using an esophageal cancer cell line, Eca109, and a normal esophageal epithelial cell line to identify novel ligands that selectively bind the surface of esophageal cancer cells with high affinity. Results Two polypeptides were isolated that exhibited higher binding affinities and specificity for the Eca109 cells. These peptides were further validated using enzyme-linked immunosorbent assays (ELISAs), immunofluorescence assays, and immunohistochemistry assays. Conclusion Two polypeptides with high binding affinities to esophageal cancer cells were isolated from the Ph.D.-12TM Phage Display Peptide Library. Further studies are needed to characterize the biological effects of these polypeptides and to explore the potential for these peptides to be used for the early screening of esophageal cancer or for cell-targeted therapies that would reduce the toxic side effects of cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - You-Bin Cui
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, 130021 Changchun, China.
| |
Collapse
|
33
|
Sørensen MD, Melchjorsen CJ, Mandrup OA, Kristensen P. Raising antibodies against circulating foetal cells from maternal peripheral blood. Prenat Diagn 2013; 33:284-91. [PMID: 23390071 DOI: 10.1002/pd.4060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Cells of foetal origin circulating in the maternal peripheral bloodstream present a unique source for non-invasive prenatal diagnostics. The aims of this study were to raise antibodies against identified circulating foetal cells from the maternal blood, test the properties of these antibodies and to determine the foetal cell type recognised by the antibodies. METHOD Cells from a male foetus were identified in a maternal blood sample by FISH analysis of the X- and Y- chromosomes. The identified cells were subjected to phage display selection using a novel single cell selection strategy. Selected antibodies were tested by immunocytochemistry on foetal and adult tissue arrays, an endothelial cell line, and peripheral blood mononuclear cells. RESULTS Three identified foetal cells subjected to antibody selection, yielded a total of 12 antibodies. Three antibodies gave distinct staining patterns on tissue arrays, and endothelial cells. One antibody, SF1.3, shows specific staining of a subpopulation of peripheral blood mononuclear cells, including a fraction of CD34 positive cells. CONCLUSION These findings indicate that the identified foetal cells could have been progenitor cells of haematopoietic origin. The antibody SF1.3 could be a potential tool toward non-invasive prenatal diagnostics.
Collapse
|
34
|
Zhou Y, Zhao L, Marks JD. Selection and characterization of cell binding and internalizing phage antibodies. Arch Biochem Biophys 2012; 526:107-13. [PMID: 22627065 DOI: 10.1016/j.abb.2012.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 05/09/2012] [Accepted: 05/13/2012] [Indexed: 12/25/2022]
Abstract
Many therapeutic targets are cell surface receptors, which can be challenging antigens for antibody generation. For many therapeutic applications, one needs antibodies that not only bind the cell surface receptor but also are internalized into the cell. This allows use of the antibody to deliver various payloads into the cell to achieve a therapeutic effect. Phage antibody technology has proven a powerful tool for the generation and optimization of human antibodies to any antigen. While applied to the generation of antibodies to purified proteins, it is possible to directly select cell binding and internalizing antibodies on cells. Potential advantages of this approach include: cell surface receptors are in native conformation on intact cells while this might not be so for recombinant proteins; antibodies can be selected for both cell binding and internalization properties; the antibodies can be used to identify their tumor associated antigens; and such antibodies can be used for human treatment directly since they are human in sequence. This review will discuss the factors that impact the successful selection of cell binding and internalizing antibodies. These factors include the cell types used for selection, the impact of different phage antibody library formats, and the specific selection protocols used.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
| | | | | |
Collapse
|
35
|
Iyer AK, Lan X, Zhu X, Su Y, Feng J, Zhang X, Gao D, Seo Y, Vanbrocklin HF, Broaddus VC, Liu B, He J. Novel human single chain antibody fragments that are rapidly internalizing effectively target epithelioid and sarcomatoid mesotheliomas. Cancer Res 2011; 71:2428-32. [PMID: 21447742 DOI: 10.1158/0008-5472.can-10-3484] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Human antibodies targeting all subtypes of mesothelioma could be useful to image and treat this deadly disease. Here we report tumor targeting of a novel internalizing human single chain antibody fragment (scFv) labeled with (⁹⁹m)Tc ((⁹⁹m)Tc-M40) in murine models of mesothelioma of both epithelioid (M28) and sarcomatoid (VAMT-1) origins. (⁹⁹m)Tc-M40 was taken up rapidly and specifically by both subtype tumor cells in vitro, with 68% to 92% internalized within 1 hour. The specificity of binding was evidenced by blocking (up to 95%) with 10-fold excess of unlabeled M40. In animal studies, tumors of both subtypes were clearly visualized by SPECT/CT as early as 1 hour postinjection of (⁹⁹m)Tc-M40. Tumor uptake measured as percent of injected dose per gram tissue (%ID/g) at 3 hours was 4.38 and 5.84 for M28 and VAMT-1 tumors, respectively, significantly greater than all organs or tissues studied (liver, 2.62%ID/g; other organs or tissues <1.7%ID/g), except the kidneys (130.7%ID/g), giving tumor-to-blood ratios of 5:1 and 7:1 and tumor-to-muscle ratios of 45:1 and 60:1, for M28 and VAMT-1, respectively. The target-mediated uptake was confirmed by a nearly 70% reduction in tumor activity following administration of 10-fold excess of unlabeled scFv. Taken together, these results indicate that M40 can rapidly and specifically target epithelioid and sarcomatoid tumor cells, demonstrating the potential of this agent as a versatile targeting ligand for imaging and therapy of all subtypes of mesothelioma.
Collapse
Affiliation(s)
- Arun K Iyer
- Center for Molecular and Functional Imaging, Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Selection of antibodies against a single rare cell present in a heterogeneous population using phage display. Nat Protoc 2011; 6:509-22. [PMID: 21455187 DOI: 10.1038/nprot.2011.311] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Here we describe a new method applying phage-displayed antibody libraries to the selection of antibodies against a single identified cell on a glass slide. This is the only described method that has successfully achieved selection of antibodies against a single rare cell in a heterogeneous population of cells. The phage library is incubated with the slide containing the identified rare cell of interest; incubation is followed by UV irradiation while protecting the target cell with a minute disc. The UV light inactivates all phages outside the shielded area by cross-linking the DNA constituting their genomes. The expected yield is between one and ten phage particles from a single cell selection. The encoded antibodies are subsequently produced monoclonally and tested for specificity. This method can be applied within a week to carry out ten or more individual cell selections. Including subsequent testing of antibody specificity, a specific antibody can be identified within 2 months.
Collapse
|
37
|
Sørensen MD, Agerholm IE, Christensen B, Kølvraa S, Kristensen P. Microselection--affinity selecting antibodies against a single rare cell in a heterogeneous population. J Cell Mol Med 2011; 14:1953-61. [PMID: 20726925 PMCID: PMC3823277 DOI: 10.1111/j.1582-4934.2010.00896.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Rare cells not normally present in the peripheral bloodstream, such as circulating tumour cells, have potential applications for development of non-invasive methods for diagnostics or follow up. Obtaining these cells however require some means of discrimination, achievable by cell type specific antibodies. Here we have generated a microselection method allowing antibody selection, by phage display, targeting a single cell in a heterogeneous population. One K562 cell (female origin) was positioned on glass slide among millions of lymphocytes from male donor, identifying the K562 cell by FISH (XX). Several single cell selections were performed on such individual slides. The phage particles bound to the target cell is protected by a minute disc, while inactivating all remaining phage by UV-irradiation; leaving only the phage bound to the target cell viable. We hereby retrieved up to eight antibodies per single cell selection, including three highly K562 cell type specific.
Collapse
|
38
|
A novel human recombinant single-chain antibody targeting CD166/ALCAM inhibits cancer cell invasion in vitro and in vivo tumour growth. Cancer Immunol Immunother 2010; 59:1665-74. [PMID: 20635083 PMCID: PMC2929338 DOI: 10.1007/s00262-010-0892-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 06/30/2010] [Indexed: 11/17/2022]
Abstract
Screening a phage-display single-chain antibody library for binding to the breast cancer cell line PM-1 an antibody, scFv173, recognising activated leukocyte cell adhesion molecule (ALCAM, CD166) was isolated and its binding profile was characterized. Positive ALCAM immunohistochemical staining of frozen human tumour sections was observed. No ALCAM staining was observed in the majority of tested normal human tissues (nine of ten). Flow cytometry analyses revealed binding to 22 of 26 cancer cell lines of various origins and no binding to normal blood and bone marrow cells. Antibody binding inhibited invasion of the breast cancer cell line MDA-MB-231 by 50% in an in vitro Matrigel-coated membrane invasion assay. Reduced growth of tumours in nude mice was observed in an in vivo model in which the mice were injected subcutaneously with colorectal carcinoma HCT 116 cells and treated with scFv173 when compared to control. In summary, we have characterized a novel fully human scFv antibody recognising ALCAM on cancer cells and in tumour tissues that reduces cancer cell invasion and tumour growth in accordance with the hypothesised role for ALCAM in cell growth and migration control.
Collapse
|
39
|
Chen Y, Zhu X, Zhang X, Liu B, Huang L. Nanoparticles modified with tumor-targeting scFv deliver siRNA and miRNA for cancer therapy. Mol Ther 2010; 18:1650-6. [PMID: 20606648 DOI: 10.1038/mt.2010.136] [Citation(s) in RCA: 390] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Targeted delivery of RNA-based therapeutics for cancer therapy remains a challenge. We have developed a LPH (liposome-polycation-hyaluronic acid) nanoparticle formulation modified with tumor-targeting single-chain antibody fragment (scFv) for systemic delivery of small interfering RNA (siRNA) and microRNA (miRNA) into experimental lung metastasis of murine B16F10 melanoma. The siRNAs delivered by the scFv targeted nanoparticles efficiently downregulated the target genes (c-Myc/MDM2/VEGF) in the lung metastasis. Two daily intravenous injections of the combined siRNAs in the GC4-targeted nanoparticles significantly reduced the tumor load in the lung. miRNA-34a (miR-34a) induced apoptosis, inhibited survivin expression, and downregulated MAPK pathway in B16F10 cells. miR-34a delivered by the GC4-targeted nanoparticles significantly downregulated the survivin expression in the metastatic tumor and reduced tumor load in the lung. When miR-34a and siRNAs were co-formulated in GC4-targeted nanoparticles, an enhanced anticancer effect was observed.
Collapse
Affiliation(s)
- Yunching Chen
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | |
Collapse
|
40
|
Zhu X, Bidlingmaier S, Hashizume R, James CD, Berger MS, Liu B. Identification of internalizing human single-chain antibodies targeting brain tumor sphere cells. Mol Cancer Ther 2010; 9:2131-41. [PMID: 20587664 DOI: 10.1158/1535-7163.mct-09-1059] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive form of primary brain tumor for which there is no curative treatment to date. Resistance to conventional therapies and tumor recurrence pose major challenges to treatment and management of this disease, and therefore new therapeutic strategies need to be developed. Previous studies by other investigators have shown that a subpopulation of GBM cells can grow as neurosphere-like cells when cultured in restrictive medium and exhibits enhanced tumor-initiating ability and resistance to therapy. We report here the identification of internalizing human single-chain antibodies (scFv) targeting GBM tumor sphere cells. We selected a large naive phage antibody display library on the glycosylation-dependent CD133 epitope-positive subpopulation of GBM cells grown as tumor spheres and identified internalizing scFvs that target tumor sphere cells broadly, as well as scFvs that target the CD133-positive subpopulation. These scFvs were found to be efficiently internalized by GBM tumor sphere cells. One scFv GC4 inhibited self-renewal of GBM tumor sphere cells in vitro. We have further developed a full-length human IgG1 based on this scFv, and found that it potently inhibits proliferation of GBM tumor sphere cells and GBM cells grown in regular nonselective medium. Taken together, these results show that internalizing human scFvs targeting brain tumor sphere cells can be readily identified from a phage antibody display library, which could be useful for further development of novel therapies that target subpopulations of GBM cells to combat recurrence and resistance to treatment.
Collapse
Affiliation(s)
- Xiaodong Zhu
- Department of Anesthesia, University of California at San Francisco, San Francisco, California 94110, USA
| | | | | | | | | | | |
Collapse
|
41
|
He J, Wang Y, Feng J, Zhu X, Lan X, Iyer AK, Zhang N, Seo Y, VanBrocklin HF, Liu B. Targeting prostate cancer cells in vivo using a rapidly internalizing novel human single-chain antibody fragment. J Nucl Med 2010; 51:427-32. [PMID: 20150269 DOI: 10.2967/jnumed.109.069492] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED Human antibodies targeting prostate cancer cell surface epitopes may be useful for imaging and therapy. The objective of this study was to evaluate the tumor targeting of an internalizing human antibody fragment, a small-size platform, to provide high contrast in a mouse model of human prostate carcinoma. METHODS A prostate tumor-targeting single-chain antibody fragment (scFv), UA20, along with a nonbinding control scFv, N3M2, were labeled with (99m)Tc and evaluated for binding and rapid internalization into human prostate tumor cells in vitro and tumor homing in vivo using xenograft models. For the in vitro studies, the labeled UA20 scFv was incubated at 37 degrees C for 1 h with metastatic prostate cancer cells (DU145) to assess the total cellular uptake versus intracellular uptake. For the animal studies, labeled UA20 and N3M2 scFvs were administered to athymic mice implanted subcutaneously with DU145 cells. Mice were imaged with small-animal SPECT/CT with concomitant biodistribution at 1 and 3 h after injection. RESULTS The UA20 scFv was labeled in 55%-65% yield and remained stable in phosphate buffer within 24 h. The labeled UA20 scFv was taken up specifically by prostate tumor cells. Internalization was rapid, because incubation at 37 degrees C for less than 1 h resulted in 93% internalization of total cell-associated scFvs. In animal studies, SPECT/CT showed significant tumor uptake as early as 1 h after injection. At 3 h after injection, tumor uptake was 4.4 percentage injected dose per gram (%ID/g), significantly greater than all organs or tissues studied (liver, 2.7 %ID/g; other organs or tissues, <1 %ID/g), except the kidneys (81.4 %ID/g), giving tumor-to-blood and tumor-to-muscle ratios of 12:1 and 70:1, respectively. In contrast, the control antibody exhibited a tumor uptake of only 0.26 %ID/g, similar to that of muscle and fat. Tumor-specific targeting was evidenced by reduced tumor uptake of nearly 70% on administration of a 10-fold excess of unlabeled UA20 scFv. Kidney uptake was nonspecific, consistent with the route of excretion by scFvs. CONCLUSION The UA20 scFv showed rapid and specific internalization in prostate tumor cells in vitro and accumulation in prostate tumor xenografts in vivo, demonstrating the potential for future development for prostate cancer imaging and targeted therapy.
Collapse
Affiliation(s)
- Jiang He
- Department of Radiology and Biomedical Imaging, Center for Molecular and Functional Imaging, University of California at San Francisco, San Francisco, California 94143, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Confronting Cellular Heterogeneity in Studies of Protein Metabolism and Homeostasis in Aging Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 694:234-44. [DOI: 10.1007/978-1-4419-7002-2_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
43
|
Zhao Q, Chan YW, Lee SST, Cheung WT. One-step expression and purification of single-chain variable antibody fragment using an improved hexahistidine tag phagemid vector. Protein Expr Purif 2009; 68:190-5. [PMID: 19683057 DOI: 10.1016/j.pep.2009.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Revised: 07/23/2009] [Accepted: 08/10/2009] [Indexed: 12/17/2022]
Abstract
Millions of candidate clones are commonly obtained following rounds of phage-displayed antibody library panning, and expression of those selected single-chain variable fragment (scFv) is required for secondary functional screening to identify positive clones. Large scale functional screening is often hampered by the time-consuming and labor-intensive subcloning of those candidate scFv clones into a bacterial expression vector carrying an affinity tag for scFv purification and detection. To overcome the limitations and to develop a multiplex approach, an improved hexahistidine tag phagemid vector was constructed for one-step scFv expression and purification. By using hexahistidine as an affinity tag, soluble scFvs can be rapidly and cost-effectively captured from Escherichia coli periplasmic extracts. For proof-of-concept, feasibility of the improved phagemid vector was examined against two scFvs, L17E4d targeting a cell surface antigen and L18Hh5 recognizing a monoclonal antibody (mAb). Using 1 ml of Ni-NTA agarose, 0.2-0.5 mg of soluble scFv was obtained from 1 L of bacteria culture, and the purified scFvs bound specifically to their target antigens with high affinity. Moreover, using two randomly selected hapten-specific scFv phage clones, it was demonstrated that the display of scFvs on phage surface was not affected by the hexahistidine affinity tag. These results suggest the improved phagemid vector allows the shuttle of phage-displayed antibody library panning and functional scFv production. Importantly, the improved phagemid vector can be easily adapted for multiplex screening.
Collapse
Affiliation(s)
- Qi Zhao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | | | | | | |
Collapse
|
44
|
Phage-display selection on tumor histological specimens with laser capture microdissection. J Immunol Methods 2009; 347:46-53. [PMID: 19538966 DOI: 10.1016/j.jim.2009.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Revised: 06/03/2009] [Accepted: 06/05/2009] [Indexed: 11/22/2022]
Abstract
A method was developed to obtain phage-display ligands that bind to a select population of cells in histological specimens of freshly harvested solid human cancers. It combines phage-display panning with laser capture microdissection (LCM). This method allows selection of phage ligands bound to subpopulations of specific cells contained in tumor tissue on histological sections. Naïve phage scFv library was incubated directly on a histological section of human breast cancer that was snap frozen immediately after surgical resection. Tumor and stromal cells were captured by LCM and bound phages were recovered by bacterial infection. Individual phage clones selected after panning were evaluated for their binding ability by immunofluorescence staining on tumor tissue from the same patient. One phage-display antibody clone selected on tumor stroma showed selective binding on tumor stroma but did not bind to malignant cell population. The expressed scFv of this clone showed no significant binding to normal tissue, or 13 other breast cancers, or 4 colon cancer samples. Using the same method, phage display antibody clones were selected on tumor cells which showed binding to tumor cells and normal tissue. This method is applicable for selection of ligands to virtually any portion of a histological specimen amenable to LCM. This may speed the process of generating ligands to any subset of cells or noncellular feature present on histological specimens.
Collapse
|
45
|
Human antibodies targeting cell surface antigens overexpressed by the hormone refractory metastatic prostate cancer cells: ICAM-1 is a tumor antigen that mediates prostate cancer cell invasion. J Mol Med (Berl) 2009; 87:507-14. [PMID: 19219419 PMCID: PMC2796542 DOI: 10.1007/s00109-009-0446-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Revised: 12/09/2008] [Accepted: 01/19/2009] [Indexed: 12/24/2022]
Abstract
Transition from hormone-sensitive to hormone-refractory metastatic tumor types poses a major challenge for prostate cancer treatment. Tumor antigens that are differentially expressed during this transition are likely to play important roles in imparting prostate cancer cells with the ability to grow in a hormone-deprived environment and to metastasize to distal sites such as the bone and thus, are likely targets for therapeutic intervention. To identify those molecules and particularly cell surface antigens that accompany this transition, we studied the changes in cell surface antigenic profiles between a hormone-sensitive prostate cancer line LNCaP and its hormone-refractory derivative C4-2B, using an antibody library-based affinity proteomic approach. We selected a naïve phage antibody display library to identify human single-chain antibodies that bind specifically to C4-2B but not LNCaP. Using mass spectrometry, we identified one of the antibody-targeted antigens as the ICAM-1/CD54/human rhinovirus receptor. Recombinant IgG1 derived from this single-chain antibody binds to a neutralizing epitope of ICAM-1 and blocks C4-2B cell invasion through extracellular matrix in vitro. ICAM-1 is thus differentially expressed during the transition of the hormone-sensitive prostate cancer cell line LNCaP to its hormone-refractory derivative C4-2B, plays an important role in imparting the C4-2B line with the ability to invade, and may therefore be a target for therapeutic intervention.
Collapse
|
46
|
Kubo N, Akita N, Shimizu A, Kitahara H, Parker AL, Miyagawa S. Identification of oligopeptide binding to colon cancer cells separated from patients using laser capture microdissection. J Drug Target 2008; 16:396-404. [PMID: 18569284 DOI: 10.1080/10611860802088796] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The development of intravascular conjugates that efficiently deliver genes or drugs to tumors is limited by the lack of efficacious targeting ligands. Small targeting peptides, such as those iterated by phage display technology, offer enormous potential for these applications. The majority of reports published to date have focused on the identification of peptides isolated for their ability to bind to human cancer cell lines in vitro, and have failed to account for the loss of polarization and de-differentiation of such cells from their in vivo state. Here, we report a novel approach for the identification of peptides capable of binding specifically to cancer cells derived from clinically resected human colon cancer. In this strategy, laser capture microdissection (LCM) is performed on a surgically resected colon cancer specimen to separate only cancer cells from the specimen. Subsequently, biopanning was performed on the LCM-selected colon cancer cells to identify peptide sequences that bound specifically to them. A peptide containing the SPT motif was selected as the most promising consensus sequence binding specifically to the LCM-selected colon cancer cells. Phage clones displaying the SPT motif demonstrated 9-fold higher binding to colon cancer cells derived from a patient than insertless phage (p < 0.05), while, recovery of the SPT phage from the colon cancer cell lines DLD-1 and HCT-15 was 7-fold higher than that of the control insertless phage (p < 0.05). The binding of SPT phage to colon cancer cells from the patient was confirmed by immunofluorescence. Additionally, a synthesized SPT-containing peptide (SPTKSNS) showed binding activity in the absence of mitogenic effects on colon cancer cells in vitro. In summary, we have introduced LCM into a biopanning procedure and identified a small peptide that binds preferentially to colon cancer cells derived from a clinically resected sample. This procedure could be applicable for the design of customized cancer cell targeting methodologies using clinical biopsy samples from human subjects.
Collapse
Affiliation(s)
- Naoki Kubo
- Department of Surgery, Shinshu University School of Medicine, Asahi, Matsumoto, Japan.
| | | | | | | | | | | |
Collapse
|
47
|
An F, Drummond DC, Wilson S, Kirpotin DB, Nishimura SL, Broaddus VC, Liu B. Targeted drug delivery to mesothelioma cells using functionally selected internalizing human single-chain antibodies. Mol Cancer Ther 2008; 7:569-78. [PMID: 18319332 DOI: 10.1158/1535-7163.mct-07-2132] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Mesothelioma is a malignancy of the mesothelium and current treatments are generally ineffective. One promising area of anticancer drug development is to explore tumor susceptibility to targeted therapy. To achieve efficient, targeted intracellular delivery of therapeutic agents to mesothelioma cells, we selected a naive human single-chain (scFv) phage antibody display library directly on the surface of live mesothelioma cells to identify internalizing antibodies that target mesothelioma-associated cell surface antigens. We have identified a panel of internalizing scFvs that bind to mesothelioma cell lines derived from both epithelioid (M28) and sarcomatous (VAMT-1) types of this disease. Most importantly, these antibodies stain mesothelioma cells in situ and therefore define a panel of clinically represented tumor antigens. We have further exploited the internalizing function of these scFvs to achieve targeted intracellular drug delivery to mesothelioma cells. We showed that scFv-targeted immunoliposomes were efficiently and specifically taken up by both epithelioid and sarcomatous mesothelioma cells, but not control cells, and immunoliposomes encapsulating the small-molecule drug topotecan caused targeted killing of both types of mesothelioma cells in vitro.
Collapse
Affiliation(s)
- Feng An
- Department of Anesthesia, University of California San Francisco, San Francisco, CA 94110, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Roth A, Drummond DC, Conrad F, Hayes ME, Kirpotin DB, Benz CC, Marks JD, Liu B. Anti-CD166 single chain antibody-mediated intracellular delivery of liposomal drugs to prostate cancer cells. Mol Cancer Ther 2008; 6:2737-46. [PMID: 17938267 DOI: 10.1158/1535-7163.mct-07-0140] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Targeted delivery of small-molecule drugs has the potential to enhance selective killing of tumor cells. We have identified previously an internalizing single chain [single chain variable fragment (scFv)] antibody that targets prostate cancer cells and identified the target antigen as CD166. We report here the development of immunoliposomes using this anti-CD166 scFv (H3). We studied the effects of a panel of intracellularly delivered, anti-CD166 immunoliposomal small-molecule drugs on prostate cancer cells. Immunoliposomal formulations of topotecan, vinorelbine, and doxorubicin each showed efficient and targeted uptake by three prostate cancer cell lines (Du-145, PC3, and LNCaP). H3-immunoliposomal topotecan was the most effective in cytotoxicity assays on all three tumor cell lines, showing improved cytotoxic activity compared with nontargeted liposomal topotecan. Other drugs such as liposomal doxorubicin were highly effective against LNCaP but not PC3 or Du-145 cells, despite efficient intracellular delivery. Post-internalization events thus modulate the overall efficacy of intracellularly delivered liposomal drugs, contributing in some cases to the lower than expected activity in a cell line-dependent manner. Further studies on intracellular tracking of endocytosed liposomal drugs will help identify and overcome the barriers limiting the potency of liposomal drugs.
Collapse
Affiliation(s)
- Audrey Roth
- Department of Anesthesia, University of California at San Francisco, Room 3C38, 1001 Potrero Avenue, San Francisco, CA 94110, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
|