Purpose:

Human B7-H3 (hB7-H3) is a promising molecular imaging target differentially expressed on the neovasculature of breast cancer and has been validated for preclinical ultrasound (US) imaging with anti–B7-H3-antibody-functionalized microbubbles (MB). However, smaller ligands such as affibodies (ABY) are more suitable for the design of clinical-grade targeted MB.

Experimental Design:

Binding of ABYB7-H3 was confirmed with soluble and cell-surface B7-H3 by flow cytometry. MB were functionalized with ABYB7-H3 or anti–B7-H3-antibody (AbB7-H3). Control and targeted MB were tested for binding to hB7-H3–expressing cells (MS1hB7-H3) under shear stress conditions. US imaging was performed with MBABY-B7-H3 in an orthotopic mouse model of human MDA-MB-231 coimplanted with MS1hB7-H3 or control MS1WT cells and a transgenic mouse model of breast cancer development.

Results:

ABYB7-H3 specifically binds to MS1hB7-H3 and murine-B7-H3–expressing monocytes. MBABY-B7-H3 (8.5 ± 1.4 MB/cell) and MBAb-B7-H3 (9.8 ± 1.3 MB/cell) showed significantly higher (P < 0.0001) binding to the MS1hB7-H3 cells compared with control MBNon-targeted (0.5 ± 0.1 MB/cell) under shear stress conditions. In vivo, MBABY-B7-H3 produced significantly higher (P < 0.04) imaging signal in orthotopic tumors coengrafted with MS1hB7-H3 (8.4 ± 3.3 a.u.) compared with tumors with MS1WT cells (1.4 ± 1.0 a.u.). In the transgenic mouse tumors, MBABY-B7-H3 (9.6 ± 2.0 a.u.) produced higher (P < 0.0002) imaging signal compared with MBNon-targeted (1.3 ± 0.3 a.u.), whereas MBABY-B7-H3 signal in normal mammary glands and tumors with B7-H3 blocking significantly reduced (P < 0.02) imaging signal.

Conclusions:

MBABY-B7-H3 enhances B7-H3 molecular signal in breast tumors, improving cancer detection, while offering the advantages of a small size ligand and easier production for clinical imaging.

Translational Relevance

Breast cancer is the second leading cause of cancer deaths in women in the United States, and its early detection is key to improving survival. Although mammography is currently the modality of choice for breast cancer screening, its diagnostic accuracy is limited in women with dense breast tissue. Ultrasound (US) is often performed as a second-line test for dense breast tissue; however, due to its low specificity, US results in many false-positive findings, leading to unnecessary biopsies. The ability of clinically translatable US molecular imaging to differentiate benign and malignant lesions in dense breast tissue is critically important, as it could improve the detection of clinically relevant disease while reducing overall costs, unnecessary biopsies, and patient anxiety associated with false-positive recalls. Toward this aim, we demonstrate the efficacy of affibody-based US molecular imaging of vascular B7-H3 expression in breast cancer animal models.

Breast cancer is the second leading cause of cancer-related deaths and the most common site for cancer development at 30% of all new cancer cases in women in the United States, with an estimated 41,760 deaths and 268,600 new cases diagnosed in 2019 (1). This incidence is expected to grow by more than 50% by 2030 (2). Detection during early, localized stages of the disease significantly improves survival with 5-year survival rates of 99% compared with 27% for highly advanced stages (3).

Mammography is the first-line method in breast cancer screening programs. Clinically detected lesions have a median size of 2.6 cm, whereas those found with mammography screening have a median size of 1.5 cm (4), with digital mammogram analyses further increasing screening sensitivity (5). However, mammograms often result in overdiagnosis and unnecessary biopsies with one-half of the women experiencing false positives during the course of multiple screenings (6). Increased frequency of mammography exams along with factors such as age and breast density lowers the overall specificity of screening and results in more false positives. Women with extremely or heterogeneously dense breasts have a 4- to 6-fold greater chance of developing breast cancer compared with women with fatty breasts (7). Furthermore, women with dense breasts frequently have worse prognosis due to late-stage disease detection (8, 9). Mammographically dense tissues decrease detection of malignant lesions because dense tissues appear opaque, which can obscure or mimic malignant lesions and to some extent calcifications, and mislead even experienced radiologists.

Currently, alternative imaging tools are applied whenever mammography alone is insufficient for radiological detection of breast lesions. MRI is used to screen high-risk candidates with familial history of breast cancer, but is expensive, not readily available, and is not suitable for everyone. At present, supplemental screening by ultrasound (US) is recommended to assess suspicious lesions observed in mammograms and is available for high-risk patients with contraindications to MRI (10). US is widely available, portable, noninvasive, cost-effective, and free of ionizing radiation. In a report, supplemental screening with US detected 4.4 additional cancers per 1,000 exams in women with dense breasts and negative mammography, but it also increased false positives (11). Although US is helpful in detecting breast lesions, it is associated with low positive predictive value (5.6%–8.6%; refs. 12, 13) and a low sensitivity (as low as 17%; ref. 14). As a supplemental tool, molecular imaging with US contrast agents would enable highly accurate and sensitive noninvasive methods of differentiating cancer from benign lesions in women with dense breasts.

Contrast-mode US (also called contrast-enhanced US) using microbubbles (MB) as blood-pool agents is often applied to assess abnormal tissue perfusion patterns in tumors due to altered angiogenesis (15, 16). For example, Bar-Zion and colleagues used contrast-enhanced US imaging to monitor tumor perfusion changes after treatment by a vascular disrupting agent in a breast cancer mouse model (16). In addition, molecularly targeted contrast MB that bind to proteins expressed on the tumor neovasculature is an emerging imaging approach with large potential for improving diagnostic accuracy and characterization of focal breast lesions (17, 18).

MB are gas-filled lipid- or protein-stabilized particles and enhance US imaging contrast due to differential response to changes in acoustic pressure. Gas-filled MB that are a few micrometers in size (1–4 μm) remain within the blood vessel lumen, which renders molecularly targeted MB uniquely suitable for US imaging of molecular markers expressed on the neovasculature (18–21). It is of high importance to identify molecular markers that are differentially expressed on tumor-associated neovasculature compared with that of normal tissue or benign breast lesions. Although several cancer-specific vascular markers have been used in contrast-enhanced US imaging of tumors (18, 19), our group and others have extensively explored B7-H3 (CD276) as a promising breast cancer US molecular imaging target. B7-H3, a T-cell modulator, has highly specific overexpression on vascular endothelial cells of different subtypes of human breast cancer and high-grade ductal carcinoma in situ (DCIS) compared with normal breast tissue and benign lesions (22, 23). B7-H3 is a cell-surface receptor protein and highly correlated with tumor drug resistance, metastasis, and immune-regulation (24–26).

Monoclonal antibodies produced by hybridoma technology have primarily been used as target-binding ligands to functionalize the surface of MB (27), although small protein scaffolds have been used as well for other targets (28, 29). Previously, an anti–B7-H3 antibody-conjugated MB was utilized for preclinical US molecular imaging of mammary tumors (22). In this study, US molecular imaging signal with antibody-conjugated MB was shown to be highly correlated with pathology-based B7-H3 expression in the mammary tumors. Unfortunately, antibody-based ligands can be problematic for clinical translation due to inefficient and random conjugation, costly and time-consuming production, and potential immune response, especially with repeated dosing (30). Smaller protein scaffolds enable efficient, site-specific conjugation and prokaryotic production and can replace antibodies (∼150 kDa) for MB functionalization. Recently, affibodies (ABY) have been shown to be a promising platform for designing binding ligands for molecular imaging (31, 32). The ABY is a 58-amino-acid protein (∼7 kDa) derived from the three alpha helix bundle Z domain of Staphylococcus aureus protein A. Compared with antibodies, ABY exhibit faster renal clearance (33), greater stability at a wide range of physiologic pH and temperature, are cost-effective for larger-scale production, and enable site-specific conjugation (34). ABY molecules are an alternative to full-length antibodies in diagnostic applications and have been validated preclinically and translated to clinical trials (35, 36).

The purpose of this proof-of-principle study is to utilize a recently engineered B7-H3–specific ABY protein (ABYB7-H3) conjugated to contrast MB (37), to validate their use in vitro for endothelial cell binding under flow shear stress conditions, and to assess specificity and sensitivity for US molecular imaging of tumor neovasculature in two different mouse models of breast cancer: human xenografts and transgenic models.

Human B7-H3 expression analysis

Human breast cancer samples for tissue staining were collected as described elsewhere (22) and were obtained with written-informed consent and Institutional Review Board approval. All studies involving human subjects were conducted in accordance with ethical guidelines such as the Declaration of Helsinki. RNA sequencing (RNA-seq) expression data for CD276 (B7-H3) and PECAM1 (CD31) in patient breast-invasive carcinoma were downloaded from The Cancer Genome Atlas (TCGA) database and compared for PAM50 gene-expression–based intrinsic breast cancer subtypes using UCSC Xena Browser (38).

Production of recombinant ABYB7-H3

ABYB7-H3 ligand was recombinantly expressed in Escherichia coli and purified by a HisTrap FF column (GE Healthcare Biosciences). For further details on production of ABY and its B7-H3–binding activity, please refer to Supplementary Methods.

Preparation of targeted MB

Commercially available perfluorocarbon-filled, lipid-shelled, streptavidin-coated contrast MB (VisualSonics) with a mean diameter of 1.5 μm (range, 1–3 μm) were reconstituted in 1 mL sterile saline (0.9% sodium chloride; ref. 39). Three types of MB were prepared: two MB with the ability to target both hB7-H3 (for xenograft tumor models) and mB7-H3 (for transgenic tumor models), made by either conjugating MB with biotinylated ABYB7-H3 (MBABY-B7-H3) or a commercially available biotinylated anti–B7-H3 antibody (eBiosciences, clone M3.2D7, MBAb-B7-H3), and unconjugated MB (MBNon-targeted).

To create each targeted MB, lyophilized streptavidin-coated MB were suspended in 1 mL of sterile saline according to the published recommendations (40). Six microgram of biotinylated ABY or antibody were incubated per 5 × 107 MB in 50 μL for 10 minutes at room temperature and washed before use in in vitro and in vivo experiments (19). To separate unconjugated ligands, the MB solution vial was centrifuged at 300 g in 4°C for 2 minutes followed by removal of clear liquid from the bottom of the vial using a syringe needle without disturbing the top MB layer. MB were then resuspended in PBS and washed similarly two more times (19). For the confirmation of ABY conjugation to MB, biotin-ABY was also conjugated to AF647 NHS ester (ThermoFisher Scientific) dye, purified, and then incubated with streptavidin MB for 10 minutes. Nonbound ligands were removed by centrifugation. ABY-coated MB were then assessed for AF647 signal by flow cytometry (Guava easyCyte, Luminex). The average number of biotinylated-ABY or -antibody, when occupying all the streptavidin molecules in our tested concentrations, are approximately 7,600 per square micrometer MB surface (22).

Flow chamber cell attachment assay

Binding specificity of MBABY-B7-H3 and MBAb-B7-H3 to the target B7-H3 was first assessed in cell culture experiments under flow shear stress conditions simulating flow in blood capillaries by using a flow chamber experimental set-up. Please see Supplementary Methods for more details.

Two mouse models of breast cancer

All experiments were approved by the Institutional Administrative Panel on Laboratory Animal Care. An orthotopic human breast cancer model with tumors from MDA-MB-231 cells stably expressing firefly luciferase (f-luc) reporter gene mixed in matrigel with either MS1WT or MS1hB7-H3 cells expressing renilla luciferase (r-luc) reporter gene was used for establishing tumors in the contralateral flanks of nude (nu/nu) mice (The Jackson Laboratory). Reporter expression details are provided in the Supplementary Document. Note that 1 × 106 MDA-MB-231/f-luc cells were mixed with 5 × 106 MS1/r-luc cells and implanted on the fourth mammary glands: MS1hB7-H3/r-luc cell mixture on the left flank and MS1WT/r-luc cell mixture on the right flank. Imaging was performed in the nude mice after 2 weeks of tumor cell engraftment with a mean size of 4 mm (range, 3–5 mm).

In addition, the transgenic mouse model of breast cancer development FVB/N-Tg(MMTV-PyMT)634Mul was used (The Jackson Laboratory; ref. 22). The mammary tissue of this transgenic mouse model progresses through four distinct histologic stages from normal mammary tissues, hyperplasia, ductal carcinoma in situ, and finally invasive breast carcinoma, which highly recapitulates human breast cancer. For this study, female mice (mean age, 7 weeks; range, 4–10 weeks) with 10 mammary glands with invasive breast carcinoma were imaged with a mean size of 7 mm (range 5–9 mm) by US molecular imaging. The litter mates with normal mammary glands were used as controls.

In vivo imaging

Bioluminescence imaging

Mice coinjected with MDA-MB-231/f-luc cells and MS1/r-luc cells were tested for successful implantation by bioluminescence imaging in live animals after 2 weeks of engraftment. Mice were subjected to i.p. injection of 50 μL D-luciferin (30 mg/mL) substrate followed by anesthesia in 2% isoflurane in room air, and bioluminescence imaging (Lago in vivo Imaging System, Spectral Instruments Imaging) to confirm the growth of MDA-MB-231 tumors. After a 24-hour interval, anesthetized mice were injected with 150 μL of coelenterazine (5 mg/mL) substrate via tail vein injection followed by bioluminescence imaging to confirm the presence of MS1 cells within these tumors.

US molecular imaging

Contrast-enhanced US imaging was performed using a dedicated small-animal high-resolution US imaging system (Vevo 2100; VisualSonics). All mice were kept anesthetized with 2% isoflurane in room air at 2 L/min on a heated stage at 37°C throughout the US molecular imaging sessions. Image acquisition was performed in the transverse plane using a high-resolution transducer (MS250; center frequency, 18 MHz, lateral and axial resolution of 165 μm and 75 μm, respectively; focal length, 8 mm; transmit power, 10%; mechanical index, 0.2; dynamic range, 40 dB). Imaging was performed by fixing the transducer with a clamp and placing the acoustic focus at the center of the mammary tumors in the plane showing the largest transverse cross-section. The same US settings and equipment were used for all imaging experiments.

Recommendations for using targeted MB (MicroMarker, VisualSonics) for small animal imaging were followed as described elsewhere (40). US molecular signal (i.e., US contrast signal from vessel-bound MBABY-B7-H3) was obtained by the destruction–subtraction technique (41), and its signal specificity confirmed by comparisons with a positive control, MB coated with anti–B7-H3 antibody (MBAb-B7-H3), and a negative control, nonfunctionalized MB (MBNon-targeted). Before i.v. injections with a catheter (12 cm PU tubing and 27G butterfly needle; VisualSonics), MB concentration was determined in a particle counter (Z2 Particle Counter, Beckman Coulter). A small sample volume of MB solution was diluted in 10 mL diluent (Isoton II; Beckman Coulter) and concentration determined based on a size range calibration of 1–5 μm. A total of 5 × 107 MB were mixed in approximately 100 μL final volume/injection and bolus administered i.v. through the tail vein followed by another 20 μL saline flush. MB injection was performed manually at a low constant pressure (100 μL in 6 seconds as recommended by the manufacturer) and tissue contrast signal bias between animals minimized by random injection order of different MB constructs. Targeted MB were allowed to attach to B7-H3 on the tumor neovasculature. After 4 minutes, 200 imaging frames were captured and averaged to obtain imaging signal from adherent and freely circulating MB. This was followed by a 1-second continuous high-power destructive pulse of 3.7 MPa (transmit power, 100%; mechanical index, 0.63), which destroyed all MB within the image. After the destruction pulse, another 200 imaging frames were acquired and averaged to capture the signal from the replenishment of freely circulating MB into the tumor area. To determine US molecular imaging signal, the averaged images before and after bursting MB were subtracted as differential targeted enhancement (dTE). dTE image, representing B7-H3 signal, was displayed as a colored overlay on the contrast-mode. The time between injection and imaging with different MB constructs was 20 minutes to allow for freely circulating MB to clear from any previous injections. In addition, destruction–replenishment curves for MB-associated contrast signal before and after the ultrasonic destructive pulse were formed by perfusion modeling in which amplitude-related parameters are expressed relative to plotting as a function of time expressed in seconds.

All US data analyses, including breathing motion correction, defining the region of interest in mammary tissue, dTE, and destruction–replenishment curves were performed in Vevo LAB (Visual Sonics) software (22).

Statistical analysis

The Student t test was used to compare statistical significance between the experimental groups, and all data were expressed as mean ± SEM. Experiments were considered significantly different if the P value was less than 0.05. ROC curve was generated (Prism 8, GraphPad) to determine the diagnostic accuracy of MBABY-B7-H3 in differentiating normal tissue from malignant tumors by US molecular imaging.

B7-H3 expression in breast cancer

To compare gene expression of CD276 (B7-H3) in patient breast cancer samples against a pan-endothelial marker, PECAM1 (CD31), RNA-seq expression data across PAM50 molecular breast cancer subtypes in the TCGA database were analyzed using the UCSC Xena web-based tool (38). Analysis showed higher CD276 expression in all breast cancer subtypes compared with the normal tissue, whereas the PECAM1 expression was higher in normal tissue compared with cancer tissue (Fig. 1A). As B7-H3 is expressed by both vascular endothelial cells and neoplastic cells, we tested its protein expression in patient tissue sections by IHC staining. In the representative samples, B7-H3 staining was observed in structures morphologically resembling blood vessels as well as the cancer cells independent of disease subtypes as seen by its increased staining in luminal, triple-negative, and Her2+ breast cancer samples compared with normal mammary tissue (Fig. 1B; ref. 22). Based on endothelial expression of B7-H3 in clinical tissue samples, experimental plans for clinically translatable targeted MB contrast agent development and its use in contrast-enhanced US imaging of mouse models of breast cancer were formulated as the work flow in Fig. 1C.

Figure 1.

B7-H3 expression and study design of US molecular imaging in breast cancer. A, Box-plots analysis of CD276 (B7-H3) and PECAM1 (CD31) RNA-seq expression data from TCGA breast-invasive carcinoma database showing increased CD276 expression in LumA (n = 434), LumB (n = 194), Basal (n = 142), and Her2+ (n = 67) subtypes compared with normal breast tissue samples (n = 119). In contrast, PECAM1 expression is higher in normal tissue compared with all breast cancer subtypes. B, Representative IHC staining of B7-H3 in normal and breast cancer tissue sections showing its expression in structures morphologically resembling blood vessels (arrows) and cancer cells. Extensive staining of clinical samples was shown previously (22). C, Experimental overview including (I) MB targeting with affibody (ABY), (II) in vitro MB cell attachment assay, (III and IV) in vivo US imaging of mammary tumors in orthotopic and transgenic breast cancer mouse models, and (V) B7-H3 expression on the neovasculature of both mouse models was confirmed by ex vivo immunofluorescence.

Figure 1.

B7-H3 expression and study design of US molecular imaging in breast cancer. A, Box-plots analysis of CD276 (B7-H3) and PECAM1 (CD31) RNA-seq expression data from TCGA breast-invasive carcinoma database showing increased CD276 expression in LumA (n = 434), LumB (n = 194), Basal (n = 142), and Her2+ (n = 67) subtypes compared with normal breast tissue samples (n = 119). In contrast, PECAM1 expression is higher in normal tissue compared with all breast cancer subtypes. B, Representative IHC staining of B7-H3 in normal and breast cancer tissue sections showing its expression in structures morphologically resembling blood vessels (arrows) and cancer cells. Extensive staining of clinical samples was shown previously (22). C, Experimental overview including (I) MB targeting with affibody (ABY), (II) in vitro MB cell attachment assay, (III and IV) in vivo US imaging of mammary tumors in orthotopic and transgenic breast cancer mouse models, and (V) B7-H3 expression on the neovasculature of both mouse models was confirmed by ex vivo immunofluorescence.

Close modal

Production of ABYB7-H3

Development of a B7-H3–binding ligand from an ABY yeast display library is described elsewhere (37). Here, an ABY ligand clone termed ABYB7-H3 with a Kd of 310 ± 100 nmol/L is used. ABYB7-H3 ligand was recombinantly expressed in E. coli, purified, and functionalized with approximately 1:1 molar ratio of biotin (Supplementary Fig. S1). SDS-PAGE analysis of biologically produced ABY showed the predicted size of the ABY (expected: 7,490 Da), which was measured to be 7,546 Da by mass spectrometry (Fig. 2A). No impurities were observed in the purified ABY by SDS gel or mass spectrometry. In order to confirm binding specificity of recombinantly produced ABYB7-H3 to its target, biotinylated-ABY was immobilized to streptavidin-magnetic beads and incubated with 3 μmol/L of recombinant soluble human B7-H3 ectodomain protein. Unconjugated beads (negative control) showed 1% binding compared with ABYB7-H3–conjugated beads, which showed 68% binding to the soluble B7-H3. The binding of ABYB7-H3 is comparable to the beads conjugated to biotinylated anti–B7-H3 antibody positive control, which had 81% binding (Fig. 2B).

Figure 2.

Production and validation of ABYB7-H3. A, Left plot: Coomassie Blue staining of SDS-PAGE showing high purity and expected size (7,490 Da) of finalized ABYB7-H3 produced in E. Coli. Right plot: Mass spectrometric analysis of ABYB7-H3 showing a mass-to-charge peak (m/z = 7,546) corresponding to ABY molecular weight. A doubly charged peak (m/z = 3,763) is also present. Amino acid sequence of ABYB7-H3 is shown on the bottom. B, Flow cytometry of biotin-ABYB7-H3–conjugated streptavidin–microbeads complex showing binding to soluble recombinant B7-H3 (3 μmol/L) as detected by APC-conjugated anti–B7-H3 antibody. Unconjugated beads (negative control) and biotinylated anti–B7-H3 Ab-conjugated beads (positive control) were also tested for binding to soluble B7-H3 antigen.

Figure 2.

Production and validation of ABYB7-H3. A, Left plot: Coomassie Blue staining of SDS-PAGE showing high purity and expected size (7,490 Da) of finalized ABYB7-H3 produced in E. Coli. Right plot: Mass spectrometric analysis of ABYB7-H3 showing a mass-to-charge peak (m/z = 7,546) corresponding to ABY molecular weight. A doubly charged peak (m/z = 3,763) is also present. Amino acid sequence of ABYB7-H3 is shown on the bottom. B, Flow cytometry of biotin-ABYB7-H3–conjugated streptavidin–microbeads complex showing binding to soluble recombinant B7-H3 (3 μmol/L) as detected by APC-conjugated anti–B7-H3 antibody. Unconjugated beads (negative control) and biotinylated anti–B7-H3 Ab-conjugated beads (positive control) were also tested for binding to soluble B7-H3 antigen.

Close modal

ABYB7-H3 binds specifically to B7-H3

ABYB7-H3 binding to cells predominantly expressing 4Ig-B7-H3 (hB7-H3) isoform and mouse cells expressing 2Ig-B7-H3 (mB7-H3) isoform (42) was tested. B7-H3 total protein as well as cell-surface expression was confirmed in MS1 mouse endothelial cells engineered to overexpress hB7-H3 (MS1hB7-H3) by Western blotting and flow cytometry (Fig. 3A and B); only MS1hB7-H3 cells expressed B7-H3 protein, whereas both MS1WT and MS1hB7-H3 cells endogenously expressed CD31 protein, a common vascular endothelial marker, by Western blot. Biotinylated ABYB7-H3 bound specifically to MS1hB7-H3 cells but not to MS1WT cells as detected by streptavidin-AF647 dye in flow cytometry (Fig. 3B). Similarly, ABYB7-H3 binds to the mouse monocyte cell line, RAW264.7, which endogenously expresses moderate levels of cell-surface mB7-H3 (Fig. 3C). ABYB7-H3 dose-dependent increases in binding signal in both MS1hB7-H3 (0–10 μmol/L) and RAW264.7 (0–25 μmol/L) cells were observed, but not in the B7-H3–negative cells, MS1WT (Supplementary Fig. S2).

Figure 3.

ABYB7-H3 binds to B7-H3–positive cells with high specificity in vitro. A, Human B7-H3 protein overexpression in the cell lysate of stably transfected murine endothelial cells, MS1hB7-H3, compared with that of MS1 wild-type (MS1WT) cells by Western blot with fluorescent detection. CD31 is an endothelial cell–specific maker, and actin β is a protein loading control. Numbers below the protein bands indicate radiance (p/sec/cm2/sr) of the expressed proteins. B, Left plot: Histogram showing cell-surface hB7-H3 receptor expression in MS1hB7-H3 cells compared with MS1WT by flow cytometry using anti–hB7-H3-APC antibody (Ab) or no antibody control (Ctl.). Middle and right plots: Biotin-ABYB7-H3 (10 μmol/L) binding specifically to MS1hB7-H3 cells but not the MS1WT cells as detected by streptavidin-AF647 dye. C, Cell-surface staining of endogenous mB7-H3 in the mouse monocyte cell line, RAW264.7, using anti–mB7-H3 antibody compared with IgG control (Ctl.) antibody; histogram representation of biotin-ABYB7-H3 (10 μmol/L) binding to cells. D, Histograms showing anti–hB7-H3 antibody or biotin-ABYB7-H3 (10 μmol/L) staining of THP1 human monocytic cell line chemically induced for B7-H3 expression with PMA (0 and 10 ng/mL). Unstained control (Ctl.) cells used as reference. E, Immunofluorescence staining with ABYB7-H3-AF647 (red) and a nuclear marker, DAPI (blue), in a human breast tumor and normal breast tissue sections. Scale bar, 100 μm. Zoomed images of insets are shown on the right plot.

Figure 3.

ABYB7-H3 binds to B7-H3–positive cells with high specificity in vitro. A, Human B7-H3 protein overexpression in the cell lysate of stably transfected murine endothelial cells, MS1hB7-H3, compared with that of MS1 wild-type (MS1WT) cells by Western blot with fluorescent detection. CD31 is an endothelial cell–specific maker, and actin β is a protein loading control. Numbers below the protein bands indicate radiance (p/sec/cm2/sr) of the expressed proteins. B, Left plot: Histogram showing cell-surface hB7-H3 receptor expression in MS1hB7-H3 cells compared with MS1WT by flow cytometry using anti–hB7-H3-APC antibody (Ab) or no antibody control (Ctl.). Middle and right plots: Biotin-ABYB7-H3 (10 μmol/L) binding specifically to MS1hB7-H3 cells but not the MS1WT cells as detected by streptavidin-AF647 dye. C, Cell-surface staining of endogenous mB7-H3 in the mouse monocyte cell line, RAW264.7, using anti–mB7-H3 antibody compared with IgG control (Ctl.) antibody; histogram representation of biotin-ABYB7-H3 (10 μmol/L) binding to cells. D, Histograms showing anti–hB7-H3 antibody or biotin-ABYB7-H3 (10 μmol/L) staining of THP1 human monocytic cell line chemically induced for B7-H3 expression with PMA (0 and 10 ng/mL). Unstained control (Ctl.) cells used as reference. E, Immunofluorescence staining with ABYB7-H3-AF647 (red) and a nuclear marker, DAPI (blue), in a human breast tumor and normal breast tissue sections. Scale bar, 100 μm. Zoomed images of insets are shown on the right plot.

Close modal

Because ABYB7-H3 binding to endogenously expressed hB7-H3 is critical for its clinical translation, a human monocytic cell line, THP1, was chemically induced for hB7-H3 expression by phorbol myristate acetate (PMA) to test for ABY binding (43). THP1 cells express a basal level of cell-surface hB7-H3, which was further increased upon stimulation by PMA (10 ng/mL) as detected by APC-conjugated anti–B7-H3 antibody using flow cytometry (Fig. 3D). The increase in PMA-induced B7-H3 expression correlated with increased biotinylated ABYB7-H3 binding to THP1 cells as detected by streptavidin AF647 (Fig. 3D).

As posttranslational modifications, including glycosylation of tissue-expressed B7-H3 receptors (44), can limit accessibility of engineered ligands or antibodies to the receptor binding pockets, ABY binding was tested in human breast tissue sections by immunofluorescence staining. ABYB7-H3 conjugated to AF647 dye stained positive for B7-H3 expressed on the cell surface on the tumor tissue section (Fig. 3E). In contrast, ABYB7-H3 did not bind to normal breast tissue sections. In addition, ABYB7-H3 IHC staining of human breast carcinoma tissue sections positively stained tumor epithelia and blood vessels that were consistent with anti–hB7-H3 antibody staining results (Supplementary Fig. S3).

ABYB7-H3–coated MB (MBABY-B7-H3) attach to B7-H3 expressed on endothelial cells

To mimic the in vivo shear stress that would occur on bound MB by blood flow in the capillaries, an in vitro flow chamber cell attachment assay of MB using MS1hB7-H3 cells was performed (Fig. 4A). Biotinylated ABY was conjugated to streptavidin MB (MBABY-B7-H3). A positive control group was included with biotinylated anti–B7-H3 antibody-conjugated MB (MBAb-B7-H3), which were validated in previous studies (22). Unconjugated MB (MBNon-targeted) served as negative control for quantification of nonspecific binding to cells. The number of MBABY-B7-H3 (8.5 ± 1.4 MB/cell) and MBAb-B7-H3 (9.8 ± 1.3 MB/cell) attached to MS1hB7-H3 cells was significantly higher (P < 0.0001) compared with the MBNon-targeted (0.5 ± 0.1 MB/cell; Fig. 4B and C). In contrast, a low number of all MB constructs attached to MS1WT cells (Supplementary Fig. S4). There was no significant difference between MBABY-B7-H3 (1.5 ± 0.3 MB/cell; P < 0.00001 vs. MS1hB7-H3) and MBNon-targeted (1.3 ± 0.3 MB/cell) attachment to the MS1WT cells. Furthermore, to determine the specificity of MBABY-B7-H3 to the cell-surface B7-H3, a receptor-blocking study was performed by incubating MS1hB7-H3 cells with free ABYB7-H3 (5 μg/mL) prior to the cell attachment assay with MBABY-B7-H3. Blocking significantly (P < 0.009) decreased the MBABY-B7-H3 attachment to cells (10.9 ± 2.9 MB/cell) compared with cells without blocking (26.3 ± 4.4 MB/cell; Fig. 4D). Overall, these results suggest that MBABY-B7-H3 is capable of binding specifically to human B7-H3–expressing cells under flow shear stress conditions.

Figure 4.

MBABY-B7-H3 specifically bind to MS1hB7-H3 cells under flow shear stress condition. A, Schematic diagram of MB biofunctionalization with ABYB7-H3 (MBABY-B7-H3) or anti–B7-H3 antibody (MBAb-B7-H3) and flow chamber cell attachment of MS1hB7-H3 cells grown on glass slides. MBNon-targeted served as control. B, Representative photomicrographs (20X magnification) of MS1hB7-H3 cells showing increased attachment (white arrows) of MBAb-B7-H3 and MBABY-B7-H3 compared with MBNon-targeted. Arrows point to the attached MB on cells. Scale bar, 10 μm. C, Bar graph quantification showing significantly higher MBAb-B7-H3 (*, P < 0.0001) and MBABY-B7-H3 (*, P < 0.0001) counts per MS1hB7-H3 cell compared with MBNon-targeted. D, Significant decrease (*, P < 0.009) in MBABY-B7-H3 attachment after B7-H3 receptor blocking with free ABYB7-H3 (5 μg/mL) 1 hour prior to cell attachment assay in comparison with the nonblocking group. Moderate changes in cell density across experimental sets (e.g., C vs. D) affect the magnitude of captured MB, but each experimental set is performed with cells cultured in parallel for rigorous comparison within a set.

Figure 4.

MBABY-B7-H3 specifically bind to MS1hB7-H3 cells under flow shear stress condition. A, Schematic diagram of MB biofunctionalization with ABYB7-H3 (MBABY-B7-H3) or anti–B7-H3 antibody (MBAb-B7-H3) and flow chamber cell attachment of MS1hB7-H3 cells grown on glass slides. MBNon-targeted served as control. B, Representative photomicrographs (20X magnification) of MS1hB7-H3 cells showing increased attachment (white arrows) of MBAb-B7-H3 and MBABY-B7-H3 compared with MBNon-targeted. Arrows point to the attached MB on cells. Scale bar, 10 μm. C, Bar graph quantification showing significantly higher MBAb-B7-H3 (*, P < 0.0001) and MBABY-B7-H3 (*, P < 0.0001) counts per MS1hB7-H3 cell compared with MBNon-targeted. D, Significant decrease (*, P < 0.009) in MBABY-B7-H3 attachment after B7-H3 receptor blocking with free ABYB7-H3 (5 μg/mL) 1 hour prior to cell attachment assay in comparison with the nonblocking group. Moderate changes in cell density across experimental sets (e.g., C vs. D) affect the magnitude of captured MB, but each experimental set is performed with cells cultured in parallel for rigorous comparison within a set.

Close modal

MBABY-B7-H3 enhance B7-H3 US molecular signal in breast tumors

To evaluate binding of MBABY-B7-H3 to both human and murine B7-H3 in vivo, both an orthotopic human breast cancer xenograft expressing hB7-H3 on its vasculature as well as a transgenic mouse model of spontaneous breast cancer expressing mB7-H3 on its neovasculature were used. US molecular imaging of tumors was conducted with all MB constructs in mice. After 2 weeks of tumor and endothelial cell coengraftments in nude mice, in vivo bioluminescence imaging confirmed signal from MDA-MB-231/f-luc cancer cells from both flanks, MS1hB7-H3/r-luc endothelial cells on the left flank, and MS1WT/r-luc endothelial cells on the right flank (Fig. 5A). Luciferase reporter gene introduction into MS1 cells did not alter B7-H3 expression, and bioluminescence activity was confirmed in vitro prior to in vivo use (Supplementary Fig. S5). US molecular imaging of the left tumors, consisting of MS1hB7-H3 cells, produced significantly higher molecular imaging signal with MBABY-B7-H3 (8.4 ± 3.3 a.u.; n = 12 tumors, *, P < 0.04) compared with the MBNon-targeted (1.4 ± 1.0 a.u., n = 14 tumors). The positive control MBAb-B7-H3 also produced high molecular imaging signal (8.2 ± 1.3 a.u.; n = 6, *, P < 0.001 vs. MBNon-targeted), similar to that from MBABY-B7-H3 (Fig. 5B and C). In order to confirm that the B7-H3 molecular imaging signal quantified by the dTE method was from the stationary MB attached to the tumor vasculature, we examined the destruction–replenishment curve for each contrast agent. MB-associated echo power after the ultrasonic bursts showed a decrease only in the MBABY-B7-H3 imaging group of tumors consisting of MS1hB7-H3 cells (Fig. 5C). In the tumors consisting of MS1WT cells on the right flanks, the molecular imaging signals with MBABY-B7-H3 (1.6 ± 0.6 a.u.), MBAb-B7-H3 (3.1 ± 0.9 a.u.), and MBNon-targeted (0.6 ± 0.2 a.u.) were low. Of all the MB constructs tested for molecular imaging signal in the tumors with MS1WT cells, MBAb-B7-H3 produced the highest background imaging signal (*, P < 0.001 vs. MBNon-targeted). MBABY-B7-H3 (**, P < 0.05) and MBAb-B7-H3 (**, P < 0.008), but not MBNon-targeted, produced higher imaging signal in tumors consisting of MS1hB7-H3 cells compared with their respective MB constructs in the tumors consisting of MS1WT cells (Fig. 5B and C). Immunofluorescence costaining with anti-mouse CD31 and anti-human B7-H3 antibodies confirmed the integration of MS1 cells on the mouse blood vessels as well as nonvascular compartments of the engrafted tumors (Fig. 5D). hB7-H3 expression was absent on the tumors engrafted with MS1WT cells.

Figure 5.

MBABY-B7-H3 enhances US molecular imaging signal of human MDA-MB-231 orthotopic breast tumors in mice. A, Representative bioluminescence imaging signal of nude mice coimplanted with human breast cancer cell line, MDA-MB-231/f-luc (firefly luciferase reporter; left image), and MS1/r-luc endothelial cells (renilla luciferase reporter; right image). Left flank tumor consisted of MS1hB7-H3/r-luc cells, and right flank tumor consisted of MS1WT/r-luc cells. B, US molecular imaging signal of tumors with the administration of various MB constructs was quantified and analyzed within the same tumor group (*) or between the two tumor groups (**). Imaging within the tumor group coimplanted with MS1hB7-H3 cells, MBABY-B7-H3 (*, P < 0.04; n = 12), and MBAb-B7-H3 (*, P < 0.001; n = 6) produced significantly higher imaging signal compared with MBNon-targeted (n = 14). Between the two tumor groups coimplanted with either MS1hB7-H3 or MS1WT cells, US molecular imaging signal with MBABY-B7-H3 (**, P < 0.05) and MBAb-B7-H3 (**, P < 0.008) was also significantly higher in tumors coimplanted with MS1hB7-H3 cells compared with tumors coimplanted with MS1WT cells. B7-H3 imaging signal in the tumors with MS1WT cells was low with all the MB constructs with MBAb-B7-H3 producing a significantly higher (*, P < 0.001) signal compared with MBNon-targeted. C, Representative B-mode/dTE US images and corresponding destruction–replenishment curves from tumors with MBABY-B7-H3 and MBNon-targeted. B-mode images were used to draw region of interest (green outline) around the tumor for signal quantification by dTE. Scale bar, 1 mm. MB destruction–replenishment curves show a decrease in echo power from MBABY-B7-H3 only in tumors with MS1hB7-H3 cells after the ultrasonic bursts are applied (gray zone). D, Individual immunofluorescence channels and composite images showing staining of extracted tumor sections confirming integration of MS1hB7-H3 cells in tumor blood vessels by anti-human B7-H3 (red) and anti-mouse CD31 (green) costaining. Tumors coimplanted with MS1hB7-H3 but not the MS1WT cells stain for human B7-H3 in CD31-positive endothelial cells as indicated by white arrows. Scale bar, 100 μm.

Figure 5.

MBABY-B7-H3 enhances US molecular imaging signal of human MDA-MB-231 orthotopic breast tumors in mice. A, Representative bioluminescence imaging signal of nude mice coimplanted with human breast cancer cell line, MDA-MB-231/f-luc (firefly luciferase reporter; left image), and MS1/r-luc endothelial cells (renilla luciferase reporter; right image). Left flank tumor consisted of MS1hB7-H3/r-luc cells, and right flank tumor consisted of MS1WT/r-luc cells. B, US molecular imaging signal of tumors with the administration of various MB constructs was quantified and analyzed within the same tumor group (*) or between the two tumor groups (**). Imaging within the tumor group coimplanted with MS1hB7-H3 cells, MBABY-B7-H3 (*, P < 0.04; n = 12), and MBAb-B7-H3 (*, P < 0.001; n = 6) produced significantly higher imaging signal compared with MBNon-targeted (n = 14). Between the two tumor groups coimplanted with either MS1hB7-H3 or MS1WT cells, US molecular imaging signal with MBABY-B7-H3 (**, P < 0.05) and MBAb-B7-H3 (**, P < 0.008) was also significantly higher in tumors coimplanted with MS1hB7-H3 cells compared with tumors coimplanted with MS1WT cells. B7-H3 imaging signal in the tumors with MS1WT cells was low with all the MB constructs with MBAb-B7-H3 producing a significantly higher (*, P < 0.001) signal compared with MBNon-targeted. C, Representative B-mode/dTE US images and corresponding destruction–replenishment curves from tumors with MBABY-B7-H3 and MBNon-targeted. B-mode images were used to draw region of interest (green outline) around the tumor for signal quantification by dTE. Scale bar, 1 mm. MB destruction–replenishment curves show a decrease in echo power from MBABY-B7-H3 only in tumors with MS1hB7-H3 cells after the ultrasonic bursts are applied (gray zone). D, Individual immunofluorescence channels and composite images showing staining of extracted tumor sections confirming integration of MS1hB7-H3 cells in tumor blood vessels by anti-human B7-H3 (red) and anti-mouse CD31 (green) costaining. Tumors coimplanted with MS1hB7-H3 but not the MS1WT cells stain for human B7-H3 in CD31-positive endothelial cells as indicated by white arrows. Scale bar, 100 μm.

Close modal

After evaluation in the xenograft breast cancer model, we also tested targeted MB in a MMTV-PyMT transgenic mouse model that spontaneously develops mammary tumors, which progress into highly invasive disease over time (22). MBABY-B7-H3 (9.6 ± 2.0 a.u.; n = 47 tumors, *, P < 0.0002) and MBAb-B7-H3 (7.2 ± 1.8 a.u.; n = 45 tumors, *, P < 0.001) produced significantly higher US molecular imaging signal from mammary tumors compared with the MBNon-targeted (1.3 ± 0.3 a.u.; n = 41 tumors; Fig. 6A and C). In vivo blocking of the mB7-H3 receptors in the transgenic mice with free ABY (150 μg) 24 hours prior to tumor imaging resulted in significantly reduced molecular imaging signal (0.4 ± 0.2 a.u.; n = 5, *, P < 0.02) from tumors compared with the same animals imaged before blocking (4.3 ± 1.0 a.u.) with the MBABY-B7-H3 (Fig. 6B and C). Destruction–replenishment curve analysis showed a decrease in MB-associated echo power after the ultrasonic bursts only in the tumor imaging groups with MBABY-B7-H3 and MBAb-B7-H3 but not in the MBNon-targeted or blocking groups (Fig. 6C). Also, as a negative control for nonangiogenic B7-H3–negative blood vessels, normal mammary glands in age-matched control littermates were scanned after intravenous administration of MBABY-B7-H3, MBAb-B7-H3, and MBNon-targeted. US imaging signal in normal mammary gland was low with all MB constructs [MBABY-B7-H3 (2.3 ± 0.5 a.u.; n = 14 glands), MBAb-B7-H3 (2.5 ± 0.8 a.u.; n = 10 glands), and MBNon-targeted (2.6 ± 0.6 a.u.; n = 15 glands); Fig. 6D]. B7-H3 expression on the CD31-positive vasculature of the transgenic mammary tumors was confirmed by immunofluorescence staining with anti–B7-H3 antibody (Supplementary Fig. S6). B7-H3 was not expressed in CD31-positive vasculature in the tissue sections derived from normal mammary glands. ROC analysis was performed to assess the ability of MBABY-B7-H3 to distinguish invasive cancer (n = 47) from the normal breast tissue (n = 44) by US molecular imaging. Overall, MBABY-B7-H3 contrast agent allowed the differentiation of normal tissue from breast cancer with high diagnostic accuracy, with an area under the ROC curve (AUC) of 0.90 [95% confidence interval (CI), 0.82–0.95; Fig. 6E]. These results confirm that ABY is specific to both human and murine B7-H3 expressed on tumor endothelial cells in vivo and that MBABY-B7-H3 provides B7-H3–specific molecular imaging signal of breast tumors compared with the normal mammary tissue.

Figure 6.

MBABY-B7-H3 enhances US molecular imaging signal of mouse mammary tumors in a transgenic breast cancer model. A, Quantification of US molecular imaging signal of mammary tumors with the administration of various MB constructs. B7-H3–targeted imaging with MBABY-B7-H3 (*, P < 0.0002; n = 47) or MBAb-B7-H3 (*, P < 0.001; n = 45) produced significantly higher imaging signal in tumors compared with MBNon-targeted (n = 41). B, Quantification of US molecular imaging signal with MBABY-B7-H3 before and after in vivo B7-H3 receptor blocking overnight with 150 μg free ABYB7-H3 (n = 5). C, Representative B-mode/dTE US images and corresponding destruction–replenishment curves from tumors with administration of various MB constructs including MBABY-B7-H3 imaging postreceptor blocking. B-mode images were used to draw region of interest (green border) around the tumor for signal quantification by dTE. Scale bar, 1 mm. Corresponding MB destruction–replenishment curves show a decrease in echo power from targeted MB in tumors but not in the control groups after the ultrasonic bursts are applied (gray zone). D, Top plot: Quantification of normal mammary gland US imaging signal with MBABY-B7-H3 (n = 14) and MBAb-B7-H3 (n = 10) compared with MBNon-targeted (n = 15); Bottom plot: Representative dTE images of a mammary tumor with MBABY-B7-H3 and normal glands with all MB constructs. Scale bar, 1 mm. E, ROC curve in distinguishing normal tissue (n = 44) from breast cancer (n = 47) with MBABY-B7-H3–based US molecular imaging signal. Area under the ROC curve (AUC) is 0.90 (95% CI, 0.82–0.95).

Figure 6.

MBABY-B7-H3 enhances US molecular imaging signal of mouse mammary tumors in a transgenic breast cancer model. A, Quantification of US molecular imaging signal of mammary tumors with the administration of various MB constructs. B7-H3–targeted imaging with MBABY-B7-H3 (*, P < 0.0002; n = 47) or MBAb-B7-H3 (*, P < 0.001; n = 45) produced significantly higher imaging signal in tumors compared with MBNon-targeted (n = 41). B, Quantification of US molecular imaging signal with MBABY-B7-H3 before and after in vivo B7-H3 receptor blocking overnight with 150 μg free ABYB7-H3 (n = 5). C, Representative B-mode/dTE US images and corresponding destruction–replenishment curves from tumors with administration of various MB constructs including MBABY-B7-H3 imaging postreceptor blocking. B-mode images were used to draw region of interest (green border) around the tumor for signal quantification by dTE. Scale bar, 1 mm. Corresponding MB destruction–replenishment curves show a decrease in echo power from targeted MB in tumors but not in the control groups after the ultrasonic bursts are applied (gray zone). D, Top plot: Quantification of normal mammary gland US imaging signal with MBABY-B7-H3 (n = 14) and MBAb-B7-H3 (n = 10) compared with MBNon-targeted (n = 15); Bottom plot: Representative dTE images of a mammary tumor with MBABY-B7-H3 and normal glands with all MB constructs. Scale bar, 1 mm. E, ROC curve in distinguishing normal tissue (n = 44) from breast cancer (n = 47) with MBABY-B7-H3–based US molecular imaging signal. Area under the ROC curve (AUC) is 0.90 (95% CI, 0.82–0.95).

Close modal

The purpose of this study was to develop a clinically translatable contrast agent for US molecular imaging of breast cancer. An engineered ABY protein ligand for a breast cancer–associated vascular marker, B7-H3, was applied to MB-based US molecular imaging for mammary tumor detection in mouse models. In vitro, ABYB7-H3 binds specifically to its soluble and cell-surface target overexpressed on the endothelial cells and mouse RAW264.7 or human THP1 monocyte cells that are known to endogenously express B7-H3. ABYB7-H3 also binds specifically to B7-H3–positive tumor epithelial and endothelial cells of human breast carcinoma samples. Targeting of MB contrast agent by biofunctionalization with surface conjugation of ABYB7-H3 significantly increased their attachment to endothelial cells under flow shear stress conditions. In vivo, B7-H3 targeting of MB (MBABY-B7-H3) significantly improved the blood vessel–associated US molecular imaging signal in tumors but not in the normal mammary glands. In vivo blocking of B7-H3 receptor significantly reduced the molecular imaging signal achieved with MBABY-B7-H3 further validating its molecular specificity. The sensitivity and specificity of this molecular contrast agent is encouraging for clinical translation for US detection of vascular B7-H3–expressing breast tumors, which could increase the sensitivity of US as a complementary imaging modality for accurate diagnosis of breast cancer in women, including those with mammographically dense breasts.

In recent years, US molecular imaging of tumor angiogenesis and inflammatory processes using MB-based contrast agents has made significant progress in biomarker-based detection of underlying pathology (45). For this study, B7-H3 (CD276) was selected as a biomarker for US molecular imaging as the expression of B7-H3 in tumor endothelial and epithelial compartments of breast cancer has been extensively reported in the literature (22, 23, 26, 46). In our analysis, B7-H3 expression in the blood vessels as well as the cancer cells in human tissue sections of various breast cancer subtypes was upregulated compared with normal breast tissues (22). As B7-H3 expression is a subtype-independent biomarker of breast cancer angiogenesis and is downregulated in benign tumors of the breast tissue, it is an ideal biomarker for the development of a clinically translatable US contrast agent. As an example, a targeted MB contrast agent against another validated vascular marker, VEGFR2/KDR, has been used in antiangiogenic therapy monitoring of colon cancer (47) and recently tested in the first-in-human trials (48) with US signal correlating with histologic VEGFR2 expression of patient tissue sections of breast and ovarian cancer. The current work aims to expand the capacity for molecular profiling of breast cancer.

Previously, an US contrast agent composed of MB functionalized with anti–B7-H3 antibody improved US imaging signal of mammary tumors expressing B7-H3 in the vasculature (22). Instead of an antibody, the use of ABY is economical for large-scale synthetic production, which is a major advantage over the costs associated with development and production of humanized monoclonal antibodies. ABY have high stability, solubility, and ability to withstand high temperatures (90°C) or acidic and alkaline conditions (pH 2.5 and pH 11, respectively; ref. 37). Moreover, the small, single-domain architectures of ABY allow for efficient site-specific chemical conjugations via incorporation of terminal amino acids such as a cysteine to functionalize contrast MB. ABYB7-H3 was developed based on multiple criteria encompassing affinity, specificity, solubility, and thermal stability that are crucial for its optimal function in vivo. ABY scaffolds are of clinical value for biomarker detection and safe for imaging use in humans (32). ABYB7-H3 recognized both exogenously overexpressed human B7-H3 in endothelial cells as well as endogenously expressed mouse/human B7-H3 by monocyte cell lines in our in vitro experiments. ABYB7-H3 recognized B7-H3 expressed in tissue sections of patients with breast cancer. MBABY-B7-H3 bound specifically to the endothelial cells expressing human B7-H3 under flow shear stress conditions in vitro and tumor blood vessels of breast cancer mouse models. A crucial requirement for targeted MB is that they not only bind to the desired target but bind under shear stress from the expected forces on the MB from blood flow in capillaries. We have shown in an in vivo murine model of breast cancer that MBABY-B7-H3 enhanced US molecular imaging signal of MDA-MB-231 orthotopic tumors consisting of MS1hB7-H3 but not those consisting of MS1WT cells. MBABY-B7-H3 showed lower nonspecific imaging signal compared with MBAb-B7-H3 in tumors implanted with MS1WT cells suggesting its ability to reduce background contrast signal in US. Immunofluorescence staining of tumor sections showed hB7-H3 staining in the CD31-positive tumor endothelium indicating incorporation of MS1hB7-H3 cells in angiogenic vessels during the growth of orthotopic tumors. Furthermore, MBABY-B7-H3 significantly increased imaging signal of spontaneously developed mammary tumors in transgenic mice but not the normal glands of control mice. Immunostaining of B7-H3 showed expression in CD31-positive blood vessels of mammary tumor tissue sections, while staining was negative in normal glandular tissue. Anecdotal evidence suggested that the in vivo B7-H3 receptor blocking with excess ABYB7-H3 prior to imaging with MBABY-B7-H3 significantly reduced US molecular imaging signal from tumors. These results indicate that MBABY-B7-H3 not only differentiates a malignant tumor within the normal mammary tissue but also generates a highly specific molecular B7-H3 signal in tumors. Due to its affinity for both mouse and human isoforms of B7-H3, ABYB7-H3 allows for simultaneous optimization of MB contrast development for human use and testing in translational mouse models of breast cancer. However, additional imaging studies are necessary in a larger number of animals expressing human B7-H3 in the tumor-associated blood vessels to determine sensitivity and specificity of MBABY-B7-H3.

Although a clinical grade MBABY-B7-H3 contrast agent may be suitable for breast cancer screening, it will be an ideal development for ultrasonic detection of mammographically occult malignancy in women with dense breast tissue. Full clinical translation of MBABY-B7-H3 for breast cancer detection and screening needs additional work in many other fronts, such as safety testing of targeted MB use in patients, fine tuning the acoustic parameters for the MB constructs, and image processing or pulse sequencing for use in newer clinical US systems, such as the automated breast volume scanners that are capable of producing 3D images and lowering operator dependency or software beamforming systems that are capable of integrating new and more sensitive molecular imaging techniques. To date, the use of targeted and nontargeted contrast MB has shown a very low number of adverse events in humans and had no nephrotoxic effects, which also means a patient is not required to perform renal function tests prior to MB administration as required before CT and MRI imaging methods (45, 48). In addition, MB show a low systemic toxicity profile in human subjects based on recent clinical trials on inoperable pancreatic cancer patients (49). In the first-in-human trial with MB targeted to VEGFR2 (BR55; ClinicalTrials.gov identifiers: NCT01253213, NCT02142608, EudraCT2012-000699-40), 12 of 45 patients reported 15 adverse events that were mild in intensity with no serious complications (48).

Limitations of the current contrast agent and imaging technique need to be addressed for full clinical translation. In the current study, MB were conjugated to ABYB7-H3 with biotin–streptavidin chemistry. This conjugation chemistry is immunogenic in humans, and streptavidin can also bind nonspecifically to biotin present in the human body. Less immunogenic chemical approaches are available for conjugating ABYB7-H3 to the MB such as cysteine–maleimide conjugation used in the FDA-approved antibody–drug conjugates (50) and synthesis of MB from preformed lipid–ligand conjugates increasing their shelf life. Also, although this study was conducted with an adequate affinity ABY, a higher affinity ABY (Kd = 0.9 ± 0.6 nmol/L) was developed (37), which can further increase the US molecular signal sensitivity and specificity. In preclinical US molecular imaging, high-frequency US is used to delineate targeted imaging signal, which cannot be integrated in low-frequency US systems used in clinic. Preclinical US systems are also poor in distinguishing background tissue signal from MB signal due to high echogenicity of tissue in a heterogeneous tumor microenvironment. Clinical-frequency, real-time molecular imaging methods that do not require the destruction–subtraction technique used here and techniques to improve the sensitivity of bound MB signals to background tissue signal and nonbound MB signals are necessary for full clinical translation.

Our work is one step closer to achieving clinically translatable targeted MB for accurate breast cancer detection by US molecular imaging. The use of B7-H3–targeted US imaging can be expanded to monitoring breast cancer antiangiogenic therapy, determining disease progression with noninvasive quantification of vascular B7-H3 expression as a proxy for tumor pathologic state, and creating multitargeted MB with other relevant disease markers for high contrast clinical images. More immediately, MBABY-B7-H3 will aid in the earlier cancer detection of breast cancer as a supporting tool for mammography.

R. Bam and L. Abou-Elkacem are listed as coinventors on a provisional patent application on Affibody Proteins Specific for B7-H3 (CD276) that is owned by Stanford University. No potential conflicts of interest were disclosed by the other authors.

Conception and design: R. Bam, P.S. Lown, K.E. Wilson, A.M. Lutz, R. Paulmurugan, B.J. Hackel, L. Abou-Elkacem

Development of methodology: R. Bam, A.M. Lutz, R. Paulmurugan, L. Abou-Elkacem

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): R. Bam, K. Sharma, K.E. Wilson, L. Abou-Elkacem

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): R. Bam, P.S. Lown, L.A. Stern, K. Sharma, K.E. Wilson, G.R. Bean, B.J. Hackel, J. Dahl, L. Abou-Elkacem

Writing, review, and/or revision of the manuscript: R. Bam, P.S. Lown, L.A. Stern, K.E. Wilson, G.R. Bean, A.M. Lutz, R. Paulmurugan, B.J. Hackel, J. Dahl, L. Abou-Elkacem

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): R. Bam, P.S. Lown, A.M. Lutz, R. Paulmurugan, L. Abou-Elkacem

Study supervision: R. Bam, R. Paulmurugan, B.J. Hackel, J. Dahl, L. Abou-Elkacem

Other (I am the first author of this work): R. Bam

We acknowledge the guidance and mentorship of the late Dr. Juergen K Willmann. We thank the Preclinical Imaging Core Facility and Proteomics Resource Facility at the Canary Center for Cancer Early Detection, Stanford University. IHC staining of human breast cancer specimens was performed at Human Pathology/Histology Service Center, Stanford University. This study was funded by the NCI under grant number R41-CA213544 and a Stanford Women's Cancer Center Innovation Award from the Stanford Cancer Institute, a NCI-designated Comprehensive Cancer Center.

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.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2019
.
CA Cancer J Clin
2019
;
69
:
7
34
.
2.
Rosenberg
PS
,
Barker
KA
,
Anderson
WF
. 
Estrogen receptor status and the future burden of invasive and in situ breast cancers in the United States
.
J Natl Cancer Inst
2015
;
107
:
pii
:
djv159
.
3.
Howlader
N
,
Noone
AM
,
Krapcho
M
,
Miller
D
,
Brest
A
,
Yu
M
, et al
SEER cancer statistics review, 1975–2016
.
Bethesda, MD
:
National Cancer Institute
; 
2019
.
4.
Berg
WA
,
Bandos
AI
,
Mendelson
EB
,
Lehrer
D
,
Jong
RA
,
Pisano
ED
. 
Ultrasound as the primary screening test for breast cancer: analysis from acrin 6666
.
J Natl Cancer Inst
2016
;
108
:
pii
:
djv367
.
5.
Plevritis
SK
,
Munoz
D
,
Kurian
AW
,
Stout
NK
,
Alagoz
O
,
Near
AM
, et al
Association of screening and treatment with breast cancer mortality by molecular subtype in US Women, 2000–2012
.
JAMA
2018
;
319
:
154
64
.
6.
Elmore
JG
,
Barton
MB
,
Moceri
VM
,
Polk
S
,
Arena
PJ
,
Fletcher
SW
. 
Ten-year risk of false positive screening mammograms and clinical breast examinations
.
N Engl J Med
1998
;
338
:
1089
96
.
7.
Vourtsis
A
,
Berg
WA
. 
Breast density implications and supplemental screening
.
Eur Radiol
2019
;
29
:
1762
77
.
8.
Checka
CM
,
Chun
JE
,
Schnabel
FR
,
Lee
J
,
Toth
H
. 
The relationship of mammographic density and age: implications for breast cancer screening
.
Am J Roentgenol
2012
;
198
:
292
5
.
9.
Brentnall
AR
,
Cuzick
J
,
Buist
DSM
,
Bowles
A
. 
Long-term accuracy of breast cancer risk assessment combining classic risk factors and breast density
.
JAMA Oncol
2018
;
4
:
e180174
.
10.
Schneble
EJ
,
Graham
LJ
,
Shupe
MP
,
Flynt
FL
,
Banks
KP
,
Aaron
D
, et al
Current approaches and challenges in early detection of breast cancer recurrence
.
J Cancer
2014
;
5
:
281
90
.
11.
Melnikow
J
,
Fenton
JJ
,
Whitlock
EP
,
Miglioretti
DL
,
Weyrich
MS
,
Thompson
JH
, et al
Supplemental screening for breast cancer in women with dense breasts: a systematic review for the U.S. Preventive services task force
.
Ann Intern Med
2016
;
164
:
268
78
.
12.
Berg
WA
,
Blume
JD
,
Cormack
JB
,
Mendelson
EB
,
Lehrer
D
,
Bohm-Velez
M
, et al
Combined screening with ultrasound and mammography vs. mammography alone in women at elevated risk of breast cancer
.
JAMA
2008
;
299
:
2151
63
.
13.
Hooley
RJ
,
Greenberg
KL
,
Stackhouse
RM
,
Geisel
JL
,
Butler
RS
,
Philpotts
LE
. 
Screening US in patients with mammographically dense breasts: initial experience with Connecticut Public Act 09-41
.
Radiology
2012
;
265
:
59
69
.
14.
Weinstein
SP
,
Localio
AR
,
Conant
EF
,
Rosen
M
,
Thomas
KM
,
Schnall
MD
. 
Multimodality screening of high-risk women: a prospective cohort study
.
J Clin Oncol
2009
;
27
:
6124
8
.
15.
Unnikrishnan
S
,
Klibanov
AL
. 
Microbubbles as ultrasound contrast agents for molecular imaging: preparation and application
.
Am J Roentgenol
2012
;
199
:
292
9
.
16.
Bar-Zion
A
,
Yin
M
,
Adam
D
,
Foster
FS
. 
Functional flow patterns and static blood pooling in tumors revealed by combined contrast-enhanced ultrasound and photoacoustic imaging
.
Cancer Res
2016
;
76
:
4320
31
.
17.
Bzyl
J
,
Lederle
W
,
Rix
A
,
Grouls
C
,
Tardy
I
,
Pochon
S
, et al
Molecular and functional ultrasound imaging in differently aggressive breast cancer xenografts using two novel ultrasound contrast agents (BR55 and BR38)
.
Eur Radiol
2011
;
21
:
1988
95
.
18.
Bachawal
SV
,
Jensen
KC
,
Lutz
AM
,
Gambhir
SS
,
Tranquart
F
,
Tian
L
, et al
Earlier detection of breast cancer with ultrasound molecular imaging in a transgenic mouse model
.
Cancer Res
2013
;
73
:
1689
98
.
19.
Abou-elkacem
L
,
Wang
H
,
Chowdhury
SM
,
Kimura
RH
. 
Thy1-targeted microbubbles for ultrasound molecular imaging of pancreatic ductal adenocarcinoma
.
Clin Cancer Res
2018
;
24
:
1574
86
.
20.
Wang
S
,
Hossack
JA
,
Klibanov
AL
. 
Targeting of microbubbles - contrast agents for ultrasound molecular imaging
.
J Drug Target
2018
;
26
:
420
34
.
21.
Kiessling
F
,
Fokong
S
,
Koczera
P
,
Lederle
W
,
Lammers
T
. 
Ultrasound microbubbles for molecular diagnosis, therapy, and theranostics
.
J Nucl Med
2012
;
53
:
345
8
.
22.
Bachawal
SV
,
Jensen
KC
,
Wilson
KE
,
Tian
L
,
Lutz
AM
,
Willmann
JK
. 
Breast cancer detection by B7-H3-targeted ultrasound molecular imaging
.
Cancer Res
2015
;
75
:
2501
9
.
23.
Seaman
S
,
Zhu
Z
,
Saha
S
,
Zhang
XM
,
Yang
MY
,
Hilton
MB
, et al
Eradication of tumors through simultaneous ablation of CD276/B7-H3-positive tumor cells
.
Cancer Cell
2017
;
31
:
501
15
.
24.
Dong
P
,
Watari
H
. 
B7H3 as a promoter of metastasis and promising therapeutic target
.
Front Oncol
2018
;
8
:
1
8
.
25.
Majzner
RG
,
Theruvath
JL
,
Nellan
A
,
Heitzeneder
S
,
Cui
Y
,
Mount
CW
, et al
CAR T cells targeting B7-H3, a pan-cancer antigen, demonstrate potent preclinical activity against pediatric solid tumors and brain tumors
.
Clin Cancer Res
2019
;
25
:
2560
74
.
26.
Arigami
T
,
Narita
N
,
Mizuno
R
,
Nguyen
L
,
Ye
X
,
Chung
A
, et al
B7–H3 ligand expression by primary breast cancer and associated with regional nodal metastasis
.
Ann Surg
2010
;
252
:
1044
51
.
27.
Klibanov
AL
. 
Microbubble contrast agents: targeted ultrasound imaging and ultrasound-assisted drug-delivery applications
.
Invest Radiol
2006
;
41
:
354
62
.
28.
Willmann
JK
,
Kimura
RH
,
Deshpande
N
,
Lutz
AM
,
Cochran
JR
,
Gambhir
SS
. 
Targeted contrast-enhanced ultrasound imaging of tumor angiogenesis with contrast microbubbles conjugated to integrin-binding knottin peptides
.
J Nucl Med
2010
;
51
:
433
40
.
29.
Abou-Elkacem
L
,
Wilson
KE
,
Johnson
SM
,
Chowdhury
SM
,
Bachawal
S
,
Hackel
BJ
, et al
Ultrasound molecular imaging of the breast cancer neovasculature using engineered fibronectin scaffold ligands: a novel class of targeted contrast ultrasound agent
.
Theranostics
2016
;
6
:
1740
52
.
30.
Descotes
J
. 
Immunotoxicity of monoclonal antibodies
.
MAbs
2009
;
1
:
104
11
.
31.
Löfblom
J
,
Feldwisch
J
,
Tolmachev
V
,
Carlsson
J
,
Ståhl
S
,
Frejd
FY
. 
Affibody molecules: engineered proteins for therapeutic, diagnostic and biotechnological applications
.
FEBS Lett
2010
;
584
:
2670
80
.
32.
Velikyan
I
,
Schweighöfer
P
,
Feldwisch
J
,
Seemann
J
,
Frejd
FY
,
Lindman
H
, et al
Diagnostic HER2-binding radiopharmaceutical, [68 Ga] Ga-ABY-025, for routine clinical use in breast cancer patients
.
Am J Nucl Med Mol Imaging
2019
;
9
:
12
23
.
33.
Schmidt
MM
,
Wittrup
KD
. 
A modeling analysis of the effects of molecular size and binding affinity on tumor targeting
.
Mol Cancer Ther
2009
;
8
:
2861
72
.
34.
Frejd
FY
,
Kim
KT
. 
Affibody molecules as engineered protein drugs
.
Exp Mol Med
2017
;
49
:
e306
.
35.
Lubberink
M
,
Garske-rom
U
,
Carlsson
J
,
Lindman
H
. 
Cancer metastases using the 111 In-ABY-025 affibody molecule
.
J Nucl Med
2014
;
55
:
730
6
.
36.
Krasniqi
A
,
Devoogdt
N
,
Frejd
FY
,
Keyaerts
M
,
Tolmachev
V
,
Cellular
IV
, et al
Same-day imaging using small proteins : clinical experience and translational prospects in oncology
.
J Nucl Med
2018
;
59
:
885
91
.
37.
Stern
LA
,
Lown
P
,
Kobe
AC
,
Abou-Elkacem
L
,
Willmann
JK
,
Hackel
BJ
. 
Cellular-based selections aid yeast-display discovery of genuine cell-binding ligands: targeting oncology vascular biomarker CD276
.
ACS Comb Sci
2018
;
21
:
207
22
.
38.
Goldman
M
,
Craft
B
,
Kamath
A
,
Brooks
A
,
Zhu
J
,
Haussler
D
. 
The UCSC Xena Platform for cancer genomics data visualization and interpretation
.
bioRxiv
2018
;
326470
.
39.
Willmann
JK
,
Paulmurugan
R
,
Chen
K
,
Gheysens
O
,
Rodriguez-Porcel
M
,
Lutz
A
, et al
US imaging of tumor angiogenesis with microbubbles targeted to vascular endothelial growth factor receptor type 2 in mice
.
Radiology
2008
;
246
:
508
18
.
40.
Hyvelin
J-M
,
Tardy
I
,
Arbogast
C
,
Costa
M
,
Emmel
P
,
Helbert
A
, et al
Use of ultrasound contrast agent microbubbles in preclinical research: recommendations for small animal imaging
.
Invest Radiol
2013
;
48
:
570
83
.
41.
Lindner
JR
,
Song
J
,
Christiansen
J
,
Klibanov
AL
,
Xu
F
,
Ley
K
. 
Ultrasound assessment of inflammation and renal tissue injury with microbubbles targeted to P-selectin
.
Circulation
2001
;
104
:
2107
12
.
42.
Sun
M
,
Richards
S
,
Prasad
DV
,
Mai
XM
,
Rudensky
A
,
Dong
C
. 
Characterization of mouse and human B7-H3 genes
.
J Immunol
2002
;
168
:
6294
7
.
43.
Chapoval
AI
,
Ni
J
,
Lau
JS
,
Wilcox
RA
,
Flies
DB
,
Liu
D
, et al
B7-H3: a costimulatory molecule for T cell activation and IFN-γ production
.
Nat Immunol
2001
;
2
:
269
74
.
44.
Turtoi
A
,
Dumont
B
,
Gre
Y
,
Blomme
A
,
Mazzucchelli
G
,
Delvenne
P
, et al
Novel comprehensive approach for accessible biomarker identification and absolute quantification from precious human tissues
.
J Proteome Res
2011
;
10
:
3160
82
.
45.
Deshpande
N
,
Needles
A
,
Willmann
JK
. 
Molecular ultrasound imaging: current status and future directions
.
Clin Radiol
2010
;
65
:
567
81
.
46.
Lim
S
,
Liu
H
,
Da Silva
LM
,
Arora
R
,
Liu
Z
,
Phillips
JB
, et al
Immunoregulatory protein B7-H3 reprograms glucose metabolism in cancer cells by ROS-mediated stabilization of HIF1a
.
Cancer Res
2016
;
76
:
2231
42
.
47.
Pysz
MA
,
Foygel
K
,
Rosenberg
J
,
Gambhir
SS
,
Schneider
M
,
Willmann
JK
. 
Antiangiogenic cancer therapy : monitoring with molecular US and a clinically translatable contrast agent (BR55)
.
Radiology
2010
;
256
:
519
27
.
48.
Willmann
K
,
Bonomo
L
,
Testa
AC
,
Rinaldi
P
,
Rindi
G
,
Valluru
KS
, et al
Ultrasound molecular imaging with BR55 in patients with breast and ovarian lesions : first-in-human results
.
J Clin Oncol
2017
;
35
:
2133
40
.
49.
Dimcevski
G
,
Kotopoulis
S
,
Bjånes
T
,
Hoem
D
,
Schjøtt
J
,
Gjertsen
BT
, et al
A human clinical trial using ultrasound and microbubbles to enhance gemcitabine treatment of inoperable pancreatic cancer
.
J Control Release
2016
;
243
:
172
81
.
50.
Fontaine
SD
,
Reid
R
,
Robinson
L
,
Ashley
GW
,
Santi
DV
. 
Long-term stabilization of maleimide-thiol conjugates
.
Bioconjug Chem
2015
;
26
:
145
52
.