Prostate cancer is one of the most common malignancies worldwide, yet limited tools exist for prognostic risk stratification of the disease. Identification of new biomarkers representing intrinsic features of malignant transformation and development of prognostic imaging technologies are critical for improving treatment decisions and patient survival. In this study, we analyzed radical prostatectomy specimens from 422 patients with localized disease to define the expression pattern of methionine aminopeptidase II (MetAP2), a cytosolic metalloprotease that has been identified as a druggable target in cancer. MetAP2 was highly expressed in 54% of low-grade and 59% of high-grade cancers. Elevated levels of MetAP2 at diagnosis were associated with shorter time to recurrence. Controlled self-assembly of a synthetic small molecule enabled design of the first MetAP2-activated PET imaging tracer for monitoring MetAP2 activity in vivo. The nanoparticles assembled upon MetAP2 activation were imaged in single prostate cancer cells with post-click fluorescence labeling. The fluorine-18–labeled tracers successfully differentiated MetAP2 activity in both MetAP2-knockdown and inhibitor-treated human prostate cancer xenografts by micro-PET/CT scanning. This highly sensitive imaging technology may provide a new tool for noninvasive early-risk stratification of prostate cancer and monitoring the therapeutic effect of MetAP2 inhibitors as anticancer drugs.

Significance:

This study defines MetAP2 as an early-risk stratifier for molecular imaging of aggressive prostate cancer and describes a MetAP2-activated self-assembly small-molecule PET tracer for imaging MetAP2 activity in vivo.

Prostate cancer is the second most commonly diagnosed malignancy among men and a leading cause of cancer-related deaths worldwide (1). Most cases of prostate cancer are asymptomatic and are detected within the prostate as a result of screening. However, screening has also resulted in overdetection of indolent prostate cancers resulting in overtreatment and unnecessary morbidity, necessitating the development of prognostic models in early disease (2). Current risk stratification uses serum PSA measurements, Gleason grade on biopsy (microscopic morphology of the cancer cells), and clinical stage for prognostication, although these provide only modest prediction of clinical outcomes (3). Identification of new biomarkers suitable for development of noninvasive prognostic technology is highly desirable.

PET imaging hybridized with CT or MRI is sensitive, noninvasive, three-dimensional (3D), and has demonstrated utility in improving the diagnosis of prostate cancer, especially for recurrent and advanced disease (4). Current prostate cancer imaging tracers approved by the FDA or in clinical trial typically rely on binding of a radiolabeled ligand, inhibitor, or antibody to a membrane receptor (5). However, not all prostate cancers express the membrane receptors targeted by these imaging agents. For example, neuroendocrine prostate cancer (NEPC), a highly aggressive form of advanced prostate cancer, commonly lacks expression of prostate-specific membrane antigen (PSMA), the target of a recently approved PET tracer, gallium-68 PSMA-11 (6, 7). PET tracers that can traverse cell membrane and report on activity of molecules essential for intrinsic biochemical processes, which undergo alterations during prostate cancer initiation and progression independent of the androgen receptor (AR) signaling axis, could offer attractive imaging targets.

Methionine aminopeptidase II (MetAP2) is a cytosolic metalloprotease that catalyzes the cotranslational removal of the N-terminus initiator methionine residue from nascent proteins (8). MetAP2 overexpression stimulates cancer cell proliferation (9) and is well known as a druggable target of angiogenesis, a pathologic determinant of progression in solid tumors (10). Considerable effort over many years has been devoted to developing MetAP2 inhibitors as anticancer drugs (11–13), because its expression has been correlated with proliferation and angiogenesis in advanced prostate cancer (14, 15). High MetAP2 expression has been observed in prostatic carcinoma (9); we now report that high levels of MetAP2 are predictive for shorter time to biochemical recurrence in prostate cancer treated surgically. Analysis of MetAP2 levels in a cohort of more than 400 patient prostate cancer samples from radical prostatectomies confirmed the overexpression of MetAP2 in more than half of prostate cancer samples regardless of grade, and MetAP2 overexpression was associated with a 2-fold increased risk of biochemical recurrence within 5 years. MetAP2, therefore, represents a promising target for noninvasive functional imaging of clinically significant prostate cancer.

We have devised a strategy termed target-enabled in situ ligand aggregation (TESLA) to develop molecular probes for imaging the activity of proteolytic enzymes in vivo that can be used with a variety of imaging modalities, including fluorescence imaging, photoacoustic imaging, MRI, and PET (16–19). This strategy was inspired by the rapid condensation reaction between 6-hydroxy-2-cyanobenzothiazole (CBT) and d-cysteine in the synthesis of firefly d-luciferin under physiologic conditions (20). The cysteine can be masked at its amino and thiol groups, for example, by a peptide substrate, and linked to the CBT unit in designing a TESLA probe. Such probes can undergo self-condensation and assemble into nanoaggregates after the target enzyme activation (17, 21–23). Recently, we have further explored this condensation reaction from CBT to other aromatic nitriles, such as 2-cyanopyrimidine and aminothiols (24). In this study, we designed novel MetAP2-sensitive nanoaggregation tracers (M-SNAT; Supplementary Figs. S1–S4) and validated them for imaging the activity of MetAP2 in a series of in vitro and in vivo models. The first demonstration of MetAP2-activatable small molecule for PET imaging in preclinical models highlights its potential for noninvasive diagnosis and risk stratification of early prostate cancer.

Cell culture

PC3, DU145, 22Rv1, LNCaP, and NCI-H660 carcinoma lines were obtained from the ATCC. LNCaP-Trop2 are engineered LNCaP cells overexpressing Trop2, as reported previously (25). Culture was maintained at 37°C with 5% CO2. Cells were split using 0.25% Trypsin-EDTA. PC3, DU145, 22Rv1, LNCaP, and LNCaP-Trop2 were cultured in RPMI1640 medium supplemented with 10% FBS, 100 U/mL penicillin, 100 μg/mL streptomycin, and 1% l-glutamine. H660 cells were cultured in RPMI1640 medium supplemented with 5% FBS, 0.005 mg/mL insulin, 0.01 mg/mL transferrin, 30 nmol/L sodium selenite, 10 nmol/L hydrocortisone, 10 nmol/L beta-estradiol, and 4 mmol/L l-glutamine. Human prostate epithelial cell line, PrEC, was cultured in prostate epithelial cell basal medium (PCS-440-030, ATCC) supplemented with 100 U/mL penicillin, 100 μg/mL streptomycin, 1% l-glutamine, and a Prostate Epithelial Cell Growth Kit (ATCC, PCS-440-040). PC3, DU145, 22Rv1, LNCaP, NCI-H660, and LNCaP-Trop2 were maintained below 10 passages. PrEC was maintained below three passages. All cell lines were routinely tested for Mycoplasma contamination (MycoAlert Mycoplasma Detection Kit purchased from Lonza), with authentication performed at Stanford Functional Genomics Facility (Stanford, CA) for short tandem repeat profiling.

Preparation of human prostatectomy slides and microarrays

Prostates were collected at Stanford Hospital (Stanford, CA) under institutional review board approval and written informed consent. All patient samples were deidentified prior to inclusion in this study. Subsequently, tissues were processed at the Stanford Department of Urology tissue bank and consisted of benign prostatic hyperplasia (n = 62), benign tissue adjacent to cancer (n = 63), low-grade cancer (Gleason 3 + 3 to 3 + 4; n = 249), or high-grade cancer (Gleason 4 + 3 to 5 + 5; n = 56). Samples were sliced to display clinically relevant disease, fixed overnight, then sectioned at 4 μm, and mounted on lysine-coated glass slides. Samples were stained for MetAP2 by IHC and scored by intensity by a pathologist from 0 to 3 (0, negative; 1, uninterpretable; 2, low intensity positive staining; and 3, high intensity positive staining).

Tissue microarrays (TMA) for clinical evaluation of MetAP2 were constructed at Stanford University (Stanford, CA) using an internal cohort of patients with a median follow-up of 10 years, as described previously (26). This cohort consisted of recurrent (n = 58) and nonrecurrent patients (n = 176) and included patients with Gleason score 3 + 3 to 4 + 4. Three cores per patient were included on the TMA. MetAP2 staining was performed as described next. Samples were scored for MetAP2 intensity by a genitourinary pathologist (C.A. Kunder) who was blinded to sample information.

IHC staining of tissue slides and TMAs

Tumor slices or TMAs were heated to 65°C for 1 hour prior to deparaffinization in Clearify and rehydration. Antigen retrieval was performed for 20 minutes in 10 mmol/L sodium citrate buffer pH 6 in a steam bath (95°C) and washed, followed by 3% H2O2 for 5 minutes. Slides were blocked for 1 hour in 2.5% goat serum at room temperature. Mouse primary antibody (OriGene, concentration 1:75 and Santa Cruz Biotechnology, 1:150) was added overnight at 4°C.

Preparation and IHC staining of xenograft tissue slides

Prostate cancer cells (5 × 105) were implanted with 100% Matrigel into the rear flank of 6- to 8-week-old NOD/SCID/IL2Rγ-null (NSG) mice. Tumors were grown to approximately 500 mm3 and harvested. Tumors were divided for either protein (flash-frozen in liquid nitrogen) or histologic analysis (fixed overnight at 4°C in 10% buffered formalin, paraffin embedded, and sliced at 4 μm thickness). IHC for xenografts was performed as described above.

Western blot analysis of cells and tumors

Cells were trypsinized, pelleted, and washed three times with ice-cold PBS and lysed in RIPA buffer. Tumor tissues were cut into small pieces and ground in 1.5 mL Eppendorf tubes with microtube pestles for lysis in RIPA buffer. Protein concentrations of centrifuged whole lysates were determined with BCA assay. As described previously (27), in brief, either 40 or 50 μg of cell or tumor lysates were loaded in each lane in NuPAGE 4% to 12% bis-tris protein gels for electrophoresis at 200 V for 90 minutes. Wet transfer was performed using a Bio-Rad Transfer Kit at 300 mA for 90 minutes. The transferred nitrocellulose membrane was blocked in PBS containing 5% BSA and 0.1% Tween-20 for 1 hour. Primary antibody incubation was performed in the blocking buffer overnight at 4°C at the recommended concentration. The membrane was then washed with PBS containing 0.1% Tween-20 for 5 minutes, four times. Secondary antibody incubation (LI-COR donkey anti-mouse IgG IRDye 680 or anti-rabbit IgG IRDye 800CW, 1:10,000) was performed in the blocking buffer for 2 hours at room temperature. After washing four times with PBS containing 0.1% Tween-20, membranes were analyzed in a LI-COR Odyssey Imaging System.

MetAP2-mediated macrocyclization in vitro

M-SNAT (10 μmol/L, 10 μL of 1 mmol/L stock in DMSO) was diluted in reaction buffer containing 50 mmol/L HEPES, 100 mmol/L NaCl, and 0.1 mmol/L MgCl2 at pH 7.5 with or without 1 mmol/L tris(2-carboxyethyl)phosphine (TCEP; 2 μL of 500 mmol/L stock in water) and 7.5 μg recombinant human MetAP2 in 1 mL final concentration. Reactions were performed at 37°C for 24 hours and monitored by high-performance liquid chromatography (HPLC).

Post-click labeling of M-SNAT in cells by Cy5-azide for microscope imaging and flow cytometry analysis

For microscopic imaging, PC3, DU145, 22Rv1, PC3 MetAP2-knockdown (KD), and 22Rv1 MetAP2-KD cells were seeded at about 50% confluence on cover glasses in 6-well plates a day before incubation with 20 μmol/L M-SNAT (10 μL of 2 mmol/L stock in DMSO) in medium containing 1% FBS and TNP-470 (2 μmol/L, 2 μL of 1 mmol/L stock in DMSO). After incubation at 37°C for 24 hours, cells were washed three times with PBS, and then fixed with 10% formalin for 30 minutes. Cells were permeabilized in PBS-Triton X100 (0.1% volume/volume) at room temperature for 15 minutes, and then washed three times with PBS. Post-click assay solution was freshly prepared with 100 mmol/L ascorbic acid (100 μL of 1 mol/L stock in water, freshly prepared), 1 mmol/L CuSO4 (10 μL of 100 mmol/L stock in water), 15 μmol/L (BimC4A)3 (0.5 μL of 30 mmol/L stock in water), and 5 μmol/L Cy5-azide (1 μL of 5 mmol/L in DMF) in 1 mL PBS. Cells were incubated with assay solution for 4 hours at 37°C, and then washed three times with PBS. Cells were then stained with 300 nmol/L DAPI in PBS for 10 minutes at room temperature, washed with PBS three times, and mounted on glass slides with antifade mounting medium.

The epifluorescence microscope images were acquired by 1 × 81 Inverted Microscope (Olympus) equipped with pE-4000 Illumination Systems (CoolLED) and ORCA-Flash4.0 Digital CMOS Camera (HAMAMATSU), with excitation at 405 nm for DAPI and 650 nm for Cy5. Digital images were reconstructed by MetaMorph software (v. 7.8.11.0) and analyzed using the ImageJ (NIH) software package.

The superresolution images were acquired by OMX BLAZE 3D-structured illumination, super-resolution microscope (Applied Precision) with simultaneous excitation at 405 nm for DAPI (emission filters 435/31 nm) and 642 nm for Cy5 (emission filters 683/40 nm), according to the standard procedure in the manufacturer’s instructions, and reconstructed with OMX softWoRx image processing software. Images were further processed using Volocity (v. 6.3.0, PerkinElmer) to generate the video.

For flow cytometry analysis, PC3 cells were trypsinized and resuspended after incubation in 20 μmol/L M-SNAT and medium containing 1% FBS with or without 2 μmol/L TNP-470 for 24 hours. Cells were centrifuged at 300 × g for 10 minutes and washed with PBS once, and then resuspended and fixed in 10% formalin at room temperature for 30 minutes. Cells were washed with PBS for three times by centrifugation, and then resuspended in post-click assay buffer at 37°C for 4 hours. Cells were washed with PBS for three times, and then analyzed with a 4-laser, 12-color DxP12 Cytek Upgrade (Becton Dickinson, Cytek Biosciences) and evaluated according to their size (forward scatter, FSC) on a linear scale, excited by 640 nm later, filtered by 655 nm long pass filter and 616/661 nm band pass filter. Flow cytometry data analysis was done using FlowJo V10 software. The mean fluorescence intensity was collected and plotted against the natural autofluorescence of cells.

Cell viability assay

Cell viability assay was performed with a CellTiter 96 AQueous One Solution Cell Proliferation Assay (MTS) Kit (Promega) according to the manufacturer’s protocol. PC3 cells were incubated with M-SNAT (100 pmol/L–100 μmol/L), cisplatin (500 μmol/L), and solvent (DMSO, 1%) in a 96-well plate at 37°C for 24 hours. The second day, medium was changed, and cells were incubated with MTS reagent for additional 3 hours. The absorbance at 490 nm was measured and plotted.

Generation of lentivirus and MetAP2-KD stable lines in PC3 and 22Rv1 cells

pLKO.1 control scramble short hairpin RNA (shRNA) vector (Addgene, plasmid #1864) or pLKO.1-MetAP2 shRNAs (Millipore Sigma, TRCN0000294143: GGATCATATACAGCG CAATTT; TRCN0000286742: GCAGAAGCACATCGACAAGTT; and TRCN0000306917: GACAAAGGCAAACCGTCTAAT) were cotransfected with pMDLg/pRRE, pRSV-Rev, and pMD2.G into 293T cells based on standard calcium phosphate transfection. The media with viral particles were collected at 24 and 48 hours, and 0.45 μm filtered for usage. A total of 2 × 105 PC3 or 22Rv1 cells were seeded in 6-well plates for infection with 2 mL viral media containing polybrene (10 μg/mL). Seventy-two hours after transfection, the infected cells underwent selection using puromycin (1 μg/mL). The media with puromycin were changed every 3 days and the stable lines were generated after 9 days for further validation utilizing Western blot analysis to determine the KD efficiency of MetAP2.

Stability in mouse serum

M-SNAT (100 μmol/L, 1 μL of 2 mmol/L stock in DMSO) was diluted in 20 μL mouse serum and incubated at 37°C for 0 minute, 15 minutes, 30 minutes, 1 hour, and 2 hours. Serum proteins were denatured by mixing cold methanol (980 μL) and precipitated by centrifugation at 8,000 × g for 10 minutes. Supernatants were analyzed by HPLC and LC/MS. Percentage of tracer (relative area) was calculated as (peak area of tracer/total peak area on the HPLC chromatogram) × 100.

Animals

All experimental procedures using mice were performed in agreement with protocols approved by the Institutional Animal Care and Use Committee (IACUC) of Stanford University, as well as the USAMRMC Animal Care and Use Review in accordance with the laws of the United States and regulations of the Department of Agriculture.

For the MetAP2 inhibition model, 6- to 8-week-old male nu/nu nude mice were purchased from Charles River Laboratories. Mice were housed in the Research Animal Facility at the Clark Center of the Stanford University. A total of 5 × 105 PC3 cells were injected subcutaneously into the right upper flank region of nu/nu nude mice in 50% Matrigel. When the tumors reached approximately 100 mm3, TNP-470 (stock in DMSO) was sequentially dissolved in ethanol and PBS, and then injected subcutaneously at a dose of 30 mg/kg at a position remote from the tumor (lower left back) every other day for 4 days and then injected every day for 5 additional days, with the last injection 4 hours prior to injection of PET tracer. Tumor dimensions were measured every other day by external caliper and tumor volume was calculated using the modified ellipsoid formula (length × width2)/2.

For PET imaging of MetAP2 stable KD models, NOD-SCID-IL2Rγ (NSG; Jackson Laboratory) male mice were bred at the Canary Center (Stanford, CA) in accordance with IACUC guidelines. A total of 2.5 × 105 wild-type (WT), sh-ctrl, or sh-MetAP2 PC3 cells were injected subcutaneously into bilateral flanks of 6-week-old male NSG mice in 100% Matrigel. Tumors were measured every third day by external caliper, and tumor volume was calculated by use of the formula (length × width × height)/2. Tumors were grown to around 300–1,000 mm3 volume on average at the time of imaging.

For biodistribution studies, 1 × 106 sh-ctrl or sh-MetAP2 PC3 cells were subjected to bilateral subcutaneous injection into the flanks of 6-week-old male NSG mice in 100% Matrigel. Tumors were measured every second or third day by external caliper, and tumor volume was calculated by use of the formula (length × width × height)/2. Tumors were grown to around 300 to 600 mm3 volume on average at the time of studies.

In vivo PET/CT imaging and data analysis

In experiments using the MetAP2 inhibitor, PET/CT imaging was performed using a Siemens Inveon micro PET/CT scanner (CT attenuation corrected). Totally, 8 mice were imaged in two batches, 4 mice each. Approximately 145 μCi of [18F]-M-SNAT (radiochemical purity of 99% as determined by HPLC; first batch, ∼14 pmol and second batch, ∼26 pmol) was injected intravenously into tumor-bearing mice. For the MetAP2 stable KD mouse model, PET imaging was performed using a Siemens Inveon D-PET micro-PET scanner (transmission attenuation corrected). CT was performed using a Sofie G-Next PET-CT scanner. Totally, 8 mice were imaged in two batches, 4 mice each. Approximately 79 μCi of [18F]-M-SNAT (radiochemical purity of 99% as determined by HPLC; first batch, ∼46 pmol and second batch, ∼80 pmol) was injected intravenously into tumor-bearing mice. Dynamic scans were acquired in list mode format over 90 minutes, and sorted into 22 times of frames, 0.5-mm sinogram bins for image reconstruction (4 × 15 seconds, 4 × 60 seconds, 11 × 300 seconds, and 3 × 600 seconds). Mice were anesthetized with 2% isoflurane near the center of the FOV to ensure the highest image resolution and sensitivity. Iterative reconstruction was performed using 3D-ordered subsets expectation maximization (OSEM), followed by fast maximum posteriori (MAP) with the following parameters: MAP OSEM iterations, 2; MAP subsets, 16; and MAP iterations, 18. PET images were registered with CT images. Siemens Inveon Research Workplace software v.4.2 was used for visualization of radiotracer uptake in the tumor, and to define the 3D volumes of interest (VOI). The relative tumor or organ radioactivity concentrations were from mean pixel values within the multiple VOIs and converted to counts per milliliter per minute and then divided by the injected dose (ID) to obtain an imaging VOI-derived percentage of the injected radioactive dose per cubic centimeter of tissue (%ID/cc). We assumed the average tissue density is close to density of water and converted %ID/cc to percentage of the injected radioactive dose per gram tissue (%ID/g).

Ex vivo biodistribution studies

Exvivo biodistribution studies were performed to measure tumor- and tissue-associated radioactivity. Totally, 18 mice were analyzed in two batches, 9 mice in the first 40 μCi (∼38.28 pmol) injection batch (sh-ctrl, n = 4 and sh-MetAP2, n = 5) and 9 mice in the second 300 μCi (∼520.90 pmol) injection batch (sh-ctrl, n = 4 and sh-MetAP2, n = 5). [18F]-M-SNAT (radiochemical purity of 99% as determined by HPLC) was injected intravenously into tumor-bearing mice. Mice were sacrificed 60 minutes after injection to collect tumor, heart, lung, spleen, muscle, liver, kidney, prostate, and bone. The radioactivity in each tissue was measured using a gamma counter (Hidex automatic gamma counter) and normalized to tissue weight and the amount of radioactivity administered to each mouse, which was decay corrected to the time of radiotracer injection. Data were expressed as %ID/g values.

Statistical analysis

GraphPad Prism 7 was utilized for plotting and statistical analysis. The significant difference was determined by performing one-way or two-way ANOVA followed by Bonferroni multiple comparisons test and t tests (Mann–Whitney test, nonparametric) to determine the statistical significance with 95% confidence intervals with *, P < 0.0332; **, P < 0.0021; ***, P < 0.0002; ****, P < 0.0001; ns, not significant.

Data and materials availability

All data associated with this study are present in the article or Supplementary Materials and Methods. Materials are available and will be provided under the material transfer policies of Stanford University. These requests should be directed to the corresponding authors.

High levels of MetAP2 are predictive for biochemical recurrence in prostate cancer

We first measured the protein levels of MetAP2 in a panel of prostate cell lines, including a benign human prostate epithelial cell line, PrEC (28), and prostate cancer cell lines PC3, DU145, and 22Rv1, and their corresponding tumor xenografts. PrEC cells expressed significantly lower levels of MetAP2 when compared with PC3, DU145, and 22Rv1 prostate cancer cells and xenografts (Fig. 1A). MetAP2 was also expressed in LNCaP prostate cancer xenografts and xenografts from two different models of NEPC, NCI-H660, and NEPC driven by the Trop2 oncogene (TD-NEPC; Fig. 1B and C; ref. 25). In contrast to MetAP2, PSMA was only detected in LNCaP xenografts, suggesting that utilizing MetAP2 as a target for new imaging modalities may detect additional subsets of prostate cancer missed by PSMA tracers. Together, these findings demonstrate that MetAP2 levels are elevated in models of adenocarcinoma and NEPC.

Figure 1.

MetAP2 is highly expressed in prostate cancer cells, xenografts, and human prostate cancer and associates with biochemical recurrence. A and B, Western blot analysis of MetAP2 in prostate cell lines and xenografts, and AR (FL, full length; V7, variant) and PSMA in tumor xenografts; 40 μg of each whole-cell or tumor lysate was loaded. A total of 5 × 105 cells in 100% Matrigel were implanted subcutaneously into rear flank of NSG mice. C, Representative images of IHC staining of MetAP2 and PSMA in xenograft prostate tumor samples from PC3, DU145, 22Rv1, NCI-H660, LNCaP, and LNCaP-Trop2 (NEPC model). Scale bar, 50 μm. D, Representative images of IHC staining from prostate cancer patient samples showing the overexpression of MetAP2 in both low- and high-grade tumors, but not benign regions. IHC was done in parallel with MetAP2 primary antibodies from Santa Cruz Biotechnology (Supplementary Fig. S5) and OriGene (shown here) to demonstrate reproducibility. Scale bar, 500 μm (left) and 100 μm (right). E, Intensity of the staining of MetAP2 in patient specimens was scored from 0 to 3 by a pathologist (0, negative; 1, uncertain/excluded because of lack of sufficient tissue for evaluation; 2, weakly positive; and 3, strongly positive). Percentage of patients with none (0), low (average >1 and ≤2), and high (average >2) MetAP2 staining was presented within each group of samples (BPH, n = 62; benign, n = 63; 3 + 3 to 3 + 4, n = 249; and 4 + 3 to 5 + 4, n = 56). F, The mean intensity in BPH, benign adjacent to cancer, low-grade cancer, and high-grade cancer. Error bars, SE; ****, P < 0.0001; ns, no significance. G, Kaplan–Meier analysis showing biochemical recurrence of patients (n = 212) stratified by strong MetAP2 IHC staining in at least two of the three biopsy cores (MetAP2 high, red curve) and all the other patients (MetAP2 negative/low, blue curve).

Figure 1.

MetAP2 is highly expressed in prostate cancer cells, xenografts, and human prostate cancer and associates with biochemical recurrence. A and B, Western blot analysis of MetAP2 in prostate cell lines and xenografts, and AR (FL, full length; V7, variant) and PSMA in tumor xenografts; 40 μg of each whole-cell or tumor lysate was loaded. A total of 5 × 105 cells in 100% Matrigel were implanted subcutaneously into rear flank of NSG mice. C, Representative images of IHC staining of MetAP2 and PSMA in xenograft prostate tumor samples from PC3, DU145, 22Rv1, NCI-H660, LNCaP, and LNCaP-Trop2 (NEPC model). Scale bar, 50 μm. D, Representative images of IHC staining from prostate cancer patient samples showing the overexpression of MetAP2 in both low- and high-grade tumors, but not benign regions. IHC was done in parallel with MetAP2 primary antibodies from Santa Cruz Biotechnology (Supplementary Fig. S5) and OriGene (shown here) to demonstrate reproducibility. Scale bar, 500 μm (left) and 100 μm (right). E, Intensity of the staining of MetAP2 in patient specimens was scored from 0 to 3 by a pathologist (0, negative; 1, uncertain/excluded because of lack of sufficient tissue for evaluation; 2, weakly positive; and 3, strongly positive). Percentage of patients with none (0), low (average >1 and ≤2), and high (average >2) MetAP2 staining was presented within each group of samples (BPH, n = 62; benign, n = 63; 3 + 3 to 3 + 4, n = 249; and 4 + 3 to 5 + 4, n = 56). F, The mean intensity in BPH, benign adjacent to cancer, low-grade cancer, and high-grade cancer. Error bars, SE; ****, P < 0.0001; ns, no significance. G, Kaplan–Meier analysis showing biochemical recurrence of patients (n = 212) stratified by strong MetAP2 IHC staining in at least two of the three biopsy cores (MetAP2 high, red curve) and all the other patients (MetAP2 negative/low, blue curve).

Close modal

We further evaluated the levels of MetAP2 in human prostate cancer by IHC staining of radical prostatectomy sections and TMAs containing samples from 422 patients with prostate cancer, of which, 212 patients had 10 years of follow-up (29). As shown in Fig. 1D, benign prostate tissues had low levels of MetAP2, while low-grade prostate cancers (Gleason score 3 + 3) expressed moderate to high levels of MetAP2, and high-grade cancers (Gleason score 4 + 4 and 5 + 5) expressed high MetAP2 levels throughout the cancerous tissues (Fig. 1D and E; Supplementary Fig. S5). Benign prostatic hyperplasia (BPH) and benign adjacent to cancer tissues had significantly lower levels of MetAP2 than both low- and high-grade prostate cancers, while there was no significant difference between BPH and benign adjacent to prostate cancer (Fig. 1F). Among the 212 patients with long-term follow-up (recurrent, n = 52; nonrecurrent, n = 160), high levels of MetAP2 at the time of surgery were associated with biochemical recurrence (Fig. 1G; Supplementary Fig. S6).

Design of MetAP2-sensitive probes

Figure 2A illustrates the design of M-SNAT: the probe carries a methionine (P1, green) followed by a cysteine (P2) on the N-terminus that is connected to a 2-cyanopyrimidine (blue) through a linker. In eukaryotes, MetAP2 substrates prefer penultimate residues (P2) with a small nonpolar side chain (e.g., Gly, Ala, Ser, Thr, Val, and Cys; ref. 30). After intravenous administration, M-SNAT extravasates into tissues. In normal prostatic or BPH tissues with minimal MetAP2 activity, M-SNAT will mostly remain intact or be partially reduced and will, therefore, diffuse away freely. In prostate cancer with high MetAP2 activity, cleavage and release of the P1 methionine residue by MetAP2, as well as the reduction of the disulfide bond by intracellular glutathione (GSH), trigger the intramolecular condensation reaction. Unlike the unprocessed probes, cyclized macromolecules become rigid and tend to interact with each other to form molecular nanoaggregates (Fig. 2B). By tagging the probe with a radioisotope like fluorine-18, the location and amounts of these nanoaggregates can be detected in vivo through PET/CT imaging (Fig. 2C, top compound). For in vitro studies, a post-click reaction at the propargylglycine with an azide-conjugated far-red fluorophore, Cy5, was utilized to pinpoint the aggregated and precipitated M-SNAT in cells (Fig. 2C, middle compound). A control compound (M-SNAT-ctrl), with a methylated thiol that prevents intramolecular cyclization after methionine cleavage by MetAP2, was synthesized to examine the role of cyclization and aggregation in the activity of M-SNAT (Fig. 2C, bottom compound).

Figure 2.

Illustration of the mechanism of in vivo imaging of MetAP2 activity by [18F]-M-SNAT in human prostate cancer. A, Proposed MetAP2- and cellular GSH–controlled conversion of M-SNAT into M-SNAT-cyclized through the bioorthogonal intramolecular cyclization reaction, followed by self-assembly into nanoaggregates in situ. Blue, 2-cyanopyrimidine; brown and orange, amino and thiol groups of d-cysteine, respectively; yellow, thioethyl masking group; green, the capping methionine residue; pink, isotope molecule. B, After intravenous administration, M-SNAT extravasates into tumor tissue because of its small size. In normal or benign tissue that expresses minimal amounts of MetAP2, the methionine mostly remains and M-SNAT can diffuse away freely, leading to low signal. In aggressive cancer cells overexpressing MetAP2, after methionine cleavage, M-SNAT undergoes macrocyclization and in situ nanoaggregation, leading to enhanced probe retention and high radioactive signal. C, Structures of [18F]-M-SNAT, M-SNAT, and M-SNAT-Ctrl.

Figure 2.

Illustration of the mechanism of in vivo imaging of MetAP2 activity by [18F]-M-SNAT in human prostate cancer. A, Proposed MetAP2- and cellular GSH–controlled conversion of M-SNAT into M-SNAT-cyclized through the bioorthogonal intramolecular cyclization reaction, followed by self-assembly into nanoaggregates in situ. Blue, 2-cyanopyrimidine; brown and orange, amino and thiol groups of d-cysteine, respectively; yellow, thioethyl masking group; green, the capping methionine residue; pink, isotope molecule. B, After intravenous administration, M-SNAT extravasates into tumor tissue because of its small size. In normal or benign tissue that expresses minimal amounts of MetAP2, the methionine mostly remains and M-SNAT can diffuse away freely, leading to low signal. In aggressive cancer cells overexpressing MetAP2, after methionine cleavage, M-SNAT undergoes macrocyclization and in situ nanoaggregation, leading to enhanced probe retention and high radioactive signal. C, Structures of [18F]-M-SNAT, M-SNAT, and M-SNAT-Ctrl.

Close modal

Macrocyclization and nanoaggregation of M-SNAT in vitro

Macrocyclization of M-SNAT to produce M-SNAT-cyclized was monitored in solution using HPLC and mass spectrometry. After 24-hour incubation with recombinant human MetAP2 in the presence of TCEP to mimic the intracellular reducing environment, M-SNAT (10 μmol/L; HPLC retention time, TR = 15.5 minutes) was converted completely into M-SNAT-cyclized (TR = 11.8 minutes; Fig. 3A; Supplementary Fig. S7). In contrast, the M-SNAT-ctrl cannot cyclize due to the nonreducible methylated thiol group (Supplementary Fig. S8). Dynamic light scattering (DLS) analysis showed the formed nanoparticles with an average diameter of 861 nm (531–1281 nm; Fig. 3B), which was confirmed by transmission electron microscopy (TEM; Fig. 3C; Supplementary Fig. S9). MetAP2 catalyzed removal of the methionine residue and activated M-SNAT to undergo intramolecular macrocyclization and subsequent nanoassembly.

Figure 3.

In vitro characterization of M-SNAT. A, HPLC traces of M-SNAT (black) and M-SNAT (10 μmol/L) after incubation with TCEP (1 mmol/L) and recombinant human MetAP2 (7.5 μg/mL) at 37°C in reaction buffer for 24 hours (red). B, DLS analysis shows the formation of nanoparticles with an average diameter of 861 nm (531–1281 nm). C, TEM image of nanoaggregates after incubation of M-SNAT (10 μmol/L) with MetAP2.

Figure 3.

In vitro characterization of M-SNAT. A, HPLC traces of M-SNAT (black) and M-SNAT (10 μmol/L) after incubation with TCEP (1 mmol/L) and recombinant human MetAP2 (7.5 μg/mL) at 37°C in reaction buffer for 24 hours (red). B, DLS analysis shows the formation of nanoparticles with an average diameter of 861 nm (531–1281 nm). C, TEM image of nanoaggregates after incubation of M-SNAT (10 μmol/L) with MetAP2.

Close modal

Imaging MetAP2 activity in prostate cancer cells

The uptake and MetAP2-specific activation of M-SNAT were evaluated in PC3 and DU145 prostate cancer cells. Post-click fluorescence labeling with Cy5-azide was performed to pinpoint the nanoaggregation in cells and to avoid possible interference with probe uptake incurred by conjugating the probe with a bulky Cy5 fluorophore prior to treating the cells (Fig. 4A). A well-characterized MetAP2 inhibitor (TNP-470), which inhibits the aminopeptidase activity by alkylating the His231 residue in the active center, was used to evaluate the probe specificity (10). Under microscope, red fluorescence was observed in M-SNAT–treated PC3 and DU145 cells in a concentration-dependent manner after 24-hour incubation, while there was minimal fluorescence signal in cells with the control probe M-SNAT-ctrl (Fig. 4B; Supplementary Figs. S10–S12). When TNP-470 (2 μmol/L) was added, the fluorescence in both PC3 and DU145 cells was significantly reduced (Fig. 4B; Supplementary Fig. S11). Flow cytometry analysis of PC3 cells incubated with M-SNAT and postlabeled with Cy5-azide revealed 22.7% inhibition by TNP-470 (Fig. 4C; Supplementary Fig. S13). These data indicate the requirement of MetAP2 hydrolysis in conversion of M-SNAT to cyclized product and the macrocyclization-dependent probe nanoaggregation and retention in cells. We evaluated the toxicity of M-SNAT up to 100 μmol/L in PC3 cells and found little effect on the cell viability (Supplementary Fig. S14).

Figure 4.

Characterization of M-SNAT in prostate cancer cells. A, Illustration of the post-click labeling of MetAP2-activated M-SNAT nanoaggregation by Cy5-azide. B, Fluorescence microscopic imaging of cy5 (red, Ex650/Em670) postlabeled M-SNAT (20 μmol/L) with or without TNP470 (2 μmol/L) and M-SNAT-ctrl (20 μmol/L) in PC3 cells. Permeabilized cells were also stained with DAPI (blue, Ex405/Em470). C, Flow cytometry analysis of PBS-, M-SNAT– (20 μmol/L), and with or without TNP-470 (2 μmol/L)–treated PC3 cells. Cells were suspended, fixed, and permeabilized for post-click labeling after 24-hour incubation. The intensity of Cy5 in each group was plotted either individually or as overlay. The percentage, instead of actual counts in forward scatter, was normalized to its mode as shown in distribution. D and E, Western blot analysis of WT, KD control (Ctrl), and MetAP2-KD (M1–3) PC3 and 22Rv1 cells. A total of 50 μg of each whole-cell lysate was loaded. F and G, Growth curves of WT, KD control (sh-ctrl), and MetAP2-KD PC3 and 22Rv1 cells. Error bars, SE; *, P < 0.0332; **, P < 0.0021; ns, no significance. H, Fluorescence microscopy imaging of Cy5 postlabeled M-SNAT (20 μmol/L) in KD control (Sh-ctrl) and MetAP2-KD (M1 and M2) PC3 and 22Rv1 cells. Permeabilized cells were also stained with DAPI.

Figure 4.

Characterization of M-SNAT in prostate cancer cells. A, Illustration of the post-click labeling of MetAP2-activated M-SNAT nanoaggregation by Cy5-azide. B, Fluorescence microscopic imaging of cy5 (red, Ex650/Em670) postlabeled M-SNAT (20 μmol/L) with or without TNP470 (2 μmol/L) and M-SNAT-ctrl (20 μmol/L) in PC3 cells. Permeabilized cells were also stained with DAPI (blue, Ex405/Em470). C, Flow cytometry analysis of PBS-, M-SNAT– (20 μmol/L), and with or without TNP-470 (2 μmol/L)–treated PC3 cells. Cells were suspended, fixed, and permeabilized for post-click labeling after 24-hour incubation. The intensity of Cy5 in each group was plotted either individually or as overlay. The percentage, instead of actual counts in forward scatter, was normalized to its mode as shown in distribution. D and E, Western blot analysis of WT, KD control (Ctrl), and MetAP2-KD (M1–3) PC3 and 22Rv1 cells. A total of 50 μg of each whole-cell lysate was loaded. F and G, Growth curves of WT, KD control (sh-ctrl), and MetAP2-KD PC3 and 22Rv1 cells. Error bars, SE; *, P < 0.0332; **, P < 0.0021; ns, no significance. H, Fluorescence microscopy imaging of Cy5 postlabeled M-SNAT (20 μmol/L) in KD control (Sh-ctrl) and MetAP2-KD (M1 and M2) PC3 and 22Rv1 cells. Permeabilized cells were also stained with DAPI.

Close modal

To further validate the specificity of M-SNAT activation by MetAP2, we generated MetAP2-KD PC3 and 22Rv1 cells. Three KD lines (M1–M3) of each cell line were generated via transduction of lentivirus carrying different shRNA sequences. A scramble shRNA was used to generate KD controls. The cell lines with the highest KD (PC3-M1 and 22Rv1-M2) were selected for cell-based studies (Fig. 4D and E). Previous work has shown that cells deficient in p53 show less inhibition of proliferation by TNP-470 due to disruption of p53-dependent induction of p21Waf1/CIP1 to induce cell-cycle arrest (31, 32). As expected, MetAP2 KD slowed the growth of 22Rv1-M2 cells expressing WT p53, but had minimal impact on the growth of PC3-M1 (p53 null) cells (Fig. 4F and G; ref. 33). High doses of TNP-470, however, were reported to slow the growth of PC3 cells 30 days after tumor implantation (15). After incubation with M-SNAT for 24 hours, both PC3- and 22Rv1-KD cells showed much weaker red fluorescence than their WT counterparts (Fig. 4H; Supplementary Figs. S15 and S16). Taken together, these cell studies demonstrate specific intracellular activation and retention of M-SNAT due to MetAP2 activity.

Direct observation of the nanoparticles formed in single prostate cancer cells

Aggregated and Cy5 postlabeled M-SNAT assemblies observed in Fig. 4B were further visualized at a single-cell level using superresolution structured illumination microscope (SR-SIM), which provided both lateral (XY) and axial (Z) resolutions at twice the diffraction limit of conventional high-resolution light microscope systems, such as confocal (Fig. 5; Supplementary Movie S1). The size of particles formed in situ varied from a few hundred nanometers to more than 1 μm, consistent with the values measured by DLS and TEM (Fig. 3B and C). The postlabeled red fluorescence aggregates occurred throughout the cytoplasm with a relatively higher density surrounding the nucleus.

Figure 5.

3D projected SR-SIM imaging of cy5 postlabeled self-nanoaggregation of M-SNAT in a single PC3 cell. Representative SR-SIM image of self-aggregated fluorescence nanoparticles in PC3 cells incubated with M-SNAT (20 μmol/L) and postlabeled by Cy5. Cells were costained with DAPI. Red, aggregated M-SNAT; blue, nucleus. The yellow boxes i and ii indicate the enlarged area. Arrow points to an aggregation structure in the cell.

Figure 5.

3D projected SR-SIM imaging of cy5 postlabeled self-nanoaggregation of M-SNAT in a single PC3 cell. Representative SR-SIM image of self-aggregated fluorescence nanoparticles in PC3 cells incubated with M-SNAT (20 μmol/L) and postlabeled by Cy5. Cells were costained with DAPI. Red, aggregated M-SNAT; blue, nucleus. The yellow boxes i and ii indicate the enlarged area. Arrow points to an aggregation structure in the cell.

Close modal

[18F]-M-SNAT enables visualization of MetAP2 activity in prostate cancer xenografts

The ability of [18F]-M-SNAT to report MetAP2 activity in vivo was assessed in two different models of MetAP2 inhibition. To test the specificity of [18F]-M-SNAT for MetAP2 activity independent of tumor growth, we used a PC3 prostate cancer model in which cell growth was not affected by MetAP2 knockdown (Fig. 4F). PC3 cells (5 × 105) were implanted subcutaneously in male nude mice (15) and measured every other day for 18 days until reaching approximately 100 mm3. Starting at day 18, PBS with or without TNP-470 (30 mg/kg) was injected subcutaneously every other day for 4 days, and then daily for 5 additional days, with the last injection 4 hours prior to PET imaging. Consistent with the in vitro knockdown experiments, there were no significant differences in tumor volumes (∼400 mm3) between the inhibitor- or PBS-treated groups over the short period of treatment (Fig. 6A). [18F]-M-SNAT was prepared in a rapid two-step chemical synthesis using the labeling chemistry as described previously (Fig. 2C; Supplementary Fig. S3; ref. 20). Mice received an intravenous injection of approximately 145 μCi [18F]-M-SNAT and underwent a programed 90-minute micro-PET/CT whole-body scan. As shown in Fig. 6B and C (corresponding 3D projection in Supplementary Fig. S17), coregistered PET (sky color) and CT (gray and white) images demonstrated that PC3 xenografts in PBS-treated mice retained higher radioactivity (up to 1.6 %ID/g) than tumors in mice treated with TNP-470 (around 1 %ID/g). Tumors were resected 20 hours after tracer injection for ex vivo analysis. As antibodies used for IHC recognize epitopes upstream and distant from the active binding pocket (β sheet 1–8, from Asn223; ref. 34), changes in expression of MetAP2 by IHC were not seen after inhibitor treatment (Supplementary Fig. S18). CD31 (PECAM1) staining revealed slightly decreased neovascularization in TNP-470–treated tumors, reflecting its known antiangiogenic effect (Supplementary Fig. S18).

Figure 6.

[18F]-M-SNAT PET/CT imaging and distribution studies of PC3 tumor xenografts. A, Growth curves of PC3 tumors treated with PBS/DMSO/ethanol (ctrl; n = 4) or TNP-470/DMSO dissolved in ethanol and PBS (inhibitor; n = 4). Black arrows, time point of inhibitor injection. Error bars, SE; ns, no significance. B, Curves of the percentage of tumor-retained injection dosage per gram acquired by defining the VOIs with Inveon Research Workplace 4.2. Error bars, SE; *, P < 0.0332. C, Representative plane images after 90-minute programmed micro-PET/CT scan of PBS- or inhibitor-treated mice. Tumors were centered for comparison. White and gray indicate CT signal; sky colors (0.5%–2.5% of injected dosage per gram) indicate the mean value of accumulated PET signal. Yellow arrowheads, tumors. D, Growth curves of WT, sh-ctrl, and sh-MetAP2 tumors, n = 10. Error bars, SE; ns, no significance. E, Curves of the percentage of injection dosage per gram tumor retained acquired by defining the VOIs. WT, n = 3; sh-ctrl, n = 5; shMetAP2-M1, n = 4. Error bars, SE; **, P < 0.0021; ns, no significance. F, Representative plane images after 90-minute programmed PET/CT scan of WT, sh-ctrl, and sh-MetAP2-M1 PC3 cells implanted in mice. The WT tumor on the right flank of a representative mouse was <300 mm3, therefore, excluded from analysis. G, Western blot analysis of MetAP2 in representative WT, sh-ctrl, and all sh-MetAP2-M1 PC3 tumor xenografts. Tumors were collected 1 day after imaging; 40 μg of each tumor lysate was analyzed. H, Quantification of [18F]-M-SNAT signal (%ID/g) from imaging data in PBS-treated and sh-ctrl mice by defining the VOIs at 65 minutes. Error bars, SE. I, Quantification of [18F]-M-SNAT signal (%ID/g) from ex vivo biodistribution (Bio-D) analysis of tumor, heart, lung, spleen, muscle, liver, kidney, prostate, and bone 60 minutes after tracer administration. Tracer uptake in tumors (Sh-ctrl, n = 8 and sh-MetAP2-M1, n = 10) was compared using t tests (Mann–Whitney test). Control, ctrl.

Figure 6.

[18F]-M-SNAT PET/CT imaging and distribution studies of PC3 tumor xenografts. A, Growth curves of PC3 tumors treated with PBS/DMSO/ethanol (ctrl; n = 4) or TNP-470/DMSO dissolved in ethanol and PBS (inhibitor; n = 4). Black arrows, time point of inhibitor injection. Error bars, SE; ns, no significance. B, Curves of the percentage of tumor-retained injection dosage per gram acquired by defining the VOIs with Inveon Research Workplace 4.2. Error bars, SE; *, P < 0.0332. C, Representative plane images after 90-minute programmed micro-PET/CT scan of PBS- or inhibitor-treated mice. Tumors were centered for comparison. White and gray indicate CT signal; sky colors (0.5%–2.5% of injected dosage per gram) indicate the mean value of accumulated PET signal. Yellow arrowheads, tumors. D, Growth curves of WT, sh-ctrl, and sh-MetAP2 tumors, n = 10. Error bars, SE; ns, no significance. E, Curves of the percentage of injection dosage per gram tumor retained acquired by defining the VOIs. WT, n = 3; sh-ctrl, n = 5; shMetAP2-M1, n = 4. Error bars, SE; **, P < 0.0021; ns, no significance. F, Representative plane images after 90-minute programmed PET/CT scan of WT, sh-ctrl, and sh-MetAP2-M1 PC3 cells implanted in mice. The WT tumor on the right flank of a representative mouse was <300 mm3, therefore, excluded from analysis. G, Western blot analysis of MetAP2 in representative WT, sh-ctrl, and all sh-MetAP2-M1 PC3 tumor xenografts. Tumors were collected 1 day after imaging; 40 μg of each tumor lysate was analyzed. H, Quantification of [18F]-M-SNAT signal (%ID/g) from imaging data in PBS-treated and sh-ctrl mice by defining the VOIs at 65 minutes. Error bars, SE. I, Quantification of [18F]-M-SNAT signal (%ID/g) from ex vivo biodistribution (Bio-D) analysis of tumor, heart, lung, spleen, muscle, liver, kidney, prostate, and bone 60 minutes after tracer administration. Tracer uptake in tumors (Sh-ctrl, n = 8 and sh-MetAP2-M1, n = 10) was compared using t tests (Mann–Whitney test). Control, ctrl.

Close modal

To further evaluate the specificity of [18F]-M-SNAT for imaging MetAP2 activity, we developed MetAP2-KD PC3 xenograft models. A total of 2.5 × 105 WT, MetAP2-KD (sh-MetAP2-M1), or scramble shRNA–transfected (sh-ctrl) PC3 cells (Fig. 4D) were suspended in 100% Matrigel and subcutaneously injected bilaterally into rear flanks of male NSG mice (n = 5, each group). Tumors were measured every 3 days after implantation. As observed in vitro (Fig. 4F), growth of PC3 tumors in NSG mice was not affected by MetAP2 KD (Fig. 6D). At day 30, mice carrying WT (n = 2), sh-MetAP2-M1 (n = 3), and sh-ctrl (n = 3) tumors were injected with [18F]-M-SNAT (∼79 μCi) and imaged by a micro-PET/CT for 90 minutes. Tumors larger than 900 mm3 or smaller than 300 mm3 were excluded from analysis to minimize the impact of tumor volumes on tracer uptake. The PET signal in sh-MetAP2-M1 tumors was significantly lower than WT and sh-ctrl tumors, with less than 1% uptake in sh-MetAP2-M1 tumors compared with 1.5% uptake in both WT and sh-ctrl tumors (Fig. 6E and F; corresponding 3D projection in Supplementary Fig. S19). Ex vivo analysis confirmed the lower expression of MetAP2 in imaged sh-MetAP2-M1 tumors than WT and sh-ctrl tumors (Fig. 6G). Whole-body distribution of [18F]-M-SNAT in both models at 65 minutes postinjection was analyzed by defining the 3D VOIs and showed that most radioactivity was in the kidney and bladder, with additional uptake in the liver (Fig. 6H). We compared the uptake in liver between PBS- and inhibitor-treated groups, but found no statistical differences, suggesting that uptake in the liver was not due to MetAP2 activity (Supplementary Fig. S20). Bone, muscle, and the pelvic region, in contrast, had very little uptake of the tracer, making it ideal for imaging primary and potentially metastatic prostate cancer lesions at these sites (Fig. 6H). NSG mice have been shown to have a faster clearance of antibodies and antibody–drug conjugates compared with nude mice due to an abnormal Fc interaction (35). We compared the biodistribution of [18F]-M-SNAT in NSG and nude mice, particularly in the spleen, and found they were similar overall (Fig. 6H). The extreme immunodeficiency in NSG mice did not appear to interfere with the biodistribution of [18F]-M-SNAT.

To validate the aggregation-dependent tracer retention in vivo, we performed a titration study with two different doses of [18F]-M-SNAT in our NSG/MetAP2-KD prostate cancer xenograft model: low (40 μCi) and high (300 μCi). Because the cyclic products from MetAP2 are more hydrophobic than the tracer itself and tend to interact with each other, a higher local concentration should promote nanoaggregation of the cyclic products, the mechanism for contrast enhancement. As shown in Fig. 6I (left), a high (300 μCi, 520.90 pmol) dose of [18F]-M-SNAT showed higher tracer radioactivity (%ID/g) at 1 hour postinjection than a low dose (40 μCi, 38.28 pmol; P = 0.0019) in NSG mice bearing PC3 xenografts (sh-ctrl). This result is consistent with the proposed nanoaggregation mechanism, and also provided direct evidence that the MetAP2-activated nanoaggregation could occur at the concentration as low as 40 μCi (P = 0.0005). Consistent to the imaging data, the distribution results showed that most radioactivity was in the liver and kidney. The prostate, bone, and muscle, in contrast, had very little uptake of the tracer (Fig. 6I, right). Finally, M-SNAT seems to be relatively stable in the serum, with a gradual decline from 100% to approximately 25% in 2-hour incubation at 37°C mainly because of hydrolysis of the methionine residue (Supplementary Fig. S21).

Using the TESLA strategy, we developed a novel molecular imaging technology for noninvasive imaging of MetAP2 activity based on conversion of a small, diffusible molecule into cyclized, nondiffusible form that undergoes nanoaggregation in the cytoplasm of cancer cells. The probe chemistry is flexible and allows easy modification for generating fluorescence read out at a single-cell level, as well as a radiative output signal for detection in vivo. Because the tracer retention directly depends on MetAP2 expression and activity to produce contrast (Figs. 4 and 6), the images correlate with the catalytic activity of the enzyme in situ. This MetAP2 imaging strategy has potential for clinical translation because there are ongoing needs to develop imaging tools that identify clinically aggressive prostate cancers, and MetAP2 tends to be expressed in some high-risk localized cancer and in model systems of castrate-resistant prostate cancer and NEPC.

Several PET tracers have been studied for imaging prostate cancer in localized disease to identify lesions in the prostate, when planning localized therapy to identify lymph node metastases, at time of biochemical recurrence after definitive local therapies, such as surgery or radiotherapy, and when metastasized to bone. The 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG) PET is used in many cancers, but has limited sensitivity for imaging prostate cancer. Choline agents, [11C]-acetate, and [18F]-fluciclovine have better sensitivity than conventional imaging technologies under particular clinical settings (36). The advent of PSMA PET imaging with either gallium-68 or fluorine-18 conjugated small molecules is revolutionizing the diagnosis and management of prostate cancer (37). However, PSMA is expressed in benign prostatic epithelium, resulting in false positive findings within the prostate (38), and is incrementally upregulated in primary cancer, lymph node, and distant metastases (39). PSMA expression does not correlate with tumor aggressiveness in localized prostate cancer or with risk of recurrence after surgery (40–42). Discovery of reliable prognostic biomarkers, especially as imaging agents capable of early-stage disease stratification, remains a clinical demand that could be met, in part, through MetAP2 imaging.

MetAP2 catalyzes critical N-terminus initiator methionine hydrolysis and has been studied as a target of antiangiogenic compounds in a variety of solid tumors, including prostate cancer (43, 44). Although MetAP2 has been heavily investigated as a therapeutic target, its use as a diagnostic imaging target had yet to be explored. In localized prostate cancer, high expression of MetAP2 in 54% of low-grade and 59% of high-grade cancers, and low or absent expression in noncancerous prostatic tissues (BPH and normal adjacent tissues) make it an attractive imaging target. Furthermore, the finding that prostate cancer cases with elevated MetAP2 have twice the risk of biochemical recurrence within 5 years following surgery suggest that MetAP2 imaging could be particularly useful for identifying clinically significant localized disease, a major unmet clinical need (2). In addition, the finding that MetAP2 highly expressed in model systems of castrate resistant and NEPC merits additional study since this imaging approach identifies MetAP2 enzymatic activity, thereby identifying tumors that could be treated with MetAP2 targeted therapies.

The synthesis and radiolabeling of [18F]-M-SNAT are straightforward and ideal for large-scale preparation. We characterized [18F]-M-SNAT in both inhibitor (TNP-470) treated and stable KD PC3 xenograft models. Because TNP-470 (50–200 mg/kg) treatment in nude mice has been reported with toxicity, and even death (15), we lowered the injection dose to 30 mg/kg and performed multiple injections till the tumors reached approximately 400 mm3 on average for PET imaging. Overall, a good tolerance was observed and no obvious variances in tumor size were noticed with this short treatment plan. There are several opportunities for improving the relatively moderate uptake of [18F]-M-SNAT by tumors (up to ∼2 %ID/g). For example, increasing stability of the tracer by incorporating a more bulky disulfide on the cysteine residue of [18F]-M-SNAT might impede nonspecific hydrolysis in the serum, but likely will not impact MetAP2 hydrolysis kinetics in cancer cells where it is reduced by GSH. A previous study by Zhou and colleagues reported a tripeptide analogue containing a mercaptoacetyl residue (P2) and alanyl (or phenylalanyl) residue (P3) showed excellent efficiency for human MetAP2 [kcat/KM = 2.8 × 105 L/(mol*s); ref. 45]. Replacing the current glycyl residue by alanine may improve the hydrolytic kinetics by MetAP2, and, thus, increase signal retention.

MetAP2 PET imaging has promise to advance current imaging strategies in several ways. First, MetAP2 imaging is more likely to be active in more aggressive cancer, based on patterns of expression and, therefore, could enrich for identification of clinically significant prostate cancers. Second, loss of expression of PSMA in NEPC is often acquired after androgen deprivation therapies (7). As we observed, MetAP2 is highly expressed in model systems of castrate-resistant adenocarcinoma and NEPC, suggesting a unique role of MetAP2 PET imaging for tracking these histologic variants (Fig. 1B and C). Third, noninvasive PET imaging of MetAP2 activity could be invaluable as a companion diagnostic test to predict and monitor responses to MetAP2 inhibitors in the near future. Collectively, these data indicate that MetAP2 may be a valuable addition to the oncologist armamentarium for use as a prognostic indicator of biochemical recurrence and diagnostic imaging agent in prostate cancer.

J. Xie reports a patent for US20200085980A1 pending. Z. Chen reports a patent for caspase-3–triggered molecular self-assembling pet probes and uses thereof pending. J. Rao reports grants from NIH and U.S. Army Medical Research Acquisition Activity through the Congressionally Directed Medical Research Program during the conduct of the study, as well a patent for the imaging technology described in this article and filed by Leland Junior Stanford University pending. No disclosures were reported by the other authors.

Opinions, interpretation, conclusions, and recommendations are those of the authors and not necessarily endorsed by the U.S. Army and the funding agencies.

J. Xie: Conceptualization, software, formal analysis, funding acquisition, investigation, writing–original draft, writing–review and editing. M.A. Rice: Software, validation, investigation, methodology, writing–original draft, writing–review and editing. Z. Chen: Conceptualization, investigation. Y. Cheng: Conceptualization, investigation. E.-C. Hsu: Software, investigation, methodology. M. Chen: Software, investigation, methodology. G. Song: Investigation. L. Cui: Investigation. K. Zhou: Investigation. J.B. Castillo: Software, investigation. C.A. Zhang: Resources, software, validation, investigation. B. Shen: Resources, supervision, validation, investigation. F.T. Chin: Resources, formal analysis, supervision, validation, investigation. C.A. Kunder: Conceptualization, resources, formal analysis, supervision, validation, investigation, writing–review and editing. J.D. Brooks: Conceptualization, resources, supervision, funding acquisition, validation, writing–review and editing. T. Stoyanova: Conceptualization, resources, supervision, validation, investigation, writing–review and editing. J. Rao: Conceptualization, resources, supervision, funding acquisition, validation, writing–review and editing.

This work was supported by NIH grant U54CA199075 (Center for Cancer Nanotechnology Excellence for Translational Diagnostics). J. Xie thanks the Molecular Imaging Program at Stanford for the Molecular Imaging Young Investigator prize. M.A. Rice was supported by the U.S. Army Medical Research Acquisition Activity through the Congressionally Directed Medical Research Program (CDMRP) under award number W81XWH1810141. T. Stoyanova acknowledges the support from NIH/NCI (R37CA240822 and R03CA230819) and CDMRP (award number W81XWH1810323). M. Chen acknowledges the support from the Stanford Cancer Translational Nanotechnology Training Program funded by NCI award T32CA196585. L. Cui was supported by a CDMRP Breast Cancer Research Program Breakthrough postdoctoral fellowship award (award number W81XWH1810591). J.D. Brooks was supported by the NIH/NCI (U01CA196387). The SR-SIM was funded by an NIH High End Instrumentation grant (award number 1S10OD01227601) from the National Center for Research Resources. The authors thank J. Mulholland at Stanford Cell Sciences Imaging Facility for help on SR-SIM, Stanford Shared FACS Facility for instrumentation and assistance with flow cytometry, Functional Genomics Facility at Stanford for access to LI-COR Odyssey for Western blots, and Stanford Center for Innovation in In Vivo Imaging (SCI3) for micro-PET/CT imaging, and continued support of the Canary Foundation.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
World Health Organization
.
WHO report on cancer: setting priorities, investing wisely and providing care for all
.
Geneva (Switzerland)
:
World Health Organization
; 
2020
.
2.
Brooks
JD
. 
Managing localized prostate cancer in the era of prostate-specific antigen screening
.
Cancer
2013
;
119
:
3906
9
.
3.
Descotes
JL
. 
Diagnosis of prostate cancer
.
Asian J Urol
2019
;
6
:
129
36
.
4.
Li
R
,
Ravizzini
GC
,
Gorin
MA
,
Maurer
T
,
Eiber
M
,
Cooperberg
MR
, et al
The use of PET/CT in prostate cancer
.
Prostate Cancer Prostatic Dis
2018
;
21
:
4
21
.
5.
Evans
JD
,
Jethwa
KR
,
Ost
P
,
Williams
S
,
Kwon
ED
,
Lowe
VJ
, et al
Prostate cancer-specific PET radiotracers: a review on the clinical utility in recurrent disease
.
Pract Radiat Oncol
2018
;
8
:
28
39
.
6.
Lipianskaya
J
,
Cohen
A
,
Chen
CJ
,
Hsia
E
,
Squires
J
,
Li
Z
, et al
Androgen-deprivation therapy-induced aggressive prostate cancer with neuroendocrine differentiation
.
Asian J Androl
2014
;
16
:
541
4
.
7.
Sheikhbahaei
S
,
Werner
RA
,
Solnes
LB
,
Pienta
KJ
,
Pomper
MG
,
Gorin
MA
, et al
Prostate-specific membrane antigen (PSMA)-targeted pet imaging of prostate cancer: an update on important pitfalls
.
Semin Nucl Med
2019
;
49
:
255
70
.
8.
Li
X
,
Chang
YH
. 
Amino-terminal protein processing in Saccharomyces cerevisiae is an essential function that requires two distinct methionine aminopeptidases
.
Proc Natl Acad Sci U S A
1995
;
92
:
12357
61
.
9.
Tucker
LA
,
Zhang
Q
,
Sheppard
GS
,
Lou
P
,
Jiang
F
,
McKeegan
E
, et al
Ectopic expression of methionine aminopeptidase-2 causes cell transformation and stimulates proliferation
.
Oncogene
2008
;
27
:
3967
76
.
10.
Sin
N
,
Meng
L
,
Wang
MQ
,
Wen
JJ
,
Bornmann
WG
,
Crews
CM
. 
The anti-angiogenic agent fumagillin covalently binds and inhibits the methionine aminopeptidase, MetAP-2
.
Proc Natl Acad Sci U S A
1997
;
94
:
6099
103
.
11.
Ingber
D
,
Fujita
T
,
Kishimoto
S
,
Sudo
K
,
Kanamaru
T
,
Brem
H
, et al
Synthetic analogues of fumagillin that inhibit angiogenesis and suppress tumour growth
.
Nature
1990
;
348
:
555
7
.
12.
Benny
O
,
Fainaru
O
,
Adini
A
,
Cassiola
F
,
Bazinet
L
,
Adini
I
, et al
An orally delivered small-molecule formulation with antiangiogenic and anticancer activity
.
Nat Biotechnol
2008
;
26
:
799
807
.
13.
Heinrich
T
,
Seenisamy
J
,
Blume
B
,
Bomke
J
,
Calderini
M
,
Eckert
U
, et al
Discovery and structure-based optimization of next-generation reversible methionine aminopeptidase-2 (MetAP-2) inhibitors
.
J Med Chem
2019
;
62
:
5025
39
.
14.
Weidner
N
,
Carroll
PR
,
Flax
J
,
Blumenfeld
W
,
Folkman
J
. 
Tumor angiogenesis correlates with metastasis in invasive prostate carcinoma
.
Am J Clin Pathol
1993
;
143
:
401
9
.
15.
Yamaoka
M
,
Yamamoto
T
,
Ikeyama
S
,
Sudo
K
,
Fujita
T
. 
Angiogenesis inhibitor TNP-470 (AGM-1470) potently inhibits the tumor growth of hormone-independent human breast and prostate carcinoma cell lines
.
Cancer Res
1993
;
53
:
5233
6
.
16.
Shuhendler
AJ
,
Ye
D
,
Brewer
KD
,
Bazalova-Carter
M
,
Lee
KH
,
Kempen
P
, et al
Molecular magnetic resonance imaging of tumor response to therapy
.
Sci Rep
2015
;
5
:
14759
.
17.
Ye
D
,
Shuhendler
AJ
,
Cui
L
,
Tong
L
,
Tee
SS
,
Tikhomirov
G
, et al
Bioorthogonal cyclization-mediated in situ self-assembly of small-molecule probes for imaging caspase activity in vivo
.
Nat Chem
2014
;
6
:
519
26
.
18.
Shen
B
,
Jeon
J
,
Palner
M
,
Ye
D
,
Shuhendler
A
,
Chin
FT
, et al
Positron emission tomography imaging of drug-induced tumor apoptosis with a caspase-triggered nanoaggregation probe
.
Angew Chem Int Ed Engl
2013
;
52
:
10511
4
.
19.
Dragulescu-Andrasi
A
,
Kothapalli
SR
,
Tikhomirov
GA
,
Rao
J
,
Gambhir
SS
. 
Activatable oligomerizable imaging agents for photoacoustic imaging of furin-like activity in living subjects
.
J Am Chem Soc
2013
;
135
:
11015
22
.
20.
Oba
Y
,
Yoshida
N
,
Kanie
S
,
Ojika
M
,
Inouye
S
. 
Biosynthesis of firefly luciferin in adult lantern: decarboxylation of L-cysteine is a key step for benzothiazole ring formation in firefly luciferin synthesis
.
PLoS One
2013
;
8
:
e84023
.
21.
Liu
Y
,
Miao
Q
,
Zou
P
,
Liu
L
,
Wang
X
,
An
L
, et al
Enzyme-controlled intracellular self-assembly of (18)F nanoparticles for enhanced MicroPET imaging of tumor
.
Theranostics
2015
;
5
:
1058
67
.
22.
Qiu
L
,
Li
X
,
Lv
G
,
Seimbille
Y
,
Li
K
,
Peng
Y
, et al
Radiofluorinated smart probes for noninvasive PET imaging of legumain activity in living subjects
.
Anal Chem
2020
;
92
:
11627
34
.
23.
Wang
S
,
Gao
D
,
Li
K
,
Ye
S
,
Liu
Q
,
Peng
Y
, et al
Radiopharmacological evaluation of a caspase-3 responsive probe with optimized pharmacokinetics for PET imaging of tumor apoptosis
.
Org Biomol Chem
2020
;
18
:
3512
21
.
24.
Chen
Z
,
Chen
M
,
Cheng
Y
,
Kowada
T
,
Xie
J
,
Zheng
X
, et al
Exploring the condensation reaction between aromatic nitriles and amino thiols to optimize in situ nanoparticle formation for the imaging of proteases and glycosidases in cells
.
Angew Chem Int Ed Engl
2020
;
59
:
3272
9
.
25.
Hsu
EC
,
Rice
MA
,
Bermudez
A
,
Marques
FJG
,
Aslan
M
,
Liu
S
, et al
Trop2 is a driver of metastatic prostate cancer with neuroendocrine phenotype via PARP1
.
Proc Natl Acad Sci U S A
2020
;
117
:
2032
42
.
26.
Lapointe
J
,
Li
C
,
Higgins
JP
,
van de Rijn
M
,
Bair
E
,
Montgomery
K
, et al
Gene expression profiling identifies clinically relevant subtypes of prostate cancer
.
Proc Natl Acad Sci U S A
2004
;
101
:
811
6
.
27.
Xie
J
,
Huang
X
,
Park
MS
,
Pham
HM
,
Chan
WK
. 
Differential suppression of the aryl hydrocarbon receptor nuclear translocator-dependent function by an aryl hydrocarbon receptor PAS-A-derived inhibitory molecule
.
Biochem Pharmacol
2014
;
88
:
253
65
.
28.
Goldstein
AS
,
Drake
JM
,
Burnes
DL
,
Finley
DS
,
Zhang
H
,
Reiter
RE
, et al
Purification and direct transformation of epithelial progenitor cells from primary human prostate
.
Nat Protoc
2011
;
6
:
656
67
.
29.
Sahoo
D
,
Wei
W
,
Auman
H
,
Hurtado-Coll
A
,
Carroll
PR
,
Fazli
L
, et al
Boolean analysis identifies CD38 as a biomarker of aggressive localized prostate cancer
.
Oncotarget
2018
;
9
:
6550
61
.
30.
Xiao
Q
,
Zhang
F
,
Nacev
BA
,
Liu
JO
,
Pei
D
. 
Protein N-terminal processing: substrate specificity of Escherichia coli and human methionine aminopeptidases
.
Biochemistry
2010
;
49
:
5588
99
.
31.
Zhang
Y
,
Griffith
EC
,
Sage
J
,
Jacks
T
,
Liu
JO
. 
Cell cycle inhibition by the anti-angiogenic agent TNP-470 is mediated by p53 and p21WAF1/CIP1
.
Proc Natl Acad Sci U S A
2000
;
97
:
6427
32
.
32.
Yeh
JR
,
Mohan
R
,
Crews
CM
. 
The antiangiogenic agent TNP-470 requires p53 and p21CIP/WAF for endothelial cell growth arrest
.
Proc Natl Acad Sci U S A
2000
;
97
:
12782
7
.
33.
Chappell
WH
,
Lehmann
BD
,
Terrian
DM
,
Abrams
SL
,
Steelman
LS
,
McCubrey
JA
. 
p53 expression controls prostate cancer sensitivity to chemotherapy and the MDM2 inhibitor Nutlin-3
.
Cell Cycle
2012
;
11
:
4579
88
.
34.
Liu
S
,
Widom
J
,
Kemp
CW
,
Crews
CM
,
Clardy
J
. 
Structure of human methionine aminopeptidase-2 complexed with fumagillin
.
Science
1998
;
282
:
1324
7
.
35.
Li
F
,
Ulrich
ML
,
Shih
VF
,
Cochran
JH
,
Hunter
JH
,
Westendorf
L
, et al
Mouse strains influence clearance and efficacy of antibody and antibody-drug conjugate via Fc-FcgammaR interaction
.
Mol Cancer Ther
2019
;
18
:
780
7
.
36.
Schuster
DM
,
Nanni
C
,
Fanti
S
. 
PET tracers beyond FDG in prostate cancer
.
Semin Nucl Med
2016
;
46
:
507
21
.
37.
Zimmerman
ME
,
Meyer
AR
,
Rowe
SP
,
Gorin
MA
. 
Imaging of prostate cancer with positron emission tomography
.
Clin Adv Hematol Oncol
2019
;
17
:
455
63
.
38.
Ferraro
DA
,
Rupp
NJ
,
Donati
OF
,
Messerli
M
,
Eberli
D
,
Burger
IA
. 
68Ga-PSMA-11 PET/MR can be false positive in normal prostatic tissue
.
Clin Nucl Med
2019
;
44
:
e291
e3
.
39.
Queisser
A
,
Hagedorn
SA
,
Braun
M
,
Vogel
W
,
Duensing
S
,
Perner
S
. 
Comparison of different prostatic markers in lymph node and distant metastases of prostate cancer
.
Mod Pathol
2015
;
28
:
138
45
.
40.
Gasinska
A
,
Jaszczynski
J
,
Rychlik
U
,
Luczynska
E
,
Pogodzinski
M
,
Palaczynski
M
. 
Prognostic significance of serum PSA level and telomerase, VEGF and GLUT-1 protein expression for the biochemical recurrence in prostate cancer patients after radical prostatectomy
.
Pathol Oncol Res
2019
;
26
:
1049
56
.
41.
Minner
S
,
Wittmer
C
,
Graefen
M
,
Salomon
G
,
Steuber
T
,
Haese
A
, et al
High level PSMA expression is associated with early PSA recurrence in surgically treated prostate cancer
.
Prostate
2011
;
71
:
281
8
.
42.
Bostwick
DG
,
Pacelli
A
,
Blute
M
,
Roche
P
,
Murphy
GP
. 
Prostate specific membrane antigen expression in prostatic intraepithelial neoplasia and adenocarcinoma: a study of 184 cases
.
Cancer
1998
;
82
:
2256
61
.
43.
Kruger
EA
,
Figg
WD
. 
TNP-470: an angiogenesis inhibitor in clinical development for cancer
.
Expert Opin Investig Drugs
2000
;
9
:
1383
96
.
44.
Logothetis
CJ
,
Wu
KK
,
Finn
LD
,
Daliani
D
,
Figg
W
,
Ghaddar
H
, et al
Phase I trial of the angiogenesis inhibitor TNP-470 for progressive androgen-independent prostate cancer
.
Clin Cancer Res
2001
;
7
:
1198
203
.
45.
Zhou
Y
,
Guo
XC
,
Yi
T
,
Yoshimoto
T
,
Pei
D
. 
Two continuous spectrophotometric assays for methionine aminopeptidase
.
Anal Biochem
2000
;
280
:
159
65
.