Purpose:

Targeted therapies for cancer have accelerated the need for functional imaging strategies that inform therapeutic efficacy. This study assesses the potential of functional genetic screening to integrate therapeutic target identification with imaging probe selection through a proof-of-principle characterization of a therapy–probe pair using dynamic nuclear polarization (DNP)-enhanced magnetic resonance spectroscopic imaging (MRSI).

Experimental Design:

CRISPR-negative selection screens from a public dataset were used to identify the relative dependence of 625 cancer cell lines on 18,333 genes. Follow-up screening was performed in hepatocellular carcinoma with a focused CRISPR library targeting imaging-related genes. Hyperpolarized [1-13C]-pyruvate was injected before and after lactate dehydrogenase inhibitor (LDHi) administration in male Wistar rats with autochthonous hepatocellular carcinoma. MRSI evaluated intratumoral pyruvate metabolism, while T2-weighted segmentations quantified tumor growth.

Results:

Genetic screening data identified differential metabolic vulnerabilities in 17 unique cancer types that could be imaged with existing probes. Among these, hepatocellular carcinoma required lactate dehydrogenase (LDH) for growth more than the 29 other cancer types in this database. LDH inhibition led to a decrease in lactate generation (P < 0.001) and precipitated dose-dependent growth inhibition (P < 0.01 overall, P < 0.05 for dose dependence). Intratumoral alanine production after inhibition predicted the degree of growth reduction (P < 0.001).

Conclusions:

These findings demonstrate that DNP-MRSI of LDH activity using hyperpolarized [1-13C]-pyruvate is a theranostic strategy for hepatocellular carcinoma, enabling quantification of intratumoral LDHi pharmacodynamics and therapeutic efficacy prediction. This work lays the foundation for a novel theranostic platform wherein functional genetic screening informs imaging probe selection to quantify therapeutic efficacy on a cancer-by-cancer basis.

Translational Relevance

The increasingly central role played by targeted therapies for cancer underscores a growing need for theranostic imaging strategies to inform treatment planning. The presented data demonstrate that functional genetic screening with CRISPR-Cas9 can facilitate the selection of targeted imaging probes and therapies. As a proof of principle, this work demonstrates that hepatocellular carcinoma depends upon lactate dehydrogenase (LDH) for growth more than other cancers. Dynamic nuclear polarization–enhanced magnetic resonance spectroscopic imaging (DNP-MRSI) of LDH activity enabled the direct assessment of intratumoral pyruvate metabolism and predicted response to LDH inhibition in an autochthonous rodent model of hepatocellular carcinoma. These data suggest a role for LDH targeting in human hepatocellular carcinoma, as well as DNP-MRSI for the prediction of clinical response. More broadly, this work establishes a high-throughput theranostic paradigm that integrates probe and therapy selection to enable early measurement of on-target efficacy and response prediction on a cancer-by-cancer basis.

Targeted therapies developed through mechanism-based drug discovery are revolutionizing cancer care, enabling improved patient outcomes and motivating patient-specific treatment strategies that form the basis for precision medicine (1). Despite this promise, early experiences with these approaches support the need for integrated response assessment to further inform therapy (2). Current theranostic molecular imaging paradigms, including 18Fluorodeoxyglucose PET, enable the detection of a therapeutic target within the tumor, but fail to inform on the on-target efficacy of the therapeutic. This deficiency has limited the translation of molecular imaging approaches despite the development of a myriad of molecular probes across a spectrum of imaging modalities. Indeed, despite proven survival benefits for targeted therapies, current clinical imaging fails to quantify therapeutic response effectively (3). Integrated theranostic strategies that enable the selection of molecular imaging probes specific for targetable vulnerabilities in cancer hold the potential to overcome this fundamental limitation by providing direct and early quantifications of response.

Massively parallelized biotechnologies are transforming the discovery and development of targeted therapeutics. Among these, genetic screens using CRISPR-based genome editing enable the high-throughput and unbiased identification of potentially targetable vulnerabilities (4). In addition to identifying therapeutic dependencies, existing functional annotations may enable the simultaneous identification of functional imaging probes modified by the drug target. Until recently, existing molecular imaging technologies had limited capacity to assess in vivo enzyme function or inform drug pharmacodynamics given an inability to distinguish the parent substrate from its product. The advent of dynamic nuclear polarization–enhanced 13C nuclear magnetic resonance spectroscopic imaging (DNP-MRSI) overcomes this limitation through the ability to measure multiple chemical species simultaneously. DNP is a hyperpolarization technique that increases the fraction of nuclei aligning with an external magnetic field relative to the Boltzmann prediction of thermal equilibrium, producing up to a 105-fold increase in signal-to-noise and enabling the detection of stable 13C-labeled isotopes in real-time (5). The role of enzyme abundance and activity in influencing the conversion of 13C-labeled substrates suggests that cellular enzymatic dependencies may facilitate the selection of hyperpolarized 13C-MRSI probes that specifically measure enzymatic activity (6, 7).

This study hypothesized that CRISPR-based negative selection screening of genes encoding enzymes responsible for hyperpolarized 13C-MRS probe metabolism would simultaneously identify therapeutic targets and molecular imaging probes. This strategy holds the potential to enable a novel and high-throughput theranostic paradigm with the power to provide early measures of on-target efficacy and response prediction.

CRISPR screening

A detailed description of protocols and analyses used for CRISPR screening is provided in the Supplementary Materials and Methods.

Animal experiments

All animal experiments were performed according to an institutionally approved protocol (Institutional Animal Care and Use Committee protocol #: 803952) for the safe and humane treatment of animals.

Autochthonous rat model of hepatocellular carcinoma

Induction of autochthonous hepatocellular carcinomas in male Wistar rats was performed as described previously with diethylnitrosamine (8). One week prior to completion of the diethylnitrosamine exposure, rats underwent MRI screening approximately twice per week with T2-weighted MRI [Field of view: 70 mm × 70 mm, grid size: 256 × 256, slice thickness: 2 mm, Repetition time (TR) minimum: 1.4 s (respiratory gated), Echo time (TE): 59.1 ms, and minimum averages: 4) using an Agilent 4.7T 40 cm horizontal bore MR Spectrometer with a 25 gauss/cm gradient tube interfaced to an Agilent DirectDrive console (9). During all scans, induction and maintenance of anesthesia was achieved using approximately 2% isoflurane in O2, body temperature was maintained at 37°C with a closed loop Heating System (Small Animal Instruments), and respiration rate was monitored. Each animal was positioned within a Polarean proton-tuned (200.1 MHz) birdcage resonator. After individual hepatocellular carcinoma lesions reached approximately 100 mm3 in size, they were used for subsequent study with a lactate dehydrogenase inhibitor (LDHi, 737) graciously provided by Dr. Len Neckers (National Cancer Institute, [email protected]) and the NCI Experimental Therapeutics Program (10, 11). Following inhibitor administration, animals underwent repeated screening with T2-weighted MRI approximately twice per week.

Hyperpolarized [1-13C]-pyruvate and LDHi injections

On the day of therapeutic treatment with a LDHi, each animal was centered within a Polarean proton-tuned (200.1 MHz) birdcage resonator with a 13C-tuned (50.525 MHz) surface coil positioned over the right upper quadrant. T2-weighted imaging was performed to confirm positioning of the surface coil over the tumor of interest using a 13C-urea phantom centered within the surface coil (9). Each animal then received a baseline hyperpolarized [1-13C]-pyruvate injection followed by a LDHi injection within 15 minutes, and then a follow-up hyperpolarized [1-13C]-pyruvate injection within 90 minutes of LDHi administration (n = 9).

The composition and timing of hyperpolarized [1-13C]-pyruvate injections were identical prior to and following LDHi administration for each single animal using a concentration of 80 mmol/L hyperpolarized [1-13C]-pyruvate. Additional details about injection composition and the hyperpolarization procedure are provided in the Supplementary Materials and Methods. DNP-MRSI was performed using a 2D echo-planar spectroscopic imaging (EPSI) sequence with a spectral bandwidth of 1.1 kHz and 128 points, enabling detection of a chemical shift range of 22 ppm (9, 12). The total field of view was 60 mm × 45 mm with 12 phase-encoded steps and 16 frequency-encoded steps, enabling an in-plane resolution of 3.75 mm × 3.75 mm with a slice thickness matching the thickness of the tumor in the coronal plane (∼6–10 mm). Each image was acquired in approximately 1 second with consecutive images repeated approximately every 2 seconds for a minimum of 20 scans per study.

LDHi injections were performed at a concentration of 10 mg/mL and a dose of either 10 (n = 6) or 20 mg/kg (n = 3). All LDHi injections were performed by hand through tail vein injection at a rate of 1 mL/minute and were followed by a 2 mL saline flush.

Image reconstruction and enzymatic activity analysis

Acquired proton images and EPSI spectra were reconstructed in MATLAB 2018B (Mathworks, Inc.). To ensure accurate quantitation of the carbon data, the following corrections were employed: B1 correction, point spread function correction, polarization efficiency normalization, and baseline subtraction consistent with previously described methods (9). Chemical shifts were determined relative to a 13C-urea reference standard (165.5 ppm) present in the 13C surface coil. Intratumoral metabolites were determined using region of interest analysis based on registration of EPSI images with T2-weighted images. To ensure the entire tumor was included in each analysis (including regions encompassing only a partial voxel), the sums of intratumoral resonances were determined from a 2D interpolation of the corrected EPSI data using a cubic convolution (interpolation factor = 8). When the intratumoral signal-to-noise ratio (SNR) for an individual image was ≥2, metabolite quantifications for individual images were calculated as the integral of metabolite peaks identified on the basis of chemical shifts (pyruvate: 170.4 ppm, lactate: 183.35 ppm, and alanine: 176.5 ppm). For individual images within a time series with subthreshold SNR or for which metabolite integrals yielded negative values, metabolite quantifications were recorded as 0 for subsequent analyses. Total 13C metabolite (TCM) signal was determined as the sum of quantified pyruvate, lactate, and alanine resonances in an image. Kinetic analyses were performed by taking the area under the curve (AUC) of metabolite quantifications from the dynamic EPSI series prior to ratiometric comparisons. Nonzero AUCs were quantified according to this procedure for all metabolites in all comparisons outlined in this work. Parametric maps were generated from cubic interpolation of metabolite quantifications (interpolation factor = 8).

Statistical analyses

Statistical analyses were performed in RStudio Desktop 1.1.442; graphical representations of these data were made using GraphPad Prism 8.2.1. Shapiro–Wilk tests for normality were performed for all two-group comparisons. Normally distributed data were compared with Student t tests, while nonnormally distributed data were compared with Wilcoxon signed rank tests. Paired t tests were used for all paired data, while a two-sample t test was used for nonpaired data. T-statistics are reported for all t tests, while V-statistics are reported for all Wilcoxon signed rank tests. Linear regression was used to assess changes in tumor growth from imaging data.

CRISPR-Cas9 screening reveals theranostic targets

To evaluate the utility of functional genetic screening for therapeutic target identification and probe selection, negative selection CRISPR screens from the DepMap Public 19Q3 dataset, which collectively study the essentiality of 18,333 genes in 625 cancer lines, were analyzed (13, 14). Using the provided cancer type annotations, this analysis isolated differential vulnerabilities, identifying genes encoding proteins most essential to growth (Supplementary Table 1). These data were then filtered to isolate genes encoding differentially essential enzymes for which DNP-MRS probes were previously reported to measure enzymatic activity (Fig. 1A; Supplementary Fig. S1; Supplementary Table S1). Of the 29 different cancer types investigated, 17 demonstrated targetable dependencies that may be imaged using an established 13C-labeled DNP-MRS probe (Fig. 1A; Supplementary Fig. S1; Supplementary Tables S1–S3). In addition, analysis of the top five differentially essential genes revealed novel potential imaging targets for four of the remaining cancer types (Fig. 1A; Supplementary Fig. S1; Supplementary Table S1).

Figure 1.

Functional genetic screening enables simultaneous selection of therapeutic targets and imaging probes. A, Schematic depicting metabolic pathways that can be analyzed with DNP-MRS probes. Purple nodes correspond to previously polarized probes. Pink nodes correspond to intermediates that have either been (i) directly visualized after injection of a DNP-MRS probe or (ii) have not been directly identified but precede a downstream intermediate that has been directly visualized. Yellow nodes correspond to metabolites that may serve as future molecular imaging probes, as suggested by the data provided in Supplementary Table S1. Dashed lines correspond to an enzyme or group of enzymes connecting DNP-MRS probes and/or intermediates. Colored semi-circles correspond to cancer types that may be imaged with a given DNP-MRS probe, as described in Supplementary Table S1. Supplementary Fig. S1 situates this network of probes in the broader metabolome using Pathview (46). B, Dot plot depicting the relative essentiality of LDHA in different tumors types (as annotated in the DepMap 19Q3 dataset) based upon CERES score. Individual dots represent unique cell lines (n = 625). Blue dots highlight hepatocellular carcinoma cell lines (n = 20). Dark lines depict the median CERES score within cancer groups. C, Schematic illustrating the metabolism of hyperpolarized [1-13C]-pyruvate by LDH and alanine aminotransferase (ALT) to hyperpolarized [1-13C]-lactate and hyperpolarized [1-13C]-alanine, respectively. CNS, central nervous system; PNS, peripheral nervous system.

Figure 1.

Functional genetic screening enables simultaneous selection of therapeutic targets and imaging probes. A, Schematic depicting metabolic pathways that can be analyzed with DNP-MRS probes. Purple nodes correspond to previously polarized probes. Pink nodes correspond to intermediates that have either been (i) directly visualized after injection of a DNP-MRS probe or (ii) have not been directly identified but precede a downstream intermediate that has been directly visualized. Yellow nodes correspond to metabolites that may serve as future molecular imaging probes, as suggested by the data provided in Supplementary Table S1. Dashed lines correspond to an enzyme or group of enzymes connecting DNP-MRS probes and/or intermediates. Colored semi-circles correspond to cancer types that may be imaged with a given DNP-MRS probe, as described in Supplementary Table S1. Supplementary Fig. S1 situates this network of probes in the broader metabolome using Pathview (46). B, Dot plot depicting the relative essentiality of LDHA in different tumors types (as annotated in the DepMap 19Q3 dataset) based upon CERES score. Individual dots represent unique cell lines (n = 625). Blue dots highlight hepatocellular carcinoma cell lines (n = 20). Dark lines depict the median CERES score within cancer groups. C, Schematic illustrating the metabolism of hyperpolarized [1-13C]-pyruvate by LDH and alanine aminotransferase (ALT) to hyperpolarized [1-13C]-lactate and hyperpolarized [1-13C]-alanine, respectively. CNS, central nervous system; PNS, peripheral nervous system.

Close modal

As a proof of principle, these analyses were also performed for hepatocellular carcinoma, the most common form of primary liver cancer, which comprise a subset of the liver cancers in the DepMap database. Initial investigation of differentially essential genes identified lactate dehydrogenase A (LDHA), which encodes the predominant isoform of LDH, a known molecular imaging target, as the most differentially essential enzyme-encoding gene (P < 10−8; Table 1; Fig. 1B; refs. 13, 14). LDHA was the only DNP-MRSI imageable dependency isolated in hepatocellular carcinoma among significantly depleted genes (ref. 15; Supplementary Table S1). The identification of FGFR1, a gene encoding one of several proteins inhibited by the standard-of-care hepatocellular carcinoma therapies lenvatinib and sorafenib, as the third most differentially essential gene further corroborated the screening technique (P < 10−6; Table 1).

Table 1.

Top five differentially essential genes in 20 hepatocellular carcinoma lines, as compared with other cancer types in the DepMap 19Q3 dataset.

CERES mean
GeneHCCnon-HCCt StatisticP
HNF4A −0.146 0.075 −6.473 1.93E-10 
LDHA −0.174 0.034 −5.919 5.32E-09 
FGFR1 −0.421 −0.102 −5.225 2.36E-07 
HS2ST1 −0.273 −0.126 −4.284 2.12E-05 
XYLT2 −0.652 −0.471 −4.113 4.43E-05 
CERES mean
GeneHCCnon-HCCt StatisticP
HNF4A −0.146 0.075 −6.473 1.93E-10 
LDHA −0.174 0.034 −5.919 5.32E-09 
FGFR1 −0.421 −0.102 −5.225 2.36E-07 
HS2ST1 −0.273 −0.126 −4.284 2.12E-05 
XYLT2 −0.652 −0.471 −4.113 4.43E-05 

To validate these findings further, a focused CRISPR screening library was designed and adapted to enable high-throughput screening of theranostic probes including guides targeting genes encoding enzymes that metabolize established DNP-labeled molecular imaging substrates, and requiring as few as 4e6 cells (Fig. 1A; refs. 5, 16). Negative selection screens were performed in an established hepatocellular carcinoma cell line included in the DepMap dataset (SNU-449) and the results confirmed the essentiality of LDHA in these cells (log2 fold-change or LFC = −1.10; P < 10−4; FDR = 6.30e-5). A negative selection CRISPR screen was then performed in a primary hepatocellular carcinoma cell line (PGM-898) generated in the authors' laboratory from a patient-derived xenograft, further confirming an LDH dependency in hepatocellular carcinoma (LFC = −0.380; P < 0.05; FDR = 0.175).

Hyperpolarized [1-13C]-pyruvate quantifies response to LDH inhibition

LDH enzymatically converts pyruvate to lactate, while also regenerating NAD+ (Fig. 1C). Hyperpolarized [1-13C]-pyruvate is a molecular imaging probe that, when metabolized to form lactate, quantifies local LDH activity (5, 9, 17). To test a theranostic strategy combining LDH inhibition with DNP-MRSI assessment of LDH activity, a translational rat model of hepatocellular carcinoma was utilized wherein autochthonous hepatocellular carcinomas were chemically induced in a background of cirrhosis. To identify whether hyperpolarized [1-13C]-pyruvate could be used to measure drug pharmacodynamics, MRSI of hyperpolarized [1-13C]-pyruvate was performed before and after intravenous injection of a LDHi at either 10 (n = 6) or 20 mg/kg (n = 3; Fig. 2). As observed in prior studies with this translational model of hepatocellular carcinoma, tumor regions had up to 3-fold greater levels of signal-to-noise than the surrounding liver (9). In all cases, LDHi administration led to the near complete abrogation of intratumoral lactate production, which manifested as reductions in the lactate-to-total carbon metabolite ratio (L/TCM; t = −6.20; df = 8; P < 0.001; n = 9) and the lactate-to-pyruvate ratio (L/P; V = 45; P < 0.01; n = 9; Fig. 2 and 3; Supplementary Fig. S2). Consistent with previous reports of transport-limited metabolism of DNP probes, LDH inhibition also led to increases in the pyruvate-to-total carbon metabolite ratio (P/TCM; t = 4.40; df = 8; P < 0.01; n = 9), the alanine-to-total carbon metabolite ratio (A/TCM; t = 2.82; df = 8; P < 0.05; n = 9), the alanine-to-pyruvate ratio (A/P; t = 2.34; df = 8; P < 0.05; n = 9), and the alanine-to-lactate ratio (A/L; t = 4.35; df = 8; P < 0.01; n = 9; refs. 18, 19).

Figure 2.

DNP-MRSI quantifies intratumoral hepatocellular carcinoma LDH activity in vivo. A, DNP-MRS images following injection of hyperpolarized [1-13C]-pyruvate enable the quantification of metabolism (i) before and (ii) after administration of a LDHi, including spectra reconstructed from each voxel in the first of a series of EPSI acquisitions overlaid upon T2-weighted 1H images. B, Stacked spectra show the evolution of intratumoral pyruvate (170.4 ppm), lactate (183.35 ppm), and alanine (176.5 ppm) (i) before and (ii) after LDHi administration, demonstrating a marked reduction in lactate formation. The first spectrum in this series corresponds to the intratumoral sum of spectral data in A, where interpolation was used to quantify signal originating from partial voxels. C, Plots highlighting the evolution of metabolite integrals (i) before and (ii) after LDHi.

Figure 2.

DNP-MRSI quantifies intratumoral hepatocellular carcinoma LDH activity in vivo. A, DNP-MRS images following injection of hyperpolarized [1-13C]-pyruvate enable the quantification of metabolism (i) before and (ii) after administration of a LDHi, including spectra reconstructed from each voxel in the first of a series of EPSI acquisitions overlaid upon T2-weighted 1H images. B, Stacked spectra show the evolution of intratumoral pyruvate (170.4 ppm), lactate (183.35 ppm), and alanine (176.5 ppm) (i) before and (ii) after LDHi administration, demonstrating a marked reduction in lactate formation. The first spectrum in this series corresponds to the intratumoral sum of spectral data in A, where interpolation was used to quantify signal originating from partial voxels. C, Plots highlighting the evolution of metabolite integrals (i) before and (ii) after LDHi.

Close modal
Figure 3.

DNP-MRSI detects LDHi-induced alterations in hepatocellular carcinoma metabolism. A, Parametric maps of intratumoral pyruvate, lactate, and alanine signals before (row 1) and after (row 2) LDHi administration relative to TCM signal (P+L+A). These data demonstrate an increase in intratumoral pyruvate, a decrease in intratumoral lactate, and an increase in intratumoral alanine after LDHi administration. All maps were generated from the acquisition highlighted in Fig. 2A. B, Violin plots of changes in measured intratumoral metabolite ratios following LDHi administration, including: ↑P/TCMs (n = 9, P < 0.01), ↓L/TCMs (n = 9, P < 0.001), ↑A/TCMs (n = 9, P < 0.05), ↓L/P (n = 9, P < 0.01), ↑A/P (n = 9, P < 0.05), and ↑A/L (n = 9, P < 0.01). Normality was assessed with Shapiro–Wilk tests. P/TCM, L/TCM, A/TCM, A/P, and A/L were found to be normally distributed and were compared with a paired t test. The L/P ratio was found to be nonnormally distributed and a paired comparison was performed using a Wilcoxon signed rank test. [*, P < 0.05; **, P < 0.01; ***, P < 0.001].

Figure 3.

DNP-MRSI detects LDHi-induced alterations in hepatocellular carcinoma metabolism. A, Parametric maps of intratumoral pyruvate, lactate, and alanine signals before (row 1) and after (row 2) LDHi administration relative to TCM signal (P+L+A). These data demonstrate an increase in intratumoral pyruvate, a decrease in intratumoral lactate, and an increase in intratumoral alanine after LDHi administration. All maps were generated from the acquisition highlighted in Fig. 2A. B, Violin plots of changes in measured intratumoral metabolite ratios following LDHi administration, including: ↑P/TCMs (n = 9, P < 0.01), ↓L/TCMs (n = 9, P < 0.001), ↑A/TCMs (n = 9, P < 0.05), ↓L/P (n = 9, P < 0.01), ↑A/P (n = 9, P < 0.05), and ↑A/L (n = 9, P < 0.01). Normality was assessed with Shapiro–Wilk tests. P/TCM, L/TCM, A/TCM, A/P, and A/L were found to be normally distributed and were compared with a paired t test. The L/P ratio was found to be nonnormally distributed and a paired comparison was performed using a Wilcoxon signed rank test. [*, P < 0.05; **, P < 0.01; ***, P < 0.001].

Close modal

LDH inhibition slows tumor growth, as predicted by intratumoral hyperpolarized [1-13C]-pyruvate metabolism

Given that LDH knockout slowed hepatocellular carcinoma growth in vitro, the therapeutic efficacy of the LDHi was assessed in vivo. Tumor volumes measured from T2-weighted images acquired before and after inhibitor administration were used to quantify tumor doubling time (20). Tumor volumes from images acquired prior to therapy and those acquired within 10 days following therapy were fitted to exponential growth functions to quantify tumor doubling times. A single dose of the LDHi at either 10 (n = 6) or 20 mg/kg (n = 3) was found to significantly reduce tumor doubling times (V = 44; P < 0.01; n = 9; Fig. 4A and B). The strength of this effect increased when using a 20 mg/kg injection compared with a 10 mg/kg injection (t = 3.87; df = 3.35; P < 0.05; n = 6 at 10 mg/kg and n = 3 at 20 mg/kg; Fig. 4C).

Figure 4.

LDHi administration reduces hepatocellular carcinoma growth, as predicted by hyperpolarized [1-13C]-pyruvate metabolism to hyperpolarized [1-13C]-alanine. A, T2-weighted 1H images from 8 days prior to treatment (left), the day of treatment (center), and 8 days following treatment (right) demonstrating a reduction in tumor growth (tumor segmentations are displayed in red). B, LDHi administration reduced tumor growth, leading to an increase in the doubling time of these tumors (n = 9, P < 0.01). A Shapiro–Wilk test demonstrated these data to be nonnormally distributed and were therefore compared with a Wilcoxon signed rank test. C, Bar graph demonstrating dose-dependent tumor growth delay following LDHi administration (n = 6 at 10 mg/kg and n = 3 at 20 mg/kg; P < 0.05). These data were found to be normally distributed and were therefore compared with a two-sample t test. [*, P < 0.05; **, P < 0.01].

Figure 4.

LDHi administration reduces hepatocellular carcinoma growth, as predicted by hyperpolarized [1-13C]-pyruvate metabolism to hyperpolarized [1-13C]-alanine. A, T2-weighted 1H images from 8 days prior to treatment (left), the day of treatment (center), and 8 days following treatment (right) demonstrating a reduction in tumor growth (tumor segmentations are displayed in red). B, LDHi administration reduced tumor growth, leading to an increase in the doubling time of these tumors (n = 9, P < 0.01). A Shapiro–Wilk test demonstrated these data to be nonnormally distributed and were therefore compared with a Wilcoxon signed rank test. C, Bar graph demonstrating dose-dependent tumor growth delay following LDHi administration (n = 6 at 10 mg/kg and n = 3 at 20 mg/kg; P < 0.05). These data were found to be normally distributed and were therefore compared with a two-sample t test. [*, P < 0.05; **, P < 0.01].

Close modal

DNP-MRSI data were further analyzed to assess the ability to predict therapeutic response at the time of drug administration. Interestingly, intratumoral alanine production after LDHi administration robustly predicted the degree of growth inhibition (P < 0.001). In addition, the intratumoral level of alanine following LDHi administration was confirmed as a predictive biomarker when controlling for multiple potential covariates, including models correcting for postinhibitor pyruvate levels (P < 0.01; Table 2), preinhibitor alanine levels (P < 0.01), and postinhibitor pyruvate and lactate levels (P < 0.01). These data demonstrate that the postinhibitor alanine level directly correlates with the observed LDHi-induced reduction in tumor growth. Postinhibition intratumoral lactate, which uniformly approached the limit of detection, did not predict response, likely secondary to insufficient sensitivity.

Table 2.

Linear regression model demonstrating that postinhibitor alanine levels predict the observed tumor growth inhibition (n = 9; P < 0.01).

lm(doubling time FC ∼ pyruvate + alanine)
ParameterEstimateSEtP
Intercept 1.18E+00 2.31E-01 5.111 0.002 
Pyruvate AUC 4.26E-05 6.88E-05 0.619 0.558 
Alanine AUC 3.50E-03 6.19E-04 5.655 0.001 
lm(doubling time FC ∼ pyruvate + alanine)
ParameterEstimateSEtP
Intercept 1.18E+00 2.31E-01 5.111 0.002 
Pyruvate AUC 4.26E-05 6.88E-05 0.619 0.558 
Alanine AUC 3.50E-03 6.19E-04 5.655 0.001 

Taken together, these data demonstrate that functional genetic screening informs a theranostic paradigm leveraging DNP-MRSI to assess drug pharmacodynamics, as well as to predict therapeutic efficacy.

Functional genetic screening identified imageable metabolic vulnerabilities in 17 unique cancer types. Among these, LDH was identified as a differential vulnerability in hepatocellular carcinoma. These data demonstrate that LDH is more critical to hepatocellular carcinoma growth than it is to the growth of other cancers (21). LDHA was found to be the second most differentially essential protein-encoding gene and the single most differentially essential enzyme-encoding gene for hepatocellular carcinoma cell growth. The dependence of hepatocellular carcinoma on LDH was further validated using a focused CRISPR screen in two cell lines, including one derived from a patient biopsy. These findings underscore the growing recognition that cancer cells demonstrate differential dependencies on metabolic enzymes (14, 22, 23). Consistent with the hypothesis that differential enzyme essentiality enables a theranostic imaging approach via simultaneous identification of an imaging probe and a therapy, genetic screening accurately identified hyperpolarized [1-13C]-pyruvate and its metabolites as in vivo biomarkers of on-target efficacy of, and response to, LDH inhibition in hepatocellular carcinoma.

While the role of LDH in promoting cancer growth is well established, these findings underscore the importance of variabilities in enzymatic dependencies and activities both within and across cancer types. This variability forms the basis for clinical decision-making both with respect to the selection of targeted cancer therapies, as well as the selection of imaging studies (24, 25). Indeed, variability in the expression and activity of the GLUT1 transporter and hexokinase underlie differences in 18FDG-avidity on PET imaging and influence heterogeneity in the application of 18FDG-PET in cancer imaging (25–28). The presented data suggest that genetic screening tools can be applied to characterize these dependencies and enable the selection of optimal imaging probes on a cancer-by-cancer basis. Given inherent variability in cancer dependencies, curated screens, including those performed with the DNP-focused library, minimize experimental complexity and requisite cell number, enabling confirmation of potential therapeutic and imaging targets in patient-derived samples. Moreover, unbiased screening, including screens available within the DepMap datasets, can independently isolate alternative dependencies, suggesting avenues for future therapy and imaging probe development. Indeed, a wider, unbiased analysis limited to the top five differentially essential genes, identified potential metabolic imaging targets in four cancer types for which the focused DepMap screens did not identify a dependency on an enzyme that metabolizes an existing DNP-probe. While this study leveraged the unique capability of DNP-MRS imaging to determine enzyme function based on direct measurements of the administered substrate and its metabolites, the described paradigm is generalizable to other imaging modalities. This screening approach can be readily expanded to include targets of other molecular imaging probes including the extensive repertoire of PET-based radiotracers already developed for oncologic imaging (29).

In addition, by directly imaging the metabolic pathway targeted by a therapy, the presented data demonstrate a role for metabolic molecular imaging for predicting a clinical phenotype at the time of therapy administration. In this study, alanine generation after LDH inhibition predicted the reduction in tumor doubling time. This finding, in combination with the observed dose-dependent growth inhibition, underscores the importance of direct assessments of on-target efficacy for metabolic inhibitors. Indeed, suboptimal on-target efficacy has limited the translation of LDH inhibitors described in the literature to date (11, 30). As such, these data, in combination with previous studies, suggest the unique potential of metabolic imaging with DNP-MRSI of hyperpolarized [1-13C]-pyruvate to inform targeted therapy selection, therapeutic efficacy, as well as disease severity on a broader scale (23, 31). As a highly regulated metabolic enzyme, assessments of LDH activity in vivo with hyperpolarized [1-13C]-pyruvate MRSI may stratify patients and predict response to targeted systemic or locoregional therapies beyond LDH inhibitiors (9, 32). In hepatocellular carcinoma, hyperpolarized [1-13C]-pyruvate MRSI may predict response to lenvatinib and sorafenib given prior literature demonstrating the prognostic value of serum LDH concentration (33, 34). Similarly, a recent clinical trial in prostate cancer demonstrated that lactate generation from DNP-[1-13C]-pyruvate predicts tumor grade (35). Future preclinical and clinical studies will help assess the predictive value of hyperpolarized [1-13C]-pyruvate MRSI in this broader context. Moreover, the presented data emphasize a role for DNP-MRSI in this capacity beyond hyperpolarized [1-13C]-pyruvate. The identified dependence of hepatocellular carcinoma on FGFR1 suggests that imaging FGFR1-encoded protein activity could provide a direct assessment of lenvatinib and sorafenib efficacy in hepatocellular carcinoma. While there is not yet a DNP-labeled probe for this application, ligand-receptor–induced changes in T1 or chemical shift may guide the development of DNP-MRS probes that report on the efficacy of receptor targeting (36). Furthermore, as MRSI can be performed with multiple DNP-labeled probes simultaneously, optimization of probe combinations may facilitate diagnosis based on independent cancer dependencies. For example, the included analysis of the DepMap database suggest that DNP-[2-13C]-dihydroxyacetone, together with hyperpolarized [1-13C]-pyruvate, may enable the noninvasive distinction of colorectal cancer, which frequently metastasizes to the liver, from hepatocellular carcinoma (Fig. 1; Supplementary Fig. S1; Supplementary Table S1).

Importantly, the described data hold important implications for the treatment of hepatocellular carcinoma, the most rapidly increasing cause of cancer-related mortality in the United States (37). The majority of patients with hepatocellular carcinoma present with incurable disease at diagnosis and, despite the approval of targeted therapies, life expectancy remains less than 20 months (38). This dismal prognosis issues from challenges in developing targeted therapies and a lack of functional imaging strategies that report on therapeutic efficacy (3, 39–42). This study leveraged DNP-MRSI to visualize intratumoral metabolism of hyperpolarized [1-13C]-pyruvate, confirming that LDH inhibition prevented lactate production and reduced tumor doubling times in vivo. A single dose of a LDHi in this model of hepatocellular carcinoma slowed tumor growth in a dose-dependent manner suggesting that higher doses, repeat injections, or local administration may enable tumor regression and/or mitigate hepatocellular carcinoma recurrence. Previous studies have demonstrated that LDH inhibition may reduce tumor doubling times in several cancers and that myc-driven tumors, including hepatoblastoma, have enhanced LDH activity; however, in vivo LDH targeting in hepatocellular carcinoma has not been demonstrated previously (5, 43, 44). In addition, while the observed LDHi-induced reduction in lactate production was anticipated, the degree of LDH inhibition observed in our study is unique (23, 43, 45). These data, in combination with the genetic screening data, demonstrate that hepatocellular carcinoma is more susceptible to LDH inhibition than other cancers. Consistent with this hypothesis, a recent study of pancreatic cancer found that larger and more frequent doses of the LDHi described herein were required to observe growth inhibition of xenografts (43). This discrepancy suggests that pancreatic cancer is less dependent upon LDH for growth than hepatocellular carcinoma (Fig. 1B). Future work will determine the optimal dose, frequency, and delivery method for a LDHi by leveraging the described DNP-MRSI analysis pipeline.

In summary, this study demonstrates that biologically motivated therapeutic and imaging probe selection through genetic screening enables a high-throughput theranostic paradigm in oncology. In the era of precision medicine, wherein patient-derived samples are increasingly utilized to inform management, this approach represents a novel strategy to overcome existing barriers and facilitate the application of molecular imaging at the bench as well as at the bedside. Finally, the described data provide a blueprint for validation of this approach through the integration of hyperpolarized [1-13C]-pyruvate MRSI in clinical trials of LDH inhibitors.

N.R. Perkons reports grants from NIH NCI (F30CA232388) and grants from Society of Interventional Radiology Foundation (Allied Scientist Training Grant) during the conduct of the study, as well as non-financial support from Guerbet LLC (Travel to a research meeting) outside the submitted work. D. Ackerman reports other from Guerbet (Conference attendance) outside the submitted work. T.P.F. Gade reports grants from NIH (DP5 OD021391) during the conduct of the study, as well as grants from Society for Interventional Radiology, grants from Society for Interventional Oncology, grants from Radiological Society of North America, grants from NIH, and personal fees from Trisalus Life Sciences (Scientific Advisory Board) outside the submitted work. No potential conflicts of interest were disclosed by the other authors.

N.R. Perkons: Conceptualization, resources, data curation, software, formal analysis, funding acquisition, validation, investigation, methodology, writing-original draft, writing-review and editing. O. Johnson: Data curation, investigation, methodology, writing-review and editing. G. Pilla: Data curation, investigation, methodology, writing-review and editing. E. Profka: Data curation, investigation, methodology, writing-review and editing. M. Mercadante: Data curation, investigation, methodology, writing-review and editing. D. Ackerman: Conceptualization, supervision, funding acquisition, project administration, writing-review and editing. T.P.F. Gade: Conceptualization, resources, formal analysis, supervision, funding acquisition, methodology, writing-original draft, project administration, writing-review and editing.

This study was supported in part by the NIH, NCI, Grant F30CA232388 and Society of Interventional Radiology Foundation's Allied Scientist Grant (all to N.R. Perkons). This study was also supported in part by the NIH, Director's Office, Grant DP5OD021391 (to T.P.F. Gade).

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.
Santos
R
,
Ursu
O
,
Gaulton
A
,
Bento
AP
,
Donadi
RS
,
Bologa
CG
, et al
A comprehensive map of molecular drug targets
.
Nat Rev Drug Discov
2017
;
16
:
19
34
.
2.
Mankoff
DA
,
Farwell
MD
,
Clark
AS
,
Pryma
DA
. 
Making molecular imaging a clinical tool for precision oncology: a review
.
JAMA Oncol
2017
;
3
:
695
701
.
3.
van der Pol
CB
,
Lim
CS
,
Sirlin
CB
,
McGrath
TA
,
Salameh
JP
,
Bashir
MR
, et al
Accuracy of the liver imaging reporting and data system in computed tomography and magnetic resonance image analysis of hepatocellular carcinoma or overall malignancy—a systematic review
.
Gastroenterology
2019
;
156
:
976
86
.
4.
Shi
J
,
Wang
E
,
Milazzo
JP
,
Wang
Z
,
Kinney
JB
,
Vakoc
CR
. 
Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains
.
Nat Biotechnol
2015
;
33
:
661
7
.
5.
Keshari
KR
,
Wilson
DM
. 
Chemistry and biochemistry of 13C hyperpolarized magnetic resonance using dynamic nuclear polarization
.
Chem Soc Rev
2014
;
43
:
1627
59
.
6.
Chaumeil
MM
,
Larson
PEZ
,
Yoshihara
HAI
,
Danforth
OM
,
Vigneron
DB
,
Nelson
SJ
, et al
Non-invasive in vivo assessment of IDH1 mutational status in glioma
.
Nat Commun
2013
;
4
:
1
12
.
7.
Witney
TH
,
Kettunen
MI
,
Brindle
KM
. 
Kinetic modeling of hyperpolarized 13C label exchange between pyruvate and lactate in tumor cells
.
J Biol Chem
2011
;
286
:
24572
80
.
8.
Kiefer
RM
,
Hunt
SJ
,
Pulido
S
,
Pickup
S
,
Furth
EE
,
Soulen
MC
, et al
Relative initial weight is associated with improved survival without altering tumor latency in a translational rat model of diethylnitrosamine-induced hepatocellular carcinoma and transarterial embolization
.
J Vasc Interv Radiol
2017
;
28
:
1043
50
.
9.
Perkons
NR
,
Kiefer
RM
,
Noji
MC
,
Pourfathi
M
,
Ackerman
D
,
Siddiqui
S
, et al
Hyperpolarized metabolic imaging detects latent hepatocellular carcinoma domains surviving locoregional therapy
.
Hepatology
2019
Sep 25 [Epub ahead of print].
10.
Yeung
C
,
Gibson
AE
,
Issaq
SH
,
Oshima
N
,
Baumgart
JT
,
Edessa
LD
, et al
Targeting glycolysis through inhibition of lactate dehydrogenase impairs tumor growth in preclinical models of Ewing sarcoma
.
Cancer Res
2019
;
79
:
5060
73
.
11.
Rai
G
,
Brimacombe
KR
,
Mott
BT
,
Urban
DJ
,
Hu
X
,
Yang
SM
, et al
Discovery and optimization of potent, cell-active pyrazole-based inhibitors of lactate dehydrogenase (LDH)
.
J Med Chem
2017
;
60
:
9184
204
.
12.
Basic practical NMR concepts : A guide for the modern laboratory
.
Max T. Rogers NMR Facility
2013
. Available from: https://www2.chemistry.msu.edu/facilities/nmr/handouts/DH%20NMR%20Basics.pdf.
13.
Meyers
RM
,
Bryan
JG
,
McFarland
JM
,
Weir
BA
,
Sizemore
AE
,
Xu
H
, et al
Computational correction of copy number effect improves specificity of CRISPR-Cas9 essentiality screens in cancer cells
.
Nat Genet
2017
;
49
:
1779
84
.
14.
Broad Institute.
DepMap. 
2019
. Available from: https://depmap.org/portal/.
15.
Kudo
M
,
Finn
RS
,
Qin
S
,
Han
KH
,
Ikeda
K
,
Piscaglia
F
, et al
Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial
.
Lancet
2018
;
391
:
1163
73
.
16.
Wildenberg
J
,
Perkons
NR
,
Pilla
G
,
Kadlecek
S
,
Gade
TPF
. 
Computational pipeline for estimation of small-molecule T1 relaxation times
.
J Magn Reson
2020
;
314
:
106733
.
17.
Düwel
S
,
Durst
M
,
Gringeri C
V.
,
Kosanke
Y
,
Gross
C
,
Janich
MA
, et al
Multiparametric human hepatocellular carcinoma characterization and therapy response evaluation by hyperpolarized 13C MRSI
.
NMR Biomed
2016
;
29
:
952
60
.
18.
Takado
Y
,
Cheng
T
,
Bastiaansen
JAM
,
Yoshihara
HAI
,
Lanz
B
,
Mishkovsky
M
, et al
Hyperpolarized 13C magnetic resonance spectroscopy reveals the rate-limiting role of the blood-brain barrier in the cerebral uptake and metabolism of l-lactate in vivo
.
ACS Chem Neurosci
2018
;
9
:
2554
62
.
19.
Balzan
R
,
Fernandes
L
,
Pidial
L
,
Comment
A
,
Tavitian
B
,
Vasos
PR
. 
Pyruvate cellular uptake and enzymatic conversion probed by dissolution DNP-NMR: the impact of overexpressed membrane transporters
.
Magn Reson Chem
2017
;
55
:
579
83
.
20.
Yushkevich
PA
,
Piven
J
,
Hazlett
HC
,
Smith
RG
,
Ho
S
,
Gee
JC
, et al
User-guided 3D active contour segmentation of anatomical structures: significantly improved efficiency and reliability
.
Neuroimage
2006
;
31
:
1116
28
.
21.
Liberti
MV
,
Locasale
JW
The Warburg effect: how does it benefit cancer cells?
Trends Biochem Sci
2016
;
41
:
211
8
.
22.
Lee
P
,
Malik
D
,
Perkons
N
,
Huangyang
P
,
Khare
S
,
Rhoades
S
, et al
Targeting glutamine metabolism slows soft tissue sarcoma growth
.
Nat Commun
2020
;
11
:
1
15
.
23.
Rajeshkumar
NV
,
Dutta
P
,
Yabuuchi
S
,
De Wilde
RF
,
Martinez
GV
,
Le
A
, et al
Therapeutic targeting of the Warburg effect in pancreatic cancer relies on an absence of p53 function
.
Cancer Res
2015
;
75
:
3355
64
.
24.
McLornan
DP
,
List
A
,
Mufti
GJ
. 
Applying synthetic lethality for the selective targeting of cancer
.
N Engl J Med
2014
;
371
:
1725
35
.
25.
Pantel
AR
,
Ackerman
D
,
Lee
SC
,
Mankoff
DA
,
Gade
TPF
. 
Imaging cancer metabolism: underlying biology and emerging strategies
.
J Nucl Med
2018
;
59
:
1340
9
.
26.
Alvarez J
V.
,
Belka
GK
,
Pan
TC
,
Chen
CC
,
Blankemeyer
E
,
Alavi
A
, et al
Oncogene pathway activation in mammary tumors dictates FDG-PET uptake
.
Cancer Res
2014
;
74
:
7583
98
.
27.
Cho
MH
,
Park
CK
,
Park
M
,
Kim
WK
,
Cho
A
,
Kim
H
. 
Clinicopathologic features and molecular characteristics of glucose metabolism contributing to 18F-fluorodeoxyglucose uptake in gastrointestinal stromal tumors
.
PLoS One
2015
;
10
:
1
13
.
28.
Iwamoto
M
,
Kawada
K
,
Nakamoto
Y
,
Itatani
Y
,
Inamoto
S
,
Toda
K
, et al
Regulation of 18F-FDG accumulation in colorectal cancer cells with mutated KRAS
.
J Nucl Med
2014
;
55
:
2038
44
.
29.
Rauscher
I
,
Eiber
M
,
Souvatzoglou
M
,
Schwaiger
M
,
Beer
AJ
. 
PET/MR in oncology: non-18F-FDG tracers for routine applications
.
J Nucl Med
2014
;
55
:
25S
31S
.
30.
Shi
Y
,
Pinto
BM
. 
Human lactate dehydrogenase a inhibitors: a molecular dynamics investigation
.
PLoS One
2014
;
9:e86365
.
31.
Gutte
H
,
Hansen
AE
,
Johannesen
HH
,
Clemmensen
AE
,
Ardenkjær-Larsen
JH
,
Nielsen
CH
, et al
The use of dynamic nuclear polarization (13)C-pyruvate MRS in cancer
.
Am J Nucl Med Mol Imaging
2015
;
5
:
548
60
.
32.
Zacharias
NM
,
Baran
N
,
Shanmugavelandy
SS
,
Lee
J
,
Lujan
JV
,
Dutta
P
, et al
Assessing metabolic intervention with a glutaminase inhibitor in real-time by hyperpolarized magnetic resonance in acute myeloid leukemia
.
Mol Cancer Ther
2019
;
18
:
1937
46
.
33.
Zhang
JP
,
Wang
HB
,
Lin
YH
,
Xu
J
,
Wang
J
,
Wang
K
, et al
Lactate dehydrogenase is an important prognostic indicator for hepatocellular carcinoma after partial hepatectomy
.
Transl Oncol
2015
;
8
:
497
503
.
34.
Faloppi
L
,
Scartozzi
M
,
Bianconi
M
,
Svegliati Baroni
G
,
Toniutto
P
,
Giampieri
R
, et al
The role of LDH serum levels in predicting global outcome in HCC patients treated with sorafenib: Implications for clinical management
.
BMC Cancer
2014
;
14
:
1
8
.
35.
Granlund
KL
,
Tee
S-SS
,
Vargas
HA
,
Lyashchenko
SK
,
Reznik
E
,
Fine
S
, et al
Hyperpolarized MRI of human prostate cancer reveals increased lactate with tumor grade driven by monocarboxylate transporter 1
.
Cell Metab
2020
;
31
:
105
14
.
36.
Keshari
KR
,
Kurhanewicz
J
,
Macdonald
JM
,
Wilson
DM
. 
Generating contrast in hyperpolarized 13C MRI using ligand–receptor interactions
.
Analyst
2012
;
137
:
3427
.
37.
Torre
LA
,
Bray
F
,
Siegel
RL
,
Ferlay
J
,
Lortet-tieulent
J
,
Jemal
A
. 
Global cancer statistics, 2012
.
CA a cancer J Clin
2015
;
65
:
87
108
.
38.
Scudellari
M
. 
Drug development: try and try again
.
Nature
. 
2014
;
516
:
S4
6
.
39.
Llovet
JM
,
Di Bisceglie
AM
,
Bruix
J
,
Kramer
BS
,
Lencioni
R
,
Zhu
AX
, et al
Design and endpoints of clinical trials in hepatocellular carcinoma
.
J Natl Cancer Inst
2008
;
100
:
698
711
.
40.
Forner
A
,
Ayuso
C
,
Varela
M
,
Rimola
J
,
Hessheimer
AJ
,
De Lope
CR
, et al
Evaluation of tumor response after locoregional therapies in hepatocellular carcinoma: Are response evaluation criteria in solid tumors reliable?
Cancer
2009
;
115
:
616
23
.
41.
Bruix
J
,
Gores
GJ
,
Mazzaferro
V
. 
Hepatocellular carcinoma: clinical frontiers and perspectives
.
Gut
2014
;
63
:
844
55
.
42.
Borgheresi
A
,
Gonzalez-Aguirre
A
,
Brown
KT
,
Getrajdman
GI
,
Erinjeri
JP
,
Covey
A
, et al
Does enhancement or perfusion on preprocedure ct predict outcomes after embolization of hepatocellular carcinoma?
Acad Radiol
2018
;
25
:
1588
94
.
43.
Oshima
N
,
Ishida
R
,
Kishimoto
S
,
Beebe
K
,
Brender
J
,
Yamamoto
K
, et al
Dynamic imaging of LDH inhibition in tumors reveals rapid in vivo metabolic rewiring and vulnerability to combination therapy
.
Cell Rep
2020
;
30
:
1798
810
.
44.
Hu
S
,
Balakrishnan
A
,
Bok
RA
,
Anderton
B
,
Larson
PEZ
,
Nelson
SJ
, et al
13C-pyruvate imaging reveals alterations in glycolysis that precede c-Myc-induced tumor formation and regression
.
Cell Metab
2011
;
14
:
131
42
.
45.
Dutta
P
,
Le
A
,
Vander Jagt
DL
,
Tsukamoto
T
,
Martinez G
V.
,
Dang
C V
, et al
Evaluation of LDH-A and glutaminase inhibition in vivo by hyperpolarized13C-pyruvate magnetic resonance spectroscopy of tumors
.
Cancer Res
2013
;
73
:
4190
5
.
46.
Luo
W
,
Brouwer
C
. 
Pathview: an R/Bioconductor package for pathway-based data integration and visualization
.
Bioinformatics
2013
;
29
:
1830
1
.

Supplementary data