Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Clin Cancer Res ; 26(15): 3990-3998, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32345649

ABSTRACT

PURPOSE: Incomplete oncologic resections and damage to vital structures during colorectal cancer surgery increases morbidity and mortality. Moreover, neoadjuvant chemoradiotherapy has become the standard treatment modality for locally advanced rectal cancer, where subsequent downstaging can make identification of the primary tumor more challenging during surgery. Near-infrared (NIR) fluorescence imaging can aid surgeons by providing real-time visualization of tumors and vital structures during surgery. EXPERIMENTAL DESIGN: We present the first-in-human clinical experience of a novel NIR fluorescent peptide, cRGD-ZW800-1, for the detection of colon cancer. cRGD-ZW800-1 was engineered to have an overall zwitterionic chemical structure and neutral charge to lower nonspecific uptake and thus background fluorescent signal. We performed a phase I study in 11 healthy volunteer as well as a phase II feasibility study in 12 patients undergoing an elective colon resection, assessing 0.005, 0.015, and 0.05 mg/kg cRGD-ZW800-1 for the intraoperative visualization of colon cancer. RESULTS: cRGD-ZW800-1 appears safe, and exhibited rapid elimination into urine after a single low intravenous dose. Minimal invasive intraoperative visualization of colon cancer through full-thickness bowel wall was possible after an intravenous bolus injection of 0.05 mg/kg at least 2 hours prior to surgery. Longer intervals between injection and imaging improved the tumor-to-background ratio. CONCLUSIONS: cRGD-ZW800-1 enabled fluorescence imaging of colon cancer in both open and minimal invasive surgeries. Further development of cRGD-ZW800-1 for widespread use in cancer surgery may be warranted given the ubiquitous overexpression of various integrins on different types of tumors and their vasculature.


Subject(s)
Carcinoma/diagnosis , Colon/diagnostic imaging , Colonic Neoplasms/diagnosis , Fluorescent Dyes/administration & dosage , Optical Imaging/methods , Aged , Aged, 80 and over , Animals , Carcinoma/pathology , Carcinoma/therapy , Chemoradiotherapy, Adjuvant , Colectomy/methods , Colon/pathology , Colon/surgery , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Feasibility Studies , Female , Fluorescent Dyes/adverse effects , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Healthy Volunteers , Humans , Integrins/metabolism , Male , Mice , Middle Aged , Neoadjuvant Therapy , Optical Imaging/adverse effects , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/adverse effects , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Quaternary Ammonium Compounds/administration & dosage , Quaternary Ammonium Compounds/adverse effects , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/pharmacokinetics , Rats , Spectroscopy, Near-Infrared/methods , Sulfonic Acids/administration & dosage , Sulfonic Acids/adverse effects , Sulfonic Acids/chemistry , Sulfonic Acids/pharmacokinetics , Toxicity Tests, Acute
2.
Cancer J ; 25(2): 116-120, 2019.
Article in English | MEDLINE | ID: mdl-30896533

ABSTRACT

Transcriptome signature reversion (TSR) has been hypothesized as a promising method for discovery and use of existing noncancer drugs as potential drugs in the treatment of cancer (i.e., drug repositioning, drug repurposing). The TSR assumes that drugs with the ability to revert the gene expression associated with a diseased state back to its healthy state are potentially therapeutic candidates for that disease. This article reviews methodology of TSR and critically discusses key TSR studies. In addition, potential conceptual and computational improvements of this novel methodology are discussed as well as its current and possible future application in precision oncology trials.


Subject(s)
Drug Repositioning/methods , Gene Expression Profiling/methods , Precision Medicine/methods , Humans , Medical Oncology/methods
3.
Sci Rep ; 9(1): 2495, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30792476

ABSTRACT

To find new potentially therapeutic drugs against clear cell Renal Cell Carcinoma (ccRCC), within drugs currently prescribed for other diseases (drug repositioning), we previously searched for drugs which are expected to bring the gene expression of 500 + ccRCC samples from The Cancer Genome Atlas closer to that of healthy kidney tissue samples. An inherent limitation of this bulk RNA-seq data is that tumour samples consist of a varying mixture of cancerous and non-cancerous cells, which influences differential gene expression analyses. Here, we investigate whether the drug repositioning candidates are expected to target the genes dysregulated in ccRCC cells by studying the association with tumour purity. When all ccRCC samples are analysed together, the drug repositioning potential of identified drugs start decreasing above 80% estimated tumour purity. Because ccRCC is a highly vascular tumour, attributed to frequent loss of VHL function and subsequent activation of Hypoxia-Inducible Factor (HIF), we stratified the samples by observed activation of the HIF-pathway. After stratification, the association between estimated tumour purity and drug repositioning potential disappears for HIF-activated samples. This result suggests that the identified drug repositioning candidates specifically target the genes expressed by HIF-activated ccRCC tumour cells, instead of genes expressed by other cell types part of the tumour micro-environment.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/genetics , Gene Expression Profiling/methods , Gene Regulatory Networks/drug effects , Kidney Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Drug Repositioning , Gene Expression Regulation, Neoplastic/drug effects , Genetic Heterogeneity , Humans , Kidney Neoplasms/drug therapy , Neoplasm Staging , Sequence Analysis, RNA , Tumor Microenvironment/drug effects , Von Hippel-Lindau Tumor Suppressor Protein/genetics
4.
Sci Rep ; 8(1): 5250, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29588458

ABSTRACT

Reversal of cancer gene expression is predictive of therapeutic potential and can be used to find new indications for existing drugs (drug repositioning). Gene expression reversal potential is currently calculated, in almost all studies, by pre-aggregating all tumour samples into a single group signature or a limited number of molecular subtype signatures. Here, we investigate whether drug repositioning based on individual tumour sample gene expression signatures outperforms the use of tumour group and subtype signatures. The tumour signatures were created using 534 tumour samples and 72 matched normal samples from 530 clear cell renal cell carcinoma (ccRCC) patients. More than 20,000 drug signatures were extracted from the CMAP and LINCS databases. We show that negative enrichment of individual tumour samples correlated (Spearman's rho = 0.15) much better with the amount of differentially expressed genes in drug signatures than with the tumour group signature (Rho = 0.08) and the 4 tumour subtype signatures (Rho 0.036-0.11). Targeted drugs used against ccRCC, such as sirolimus and temsirolimus, which could not be identified with the pre-aggregated tumour signatures could be recovered using individual sample analysis. Thus, drug repositioning can be personalized by taking into account the gene expression profile of the individual's tumour sample.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Drug Repositioning/methods , Kidney Neoplasms/drug therapy , Carcinoma, Renal Cell/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kidney Neoplasms/genetics , Precision Medicine/methods , Transcriptome/drug effects
5.
MAbs ; 9(8): 1370-1378, 2017.
Article in English | MEDLINE | ID: mdl-28873009

ABSTRACT

Treatment of human epidermal growth factor receptor 2 (HER2)-driven breast cancer with tyrosine kinase inhibitor lapatinib can induce a compensatory HER3 increase, which may attenuate antitumor efficacy. Therefore, we explored in vivo HER3 tumor status assessment after lapatinib treatment with zirconium-89 (89Zr)-labeled anti-HER3 antibody mAb3481 positron emission tomography (PET). Lapatinib effects on HER3 cell surface expression and mAb3481 internalization were evaluated in human breast (BT474, SKBR3) and gastric (N87) cancer cell lines using flow cytometry. Next, in vivo effects of daily lapatinib treatment on89Zr-mAb3481 BT474 and N87 xenograft tumor uptake were studied. PET-scans (BT474 only) were made after daily lapatinib treatment for 9 days, starting 3 days prior to 89Zr-mAb3481 administration. Subsequently, ex vivo 89Zr-mAb3481 organ distribution analysis was performed and HER3 tumor levels were measured with Western blot and immunohistochemistry. In vitro, lapatinib increased membranous HER3 in BT474, SKBR3 and N87 cells, and consequently mAb3481 internalization 1.7-fold (BT474), 1.4-fold (SKBR3) and 1.4-fold (N87). 89Zr-mAb3481 BT474 tumor uptake was remarkably high at SUVmean 5.6±0.6 (51.8±7.7%ID/g) using a 10 µg 89Zr-mAb3481 protein dose in vehicle-treated mice. However, compared to vehicle, lapatinib did not affect 89Zr-mAb3481 ex vivo uptake in BT474 and N87 tumors, while HER3 tumor expression remained unchanged. In conclusion, lapatinib increased in vitro HER3 tumor cell expression, but not when these cells were xenografted. 89Zr-mAb3481 PET accurately reflected HER3 tumor status. 89Zr-mAb3481 PET showed high, HER3-specific tumor uptake, and such an approach might sensitively assess HER3 tumor heterogeneity and treatment response in patients.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Breast Neoplasms/drug therapy , Positron-Emission Tomography/methods , Quinazolines/administration & dosage , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal, Humanized/immunology , Antineoplastic Agents/administration & dosage , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/immunology , Female , Humans , Lapatinib , Male , Mice, Inbred BALB C , Mice, Nude , Radioisotopes , Receptor, ErbB-3/immunology , Tumor Burden/drug effects , Tumor Burden/immunology , Zirconium
6.
Clin Cancer Res ; 23(20): 6128-6137, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28733442

ABSTRACT

Purpose: We evaluated biodistribution and tumor targeting of 89Zr-lumretuzumab before and during treatment with lumretuzumab, a human epidermal growth factor receptor 3 (HER3)-targeting monoclonal antibody.Experimental Design: Twenty patients with histologically confirmed HER3-expressing tumors received 89Zr-lumretuzumab and underwent positron emission tomography (PET). In part A, 89Zr-lumretuzumab was given with additional, escalating doses of unlabeled lumretuzumab, and scans were performed 2, 4, and 7 days after injection to determine optimal imaging conditions. In part B, patients were scanned following tracer injection before (baseline) and after a pharmacodynamic (PD)-active lumretuzumab dose for saturation analysis. HER3 expression was determined immunohistochemically in skin biopsies. Tracer uptake was calculated as standardized uptake value (SUV).Results: Optimal PET conditions were found to be 4 and 7 days after administration of 89Zr-lumretuzumab with 100-mg unlabeled lumretuzumab. At baseline using 100-mg unlabeled lumretuzumab, the tumor SUVmax was 3.4 (±1.9) at 4 days after injection. SUVmean values for normal blood, liver, lung, and brain tissues were 4.9, 6.4, 0.9 and 0.2, respectively. Saturation analysis (n = 7) showed that 4 days after lumretuzumab administration, tumor uptake decreased by 11.9% (±8.2), 10.0% (±16.5), and 24.6% (±20.9) at PD-active doses of 400, 800, and 1,600 mg, respectively, when compared with baseline. Membranous HER3 was completely downregulated in paired skin biopsies already at and above 400-mg lumretuzumab.Conclusions: PET imaging showed biodistribution and tumor-specific 89Zr-lumretuzumab uptake. Although, PD-active lumretuzumab doses decreased 89Zr-lumretuzumab uptake, there was no clear evidence of tumor saturation by PET imaging as the tumor SUV did not plateau with increasing doses. Clin Cancer Res; 23(20); 6128-37. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Neoplasms/diagnosis , Neoplasms/drug therapy , Positron-Emission Tomography , Radiopharmaceuticals , Zirconium , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacokinetics , Drug Monitoring , Female , Gene Expression , Humans , Male , Middle Aged , Molecular Targeted Therapy , Neoplasms/metabolism , Receptor, ErbB-3/antagonists & inhibitors , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Treatment Outcome
7.
J Nucl Med ; 58(8): 1210-1215, 2017 08.
Article in English | MEDLINE | ID: mdl-28360206

ABSTRACT

The human epidermal growth factor receptor 3 (HER3) is an interesting target for antitumor therapy. For optimal HER3 signaling inhibition, a biparatopic Nanobody construct (MSB0010853) was developed that binds 2 different HER3 epitopes. In addition, MSB0010853 contains a third HER3 epitope that binds albumin to extend its circulation time. MSB0010853 is cross-reactive with HER3 and albumin of mouse origin. We aimed to gain insight into MSB0010853 biodistribution and tumor uptake by radiolabeling the Nanobody construct with 89Zr. Methods: MSB0010853 was radiolabeled with 89Zr. Dose- and time-dependent tumor uptake was studied in nude BALB/c mice bearing a subcutaneous HER3 overexpressing H441 non-small cell lung cancer xenograft. Dose-dependent biodistribution of 89Zr-MSB0010853 was assessed ex vivo at 24 h after intravenous injection. Protein doses of 5, 10, 25, 100, and 1,000 µg were used. Time-dependent biodistribution of MSB0010853 was analyzed ex vivo at 3, 6, 24, and 96 h after intravenous administration of 25 µg of 89Zr-MSB0010853. PET imaging and biodistribution were performed 24 h after administration of 25 µg of 89Zr-MSB0010853 to mice bearing human H441, FaDu (high HER3 expression), or Calu-1 (no HER3 expression) tumor xenografts. Results: Radiolabeling of MSB0010853 with 89Zr was performed with a radiochemical purity of greater than 95%. Ex vivo biodistribution showed protein dose- and time-dependent distribution of 89Zr-MSB0010853 in all organs. Uptake of 89Zr-MSB0010853 in H441 tumors was only time-dependent. Tumor could be visualized up to at least 96 h after injection. The highest mean SUV of 0.6 ± 0.2 was observed at 24 h after injection of 25 µg of 89Zr-MSB0010853. 89Zr-MSB0010853 tumor uptake correlated with HER3 expression and was highest in H441 (6.2 ± 1.1 percentage injected dose per gram [%ID/g]) and lowest in Calu-1 (2.3 ± 0.3 %ID/g) xenografts. Conclusion:89Zr-MSB0010853 organ distribution and tumor uptake in mice are time-dependent, and tumor uptake correlates with HER3 expression. In contrast to tumor uptake except for kidney uptake, organ distribution of 89Zr-MSB0010853 is protein dose-dependent for the tested doses. 89Zr-MSB0010853 PET imaging gives insight into the in vivo behavior of MSB0010853.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Positron-Emission Tomography , Receptor, ErbB-3/metabolism , Single-Chain Antibodies/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Humans , Image Processing, Computer-Assisted , Lung Neoplasms/pathology , Male , Mice , Single-Chain Antibodies/pharmacokinetics , Tissue Distribution
8.
Eur J Nucl Med Mol Imaging ; 44(8): 1328-1336, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28315949

ABSTRACT

PURPOSE: c-MET and its ligand hepatocyte growth factor are often dysregulated in human cancers. Dynamic changes in c-MET expression occur and might predict drug efficacy or emergence of resistance. Noninvasive visualization of c-MET dynamics could therefore potentially guide c-MET-directed therapies. We investigated the feasibility of 89Zr-labelled one-armed c-MET antibody onartuzumab PET for detecting relevant changes in c-MET levels induced by c-MET-mediated epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor erlotinib resistance or heat shock protein-90 (HSP90) inhibitor NVP-AUY-922 treatment in human non-small-cell lung cancer (NSCLC) xenografts. METHODS: In vitro membrane c-MET levels were determined by flow cytometry. HCC827ErlRes, an erlotinib-resistant clone with c-MET upregulation, was generated from the exon-19 EGFR-mutant human NSCLC cell line HCC827. Mice bearing HCC827 and HCC827ErlRes tumours in opposite flanks underwent 89Zr-onartuzumab PET scans. The HCC827-xenografted mice underwent 89Zr-onartuzumab PET scans before treatment and while receiving biweekly intraperitoneal injections of 100 mg/kg NVP-AUY-922 or vehicle. Ex vivo, tumour c-MET immunohistochemistry was correlated with the imaging results. RESULTS: In vitro, membrane c-MET was upregulated in HCC827ErlRes tumours by 213 ± 44% in relation to the level in HCC827 tumours, while c-MET was downregulated by 69 ± 9% in HCC827 tumours following treatment with NVP-AUY-922. In vivo, 89Zr-onartuzumab uptake was 26% higher (P < 0.05) in erlotinib-resistant HCC827ErlRes than in HCC827 xenografts, while HCC827 tumour uptake was 33% lower (P < 0.001) following NVP-AUY-922 treatment. CONCLUSION: The results show that 89Zr-onartuzumab PET effectively discriminates relevant changes in c-MET levels and could potentially be used clinically to monitor c-MET status.


Subject(s)
Antibodies, Monoclonal , Positron-Emission Tomography , Proto-Oncogene Proteins c-met/metabolism , Radioisotopes , Zirconium , Animals , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Drug Resistance, Neoplasm/drug effects , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/therapeutic use , Feasibility Studies , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Isoxazoles/pharmacology , Isoxazoles/therapeutic use , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Resorcinols/pharmacology , Resorcinols/therapeutic use , Up-Regulation/drug effects
9.
Clin Cancer Res ; 23(11): 2730-2741, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28119364

ABSTRACT

Purpose: To provide proof of principle of safety, breast tumor-specific uptake, and positive tumor margin assessment of the systemically administered near-infrared fluorescent tracer bevacizumab-IRDye800CW targeting VEGF-A in patients with breast cancer.Experimental Design: Twenty patients with primary invasive breast cancer eligible for primary surgery received 4.5 mg bevacizumab-IRDye800CW as intravenous bolus injection. Safety aspects were assessed as well as tracer uptake and tumor delineation during surgery and ex vivo in surgical specimens using an optical imaging system. Ex vivo multiplexed histopathology analyses were performed for evaluation of biodistribution of tracer uptake and coregistration of tumor tissue and healthy tissue.Results: None of the patients experienced adverse events. Tracer levels in primary tumor tissue were higher compared with those in the tumor margin (P < 0.05) and healthy tissue (P < 0.0001). VEGF-A tumor levels also correlated with tracer levels (r = 0.63, P < 0.0002). All but one tumor showed specific tracer uptake. Two of 20 surgically excised lumps contained microscopic positive margins detected ex vivo by fluorescent macro- and microscopy and confirmed at the cellular level.Conclusions: Our study shows that systemic administration of the bevacizumab-IRDye800CW tracer is safe for breast cancer guidance and confirms tumor and tumor margin uptake as evaluated by a systematic validation methodology. The findings are a step toward a phase II dose-finding study aimed at in vivo margin assessment and point to a novel drug assessment tool that provides a detailed picture of drug distribution in the tumor tissue. Clin Cancer Res; 23(11); 2730-41. ©2016 AACR.


Subject(s)
Benzenesulfonates/administration & dosage , Bevacizumab/administration & dosage , Breast Neoplasms, Male/drug therapy , Breast Neoplasms/drug therapy , Indoles/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Benzenesulfonates/adverse effects , Bevacizumab/adverse effects , Breast Neoplasms/diagnosis , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Breast Neoplasms, Male/diagnosis , Breast Neoplasms, Male/diagnostic imaging , Breast Neoplasms, Male/pathology , Cell Line, Tumor , Drug-Related Side Effects and Adverse Reactions/pathology , Feasibility Studies , Female , Humans , Indoles/adverse effects , Male , Optical Imaging , Positron-Emission Tomography , Tissue Distribution/drug effects , Vascular Endothelial Growth Factor A/genetics
10.
Mol Cancer Ther ; 16(1): 134-142, 2017 01.
Article in English | MEDLINE | ID: mdl-27760836

ABSTRACT

Antibody-drug conjugates (ADC) use monoclonal antibodies (mAb) as vehicles to deliver potent cytotoxic drugs selectively to tumor cells expressing the target. Molecular imaging with zirconium-89 (89Zr)-labeled mAbs recapitulates similar targeting biology and might help predict the efficacy of these ADCs. An anti-mesothelin antibody (AMA, MMOT0530A) was used to make comparisons between its efficacy as an ADC and its tumor uptake as measured by 89Zr immunoPET imaging. Mesothelin-targeted tumor growth inhibition by monomethyl auristatin E (MMAE), ADC AMA-MMAE (DMOT4039A), was measured in mice bearing xenografts of ovarian cancer OVCAR-3×2.1, pancreatic cancers Capan-2, HPAC, AsPC-1, and HPAF-II, or mesothelioma MSTO-211H. Ex vivo analysis of mesothelin expression was performed using immunohistochemistry. AMA-MMAE showed the greatest growth inhibition in OVCAR-3×2.1, Capan-2, and HPAC tumors, which showed target-specific tumor uptake of 89Zr-AMA. The less responsive xenografts (AsPC-1, HPAF-II, and MSTO-211H) did not show 89Zr-AMA uptake despite confirmed mesothelin expression. ImmunoPET can demonstrate the necessary delivery, binding, and internalization of an ADC antibody in vivo and this correlates with the efficacy of mesothelin-targeted ADC in tumors vulnerable to the cytotoxic drug delivered. Mol Cancer Ther; 16(1); 134-42. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , GPI-Linked Proteins/antagonists & inhibitors , Immunoconjugates/pharmacology , Positron-Emission Tomography , Radiopharmaceuticals , Zirconium , Animals , Antineoplastic Agents/pharmacokinetics , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Flow Cytometry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression , Humans , Immunoconjugates/pharmacokinetics , Mesothelin , Mice , Molecular Targeted Therapy , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/metabolism , Tumor Burden/drug effects , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
11.
Cancer Res ; 77(3): 623-631, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27879266

ABSTRACT

In vivo tumor labeling with fluorescent agents may assist endoscopic and surgical guidance for cancer therapy as well as create opportunities to directly observe cancer biology in patients. However, malignant and nonmalignant tissues are usually distinguished on fluorescence images by applying empirically determined fluorescence intensity thresholds. Here, we report the development of fSTREAM, a set of analytic methods designed to streamline the analysis of surgically excised breast tissues by collecting and statistically processing hybrid multiscale fluorescence, color, and histology readouts toward precision fluorescence imaging. fSTREAM addresses core questions of how to relate fluorescence intensity to tumor tissue and how to quantitatively assign a normalized threshold that sufficiently differentiates tumor tissue from healthy tissue. Using fSTREAM we assessed human breast tumors stained in vivo with fluorescent bevacizumab at microdose levels. Showing that detection of such levels is achievable, we validated fSTREAM for high-resolution mapping of the spatial pattern of labeled antibody and its relation to the underlying cancer pathophysiology and tumor border on a per patient basis. We demonstrated a 98% sensitivity and 79% specificity when using labeled bevacizumab to outline the tumor mass. Overall, our results illustrate a quantitative approach to relate fluorescence signals to malignant tissues and improve the theranostic application of fluorescence molecular imaging. Cancer Res; 77(3); 623-31. ©2016 AACR.


Subject(s)
Bevacizumab/pharmacokinetics , Breast Neoplasms/diagnostic imaging , Image Interpretation, Computer-Assisted/methods , Molecular Imaging/methods , Optical Imaging/methods , Aged , Antineoplastic Agents/pharmacokinetics , Benzenesulfonates/pharmacokinetics , Female , Fluorescent Dyes/pharmacokinetics , Humans , Indoles/pharmacokinetics , Middle Aged
12.
Oncotarget ; 7(42): 68111-68121, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27602494

ABSTRACT

Preclinical positron emission tomography (PET) imaging revealed a mismatch between in vivo epidermal growth factor receptor (EGFR) expression and EGFR antibody tracer tumor uptake. Shed EGFR ectodomain (sEGFR), which is present in cancer patient sera, can potentially bind tracer and therefore influence tracer kinetics. To optimize EGFR-PET, we examined the influence of sEGFR levels on tracer kinetics and tumor uptake of EGFR monoclonal antibody 89Zr-imgatuzumab in varying xenograft models. Human cancer cell lines A431 (EGFR overexpressing, epidermoid), A549 and H441 (both EGFR medium expressing, non-small cell lung cancer) were xenografted in mice. Xenografted mice received 10, 25 or 160 µg 89Zr-imgatuzumab, co-injected with equal doses 111In-IgG control. MicroPET scans were made 24, 72 and 144 h post injection, followed by biodistribution analysis. sEGFR levels in liver and plasma samples were determined by ELISA. 89Zr-imgatuzumab uptake in A431 tumors was highest (29.8 ± 5.4 %ID/g) in the 160 µg dose group. Contrary, highest uptake in A549 and H441 tumors was found at the lowest (10 µg) 89Zr-imgatuzumab dose. High 89Zr-imgatuzumab liver accumulation was found in A431 xenografted mice, which decreased with antibody dose increments. 89Zr-imgatuzumab liver uptake in A549 and H441 xenografted mice was low at all doses. sEGFR levels in liver and plasma of A431 bearing mice were up to 1000-fold higher than levels found in A549, H441 and non-tumor xenografted mice. 89Zr-imgatuzumab effectively visualizes EGFR-expressing tumors. High sEGFR levels can redirect 89Zr-imgatuzumab to the liver, in which case tumor visualization can be improved by increasing tracer antibody dose.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Bevacizumab/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Glycoproteins/pharmacokinetics , Lung Neoplasms/metabolism , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Male , Mice, Nude , Radioisotopes/pharmacokinetics , Tissue Distribution , Transplantation, Heterologous , Zirconium/pharmacokinetics
13.
Eur J Pharm Biopharm ; 104: 226-34, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27179587

ABSTRACT

There is a dire need for better visualization of cancer and analysis of specific targets in vivo. Molecular imaging with fluorescence is gaining more and more attention, as it allows detection of these targets and has advantages over radioactivity, such as no radiation dose, and lower costs. A key challenge in optical imaging however, is translation of the newly developed tracers from pre-clinical phase to clinical application. We describe the development and safety testing of clinical grade bevacizumab-800CW, an antibody-based targeted agent for non-invasive imaging of vascular endothelial growth factor A (VEGF-A). Development included implementing the manufacturing process and analytical methods according to current Good Manufacturing Practice (cGMP), formulation studies, extended characterization and stability testing. For safety pharmacology an extended single dose toxicity study in mice was performed. Bevacizumab-800CW was formulated in isotonic phosphate buffered sodium chloride solution at pH 7. The production was robust and showed a reproducible labeling efficiency, and no impurities. The binding affinity to VEGF-A remained intact. The optimized product meets all release specifications, is stable up to at least 3months and its characteristics did not significantly differ from the unlabeled bevacizumab. Toxicity testing in mice showed no remarkable findings. In conclusion, sterile bevacizumab-800CW (6mg=6ml) can be produced in stock according to current Good Manufacturing Practice. It is ready for first-in-human use.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/chemistry , Animals , Bevacizumab/adverse effects , Bevacizumab/chemistry , Fluorescent Dyes/chemistry , Humans , Mice , Spectrometry, Fluorescence
14.
Oncotarget ; 7(18): 25103-12, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27029064

ABSTRACT

The efficacy of antibody-drug conjugates (ADCs) targeted to solid tumors depends on biological processes that are hard to monitor in vivo. 89Zr-immunoPET of the ADC antibodies could help understand the performance of ADCs in the clinic by confirming the necessary penetration, binding, and internalization. This work studied monomethyl auristatin E (MMAE) ADCs against two targets in metastatic castration-resistant prostate cancer, TENB2 and STEAP1, in four patient-derived tumor models (LuCaP35V, LuCaP70, LuCaP77, LuCaP96.1). Three aspects of ADC biology were measured and compared: efficacy was measured in tumor growth inhibition studies; target expression was measured by immunohistochemistry and flow cytometry; and tumor antibody uptake was measured with 111In-mAbs and gamma counting or with 89Zr-immunoPET. Within each model, the mAb with the highest tumor uptake showed the greatest potency as an ADC. Sensitivity between models varied, with the LuCaP77 model showing weak efficacy despite high target expression and high antibody uptake. Ex vivo analysis confirmed the in vivo results, showing a correlation between expression, uptake and ADC efficacy. We conclude that 89Zr-immunoPET data can demonstrate which ADC candidates achieve the penetration, binding, and internalization necessary for efficacy in tumors sensitive to the toxic payload.


Subject(s)
Immunoconjugates/pharmacology , Positron-Emission Tomography/methods , Prostatic Neoplasms/diagnostic imaging , Animals , Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm , Antineoplastic Agents/pharmacology , Humans , Male , Membrane Proteins/antagonists & inhibitors , Mice , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Oligopeptides/pharmacology , Oxidoreductases/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Radioisotopes , Xenograft Model Antitumor Assays , Zirconium
15.
J Nucl Med ; 57(5): 812-7, 2016 05.
Article in English | MEDLINE | ID: mdl-26848172

ABSTRACT

UNLABELLED: AMG 110, a bispecific T cell engager (BiTE) antibody construct, induces T cell-mediated cancer cell death by cross-linking epithelial cell adhesion molecule (EpCAM) on tumor cells with a cluster of differentiation 3 ε (CD3ε) on T cells. We labeled AMG 110 with (89)Zr or near-infrared fluorescent dye (IRDye) 800CW to study its tumor targeting and tissue distribution. METHODS: Biodistribution and tumor uptake of (89)Zr-AMG 110 was studied up to 6 d after intravenous administration to nude BALB/c mice bearing high EpCAM-expressing HT-29 colorectal cancer xenografts. Tumor uptake of (89)Zr-AMG 110 was compared with uptake in head and neck squamous cell cancer FaDu (intermediate EpCAM) and promyelocytic leukemia HL60 (EpCAM-negative) xenografts. Intratumoral distribution in HT-29 tumors was studied using 800CW-AMG 110. RESULTS: Tumor uptake of (89)Zr-AMG 110 can be clearly visualized using small-animal PET imaging up to 72 h after injection. The highest tumor uptake of (89)Zr-AMG 110 at the 40-µg dose level was observed at 6 and 24 h (respectively, 5.35 ± 0.22 and 5.30 ± 0.20 percentage injected dose per gram; n = 3 and 4). Tumor uptake of (89)Zr-AMG 110 was EpCAM-specific and correlated with EpCAM expression. 800CW-AMG 110 accumulated at the tumor cell surface in viable EpCAM-expressing tumor tissue. CONCLUSION: PET and fluorescent imaging provided real-time information about AMG 110 distribution and tumor uptake in vivo. Our data support using (89)Zr and IRDye 800CW to evaluate tumor and tissue uptake kinetics of bispecific T cell engager antibody constructs in preclinical and clinical settings.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Epithelial Cell Adhesion Molecule/immunology , Positron-Emission Tomography/methods , Radioisotopes , T-Lymphocytes/immunology , Zirconium , Animals , HL-60 Cells , HT29 Cells , Humans , Isotope Labeling , Male , Mice , Tissue Distribution
16.
Oncotarget ; 6(39): 42081-90, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26536664

ABSTRACT

Mesothelin is a tumor differentiation antigen expressed by epithelial tumors, including pancreatic cancer. Currently, mesothelin is being targeted with an antibody-drug conjugate (ADC) consisting of a mesothelin-specific antibody coupled to a highly potent chemotherapeutic drug. Considering the toxicity of the ADC and reduced accessibility of pancreatic tumors, non-invasive imaging could provide necessary information. We therefore developed a zirconium-89 (89Zr) labeled anti-mesothelin antibody (89Zr-AMA) to study its biodistribution in human pancreatic tumor bearing mice. Biodistribution and dose-finding of 89Zr-AMA were studied 144 h after tracer injection in mice with subcutaneously xenografted HPAC. MicroPET imaging was performed 24, 72 and 144 h after tracer injection in mice bearing HPAC or Capan-2. Tumor uptake and organ distribution of 89Zr-AMA were compared with nonspecific 111In-IgG. Biodistribution analyses revealed a dose-dependent 89Zr-AMA tumor uptake. Tumor uptake of 89Zr-AMA was higher than 111In-IgG using the lowest tracer dose. MicroPET showed increased tumor uptake over 6 days, whereas activity in blood pool and other tissues decreased. Immunohistochemistry showed that mesothelin was expressed by the HPAC and CAPAN-2 tumors and fluorescence microscopy revealed that AMA-800CW was present in tumor cell cytoplasm. 89Zr-AMA tumor uptake is antigen-specific in mesothelin-expressing tumors. 89Zr-AMA PET provides non-invasive, real-time information about AMA distribution and tumor targeting.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , GPI-Linked Proteins/antagonists & inhibitors , Immunoconjugates/pharmacokinetics , Pancreatic Neoplasms/metabolism , Animals , Antibodies, Monoclonal/immunology , Benzenesulfonates/chemistry , Benzenesulfonates/pharmacokinetics , Cell Line, Tumor , Cytoplasm/metabolism , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Humans , Immunoconjugates/immunology , Immunohistochemistry , Indoles/chemistry , Indoles/pharmacokinetics , Male , Mesothelin , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/pathology , Positron-Emission Tomography/methods , Radioisotopes/chemistry , Radioisotopes/pharmacokinetics , Time Factors , Tissue Distribution , Transplantation, Heterologous , Zirconium/chemistry , Zirconium/pharmacokinetics
17.
J Clin Oncol ; 33(13): 1491-504, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25779566

ABSTRACT

More than 50 monoclonal antibodies (mAbs), including several antibody-drug conjugates, are in advanced clinical development, forming an important part of the many molecularly targeted anticancer therapeutics currently in development. Drug development is a relatively slow and expensive process, limiting the number of drugs that can be brought into late-stage trials. Development decisions could benefit from quantitative biomarkers, enabling visualization of the tissue distribution of (potentially modified) therapeutic mAbs to confirm effective whole-body target expression, engagement, and modulation and to evaluate heterogeneity across lesions and patients. Such biomarkers may be realized with positron emission tomography imaging of radioactively labeled antibodies, a process called immunoPET. This approach could potentially increase the power and value of early trials by improving patient selection, optimizing dose and schedule, and rationalizing observed drug responses. In this review, we summarize the available literature and the status of clinical trials regarding the potential of immunoPET during early anticancer drug development.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Discovery/methods , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Positron-Emission Tomography , Radiopharmaceuticals , Animals , Antibodies, Monoclonal/metabolism , Antineoplastic Agents/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Patient Selection , Predictive Value of Tests , Tissue Distribution , Treatment Outcome , Whole Body Imaging
18.
Eur J Cancer ; 50(14): 2508-16, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25027745

ABSTRACT

PURPOSE: Triple negative breast cancer (TNBC) is biologically characterised by heterogeneous presence of molecular pathways underlying it. Insulin-like growth factor receptor-1 (IGF-1R) expression and vascular endothelial growth factor-A (VEGF-A) have been identified as key factors in these pathways in TNBC. In this study, we aimed at in vivo PET imaging the effect of heat shock protein (Hsp) 90 inhibition by means of NVP-AUY922 on these pathways, with zirconium-89 ((89)Zr) labelled antibodies targeting IGF-1R and VEGF-A. MATERIALS AND METHODS: In vitro NVP-AUY922 effects on cellular IGF-1R expression and VEGF-A secretion were determined in MCF-7 and MDA-MB-231 cell lines. Moreover human TNBC bearing MDA-MB-231 mice received 50mg/kg NVP-AUY922 or vehicle q3d intraperitoneally for 21days. PET scans with (89)Zr-MAB391 and (89)Zr-bevacizumab for visualisation of IGF-1R and VEGF-A were performed before and during treatment. Ex vivo biodistribution and correlative tissue analyses were performed. RESULTS: NVP-AUY922 treatment reduced IGF-1R expression and VEGF-A excretion in both cell lines. Hsp90 inhibition lowered tumour uptake on (89)Zr-MAB391-PET by 37.3% (P<0.01) and on (89)Zr-bevacizumab-PET by 44.4% (P<0.01). This was confirmed by ex vivo biodistribution with a reduction of 41.3% injected dose (ID)/g for (89)Zr-MAB391 and 37.8% ID/g for (89)Zr-bevacizumab, while no differences were observed for other tissues. This coincided with reduced IGF-1R expression and mean vessel density in the NVP-AUY922 treated tumours. CONCLUSION: (89)Zr-MAB391 and (89)Zr-bevacizumab PET reflect effect of Hsp90 inhibitors and can therefore potentially be used to monitor therapeutic effects of Hsp90 inhibitor therapy in TNBC.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Receptor, IGF Type 1/metabolism , Resorcinols/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Down-Regulation , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , MCF-7 Cells , Male , Mice , Mice, Inbred BALB C , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
19.
MAbs ; 6(4): 1051-8, 2014.
Article in English | MEDLINE | ID: mdl-24870719

ABSTRACT

The humanized monoclonal antibody with high affinity for the human epidermal growth factor receptor (HER) 3, RG7116, is a glycoengineered, IgG1 class antibody. By labeling RG7116 with zirconium-89 ((89)Zr) we aimed to visualize in vivo HER3 expression and study the biodistribution of this antibody in human tumor-bearing mice. Biodistribution of (89)Zr-RG7116 was studied in subcutaneously xenografted FaDu tumor cells (HER3-positive). Dose-dependency of (89)Zr-RG7116 organ distribution and specific tumor uptake was assessed by administering doses ranging from 0.05 to 10 mg/kg RG7116 to SCID/Beige mice. Biodistribution was analyzed at 24 and 144 h after injection. MicroPET imaging was performed at 1, 3, and 6 days after injection of 1.0 mg/kg (89)Zr-RG7116 in the FaDu, H441, QG-56 and Calu-1 xenografts with varying HER3 expression. The excised tumors were analyzed for HER3 expression. Biodistribution analyses showed a dose- and time-dependent (89)Zr-RG7116 tumor uptake in FaDu tumors. The highest tumor uptake of (89)Zr-RG7116 was observed in the 0.05 mg/kg dose group with 27.5%ID/g at 144 h after tracer injection. MicroPET imaging revealed specific tumor uptake of (89)Zr-RG7116 in FaDu and H441 models with an increase in tumor uptake over time. Biodistribution data was consistent with the microPET findings in FaDu, H441, QG56 and Calu-1 xenografts, which correlated with HER3 expression levels. In conclusion, (89)Zr-RG7116 specifically accumulates in HER3 expressing tumors. PET imaging with this tracer provides real-time non-invasive information about RG7116 distribution, tumor targeting and tumor HER3 expression levels.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Immunoconjugates/pharmacokinetics , Isotopes/pharmacology , Neoplasms/diagnostic imaging , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacology , Receptor, ErbB-3/immunology , Zirconium/pharmacology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Heterografts , Humans , Immunoconjugates/immunology , Isotopes/immunology , Mice , Mice, SCID , Neoplasm Transplantation , Neoplasms/immunology , Radiography , Radiopharmaceuticals/immunology , Zirconium/immunology
20.
J Nucl Med ; 55(4): 665-71, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24614223

ABSTRACT

UNLABELLED: Anticalins are a novel class of biopharmaceuticals, displaying highly desirable attributes as imaging agents. The anticalin PRS-110 was rationally engineered to target the oncogene MET with high affinity and specificity. The aim of this study was to visualize MET expression and analyze biodistribution of (89)Zr-labeled PRS-110 in human tumor-bearing mice. METHODS: (89)Zr-PRS-110 was generated. For biodistribution studies (96 h after injection of tracer) 10 µg of (89)Zr-PRS-110 (with 0-490 µg of unlabeled PRS-110) were injected into BALB/c mice bearing high MET-expressing H441 non-small cell lung cancer xenografts. Further characterization with PET imaging was performed at 6, 24, 48, and 96 h after injection of 50 µg of (89)Zr-PRS-110 into mice bearing H441, primary glioblastoma U87-MG (intermediate MET), or ovarian cancer A2780 (low MET) xenografts. Drug distribution was also analyzed ex vivo using fluorescently labeled PRS-110. RESULTS: Biodistribution analyses showed a dose-dependent tumor uptake of (89)Zr-PRS-110, with the highest fractional tumor uptake at 10 µg of (89)Zr-PRS-110, with no unlabeled PRS-110. Small-animal PET imaging supported by biodistribution data revealed specific tumor uptake of (89)Zr-PRS-110 in the MET-expressing H441 and U87-MG tumors whereas the MET-negative A2780 tumor model showed a lower uptake similar to a non-MET binder anticalin control. Tumor uptake increased up to 24 h after tracer injection and remained high, whereas uptake in other organs decreased over time. Ex vivo fluorescence revealed intracellular presence of PRS-110. CONCLUSION: (89)Zr-PRS-110 specifically accumulates in MET-expressing tumors in a receptor density-dependent manner. PET imaging provides real-time noninvasive information about PRS-110 distribution and tumor accumulation in preclinical models.


Subject(s)
Proteins , Proto-Oncogene Proteins c-met/biosynthesis , Radiopharmaceuticals , Animals , Antibodies, Monoclonal, Humanized , Bevacizumab , Binding, Competitive , Cell Line, Tumor , Dose-Response Relationship, Drug , Isotope Labeling , Lipocalins , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Quality Control , Tissue Distribution , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...