1
|
Olkowski C, Fernandes B, Griffiths GL, Lin F, Choyke PL. Preclinical Imaging of Prostate Cancer. Semin Nucl Med 2023; 53:644-662. [PMID: 36882335 PMCID: PMC10440231 DOI: 10.1053/j.semnuclmed.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/01/2023] [Indexed: 03/07/2023]
Abstract
Prostate cancer remains a major cause of mortality and morbidity, affecting millions of men, with a large percentage expected to develop the disease as they reach advanced ages. Treatment and management advances have been dramatic over the past 50 years or so, and one aspect of these improvements is reflected in the multiple advances in diagnostic imaging techniques. Much attention has been focused on molecular imaging techniques that offer high sensitivity and specificity and can now more accurately assess disease status and detect recurrence earlier. During development of molecular imaging probes, single-photon emission computed tomography (SPECT) and positron emission tomography (PET) must be evaluated in preclinical models of the disease. If such agents are to be translated to the clinic, where patients undergoing these imaging modalities are injected with a molecular imaging probe, these agents must first be approved by the FDA and other regulatory agencies prior to their adoption in clinical practice. Scientists have worked assiduously to develop preclinical models of prostate cancer that are relevant to the human disease to enable testing of these probes and related targeted drugs. Challenges in developing reproducible and robust models of human disease in animals are beset with practical issues such as the lack of natural occurrence of prostate cancer in mature male animals, the difficulty of initiating disease in immune-competent animals and the sheer size differences between humans and conveniently smaller animals such as rodents. Thus, compromises in what is ideal and what can be achieved have had to be made. The workhorse of preclinical animal models has been, and remains, the investigation of human xenograft tumor models in athymic immunocompromised mice. Later models have used other immunocompromised models as they have been found and developed, including the use of directly derived patient tumor tissues, completely immunocompromised mice, orthotopic methods for inducing prostate cancer within the mouse prostate itself and metastatic models of advanced disease. These models have been developed in close parallel with advances in imaging agent chemistries, radionuclide developments, computer electronics advances, radiometric dosimetry, biotechnologies, organoid technologies, advances in in vitro diagnostics, and overall deeper understandings of disease initiation, development, immunology, and genetics. The combination of molecular models of prostatic disease with radiometric-based studies in small animals will always remain spatially limited due to the inherent resolution sensitivity limits of PET and SPECT decay processes, fundamentally set at around a 0.5 cm resolution limit. Nevertheless, it is central to researcher's efforts and to successful clinical translation that the best animal models are adopted, accepted, and scientifically verified as part of this truly interdisciplinary approach to addressing this important disease.
Collapse
Affiliation(s)
- Colleen Olkowski
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD
| | - Bruna Fernandes
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD
| | - Gary L Griffiths
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Frank Lin
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD
| | - Peter L Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD.
| |
Collapse
|
2
|
Bajgain P, Chavez AGT, Balasubramanian K, Fleckenstein L, Lulla P, Heslop HE, Vera J, Leen AM. Secreted Fas Decoys Enhance the Antitumor Activity of Engineered and Bystander T Cells in Fas Ligand-Expressing Solid Tumors. Cancer Immunol Res 2022; 10:1370-1385. [PMID: 36122411 PMCID: PMC9633434 DOI: 10.1158/2326-6066.cir-22-0115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 07/11/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022]
Abstract
T-cell immunotherapy has demonstrated remarkable clinical outcomes in certain hematologic malignancies. However, efficacy in solid tumors has been suboptimal, partially due to the hostile tumor microenvironment composed of immune-inhibitory molecules. One such suppressive agent abundantly expressed in solid tumors is Fas ligand (FasL), which can trigger apoptosis of Fas-expressing effector cells such as T cells and natural killer (NK) cells. To alleviate this FasL-induced suppression of tumor-specific immune cells in solid tumors, we describe here the development of a Fas decoy that is secreted by engineered cells upon activation and sequesters the ligand, preventing it from engaging with Fas on the surface of effector cells. We further improved the immune-stimulatory effects of this approach by creating a Fas decoy and IL15 cytokine fusion protein, which enhanced the persistence and antitumor activity of decoy-engineered as well as bystander chimeric-antigen receptor (CAR) T cells in xenograft models of pancreatic cancer. Our data indicate that secreted Fas decoys can augment the efficacy of both adoptively transferred and endogenous tumor-specific effector cells in FasL-expressing solid tumors.
Collapse
Affiliation(s)
- Pradip Bajgain
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, Texas
- Center for Cancer Research, National Cancer Institute, Frederick, MD
| | - Alejandro G. Torres Chavez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, Texas
| | - Kishore Balasubramanian
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, Texas
| | - Lindsey Fleckenstein
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, Texas
| | - Premal Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, Texas
| | - Helen E. Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, Texas
| | - Juan Vera
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, Texas
- Marker Therapeutics, Inc., Houston, Texas
| | - Ann M. Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
3
|
Kumar K, Ghosh A. Radiochemistry, Production Processes, Labeling Methods, and ImmunoPET Imaging Pharmaceuticals of Iodine-124. Molecules 2021; 26:E414. [PMID: 33466827 PMCID: PMC7830191 DOI: 10.3390/molecules26020414] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 01/01/2023] Open
Abstract
Target-specific biomolecules, monoclonal antibodies (mAb), proteins, and protein fragments are known to have high specificity and affinity for receptors associated with tumors and other pathological conditions. However, the large biomolecules have relatively intermediate to long circulation half-lives (>day) and tumor localization times. Combining superior target specificity of mAbs and high sensitivity and resolution of the PET (Positron Emission Tomography) imaging technique has created a paradigm-shifting imaging modality, ImmunoPET. In addition to metallic PET radionuclides, 124I is an attractive radionuclide for radiolabeling of mAbs as potential immunoPET imaging pharmaceuticals due to its physical properties (decay characteristics and half-life), easy and routine production by cyclotrons, and well-established methodologies for radioiodination. The objective of this report is to provide a comprehensive review of the physical properties of iodine and iodine radionuclides, production processes of 124I, various 124I-labeling methodologies for large biomolecules, mAbs, and the development of 124I-labeled immunoPET imaging pharmaceuticals for various cancer targets in preclinical and clinical environments. A summary of several production processes, including 123Te(d,n)124I, 124Te(d,2n)124I, 121Sb(α,n)124I, 123Sb(α,3n)124I, 123Sb(3He,2n)124I, natSb(α, xn)124I, natSb(3He,n)124I reactions, a detailed overview of the 124Te(p,n)124I reaction (including target selection, preparation, processing, and recovery of 124I), and a fully automated process that can be scaled up for GMP (Good Manufacturing Practices) production of large quantities of 124I is provided. Direct, using inorganic and organic oxidizing agents and enzyme catalysis, and indirect, using prosthetic groups, 124I-labeling techniques have been discussed. Significant research has been conducted, in more than the last two decades, in the development of 124I-labeled immunoPET imaging pharmaceuticals for target-specific cancer detection. Details of preclinical and clinical evaluations of the potential 124I-labeled immunoPET imaging pharmaceuticals are described here.
Collapse
Affiliation(s)
- Krishan Kumar
- Laboratory for Translational Research in Imaging Pharmaceuticals, The Wright Center of Innovation in Biomedical Imaging, Department of Radiology, The Ohio State University, Columbus, OH 43212, USA;
| | | |
Collapse
|
4
|
Gamache RF, Zettlitz KA, Tsai WTK, Collins J, Wu AM, Murphy JM. Tri-functional platform for construction of modular antibody fragments for in vivo 18F-PET or NIRF molecular imaging. Chem Sci 2020; 11:1832-1838. [PMID: 34123276 PMCID: PMC8148382 DOI: 10.1039/c9sc05007h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Positron emission tomography (PET) molecular imaging is a powerful tool for interrogating physiological and biochemical processes to understand the biology of disease and advance therapeutic developments. Near-infrared fluorescence (NIRF) optical imaging has become increasingly popular for intraoperative staging to enable cellular resolution imaging of tumor margins during surgical resection. In addition, engineered antibody fragments have emerged as promising molecular imaging agents given their exquisite target selectivity, rapid systemic clearance and site-selective chemical modification. We report a tri-functional platform for construction of a modular antibody fragment that can rapidly be labeled with radionuclides or fluorophores for PET or NIRF molecular imaging of prostate stem cell antigen (PSCA). To provide a universal approach towards the targeted delivery of PET and optical imaging agents, we have developed a tri-functional platform (TFP) for the facile construction of modular, target-specific tracers.![]()
Collapse
Affiliation(s)
- Raymond F Gamache
- Department of Chemistry and Biochemistry, University of California Los Angeles CA 90095 USA
| | - Kirstin A Zettlitz
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| | - Wen-Ting K Tsai
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| | - Jeffrey Collins
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| | - Anna M Wu
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| | - Jennifer M Murphy
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| |
Collapse
|
5
|
ImmunoPET Imaging of αvβ6 Expression Using an Engineered Anti-αvβ6 Cys-diabody Site-Specifically Radiolabeled with Cu-64: Considerations for Optimal Imaging with Antibody Fragments. Mol Imaging Biol 2017; 20:103-113. [DOI: 10.1007/s11307-017-1097-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
6
|
Mohammed S, Sukumaran S, Bajgain P, Watanabe N, Heslop HE, Rooney CM, Brenner MK, Fisher WE, Leen AM, Vera JF. Improving Chimeric Antigen Receptor-Modified T Cell Function by Reversing the Immunosuppressive Tumor Microenvironment of Pancreatic Cancer. Mol Ther 2017; 25:249-258. [PMID: 28129119 DOI: 10.1016/j.ymthe.2016.10.016] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 10/07/2016] [Accepted: 10/09/2016] [Indexed: 12/21/2022] Open
Abstract
The adoptive transfer of T cells redirected to tumor-associated antigens via transgenic expression of chimeric antigen receptors (CARs) has produced tumor responses, even in patients with refractory diseases. To target pancreatic cancer, we generated CAR T cells directed against prostate stem cell antigen (PSCA) and demonstrated specific tumor lysis. However, pancreatic tumors employ immune evasion strategies such as the production of inhibitory cytokines, which limit CAR T cell persistence and function. Thus, to protect our cells from the immunosuppressive cytokine IL-4, we generated an inverted cytokine receptor in which the IL-4 receptor exodomain was fused to the IL-7 receptor endodomain (4/7 ICR). Transgenic expression of this molecule in CAR-PSCA T cells should invert the inhibitory effects of tumor-derived IL-4 and instead promote T cell proliferation. We now demonstrate the suppressed activity of CAR T cells in tumor-milieu conditions and the ability of CAR/ICR T cells to thrive in an IL-4-rich microenvironment, resulting in enhanced antitumor activity. Importantly, CAR/ICR T cells remained both antigen and cytokine dependent. These findings support the benefit of combining the 4/7 ICR with CAR-PSCA to treat pancreatic cancer, a PSCA-expressing tumor characterized by a dense immunosuppressive environment rich in IL-4.
Collapse
Affiliation(s)
- Somala Mohammed
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA; Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sujita Sukumaran
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA; Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pradip Bajgain
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA; Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - William E Fisher
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ann M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Juan F Vera
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA.
| |
Collapse
|
7
|
Ren J, Liu X, Fang C, Jiang S, June CH, Zhao Y. Multiplex Genome Editing to Generate Universal CAR T Cells Resistant to PD1 Inhibition. Clin Cancer Res 2016; 23:2255-2266. [PMID: 27815355 DOI: 10.1158/1078-0432.ccr-16-1300] [Citation(s) in RCA: 657] [Impact Index Per Article: 82.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 09/27/2016] [Accepted: 10/23/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Using gene-disrupted allogeneic T cells as universal effector cells provides an alternative and potentially improves current chimeric antigen receptor (CAR) T-cell therapy against cancers and infectious diseases.Experimental Design: The CRISPR/Cas9 system has recently emerged as a simple and efficient way for multiplex genome engineering. By combining lentiviral delivery of CAR and electro-transfer of Cas9 mRNA and gRNAs targeting endogenous TCR, β-2 microglobulin (B2M) and PD1 simultaneously, to generate gene-disrupted allogeneic CAR T cells deficient of TCR, HLA class I molecule and PD1.Results: The CRISPR gene-edited CAR T cells showed potent antitumor activities, both in vitro and in animal models and were as potent as non-gene-edited CAR T cells. In addition, the TCR and HLA class I double deficient T cells had reduced alloreactivity and did not cause graft-versus-host disease. Finally, simultaneous triple genome editing by adding the disruption of PD1 led to enhanced in vivo antitumor activity of the gene-disrupted CAR T cells.Conclusions: Gene-disrupted allogeneic CAR and TCR T cells could provide an alternative as a universal donor to autologous T cells, which carry difficulties and high production costs. Gene-disrupted CAR and TCR T cells with disabled checkpoint molecules may be potent effector cells against cancers and infectious diseases. Clin Cancer Res; 23(9); 2255-66. ©2016 AACR.
Collapse
Affiliation(s)
- Jiangtao Ren
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaojun Liu
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chongyun Fang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shuguang Jiang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Carl H June
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania. .,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yangbing Zhao
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania. .,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
8
|
Min WK, Na KI, Yoon JH, Heo YJ, Lee D, Kim SG, Seo JH. Affinity improvement by fine tuning of single-chain variable fragment against aflatoxin B1. Food Chem 2016; 209:312-7. [PMID: 27173568 DOI: 10.1016/j.foodchem.2016.04.085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/23/2016] [Accepted: 04/19/2016] [Indexed: 11/27/2022]
Abstract
Aflatoxin B1 (AFB1) produced in Aspergillus flavus is a major hepatocarcinogen found in foods and feed. For effective immunological detection of AFB1 at low concentrations, the development of high affinity antibody for AFB1 is required. Previously, an affinity-maturated single-chain variable fragment containing 6 mutations (scFv-M37) was isolated from an artificial mutagenic library, which showed a 9-fold higher affinity than its wild type scFv. In this study, the effect of the 6 mutated residues on the affinity improvement was characterized using surface plasmon resonance analysis, which identified a deleterious mutation (VH-A110T) located on a framework region of the scFv-M37. The back mutation of VH-A110T resulted in a 3.2-fold affinity improvement, which was attributed to decrease of dissociation rate constant (kd) in interaction between AFB1 and the back mutant scFv. The biophysical analyses using circular dichroism and gel filtration revealed that the back mutation of VH-A110T caused a subtle conformational change of the scFv toward tighter binding to AFB1.
Collapse
Affiliation(s)
- Won-Ki Min
- Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul 151-742, Republic of Korea
| | - Kang-In Na
- Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul 151-742, Republic of Korea
| | - Jung-Hyun Yoon
- Department of Biomedical Science, Youngdong University, Chungbuk 370-701, Republic of Korea
| | - Yoon-Jee Heo
- Department of Biomedical Science, Youngdong University, Chungbuk 370-701, Republic of Korea
| | - Daesang Lee
- The 5th R&D Institute-3, Agency for Defense Development, Daejeon 305-152, Republic of Korea
| | - Sung-Gun Kim
- Department of Biomedical Science, Youngdong University, Chungbuk 370-701, Republic of Korea.
| | - Jin-Ho Seo
- Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|
9
|
LIU QIONG, PANG HUA, HU XIAOLI, LI WENBO, XI JIMEI, XU LU, ZHOU JING. Construction of human single-chain variable fragment antibodies of medullary thyroid carcinoma and single photon emission computed tomography/computed tomography imaging in tumor-bearing nude mice. Oncol Rep 2015; 35:171-8. [DOI: 10.3892/or.2015.4345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/04/2015] [Indexed: 11/06/2022] Open
|
10
|
Sonn GA, Behesnilian AS, Jiang ZK, Zettlitz KA, Lepin EJ, Bentolila LA, Knowles SM, Lawrence D, Wu AM, Reiter RE. Fluorescent Image-Guided Surgery with an Anti-Prostate Stem Cell Antigen (PSCA) Diabody Enables Targeted Resection of Mouse Prostate Cancer Xenografts in Real Time. Clin Cancer Res 2015; 22:1403-12. [PMID: 26490315 DOI: 10.1158/1078-0432.ccr-15-0503] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 10/06/2015] [Indexed: 01/13/2023]
Abstract
PURPOSE The inability to visualize cancer during prostatectomy contributes to positive margins, cancer recurrence, and surgical side effects. A molecularly targeted fluorescent probe offers the potential for real-time intraoperative imaging. The goal of this study was to develop a probe for image-guided prostate cancer surgery. EXPERIMENTAL DESIGN An antibody fragment (cys-diabody, cDb) against prostate stem cell antigen (PSCA) was conjugated to a far-red fluorophore, Cy5. The integrity and binding of the probe to PSCA was confirmed by gel electrophoresis, size exclusion, and flow cytometry, respectively. Subcutaneous models of PSCA-expressing xenografts were used to assess the biodistribution and in vivo kinetics, whereas an invasive intramuscular model was utilized to explore the performance of Cy5-cDb-mediated fluorescence guidance in representative surgical scenarios. Finally, a prospective, randomized study comparing surgical resection with and without fluorescent guidance was performed to determine whether this probe could reduce the incidence of positive margins. RESULTS Cy5-cDb demonstrated excellent purity, stability, and specific binding to PSCA. In vivo imaging showed maximal signal-to-background ratios at 6 hours. In mice carrying PSCA(+) and negative (-) dual xenografts, the mean fluorescence ratio of PSCA(+/-) tumors was 4.4:1. In surgical resection experiments, residual tumors <1 mm that were missed on white light surgery were identified and resected using fluorescence guidance, which reduced the incidence of positive surgical margins (0/8) compared with white light surgery alone (7/7). CONCLUSIONS Fluorescently labeled cDb enables real-time in vivo imaging of prostate cancer xenografts in mice, and facilitates more complete tumor removal than conventional white light surgery alone.
Collapse
Affiliation(s)
- Geoffrey A Sonn
- Department of Urology, University of California, Los Angeles, California
| | | | - Ziyue Karen Jiang
- Department of Urology, University of California, Los Angeles, California
| | - Kirstin A Zettlitz
- Molecular and Medical Pharmacology, University of California, Los Angeles, California. Crump Institute for Molecular Imaging, University of California, Los Angeles, California
| | - Eric J Lepin
- Molecular and Medical Pharmacology, University of California, Los Angeles, California. Crump Institute for Molecular Imaging, University of California, Los Angeles, California
| | - Laurent A Bentolila
- Deparment of Chemistry and Biochemistry, University of California, Los Angeles, California. California NanoSystems Institute, University of California, Los Angeles, California
| | - Scott M Knowles
- Molecular and Medical Pharmacology, University of California, Los Angeles, California. Crump Institute for Molecular Imaging, University of California, Los Angeles, California
| | - Daniel Lawrence
- Molecular and Medical Pharmacology, University of California, Los Angeles, California
| | - Anna M Wu
- Molecular and Medical Pharmacology, University of California, Los Angeles, California. Crump Institute for Molecular Imaging, University of California, Los Angeles, California
| | - Robert E Reiter
- Department of Urology, University of California, Los Angeles, California.
| |
Collapse
|
11
|
Abate-Daga D, Lagisetty KH, Tran E, Zheng Z, Gattinoni L, Yu Z, Burns WR, Miermont AM, Teper Y, Rudloff U, Restifo NP, Feldman SA, Rosenberg SA, Morgan RA. A novel chimeric antigen receptor against prostate stem cell antigen mediates tumor destruction in a humanized mouse model of pancreatic cancer. Hum Gene Ther 2015; 25:1003-12. [PMID: 24694017 DOI: 10.1089/hum.2013.209] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Despite advances in the understanding of its molecular pathophysiology, pancreatic cancer remains largely incurable, highlighting the need for novel therapies. We developed a chimeric antigen receptor (CAR) specific for prostate stem cell antigen (PSCA), a glycoprotein that is overexpressed in pancreatic cancer starting at early stages of malignant transformation. To optimize the CAR design, we used antigen-recognition domains derived from mouse or human antibodies, and intracellular signaling domains containing one or two T cell costimulatory elements, in addition to CD3zeta. Comparing multiple constructs established that the CAR based on human monoclonal antibody Ha1-4.117 had the greatest reactivity in vitro. To further analyze this CAR, we developed a human pancreatic cancer xenograft model and adoptively transferred CAR-engineered T cells into animals with established tumors. CAR-engineered human lymphocytes induced significant antitumor activity, and unlike what has been described for other CARs, a second-generation CAR (containing CD28 cosignaling domain) induced a more potent antitumor effect than a third-generation CAR (containing CD28 and 41BB cosignaling domains). While our results provide evidence to support PSCA as a target antigen for CAR-based immunotherapy of pancreatic cancer, the expression of PSCA on selected normal tissues could be a source of limiting toxicity.
Collapse
Affiliation(s)
- Daniel Abate-Daga
- Surgery Branch, National Cancer Institute , National Institutes of Health, Bethesda, MD 20892
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Li K, Zettlitz KA, Lipianskaya J, Zhou Y, Marks JD, Mallick P, Reiter RE, Wu AM. A fully human scFv phage display library for rapid antibody fragment reformatting. Protein Eng Des Sel 2015; 28:307-16. [PMID: 25991864 DOI: 10.1093/protein/gzv024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 04/15/2015] [Indexed: 12/21/2022] Open
Abstract
Phage display libraries of human single-chain variable fragments (scFvs) are a reliable source of fully human antibodies for scientific and clinical applications. Frequently, scFvs form the basis of larger, bivalent formats to increase valency and avidity. A small and versatile bivalent antibody fragment is the diabody, a cross-paired scFv dimer (∼55 kDa). However, generation of diabodies from selected scFvs requires decreasing the length of the interdomain scFv linker, typically by overlap PCR. To simplify this process, we designed two scFv linkers with integrated restriction sites for easy linker length reduction (17-residue to 7-residue or 18-residue to 5-residue, respectively) and generated two fully human scFv phage display libraries. The larger library (9 × 10(9) functional members) was employed for selection against a model antigen, human N-cadherin, yielding novel scFv clones with low nanomolar monovalent affinities. ScFv clones from both libraries were reformatted into diabodies by restriction enzyme digestion and re-ligation. Size-exclusion chromatography analysis confirmed the proper dimerization of most of the diabodies. In conclusion, these specially designed scFv phage display libraries allow us to rapidly reformat the selected scFvs into diabodies, which can greatly accelerate early stage antibody development when bivalent fragments are needed for candidate screening.
Collapse
Affiliation(s)
- Keyu Li
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 570 Westwood Plaza, Box 951770, Los Angeles, CA 90095, USA
| | - Kirstin A Zettlitz
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 570 Westwood Plaza, Box 951770, Los Angeles, CA 90095, USA
| | - Julia Lipianskaya
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 570 Westwood Plaza, Box 951770, Los Angeles, CA 90095, USA
| | - Yu Zhou
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco General Hospital, 1001 Potrero Ave, Rm 3C-38, San Francisco, CA 94110, USA
| | - James D Marks
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco General Hospital, 1001 Potrero Ave, Rm 3C-38, San Francisco, CA 94110, USA
| | - Parag Mallick
- Canary Center for Cancer Early Detection, Stanford University, Palo Alto, CA 94304, USA
| | - Robert E Reiter
- Department of Urology, UCLA, Los Angeles, CA 90095, USA Molecular Biology Institute at UCLA, Los Angeles, CA 90095, USA Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA 90095, USA
| | - Anna M Wu
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 570 Westwood Plaza, Box 951770, Los Angeles, CA 90095, USA Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
13
|
Biological evaluation of 131I- and CF750-labeled Dmab(scFv)-Fc antibodies for xenograft imaging of CD25-positive tumors. BIOMED RESEARCH INTERNATIONAL 2014; 2014:459676. [PMID: 24864244 PMCID: PMC4017786 DOI: 10.1155/2014/459676] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/17/2014] [Accepted: 03/17/2014] [Indexed: 02/05/2023]
Abstract
A Dmab(scFv)-Fc antibody containing the single chain variable fragment of a humanized daclizumab antibody and the Fc fragment of a human IgG1 antibody was produced via recombinant expression in Pichia pastoris. The Dmab(scFv)-Fc antibody forms a dimer in solution, and it specifically binds CD25-positive tumor cells and tumor tissues. For tumor imaging, the Dmab(scFv)-Fc antibody was labeled with the 131I isotope and CF750 fluorescent dye, respectively. After intravenous injection of mice bearing CD25-positive tumor xenografts, tumor uptake of the (131)I-Dmab(scFv)-Fc antibody was visible at 1 h, and clear images were obtained at 5 h using SPECT/CT. After systemic administration of the CF750-Dmab(scFv)-Fc antibody, tumor uptake was present as early as 1 h, and tumor xenografts could be kinetically imaged within 9 h after injection. These results indicate that the Dmab(scFv)-Fc antibody rapidly and specifically targets CD25-positive tumor cells, suggesting the potential of this antibody as an imaging agent for the diagnosis of lymphomatous-type ATLL.
Collapse
|
14
|
Antiproliferative and apoptotic effects of a specific antiprostate stem cell single chain antibody on human prostate cancer cells. JOURNAL OF ONCOLOGY 2013; 2013:839831. [PMID: 24391668 PMCID: PMC3872421 DOI: 10.1155/2013/839831] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 11/14/2013] [Indexed: 02/04/2023]
Abstract
Prostate stem cell antigen (PSCA) is a highly glycosylated cell surface protein which is overexpressed in several malignancies including prostate, pancreas, and urinary bladder cancers. Tumor suppression has been reported by anti-PSCA antibody. Small and high affinity single chain antibodies (scFv) have been introduced as effective agents for cancer immunotargeting approaches. In the present study, we used a phage antibody display library of scFv and selected two antibodies against two immunodominant epitopes of PSCA by panning process. The reactivity of the scFvs for the corresponding epitopes was determined by phage ELISA. The binding specificity of antibodies to PSCA-expressing prostate cancer cell line, DU-145, was analyzed by flow cytometry. The antiproliferative and apoptotic induction effects were evaluated by MTT and Annexin-V assays, respectively. Results represented functional scFv C5-II which could bind specifically to DU-145 cells and significantly inhibited the proliferation of these cells (61%) with no effect on PSCA-negative cells. The antibody also induced apoptosis in the PSCA expressing cells. The percentage of the apoptotic cells after 24 hrs of exposure to 500 scFv/cell was 33.80%. These results demonstrate that the functional anti-PSCA scFv C5-II has the potential to be considered as a new agent for targeted therapy of prostate cancer.
Collapse
|
15
|
Leyton JV, Olafsen T, Lepin EJ, Hahm S, Fonge H, Reiter RE, Wu AM. Positron Emission Tomographic Imaging of Iodine 124 Anti–Prostate Stem Cell Antigen–Engineered Antibody Fragments in LAPC-9 Tumor–Bearing Severe Combined Immunodeficiency Mice. Mol Imaging 2013. [DOI: 10.2310/7290.2012.00033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Jeffrey V. Leyton
- From the Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA; Department of Urology, UCLA, 66-134 Center for the Health Sciences, Los Angeles, CA; and Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON
| | - Tove Olafsen
- From the Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA; Department of Urology, UCLA, 66-134 Center for the Health Sciences, Los Angeles, CA; and Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON
| | - Eric J.M. Lepin
- From the Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA; Department of Urology, UCLA, 66-134 Center for the Health Sciences, Los Angeles, CA; and Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON
| | - Scott Hahm
- From the Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA; Department of Urology, UCLA, 66-134 Center for the Health Sciences, Los Angeles, CA; and Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON
| | - Humphrey Fonge
- From the Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA; Department of Urology, UCLA, 66-134 Center for the Health Sciences, Los Angeles, CA; and Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON
| | - Robert E. Reiter
- From the Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA; Department of Urology, UCLA, 66-134 Center for the Health Sciences, Los Angeles, CA; and Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON
| | - Anna M. Wu
- From the Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA; Department of Urology, UCLA, 66-134 Center for the Health Sciences, Los Angeles, CA; and Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON
| |
Collapse
|
16
|
Knowles SM, Wu AM. Advances in immuno-positron emission tomography: antibodies for molecular imaging in oncology. J Clin Oncol 2012; 30:3884-92. [PMID: 22987087 PMCID: PMC3478579 DOI: 10.1200/jco.2012.42.4887] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Accepted: 07/20/2012] [Indexed: 01/20/2023] Open
Abstract
Identification of cancer cell-surface biomarkers and advances in antibody engineering have led to a sharp increase in the development of therapeutic antibodies. These same advances have led to a new generation of radiolabeled antibodies and antibody fragments that can be used as cancer-specific imaging agents, allowing quantitative imaging of cell-surface protein expression in vivo. Immuno-positron emission tomography (immunoPET) imaging with intact antibodies has shown success clinically in diagnosing and staging cancer. Engineered antibody fragments, such as diabodies, minibodies, and single-chain Fv (scFv) -Fc, have been successfully employed for immunoPET imaging of cancer cell-surface biomarkers in preclinical models and are poised to bring same-day imaging into clinical development. ImmunoPET can potentially provide a noninvasive approach for obtaining target-specific information useful for titrating doses for radioimmunotherapy, for patient risk stratification and selection of targeted therapies, for evaluating response to therapy, and for predicting adverse effects, thus contributing to the ongoing development of personalized cancer treatment.
Collapse
Affiliation(s)
- Scott M. Knowles
- All authors: David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA
| | - Anna M. Wu
- All authors: David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA
| |
Collapse
|
17
|
An engineered cysteine-modified diabody for imaging activated leukocyte cell adhesion molecule (ALCAM)-positive tumors. Mol Imaging Biol 2012; 14:336-47. [PMID: 21630083 DOI: 10.1007/s11307-011-0500-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE The purpose of this study was to generate and evaluate a positron emission tomography (PET) radiotracer targeting activated leukocyte cell adhesion molecule (ALCAM). PROCEDURES A human anti-ALCAM single chain variable fragment was reformatted to produce a covalent dimer, termed a cys-diabody (CysDb). Purified CysDb was characterized by gel electrophoresis and size exclusion chromatography, and immunoreactivity was assessed by flow cytometry and immunofluorescence. Targeting and imaging of ALCAM-positive tumors using (64)Cu-DOTA-CysDb were evaluated in mice bearing human pancreatic adenocarcinoma xenografts (HPAF-II or BxPC-3). RESULTS CysDb binds specifically to ALCAM-positive cells in vitro with an apparent affinity in the range of 1-3 nM. MicroPET images at 4 h showed specific targeting of positive tumors in vivo, a finding confirmed by biodistribution analysis, with positive/negative tumor ratios of 1.9 ± 0.6 and 2.4 ± 0.6, and positive tumor/blood ratios of 2.5 ± 0.9 and 2.9 ± 0.6 (HPAF-II and BxPC-3, respectively). CONCLUSIONS Successful imaging with (64)Cu-DOTA-CysDb in animal models suggests further investigation of ALCAM as an imaging biomarker is warranted.
Collapse
|
18
|
Abstract
OBJECTIVE Recent advances in the fundamental understanding of the complex biology of prostate cancer have provided an increasing number of potential targets for imaging and treatment. The imaging evaluation of prostate cancer needs to be tailored to the various phases of this remarkably heterogeneous disease. CONCLUSION In this article, I review the current state of affairs on a range of PET radiotracers for potential use in the imaging evaluation of men with prostate cancer.
Collapse
|
19
|
Rodríguez-Rodríguez ER, Ledezma-Candanoza LM, Contreras-Ferrat LG, Olamendi-Portugal T, Possani LD, Becerril B, Riaño-Umbarila L. A Single Mutation in Framework 2 of the Heavy Variable Domain Improves the Properties of a Diabody and a Related Single-Chain Antibody. J Mol Biol 2012; 423:337-50. [DOI: 10.1016/j.jmb.2012.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 07/02/2012] [Accepted: 07/06/2012] [Indexed: 10/28/2022]
|
20
|
Abstract
Combination chemotherapy and nanoparticle drug delivery are two areas that have shown significant promise in cancer treatment. Combined therapy of two or more drugs promotes synergism among the different drugs against cancer cells and suppresses drug resistance through distinct mechanisms of action. Nanoparticle drug delivery, on the other hand, enhances therapeutic effectiveness and reduces side effects of the drug payloads by improving their pharmacokinetics. These two active research fields have been recently merged to further improve the efficacy of cancer therapeutics. This review article summarizes the recent efforts in developing nanoparticle platforms to concurrently deliver multiple types of drugs for combination chemotherapy. We also highlight the challenges and design specifications that need to be considered in optimizing nanoparticle-based combination chemotherapy.
Collapse
|
21
|
Molecular imaging in tracking tumor stem-like cells. J Biomed Biotechnol 2012; 2012:420364. [PMID: 22570529 PMCID: PMC3335324 DOI: 10.1155/2012/420364] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 02/10/2012] [Indexed: 12/18/2022] Open
Abstract
Cancer remains a major public health problem in many countries. It was found to contain a subset of cancer stem cells (CSCs) that are capable of proliferation and self-renewal, and differentiation into various types of cancer cells. CSCs often display characteristics of chemotherapy resistance and radiotherapy resistance. Numerous putative biomarkers of CSCs are currently identified including CD133, CD44, CD24, ALDH (aldehyde dehydrogenase), and ABCG2. Interestingly, no single marker is exclusively expressed by CSCs. Thus, the various combinations of different biomarkers will be possible to identify CSCs, and considerable work is being done to recognize new ones. In order to demonstrate the mechanisms of resistance and response to therapy and predict the outcome as well as prognosis, the ways to track and identify CSCs will be extremely important. The technologies of molecular imaging will reveal mechanisms of cancer progression and provide visual targets for novel therapeutics. Limited studies were investigated on the detection of various types of CSCs by molecular imaging. Although the tracking of circulating CSCs is still hampered by technological challenges, personalized diagnosis and therapies of cancers are expected to be established based on increased understanding of molecular imaging of cancer stem-like cells biomarkers.
Collapse
|
22
|
Hall MA, Kwon S, Robinson H, Lachance PA, Azhdarinia A, Ranganathan R, Price RE, Chan W, Sevick-Muraca EM. Imaging prostate cancer lymph node metastases with a multimodality contrast agent. Prostate 2012; 72:129-46. [PMID: 21538422 DOI: 10.1002/pros.21413] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 04/06/2011] [Indexed: 01/05/2023]
Abstract
BACKGROUND Methods to detect lymph node (LN) metastases in prostate cancer (PCa) are limited. Pelvic LN dissection is commonly performed during prostatectomy, but often followed by morbid complications. More refined methods for detecting LN invasion are needed. METHODS We developed a dual-labeled targeting agent having a near-infrared (NIR) fluorophore for intraoperative guidance, and a conventional radiotracer for detection of LN metastasis. Nu/Nu mice were orthotopically implanted with DsRed-expressing human PCa (PC3) cells. Antibody (Ab) specific for epithelial cell adhesion molecule was conjugated to DOTA, IRDye 800CW, and radiolabeled with (64) Cu. Dual-labeled Ab was administered intravenously at 10-12 weeks post-implantation, and positron emission tomography/computed tomography (PET/CT) and fluorescence imaging were performed within 18-24 hr. RESULTS Metastasis to lumbar LNs was detected by DsRed fluorescence imaging, as well as pathology, in 75% of mice having pathology-confirmed primary prostate tumors. These metastases were also detected by NIR fluorescence imaging. In some cases, metastases to sciatic, medial, renal, and axillary nodes were also detected. For all LNs examined, no significant differences were found between the percentages of metastases detected by NIR imaging (63%) and µPET/CT (64%) (P = 0.93), or between those detected by DsRed imaging (25%) and pathological examination (19%) (P = 0.12). CONCLUSION This study demonstrates that a multimodality contrast agent is useful for early detection of metastatic disease, and has applications for intraoperative PCa treatment. Further agent optimization is necessary to enhance specificity, and provide validation for prostate and other LN metastasizing epithelial cancers.
Collapse
Affiliation(s)
- Mary A Hall
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Katari UL, Keirnan JM, Worth AC, Hodges SE, Leen AM, Fisher WE, Vera JF. Engineered T cells for pancreatic cancer treatment. HPB (Oxford) 2011; 13:643-50. [PMID: 21843265 PMCID: PMC3183449 DOI: 10.1111/j.1477-2574.2011.00344.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Conventional chemotherapy and radiotherapy produce marginal survival benefits in pancreatic cancer, underscoring the need for novel therapies. The aim of this study is to develop an adoptive T cell transfer approach to target tumours expressing prostate stem cell antigen (PSCA), a tumour-associated antigen that is frequently expressed by pancreatic cancer cells. METHODS Expression of PSCA on cell lines and primary tumour samples was confirmed by immunohistochemistry. Healthy donor- and patient-derived T cells were isolated, activated in vitro using CD3/CD28, and transduced with a retroviral vector encoding a chimeric antigen receptor (CAR) targeting PSCA. The ability of these cells to kill tumour cells was analysed by chromium-51 (Cr(51)) release. RESULTS Prostate stem cell antigen was expressed on >70% of the primary tumour samples screened. Activated, CAR-modified T cells could be readily generated in clinically relevant numbers and were specifically able to kill PSCA-expressing pancreatic cancer cell lines with no non-specific killing of PSCA-negative target cells, thus indicating the potential efficacy and safety of this approach. CONCLUSIONS Prostate stem cell antigen is frequently expressed on pancreatic cancer cells and can be targeted for immune-mediated destruction using CAR-modified, adoptively transferred T cells. The safety and efficacy of this approach indicate that it deserves further study and may represent a promising novel treatment for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Usha L Katari
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and the Methodist HospitalHouston, TX, USA
| | - Jacqueline M Keirnan
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and the Methodist HospitalHouston, TX, USA
| | - Anna C Worth
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and the Methodist HospitalHouston, TX, USA
| | - Sally E Hodges
- Elkin's Pancreas Center, Baylor College of MedicineHouston, TX, USA
| | - Ann M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and the Methodist HospitalHouston, TX, USA
| | - William E Fisher
- Elkin's Pancreas Center, Baylor College of MedicineHouston, TX, USA
| | - Juan F Vera
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and the Methodist HospitalHouston, TX, USA
| |
Collapse
|
24
|
Liu K, Wang MW, Lin WY, Phung DL, Girgis MD, Wu AM, Tomlinson JS, Shen CKF. Molecular Imaging Probe Development using Microfluidics. Curr Org Synth 2011; 8:473-487. [PMID: 22977436 DOI: 10.2174/157017911796117205] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In this manuscript, we review the latest advancement of microfluidics in molecular imaging probe development. Due to increasing needs for medical imaging, high demand for many types of molecular imaging probes will have to be met by exploiting novel chemistry/radiochemistry and engineering technologies to improve the production and development of suitable probes. The microfluidic-based probe synthesis is currently attracting a great deal of interest because of their potential to deliver many advantages over conventional systems. Numerous chemical reactions have been successfully performed in micro-reactors and the results convincingly demonstrate with great benefits to aid synthetic procedures, such as purer products, higher yields, shorter reaction times compared to the corresponding batch/macroscale reactions, and more benign reaction conditions. Several 'proof-of-principle' examples of molecular imaging probe syntheses using microfluidics, along with basics of device architecture and operation, and their potential limitations are discussed here.
Collapse
Affiliation(s)
- Kan Liu
- College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, 430073, China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Liu K, Lepin EJ, Wang MW, Guo F, Lin WY, Chen YC, Sirk SJ, Olma S, Phelps ME, Zhao XZ, Tseng HR, van Dam RM, Wu AM, Shen CKF. Microfluidic-based 18F-labeling of biomolecules for immuno-positron emission tomography. Mol Imaging 2011; 10:168-7. [PMID: 21496447 PMCID: PMC3163899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023] Open
Abstract
Methods for tagging biomolecules with fluorine 18 as immuno-positron emission tomography (immunoPET) tracers require tedious optimization of radiolabeling conditions and can consume large amounts of scarce biomolecules. We describe an improved method using a digital microfluidic droplet generation (DMDG) chip, which provides computer-controlled metering and mixing of 18F tag, biomolecule, and buffer in defined ratios, allowing rapid scouting of reaction conditions in nanoliter volumes. The identified optimized conditions were then translated to bench-scale 18F labeling of a cancer-specific engineered antibody fragments, enabling microPET imaging of tumors in xenografted mice at 0.5 to 4 hours postinjection.
Collapse
Affiliation(s)
- Kan Liu
- College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, 430000, China
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
- California Nanosystems Institute, 570 Westwood Plaza, Los Angeles, California 90095, USA
| | - Eric J. Lepin
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
- California Nanosystems Institute, 570 Westwood Plaza, Los Angeles, California 90095, USA
| | - Ming-Wei Wang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
- California Nanosystems Institute, 570 Westwood Plaza, Los Angeles, California 90095, USA
- PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, 200032, China
| | - Feng Guo
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
- California Nanosystems Institute, 570 Westwood Plaza, Los Angeles, California 90095, USA
- Department of Physics, School of Physics, and Center of Nanoscience and Nanotechnology, Wuhan University, Wuhan 430072, China
| | - Wei-Yu Lin
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
- California Nanosystems Institute, 570 Westwood Plaza, Los Angeles, California 90095, USA
| | - Yi-Chun Chen
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
- California Nanosystems Institute, 570 Westwood Plaza, Los Angeles, California 90095, USA
| | - Shannon J. Sirk
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
- California Nanosystems Institute, 570 Westwood Plaza, Los Angeles, California 90095, USA
| | - Sebastian Olma
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
| | - Michael E. Phelps
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
- California Nanosystems Institute, 570 Westwood Plaza, Los Angeles, California 90095, USA
| | - Xing-Zhong Zhao
- Department of Physics, School of Physics, and Center of Nanoscience and Nanotechnology, Wuhan University, Wuhan 430072, China
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
- California Nanosystems Institute, 570 Westwood Plaza, Los Angeles, California 90095, USA
| | - R. Michael van Dam
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
- California Nanosystems Institute, 570 Westwood Plaza, Los Angeles, California 90095, USA
| | - Anna M. Wu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
- California Nanosystems Institute, 570 Westwood Plaza, Los Angeles, California 90095, USA
| | - Clifton K.-F. Shen
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, 23–120 Center for Health Science, Los Angeles, California 90095, USA
- Crump Institute for Molecular Imaging, 570 Westwood Plaza, Los Angeles, California 90095, USA
- California Nanosystems Institute, 570 Westwood Plaza, Los Angeles, California 90095, USA
| |
Collapse
|
26
|
Liu K, Lepin EJ, Wang MW, Guo F, Lin WY, Chen YC, Sirk SJ, Olma S, Phelps ME, Zhao XZ, Tseng HR, van Dam RM, Wu AM, Shen CKF. Microfluidic-Based
18
F-Labeling of Biomolecules for Immuno–Positron Emission Tomography. Mol Imaging 2011. [DOI: 10.2310/7290.2010.00043] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Kan Liu
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - Eric J. Lepin
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - Ming-Wei Wang
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - Feng Guo
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - Wei-Yu Lin
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - Yi-Chun Chen
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - Shannon J. Sirk
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - Sebastian Olma
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - Michael E. Phelps
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - Xing-Zhong Zhao
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - Hsian-Rong Tseng
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - R. Michael van Dam
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - Anna M. Wu
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| | - Clifton K.-F. Shen
- From the College of Electronics and Information Engineering, Wuhan Textile University, Wuhan, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA; Crump Institute for Molecular Imaging, Los Angeles, CA; California Nanosystems Institute, Los Angeles, CA; PET Center and Department of Nuclear Medicine, Cancer Hospital, Fudan University, Shanghai, China; and Department of Physics, School of Physics, and Center of
| |
Collapse
|
27
|
Lopci E, Chiti A, Castellani MR, Pepe G, Antunovic L, Fanti S, Bombardieri E. Matched pairs dosimetry: 124I/131I metaiodobenzylguanidine and 124I/131I and 86Y/90Y antibodies. Eur J Nucl Med Mol Imaging 2011; 38 Suppl 1:S28-40. [PMID: 21484381 DOI: 10.1007/s00259-011-1772-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 02/22/2011] [Indexed: 11/30/2022]
Abstract
The technological advances in imaging and production of radiopharmaceuticals are driving an innovative way of evaluating the targets for antineoplastic therapies. Besides the use of imaging to better delineate the volume of external beam radiation therapy in oncology, modern imaging techniques are able to identify targets for highly specific medical therapies, using chemotherapeutic drugs and antiangiogenesis molecules. Moreover, radionuclide imaging is able to select targets for radionuclide therapy and to give the way to in vivo dose calculation to target tissues and to critical organs. This contribution reports the main studies published on matched pairs dosimetry with (124)I/(131)I- and (86)Y/(90)Y-labelled radiopharmaceuticals, with an emphasis on metaiodobenzylguanidine (MIBG) and monoclonal antibodies.
Collapse
Affiliation(s)
- Egesta Lopci
- Policlinico S.Orsola-Malpighi and University of Bologna, Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
28
|
Li L, Crow D, Turatti F, Bading JR, Anderson AL, Poku E, Yazaki PJ, Carmichael J, Leong D, Wheatcroft D, Wheatcroft MP, Raubitschek AA, Hudson PJ, Colcher D, Shively JE. Site-specific conjugation of monodispersed DOTA-PEGn to a thiolated diabody reveals the effect of increasing peg size on kidney clearance and tumor uptake with improved 64-copper PET imaging. Bioconjug Chem 2011; 22:709-16. [PMID: 21395337 DOI: 10.1021/bc100464e] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Optimal PET imaging of tumors with radiolabeled engineered antibodies requires, among other parameters, matching blood clearance and tumor uptake with the half-life of the engineered antibody. Although diabodies have favorable molecular sizes (50 kDa) for rapid blood clearance (t(1/2) = 30-60 min) and are bivalent, thereby increasing tumor uptake, they exhibit substantial kidney uptake as their major route of clearance, which is especially evident when they are labeled with the PET isotope (64)Cu (t(1/2) = 12 h). To overcome this drawback, diabodies may be conjugated to PEG, a modification that increases the apparent molecular size of the diabody and reduces kidney uptake without adversely affecting tumor uptake or the tumor to blood ratio. We show here that site-specific attachment of monodispersed PEGn of increasing molecular size (n = 12, 24, and 48) can uniformly increase the apparent molecular size of the PEG-diabody conjugate, decrease kidney uptake, and increase tumor uptake, the latter due to the increased residence time of the conjugate in the blood. Since the monodispersed PEGs were preconjugated to the chelator DOTA, the conjugates were able to bind radiometals such as (111)In and (64)Cu that can be used for SPECT and PET imaging, respectively. To allow conjugation of the DOTA-PEG to the diabody, the DOTA-PEG incorporated a terminal cysteine conjugated to a vinyl sulfone moiety. In order to control the conjugation chemistry, we have engineered a surface thiolated diabody that incorporates two cysteines per monomer (four per diabody). The thiolated diabody was expressed and purified from bacterial fermentation and only needs to be reduced prior to conjugation to the DOTA-PEGn-Cys-VS. This novel imaging agent (a diabody with DOTA-PEG48-Cys-VS attached to introduced thiols) gave up to 80%ID/g of tumor uptake with a tumor to blood ratio (T/B) of 8 at 24 h when radiolabeled with (111)In and 37.9% ID/g of tumor uptake (T/B = 8) at 44 h when radiolabeled with (64)Cu in PET imaging in an animal model. Tumor uptake was significantly improved from the 50% ID/g at 24 h observed with diabodies that were pegylated on surface lysine residues. Importantly, there was no loss of immunoreactivity of the site-specific Cys-conjugated diabody to its antigen (TAG-72) compared to the parent, unconjugated diabody. We propose that thiolated diabodies conjugated to DOTAylated monodisperse PEGs have the potential for superior SPECT and PET imaging in a clinical setting.
Collapse
Affiliation(s)
- Lin Li
- Department of Immunology, Beckman Research Institute of City of Hope , Duarte, California 91010, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Quantitative and specific molecular imaging of cancer with labeled engineered monoclonal antibody fragments. Ther Deliv 2011; 2:345-58. [DOI: 10.4155/tde.10.79] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The high target specificity of antibodies and related constructs makes them excellent scaffolds for molecular-imaging probes. Quantitative data on biodistribution and pharmacokinetics can be acquired by radiolabeling these agents. Such studies demonstrate prolonged circulation times and resulting nonspecific accumulation with high background signal using antibody-based agents. Antibody fragments demonstrate more rapid clearance, but lower tumor uptake. Optical labeling of antibodies provides a basis for developing activatable probes that can image antigens with very high specificity, potentially allowing for the simultaneous visualization of multiple targets. While radioimmunoimaging provides valuable whole-body, quantitative information, activatable optical antibody-based agents could generate real-time diagnostic and prognostic information about near-surface lesions at high-spatial and temporal resolution without requiring ionizing radiation.
Collapse
|
30
|
Abstract
Noninvasive molecular imaging approaches include nuclear, optical, magnetic resonance imaging, computed tomography, ultrasound, and photoacoustic imaging, which require accumulation of a signal delivered by a probe at the target site. Monoclonal antibodies are high affinity molecules that can be used for specific, high signal delivery to cell surface molecules. However, their long circulation time in blood makes them unsuitable as imaging probes. Efforts to improve antibodies pharmacokinetics without compromising affinity and specificity have been made through protein engineering. Antibody variants that differ in antigen binding sites and size have been generated and evaluated as imaging probes to target tissues of interest. Fast clearing fragments, such as single-chain variable fragment (scFv; 25 kDa), with 1 antigen-binding site (monovalent) demonstrated low accumulation in tumors because of the low exposure time to the target. Using scFv as building block to produce larger, bivalent fragments, such as scFv dimers (diabodies, 50 kDa) and scFv-fusion proteins (80 kDa minibodies and 105 kDa scFv-Fc), resulted in higher tumor accumulation because of their longer residence time in blood. Imaging studies with these fragments after radiolabeling have demonstrated excellent, high-contrast images in gamma cameras and positron emission tomography scanners. Several studies have also investigated antibody fragments conjugated to fluorescence (near infrared dyes), bioluminescence (luciferases), and quantum dots for optical imaging and iron oxides nanoparticles for magnetic resonance imaging. However, these studies indicate that there are several factors that influence successful targeting and imaging. These include stability of the antibody fragment, the labeling chemistry (direct or indirect), whether critical residues are modified, the number of antigen expressed on the cell, and whether the target has a rapid recycling rate or internalizes upon binding. The preclinical data presented are compelling, and it is evident that antibody-based molecular imaging tracers will play an important future role in the diagnosis and management of cancer and other diseases.
Collapse
Affiliation(s)
- Tove Olafsen
- UCLA Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA.
| | | |
Collapse
|
31
|
Saeki N, Gu J, Yoshida T, Wu X. Prostate stem cell antigen: a Jekyll and Hyde molecule? Clin Cancer Res 2010; 16:3533-8. [PMID: 20501618 DOI: 10.1158/1078-0432.ccr-09-3169] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prostate stem cell antigen (PSCA) is a glycosylphosphatidylinositol (GPI)-anchored cell surface protein. Although PSCA is thought to be involved in intracellular signaling, much remains unknown about its physiological function and regulatory mechanism in normal and cancer cells. It is up-regulated in several major cancers including prostate, bladder, and pancreatic cancers. The expression of PSCA is positively correlated with advanced clinical stage and metastasis in prostate cancers and is also associated with malignant progression of premalignant prostate lesions. Therefore, PSCA has been proposed as a biomarker of diagnosis and prognosis, as well as a target of therapy for these cancers. In addition, PSCA has also shown clinical potential in immunotherapy as a prostate-specific antigen, which, when presented by dendritic cells, may elicit strong tumor-specific immunity. In contrast, PSCA is down-regulated in esophageal and gastric cancer and may have a tumor-suppressing function in the gastric epithelium. Recent exciting findings that genetic variations of PSCA conferred increased risks of gastric cancer and bladder cancer have opened up a new avenue of research about the pathological function of PSCA. PSCA seems to be a Jekyll and Hyde molecule that plays differential roles, tumor promoting or suppressing, depending on the cellular context.
Collapse
Affiliation(s)
- Norihisa Saeki
- Genetics Division, National Cancer Center Research Institute, Tokyo, Japan
| | | | | | | |
Collapse
|
32
|
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: 1.9] [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
|
33
|
Olafsen T, Sirk SJ, Betting DJ, Kenanova VE, Bauer KB, Ladno W, Raubitschek AA, Timmerman JM, Wu AM. ImmunoPET imaging of B-cell lymphoma using 124I-anti-CD20 scFv dimers (diabodies). Protein Eng Des Sel 2010; 23:243-9. [PMID: 20053640 DOI: 10.1093/protein/gzp081] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Rapid clearing engineered antibody fragments for immunoPET promise high sensitivity at early time points. Here, tumor targeting of anti-CD20 diabodies (scFv dimers) for detection of low-grade B-cell lymphomas were evaluated. In addition, the effect of linker length on oligomerization of the diabody was investigated. Four rituximab scFv variants in the V(L)-V(H) orientation with different linker lengths between the V domains (scFv-1, scFv-3, scFv-5, scFv-8), plus the scFv-5 with a C-terminal cysteine (Cys-Db) for site-specific modification were generated. The scFv-8 and Cys-Db were radioiodinated with (124)I for PET imaging, and biodistribution of (131)I-Cys-Db was carried out at 2, 4 10 and 20 h. The five anti-CD20 scFv variants were expressed as fully functional dimers. Shortening the linker to three or one residue did not produce higher order of multimers. Both (124)I-labeled scFv-8 and Cys-Db exhibited similar tumor targeting at 8 h post injection, with significantly higher uptakes than in control tumors (P < 0.05). At 20 h, less than 1% ID/g of (131)I-labeled Cys-Db was present in tumors and tissues. Specific tumor targeting and high contrast images were achieved with the anti-CD20 diabodies. These agents extend the repertoire of reagents that can potentially be used to improve detection of low-grade lymphomas.
Collapse
Affiliation(s)
- Tove Olafsen
- Department of Molecular and Medical Pharmacology, UCLA Crump Institute for Molecular Imaging, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Positron emission tomography imaging of prostate cancer. Amino Acids 2009; 39:11-27. [PMID: 19946787 DOI: 10.1007/s00726-009-0394-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Accepted: 11/11/2009] [Indexed: 01/01/2023]
Abstract
Prostate cancer (PCa) is the second leading cause of cancer death among men in the United States. Positron emission tomography (PET), a non-invasive, sensitive, and quantitative imaging technique, can facilitate personalized management of PCa patients. There are two critical needs for PET imaging of PCa, early detection of primary lesions and accurate imaging of PCa bone metastasis, the predominant cause of death in PCa. Because the most widely used PET tracer in the clinic, (18)F-fluoro-2-deoxy-2-D-glucose ((18)F-FDG), does not meet these needs, a wide variety of PET tracers have been developed for PCa imaging that span an enormous size range from small molecules to intact antibodies. In this review, we will first summarize small-molecule-based PET tracers for PCa imaging, which measure certain biological events, such as cell membrane metabolism, fatty acid synthesis, and receptor expression. Next, we will discuss radiolabeled amino acid derivatives (e.g. methionine, leucine, tryptophan, and cysteine analogs), which are primarily based on the increased amino acid transport of PCa cells. Peptide-based tracers for PET imaging of PCa, mostly based on the bombesin peptide and its derivatives which bind to the gastrin-releasing peptide receptor, will then be presented in detail. We will also cover radiolabeled antibodies and antibody fragments (e.g. diabodies and minibodies) for PET imaging of PCa, targeting integrin alpha(v)beta(3), EphA2, the epidermal growth factor receptor, or the prostate stem cell antigen. Lastly, we will identify future directions for the development of novel PET tracers for PCa imaging, which may eventually lead to personalized management of PCa patients.
Collapse
|
35
|
Ahlskog JKJ, Schliemann C, Mårlind J, Qureshi U, Ammar A, Pedley RB, Neri D. Human monoclonal antibodies targeting carbonic anhydrase IX for the molecular imaging of hypoxic regions in solid tumours. Br J Cancer 2009; 101:645-57. [PMID: 19623173 PMCID: PMC2736829 DOI: 10.1038/sj.bjc.6605200] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background: Hypoxia, which is commonly observed in areas of primary tumours and of metastases, influences response to treatment. However, its characterisation has so far mainly been restricted to the ex vivo analysis of tumour sections using monoclonal antibodies specific to carbonic anhydrase IX (CA IX) or by pimonidazole staining, after the intravenous administration of this 2-nitroimidazole compound in experimental animal models. Methods: In this study, we describe the generation of high-affinity human monoclonal antibodies (A3 and CC7) specific to human CA IX, using phage technology. Results: These antibodies were able to stain CA IX ex vivo and to target the cognate antigen in vivo. In one of the two animal models of colorectal cancer studied (LS174T), CA IX imaging closely matched pimonidazole staining, with a preferential staining of tumour areas characterised by little vascularity and low perfusion. In contrast, in a second animal model (SW1222), distinct staining patterns were observed for pimonidazole and CA IX targeting. We observed a complementary pattern of tumour regions targeted in vivo by the clinical-stage vascular-targeting antibody L19 and the anti-CA IX antibody A3, indicating that a homogenous pattern of in vivo tumour targeting could be achieved by a combination of the two antibodies. Conclusion: The new human anti-CA IX antibodies are expected to be non-immunogenic in patients with cancer and may serve as broadly applicable reagents for the non-invasive imaging of hypoxia and for pharmacodelivery applications.
Collapse
Affiliation(s)
- J K J Ahlskog
- Department of Chemistry and Applied Biosciences, ETH Zürich, Wolfgang-Pauli-Strasse 10, Zurich CH-8093, Switzerland
| | | | | | | | | | | | | |
Collapse
|