ABSTRACT
PURPOSE: Programmed cell death protein ligand 1 (PD-L1) is a crucial biomarker for immunotherapy. However, nearly 70% of patients do not respond to PD-L1 immune checkpoint therapy. Accurate monitoring of PD-L1 expression and quantification of target binding during treatment are essential. In this study, a series of small-molecule radiotracers were developed to assess PD-L1 expression and direct immunotherapy. METHODS: Radiotracers of [68Ga]Ga-D-PMED, [68Ga]Ga-D-PEG-PMED, and [68Ga]Ga-D-pep-PMED were designed based on a 2-methyl-3-biphenyl methanol scaffold and successfully synthesized. Cellular experiments and molecular docking assays were performed to determine their specificity for PD-L1. PD-L1 status was investigated via positron emission tomography (PET) imaging in MC38 tumor models. PET imaging of [68Ga]Ga-D-pep-PMED was performed to noninvasively quantify PD-L1 blocking using an anti-mouse PD-L1 antibody (PD-L1 mAb). RESULTS: The radiosyntheses of [68Ga]Ga-D-PMED, [68Ga]Ga-D-PEG-PMED, and [68Ga]Ga-D-pep-PMED were achieved with radiochemical yields of 87 ± 6%, 82 ± 4%, and 79 ± 9%, respectively. In vitro competition assays demonstrated their high affinities (the IC50 values of [68Ga]Ga-D-PMED, [68Ga]Ga-D-PEG-PMED, and [68Ga]Ga-D-pep-PMED were 90.66 ± 1.24, 160.8 ± 1.35, and 51.6 ± 1.32 nM, respectively). At 120 min postinjection (p.i.) of the radiotracers, MC38 tumors displayed optimized tumor-to-muscle ratios for all radioligands. Owing to its hydrophilic modification, [68Ga]Ga-D-pep-PMED had the highest target-to-nontarget (T/NT) ratio of approximately 6.2 ± 1.2. Interestingly, the tumor/liver ratio was hardly affected by different concentrations of the inhibitor BMS202. We then evaluated the impacts of dose and time on accessible PD-L1 levels in the tumor during anti-mouse PD-L1 antibody treatment. The tumor uptake of [68Ga]Ga-D-pep-PMED significantly decreased with increasing PD-L1 mAb dose. Moreover, after 8 days of treatment with a single antibody, the uptake of [68Ga]Ga-D-pep-PMED in the tumor significantly increased but remained lower than that in the saline group. CONCLUSION: PET imaging with [68Ga]Ga-D-pep-PMED, a small-molecule radiotracer, is a promising tool for evaluating PD-L1 expression and quantifying the target blockade of PD-L1 to assist in the development of effective therapeutic regimens.
Subject(s)
Acetamides , B7-H1 Antigen , Positron-Emission Tomography , Pyridines , Immunotherapy , B7-H1 Antigen/analysis , B7-H1 Antigen/antagonists & inhibitors , Humans , Animals , Mice , Cell Line, Tumor , A549 Cells , Organometallic Compounds , Gallium Radioisotopes , Acetamides/chemistry , Pyridines/chemistryABSTRACT
Cytokeratin-14 (CK14), also known as keratin 14, is mainly expressed in the basal layer of stratified squamous epithelium. It has a critical role in maintaining cell morphology and resisting external mechanical stress. High levels of CK14 have been found in multiple types of tumors, especially basal-like breast cancer (BLBC). In this study, an anti-CK14 monoclonal antibody was successfully produced, purified, and labeled with 99mTc to evaluate the feasibility of visualizing the CK14 level in BLBC. Higher CK14 levels were found in MDA-MB-468 cells and tumors compared with the levels in MDA-MB-231 cells and tumors as revealed by Western blotting and immunohistochemistry experiments. The high binding specificity of 99mTc-HYNIC-Anti-CK14 mAb to CK14+ BLBC cells was verified by cell uptake and blocking studies. Single-photon emission computed tomography (SPECT) images exhibited higher radioactivity accumulation in MDA-MB-468 tumors compared with MDA-MB-231 tumors. The signal in MDA-MB-468 tumors decreased significantly when 100-fold excess amounts of anti-CK14 mAb were injected 1 h prior to SPECT, further validating the high specificity of the tracer. Biodistribution study results were consistent with SPECT imaging. In conclusion, we successfully constructed a CK14 targeting tracer, 99mTc-HYNIC-Anti-CK14 mAb, which has a high binding ability to CK14+ tumors, signifying its potential value in the immunoSPECT imaging of BLBC.
Subject(s)
Breast Neoplasms , Antibodies, Monoclonal , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Keratin-14 , Tissue Distribution , Tomography, Emission-Computed, Single-Photon/methodsABSTRACT
INTRODUCTION: This pilot study evaluated the imaging performance of pretargeted immunological positron emission tomography (immuno-PET) using an anti-carcinoembryonic antigen (CEA) recombinant bispecific monoclonal antibody (BsMAb), TF2 and the [68Ga]Ga-labelled HSG peptide, IMP288, in patients with metastatic colorectal carcinoma (CRC). PATIENTS AND METHODS: Patients requiring diagnostic workup of CRC metastases or in case of elevated CEA for surveillance were prospectively studied. They had to present with elevated CEA serum titre or positive CEA tumour staining by immunohistochemistry of a previous biopsy or surgical specimen. All patients underwent endoscopic ultrasound (EUS), chest-abdominal-pelvic computed tomography (CT), abdominal magnetic resonance imaging (MRI) and positron emission tomography using [18F]fluorodeoxyglucose (FDG-PET). For immuno-PET, patients received intravenously 120 nmol of TF2 followed 30 h later by 150 MBq of [68Ga]Ga-labelled IMP288, both I.V. The gold standard was histology and imaging after 6-month follow-up. RESULTS: Eleven patients were included. No adverse effects were reported after BsMAb and peptide injections. In a per-patient analysis, immuno-PET was positive in 9/11 patients. On a per-lesion analysis, 12 of 14 lesions were positive with immuno-PET. Median SUVmax, MTV and TLG were 7.65 [3.98-13.94, SD 3.37], 8.63 cm3 [1.98-46.64; SD 14.83] and 37.90 cm3 [8.07-127.5; SD 43.47] respectively for immuno-PET lesions. Based on a per-lesion analysis, the sensitivity, specificity, positive-predictive value and negative-predictive value were, respectively, 82%, 25%, 82% and 25% for the combination of EUS/CT/MRI; 76%, 67%, 87% and 33% for FDG-PET; and 88%, 100%, 100% and 67% for immuno-PET. Immuno-PET had an impact on management in 2 patients. CONCLUSION: This pilot study showed that pretargeted immuno-PET using anti-CEA/anti-IMP288 BsMAb and a [68Ga]Ga-labelled hapten was safe and feasible, with promising diagnostic performance. TRIAL REGISTRATION: ClinicalTrials.gov NCT02587247 Registered 27 October 2015.
Subject(s)
Colorectal Neoplasms , Gallium Radioisotopes , Antibodies, Monoclonal , Carcinoembryonic Antigen , Colorectal Neoplasms/diagnostic imaging , Fluorodeoxyglucose F18 , Heterocyclic Compounds, 1-Ring , Humans , Oligopeptides , Pilot Projects , Positron-Emission TomographyABSTRACT
Mesothelin is a molecular biomarker of many types of solid cancers, which may represent a highly promising new target in the development of cancer-targeted diagnostic agents. A human anti-mesothelin antibody with a low molecular weight, ET210sc, was applied; this antibody has potent affinity and can penetrate tissue quickly and stably without causing immunoreactions. We developed a new 124/131I-labeled radiotracer of ET210sc. The 124/131I-labeled ET210sc radiotracer showed excellent radiochemical quality (with over 99% radiolabeling yield, 0.07 GBq/µmol specific activity) and remarkable stability in phosphate-buffered saline (>95% at 3 days). The radiotracer retained its potent affinity (dissociation constant, Kd = 0.101 nM). The radiotracer specifically bound to mesothelin-positive cells in vitro. Interestingly, the radiotracer exhibited significant positive-to-negative tumor uptake ratios (1.5:1) 3 days postinjection. The estimated absorbed doses of each organ (e.g., 0.704 mGy/MBq for the rectum; 0.341 mGy/MBq for the spleen) met the medical safety standards for further clinical applications. Our findings provide an initial proof of concept for the potential use of 124/131I-labeled ET210sc radiotracers to detect mesothelin-overexpressing cancer. 124I-ET210sc is proposed to be an ideal imaging agent for further clinical applications.
Subject(s)
GPI-Linked Proteins/metabolism , Neoplasms/diagnostic imaging , Animals , Blotting, Western , Cell Line, Tumor , Female , Flow Cytometry , Humans , Immunohistochemistry , Iodine Isotopes/analysis , Mesothelin , Mice, Inbred BALB C , Positron-Emission Tomography , Radioimmunodetection , RadiometryABSTRACT
Recently, inhibiting the PD-1/PD-L1 checkpoint pathway utilizing anti-PD-1 or anti-PD-L1 antibodies has achieved great clinical success in cancer treatment. However, anti-PD-1 immunotherapy cannot be applied to all cancer patients, no more than 25% showed a positive response. Immunohistochemistry (IHC) is the gold standard to determine the PD-L1 expression level in malignant lesions, but a noninvasive imaging-meditated strategy is urgently required for clinical diagnosis to cover the shortcomings of invasive techniques. MX001, which is an anti-PD-L1 antibody, was labeled with Cu-64 ( t1/2 = 12.7 h) and purified by PD-10 chromatography. Comprehensive studies including positron emission tomography (PET), ex vivo biodistribution, IHC, and immunotherapy have been performed in mice bearing MC38 (PD-L1 positive (+)) and 4T1 (PD-L1 negative (-)) xenografts. PET imaging of [18F]FDG was taken before and after therapy to monitor the therapeutic efficacy. [64Cu]Cu-NOTA-MX001 exhibited 2.3 ± 1.2, 5.6 ± 2.1, 5.6 ± 1.2, 6.1 ± 1.1, 6.1 ± 0.5, and 10.2 ± 1.7%ID/g uptake in MC38 xenografts at 0.5, 12, 24, 36, 48, and 62 h post-injection (p.i.), respectively. Meanwhile, the uptake in the liver and muscle at corresponding time points was 17.5 ± 2.2, 8.4 ± 2.4, 11.3 ± 3.2, 7.2 ± 2.1, 7.9.1 ± 3.5, and 3.8 ± 1.8%ID/g, and 1.2 ± 0.5, 1.3 ± 0.4, 1.5 ± 0.5, 0.7 ± 0.1, 0.6 ± 0.2, and 0.2 ± 0.1%ID/g, respectively. The uptake of [18F]FDG in MC38 and 4T1 xenografts at 1-h p.i. was 5.3 ± 0.4 and 6.4 ± 0.6%ID/g, while the uptake of [64Cu]Cu-NOTA-MX001 was 5.6 ± 0.3 and 1.3 ± 0.4%ID/g at 12-h p.i. IHC analysis confirmed that the MC38 tumor exhibited high PD-L1 expression, and the 4T1 tumor, liver, and muscle exhibited low PD-L1 expression. In addition, MC38 xenografts were suppressed by MX001 about 88% in the immunotherapy study. MX001 was successfully developed as a fully human anti-PD-L1 antibody with a high binding affinity in mouse, monkey, and human. The in vivo pharmacokinetics of MX001 was evaluated with PET imaging after being radiolabeled with Cu-64. The uptake of [64Cu]Cu-NOTA-MX001 was clearly correlated to the PD-L1 expression on various types of cancer. Subsequent immunotherapy studies demonstrated that MX001 could effectively suppress tumor growth with positive PD-L1 expression, but had poor antitumor efficacy on tumors which exhibited low PD-L1 expression. Together with the above results, MX001 has the potential to be further developed as an antibody theranostic agent for both PET imaging and immunotherapy of cancers in clinics.
Subject(s)
Antibodies/therapeutic use , B7-H1 Antigen/metabolism , Immunotherapy/methods , Animals , B7-H1 Antigen/immunology , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Mice , Positron-Emission Tomography , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , RadioimmunodetectionABSTRACT
One of the main challenges of PET imaging with 89Zr-labeled monoclonal antibodies (mAbs) remains the long blood circulation of the radiolabeled mAbs, leading to high background signals, decreasing image quality. To overcome this limitation, here we report the use of a bioorthogonal linker cleavage approach (click-to-release chemistry) to selectively liberate [89Zr]Zr-DFO from trans-cyclooctene-functionalized trastuzumab (TCO-Tmab) in blood, following the administration of a tetrazine compound (trigger) in BT-474 tumor-bearing mice. Methods: We created a series of TCO-DFO constructs and evaluated their performance in [89Zr]Zr-DFO release from Tmab in vitro using different trigger compounds. The in vivo behavior of the best performing [89Zr]Zr-TCO-Tmab was studied in healthy mice first to determine the optimal dose of the trigger. To find the optimal time for the trigger administration, the rate of [89Zr]Zr-TCO-Tmab internalization was studied in BT-474 cancer cells. Finally, the trigger was administered 6 h or 24 h after [89Zr]Zr-TCO-Tmab- administration in tumor-bearing mice to liberate the [89Zr]Zr-DFO fragment. PET scans were obtained of tumor-bearing mice that received the trigger 6 h post-[89Zr]Zr-TCO-Tmab administration. Results: The [89Zr]Zr-TCO-Tmab and trigger pair with the best in vivo properties exhibited 83% release in 50% mouse plasma. In tumor-bearing mice the tumor-blood ratios were markedly increased from 1.0 ± 0.4 to 2.3 ± 0.6 (p = 0.0057) and from 2.5 ± 0.7 to 6.6 ± 0.9 (p < 0.0001) when the trigger was administered at 6 h and 24 h post-mAb, respectively. Same day PET imaging clearly showed uptake in the tumor combined with a strongly reduced background due to the fast clearance of the released [89Zr]Zr-DFO-containing fragment from the circulation through the kidneys. Conclusions: This is the first demonstration of the use of trans-cyclooctene-tetrazine click-to-release chemistry to release a radioactive chelator from a mAb in mice to increase tumor-to-blood ratios. Our results suggest that click-cleavable radioimmunoimaging may allow for substantially shorter intervals in PET imaging with full mAbs, reducing radiation doses and potentially even enabling same day imaging.
Subject(s)
Neoplasms , Radioimmunodetection , Animals , Mice , Trastuzumab , Antibodies, Monoclonal/chemistry , Positron-Emission Tomography/methods , Cyclooctanes/chemistry , Cell Line, Tumor , Zirconium/chemistryABSTRACT
The principle of pretargeted radioimmunoimaging and therapy has been investigated over the past 30 y in preclinical and clinical settings with the aim of reducing the radiation burden of healthy tissue for antibody-based nuclear medicine techniques. In the past few decades, 4 pretargeting methodologies have been proposed, and 2 of them-the bispecific antibody-hapten and the streptavidin-biotin platforms-have been evaluated in humans in phase 1 and 2 studies. With this review article, we aim to survey clinical pretargeting studies in order to understand the challenges that these platforms have faced in human studies and to provide an overview of how the clinical approval of the pretargeting system has proceeded in the past several decades. Additionally, we will discuss the successes of the pretargeting human studies and compare and highlight the pretargeting approaches and conditions that will advance clinical translation of the pretargeting platform in the future.
Subject(s)
Radioimmunodetection , Radioimmunotherapy , Humans , StreptavidinABSTRACT
Pretargeting parameters for the use of anti-carcinoembryonic antigen (CEA) bispecific monoclonal antibody TF2 and the 68Ga-labeled IMP288 peptide for immuno-PET have been optimized in a first-in-humans study performed on medullary thyroid carcinoma (MTC) patients (the iPET-MTC study). The aim of this post hoc analysis was to determine the sensitivity of immuno-PET in relapsing MTC patients, in comparison with conventional imaging and 18F-l-dihydroxyphenylalanine (18F-DOPA) PET/CT. Methods: Twenty-five studies were analyzed in 22 patients. All patients underwent immuno-PET 1 and 2 h after 68Ga-IMP288 injection pretargeted by TF2, in addition to neck, thoracic, abdominal, and pelvic CT; bone and liver MRI; and 18F-DOPA PET/CT. The gold standard was histology or confirmation by one other imaging method or by imaging follow-up. Results: In total, 190 lesions were confirmed by the gold standard: 89 in lymph nodes, 14 in lungs, 46 in liver, 37 in bone, and 4 in other sites (subcutaneous tissue, heart, brain, and pancreas). The number of abnormal foci detected by immuno-PET was 210. Among these, 174 (83%) were confirmed as true-positive by the gold standard. Immuno-PET showed a higher overall sensitivity (92%) than 18F-DOPA PET/CT (65%). Regarding metastatic sites, immuno-PET had a higher sensitivity than CT, 18F-DOPA PET/CT, or MRI for lymph nodes (98% vs. 83% for CT and 70% for 18F-DOPA PET/CT), liver (98% vs. 87% for CT, 65% for 18F-DOPA PET/CT, and 89% for MRI), and bone (92% vs. 64% for 18F-DOPA PET/CT and 86% for MRI), whereas sensitivity was lower for lung metastases (29% vs. 100% for CT and 14% for 18F-DOPA PET/CT). Tumor SUVmax at 60 min ranged from 1.2 to 59.0, with intra- and interpatient variability. Conclusion: This post hoc study demonstrates that anti-carcinoembryonic antigen immuno-PET is an effective procedure for detecting metastatic MTC lesions. Immuno-PET showed a higher overall sensitivity than 18F-DOPA PET/CT for disclosing metastases, except for the lung, where CT remains the most effective examination.
Subject(s)
Positron Emission Tomography Computed Tomography , Adult , Aged , Carcinoembryonic Antigen , GPI-Linked Proteins , Humans , Middle AgedABSTRACT
PURPOSE: The aim of this study was to perform radiotheranostics with radioiodinated monoclonal antibodies (mAbs) for targeting cancer stem cells (CSCs) in human colorectal cancer xenografts and evaluate the relative advantage of a cocktail containing both [131I]CD133 mAb and [131I]CD44 mAb. PROCEDURES: Tumor-bearing mice were randomly divided into eight groups: [131I]CD133mAb, [131I]CD44 mAb, [131I]IgG isotype control, radioiodinated mAb cocktail, CD133 mAb, CD44 mAb, unradioiodinated mAb cocktail, and saline groups. In vivo single photon emission computed tomography (SPECT) imaging was used to monitor dynamically changes in the CSC population after treatment. The radioactivity uptake of tumors was quantified ex vivo. The expression of CD133 and CD44 after treatment was also assessed by immunohistochemistry and western blot. Tumor growth curves and survival curves were generated to assess treatment efficacy. Cell apoptosis and proliferation in xenografts 30 days after treatment were measured by TdT-mediated dUTP-biotin nick end labeling (aka, TUNEL) and Ki67 staining. The expression levels of biomarkers in xenografts 30 days after treatment were measured by flow cytometry. RESULTS: The radioimmunoimaging (RII) with in vivo SPECT showed that the CSC-targeting radioimmunotherapy (RIT) groups ([131I]CD133 mAb, [131I]CD44 mAb, and radioiodinated mAb cocktail groups) had intense accumulations of radiolabeled agents in the tumor areas. The ex vivo biodistribution confirmed these findings. In the CSC-targeting RIT groups, immunohistochemistry and western blot indicated significant reduction of specific target expression in the xenografts. The tumor growth curves and survival curves showed that the CSC-targeting RIT significantly inhibited tumor growth and prolonged mean survival, respectively. Significantly increased apoptosis and decreased proliferation in xenografts further confirmed the therapeutic efficacy of CSC-targeting RIT. Flow cytometry showed that the decreases in CSCs correlated with the presence of the corresponding antibodies. CONCLUSIONS: Our results suggest that the CSC-targeting RIT can effectively reduce CSCs which consequently inhibits tumor development. The radioiodinated mAb cocktail may generate enhanced CSC-targeting specificity.
Subject(s)
Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/therapy , Neoplastic Stem Cells/pathology , Radiopharmaceuticals/chemistry , Xenograft Model Antitumor Assays , AC133 Antigen/metabolism , Animals , Body Weight , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/pathology , Humans , Hyaluronan Receptors/metabolism , Ki-67 Antigen/metabolism , Mice, Nude , Neoplastic Stem Cells/metabolism , Tissue Distribution , Tomography, Emission-Computed, Single-PhotonABSTRACT
OBJECTIVE: Pro-gastrin releasing peptide (ProGRP) plays an oncogenic role in small cell lung cancer (SCLC). The anti-ProGRP(31-98) monoclonal antibody D-D3 can selectively accumulate in SCLC xenografts in nude mice. This study evaluated the effectiveness of a new pretargeting procedure for the early diagnosis of SCLC. METHODS: D-D3 was radiolabeled with technetium-99m (99mTc) using a three-step pretargeting method. Mice with SCLC xenografts were treated with different labeling regimens, and the biodistribution and radioimmunoimaging were explored. The percentage injected dose per gram (%ID/g) in various organs, tumor/non-tumor (T/NT) ratio, and tumor/background (T/B) ratio were also calculated. RESULTS: In vivo distribution experiments revealed that 99mTc-DTPA-biotin was metabolized in the liver and kidney, with rapid elimination in the blood. The T/B ratio was highest in mice treated with biotinylated antibody D-D3 + avidin + 99mTc-DTPA-biotin. Single-photon emission computerized tomography imaging further confirmed that the T/B ratio was highest in this group at all time points. CONCLUSIONS: In contrast to directly labeled D-D3, pretargeting technology displayed specific enhancement and signal amplification in tumors, which could increase the target tumor uptake of 99mTc and provide a new approach for the early diagnosis of SCLC.
Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Animals , Biotin , Early Detection of Cancer , Lung Neoplasms/diagnostic imaging , Mice , Mice, Nude , Radioimmunodetection , Radiopharmaceuticals , Small Cell Lung Carcinoma/diagnostic imaging , Technetium , Technetium Tc 99m Pentetate , Tissue DistributionABSTRACT
Cancer immunotherapy is now established as a central therapeutic pillar in hematologic oncology. Cell-based therapies, with or without genetic modification ex vivo, have reached the clinic as the standard of care in limited indications and remain the subject of intense preclinical and translational development. Expanding on this, related therapeutic approaches are in development for solid-tumor and nonmalignant indications, broadening the scope of this technology. It has long been recognized that in vivo tracking of infused cellular therapies would provide unique opportunities to optimize their efficacy and aid in the assessment and management of toxicity. Recently, we have witnessed the introduction of novel tracers for passive labeling of cell products and advances in the introduction and use of reporter genes to enable longitudinal imaging. This review highlights the key developments over the last 5 y.
Subject(s)
Cell- and Tissue-Based Therapy/methods , Molecular Imaging/methods , Animals , Humans , Isotope Labeling , NanoparticlesABSTRACT
Glypican-3 (GPC3) over-expresses in hepatocellular carcinoma (HCC), but not expresses or under-expresses in normal adult hepatocytes. Therefore, GPC3 acts as a potential target for diagnosis and treatment of HCC. This study aimed to conduct radio-immunoimaging using GPC3 as a target in order, and to explore its potential for diagnosing and treating HCC. Humanized single-chain antibody scFv for HCC was established using phage antibody library. E.coli HB2151 was infected with recombinant phage antibodies that are considered to be strongly positive by phage ELISA. Then, the soluble antibodies were obtained post IPTG induction. Soluble antibodies were detected using SDS-PAGE assay. Anti-GPC3 single-chain antibodies were labeled using 131I, and then the distribution of radioactive markers in nude mice were analyzed in vivo by radio-immunoimaging. The results indicated that the size of soluble scFv products was 30 kD after purifying anti-GPC3 scFv antibodies that are successfully screened from phage antibody library. Anti-GPC3 phage antibodies could specifically bind to HCC cells. The ratios of radioactive tumor/blood and tumor/muscle for 131I labeled anti-GPC3 monoclonal antibodies were increased gradually, achieving the highest at 48 h. Radio-immunoimaging showed that the radioactive uptake of tumor sites remained the strongest at 48 h, and the ratio of target to non-target was the highest. In conclusion, the established anti-GPC3 scFv antibody had the potential to become an agent for radio-immunoimaging in diagnosing HCC and act as a targeted antibody for further radio-immunotherapy of HCC.
ABSTRACT
Lung cancer, especially non-small cell lung cancer (NSCLC), is the most common malignant tumor associated with poor prognosis. Angiogenesis plays a vital role in NSCLC, and could be used in tumor staging and therapy evaluation. CD93 (C1q receptor) is reportedly a key regulator of tumor angiogenesis. In the present study, the efficacy and specificity of a 125I-labeled CD93-specific monoclonal antibody (125I-anti-CD93 mAb) in detecting NSCLC xenografts were analyzed, and the association between CD93 expression and 125I-anti-CD93 mAb uptake by tumors was evaluated. The targeting ability of 125I-anti-CD93 mAb enabled its rapid, continuous and highly specific accumulation in CD93-expressing tumors in vivo. These results revealed the potential applicability of 125I-anti-CD93 mAb for non-invasive imaging diagnosis of CD93-positive NSCLC.
ABSTRACT
The recent clinical success of cancer immunotherapy has renewed interest in the development of tools to image the immune system. In general, immunotherapies attempt to enable the body's own immune cells to seek out and destroy malignant disease. Molecular imaging of the cells and molecules that regulate immunity could provide unique insight into the mechanisms of action, and failure, of immunotherapies. In this article, we will provide a comprehensive overview of the current state-of-the-art immunoimaging toolbox with a focus on imaging strategies and their applications toward immunotherapy.
Subject(s)
Molecular Imaging/methods , Animals , Humans , Immunity , Immunotherapy , Neoplasms/diagnostic imaging , Neoplasms/immunology , Neoplasms/therapyABSTRACT
INTRODUCTION: Zirconium-89 (89Zr, t1/2=78.4h) liquid target (LT) production offers an approach to introduce this positron-emitting isotope to cyclotron centres without the need for a separate solid target (ST) production set up. We compared the production, purification, and antibody radiolabeling yields of 89Zr-(LT) and 89Zr-(ST), and assessed the feasibility of 89Zr-(LT) for preclinical PET/CT. METHODS: 89Zr-(ST) production was performed with an 89Y foil on a TR 19 cyclotron at 13.8MeV. For LT production; an aqueous solution of yttrium nitrate (Y(NO3)3·6H2O) was irradiated on a TR 13 cyclotron at 12MeV. 89Zr was purified from the ST or LT material with hydroxamate resin, and used to radiolabel p-SCN-Bn-Deferoxamine (DFO)-conjugated Trastuzumab. MicroPET-CT imaging was performed at 1, 3 and 5days post-injection of 89Zr-DFO-Trastuzumab from ST or LT with biodistribution analysis on day 5. RESULTS: Irradiation of the ST yielded 2.88±1.07GBq/µA with a beam current of 14.0±3.8µA and irradiation time of 137±48min at end of bombardment while LT yielded 0.27±0.05GBq/µA with a beam current of 9.9±2.2µA and irradiation time of 221±29min. Radiolabeling of DFO-Trastuzumab with 89Zr-(ST) or 89Zr-(LT) was successful with purity>97% and specific activity>0.12MBq/µg (of antibody). MicroPET-CT imaging and biodistribution profiles showed similar uptake of 89Zr-(ST)-DFO-Trastuzumab and 89Zr-(LT)-DFO-Trastuzumab in tumor and all organs of interest. CONCLUSION: 89Zr-(LT) was effectively used to prepare antibody bioconjugates with specific activities suitable for small animal imaging. PET imaging and biodistribution revealed similar behaviours between bioconjugates labeled with 89Zr produced from the two target systems. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE: These results have important implications for the production of PET isotopes such as 89Zr to cyclotron facilities with only LT capabilities - such as most clinical centres - expanding the availability of 89Zr-immunoPET.
Subject(s)
Deferoxamine/chemistry , Isotope Labeling/methods , Radioisotopes/chemistry , Trastuzumab/chemistry , Zirconium/chemistry , Animals , Deferoxamine/pharmacokinetics , Female , Mice , Positron Emission Tomography Computed Tomography , Tissue Distribution , Trastuzumab/pharmacokineticsABSTRACT
ABCG2 is a multidrug resistance efflux pump expressed in many diverse tumors. The overexpression of ABCG2 is associated with resistance to a wide variety of anticancer agents, providing a noticeable setback to successful cancer therapy. Therapies targeting ABCG2 may therefore be a promising candidate for reversal of chemoresistance. The anti-ABCG2 single-chain variable fragment (scFv) antibody was constructed by phage display peptide library technology. Immunoblotting, ELISA and immunocytochemistry were used to evaluate the soluble expression and immunoreactivity of the scFv. The effects of scFv on cell function and chemosensitization were confirmed by colony formation, cell migration and CCK-8 assays. Flow cytometry was used to analyse the cell cycle and apoptosis. Radioimmunoimaging and nude mouse tumorigenicity assays were taken to determine the biodistribution and antitumor capacity of the scFv antibody. We have successfully screened out the candidate scFv antibody with an apparent molecular weight of 34 kDa. The scFv demonstrated favourable binding ability to lung adenocarcinoma cells and ABCG2 antigen, and the radioactivity was specifically aggregated at the tumor location. Furthermore, the internalized scFv resulted in antibody-mediated downregulation of ABCG2, proliferation inhibition, apoptosis and cisplatin (DDP) sensitivity. The anti-ABCG2 scFv antibody possesses good tumoraffin and antitumor activity and may therefore be an effective therapeutic agent for lung adenocarcinoma that is dependent on ABCG2 for drug resistance and survival.
ABSTRACT
Castration-resistant prostate cancer (CRPC) is the lethal form of prostate cancer, and more than 26,000 men will die from this disease in 2016. The pathophysiology of CRPC is clearly multifactorial, but most often, androgen receptor (AR) upregulation is associated with its earliest beginnings and the AR increase is part of the multimolecular complex including downstream effector proteins linked to AR (AR-axis) responsible for rapid proliferation and malignant features of the malignant cell. In both animal models and patients, glycolysis (Warburg effect) is also an early manifestation of CRPC transformation. At Memorial Sloan Kettering Cancer Center, we have focused our energies on imaging studies of the AR-axis in CRPC, using 18F-FDG, 18F-16ß-fluoro-5α-dihydrotestosterone (18F-FDHT), and a variety of radiolabeled antibodies targeting downstream effectors, such as prostate-specific membrane antigen (PSMA). Small-molecular-weight PSMA-targeting agents are not part of this review. In this review, we will focus on molecular imaging of the AR-axis in metastatic CRPC (mCRPC) and discuss our personal experience with these tracers. Our goal is to put these radiopharmaceuticals in the context of mCRPC biology and diagnosis (e.g., 18F-FDHT).
Subject(s)
Biomarkers, Tumor/metabolism , Dihydrotestosterone/analogs & derivatives , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Dihydrotestosterone/pharmacokinetics , Evidence-Based Medicine , Humans , Image Enhancement , Male , Molecular Imaging , Neoplasm Staging , Prostatic Neoplasms, Castration-Resistant , Radiopharmaceuticals/pharmacokineticsABSTRACT
INTRODUCTION: Cancer stem cells (CSCs) are a subpopulation within a tumor, which possesses the characteristics of self-renewal, differentiation, tumorigenicity, and drug resistance. The aim of this study was to target the colorectal CSC marker CD133 with an(131)I-labeled specific monoclonal antibody (AC133 mAb) in a nude mouse xenograft model. METHODS: Colorectal adenocarcinoma cells (LoVo cell line) were separated into CD133(+) and CD133(-) cells by magnetic activated cell sorting. CD133(+), CD133(-), and unsorted LoVo cells were cultured and then implanted subcutaneously into the lower limbs of nude mice (n = 5). AC133 mAb was labeled with (131)I by the iodogen method. RESULTS: The radiolabeled compound, (131)I-AC133 mAb, showed high stability, specificity, and immunoactivity in vitro. Obvious accumulation of (131)I-AC133 mAb was seen in nude mice bearing xenografts of CD133(+) and unsorted LoVo cells, but no uptake was found in mice bearing CD133(-) xenografts or specifically blocked xenografts. Biodistribution analysis showed that the tumor uptake of (131)I-AC133 mAb was 6.97 ± 1.40, 1.35 ± 0.48, 6.12 ± 1.91, and 1.61 ± 0.44% ID/g (n = 4) at day 7 after injection of (131)I-AC133 mAb in CD133(+), CD133(-), unsorted LoVo cell and specifically blocked xenografts, respectively. The results of immunofluorescence, autoradiography, and western blotting further verified the specific binding of (131)I-AC133 mAb to CD133(+) tumors. CONCLUSIONS: This study demonstrates the possibility of targeting CSCs with a radiolabeled AC133 mAb in colorectal cancer xenografts based on in vitro, ex vivo, and in vivo experiments. Our findings suggest a new method for imaging CSCs non-invasively.
Subject(s)
Antibodies, Monoclonal/immunology , Antigens, CD/immunology , Cell Transformation, Neoplastic , Colorectal Neoplasms/pathology , Glycoproteins/immunology , Neoplastic Stem Cells/diagnostic imaging , Peptides/immunology , AC133 Antigen , Animals , Antibodies, Monoclonal/pharmacokinetics , Autoradiography , Cell Line, Tumor , Colorectal Neoplasms/diagnostic imaging , Humans , Mice , Mice, Nude , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray ComputedABSTRACT
With the technological development of positron emission tomography (PET) and the advent of novel antibody-directed drug delivery systems, longer-lived positron-emitting radionuclides are moving to the forefront to take important roles in tracking the distribution of biotherapeutics such as antibodies, and for monitoring biological processes and responses. Longer half-life radionuclides possess advantages of convenient on-site preparation procedures for both clinical and non-clinical applications. The suitability of the long half-life radionuclides for imaging intact monoclonal antibodies (mAbs) and their respective fragments, which have inherently long biological half-lives, has attracted increased interest in recent years. In this review, we provide a survey of the recent literature as it applies to the development of nine-selected longer-lived positron emitters with half-lives of 9-140h (e.g., (124)I, (64)Cu, (86)Y and (89)Zr), and describe the biological behaviors of radionuclide-labeled mAbs with respect to distribution and targeting characteristics, potential toxicities, biological applications, and clinical translation potentials.
Subject(s)
Antibodies, Monoclonal , Radioisotopes , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Humans , Positron-Emission Tomography , Radioisotopes/pharmacokinetics , Radioisotopes/therapeutic useABSTRACT
AA amyloidosis results from the pathologic deposition in the kidneys and other organs of fibrils composed of N-terminal fragments of serum amyloid A protein (SAA). Given that there are only limited means to visualize these deposits, we have developed a series of mAbs, 2A4, 7D8, and 8G9, that bind specifically with nanomolar affinity to a carboxy-terminal epitope generated following proteolysis of SAA that yields the predominant component of AA amyloid deposits. Notably, these antibodies do not recognize native SAA, they retain their immunoreactivity when radiolabeled with I-125 and, after injection into AA amyloidotic mice, localize, as evidenced by autoradiography and micro-single photon emission computed tomography imaging, to histologically confirmed areas of amyloid deposition; namely, spleen, liver, and pancreas. The results of our in vitro and in vivo studies demonstrate the AA fibril-selectivity of mAbs 2A4, 7D8, and 8G9 and warrant further investigation into their role as novel diagnostic agents for patients with AA amyloidosis.