Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 May 28.
Published in final edited form as: Lancet Oncol. 2025 Jan;26(1):e34–e45. doi: 10.1016/S1470-2045(24)00395-4

Advances and Challenges in Precision imaging

Hedvig Hricak 1, Marius E Mayerhoefer 1, Ken Herrmann 1, Jason S Lewis 1, Martin G Pomper 1, Christopher P Hess 1, Katrine Riklund 1, Andrew M Scott 1, Ralph Weissleder 1
PMCID: PMC12117531  NIHMSID: NIHMS2084242  PMID: 39756454

Abstract

Technological innovations in genomics and related fields have facilitated large sequencing efforts, supported new biological discoveries in cancer, and spawned an era of liquid biopsy biomarkers. Despite these advances, precision oncology has practical limitations, partly related to cancer’s biological diversity and spatial/temporal complexity. Advanced imaging technologies are being developed to address some of the current limitations in early detection, treatment selection and planning, drug delivery, and therapeutic response, as well as difficulties posed by drug resistance, drug toxicity, disease monitoring, and metastatic evolution. We discuss key areas of advanced imaging on the horizon for improving cancer outcomes and survival. Finally, we discuss practical challenges to the broader adoption of precision imaging in the clinic and the need for a robust translational infrastructure.

Keywords: precision oncology, imaging, diagnostics

Introduction

Precision or personalized medicine is an increasingly accepted approach to cancer care, in which therapy is planned based on the distinct molecular characteristics of a given tumor. Critical to precision medicine is the concept that the right combination of drugs must be used at the right stage and time in the progression of the disease. Although precision oncology has historically been defined by tailoring treatments to genetic mutations, its clinical translation has proven more complex. Contributing to the complexity are biological factors including the variable nature of the transcriptome, proteome, tumor microenvironment, physiology, lineage plasticity, and, importantly, dynamic temporal changes due to treatment pressures. Further complicating the choice of therapies is that new mutations evolve over time, and some deleterious mutations can also occur in normal tissues. Furthermore, the host also plays a key role in determining treatment response (i.e., different responses to the same dose).

In parallel to the emergence of precision oncology, there have been remarkable advances in imaging diagnostics, radiotheranostics, and image-guided therapy. Indeed, imaging has become indispensable in the entire treatment chain of cancer care, from screening, detection, and staging through treatment selection, planning, efficacy monitoring, image-guided treatment delivery, toxicity monitoring, long-term surveillance, and drug development. Yet, imaging is generally not featured in the design and validation of precision oncology. We would like to point out that to bring personalized medicine to the next level, the field must embrace imaging technologies to map the spatial and temporal composition of tumors and surrounding host tissue (Fig. 1). In other words, to optimize the efficient use of resources and to maximize clinical utility, developments in precision imaging need to be closer aligned with precision medicine roadmaps. This is even more important given the long lead times of technology development compared to those in the more established, well-funded, and reimbursed drug development pipeline. This article discusses the most recent imaging advances that enable precision oncology and summarizes current needs to further the field.

Fig. 1: Precision imaging and its role in precision oncology.

Fig. 1:

Precision oncology has historically been defined by the use of genetic mutations to identify patient populations who will respond to a given drug/dose combination. Precision oncology has practical limitations, partly related to cancer’s biological diversity and spatial/temporal complexity. Precision imaging technologies are being developed to address some of the current limitations. These emerging imaging methods strengthen precision oncology by providing clinically relevant information not obtainable by other means (brown box).

Advanced precision Imaging: An Overview

Broadly, we define precision imaging as advanced imaging approaches that allow increased anatomic coverage, improved spatial and temporal resolution, multiplexing, high-throughput screening, targeted sampling of tissue, and precise delivery of new therapeutics (Fig. 2). Fig. 1 summarizes some of the reasons why these advances are essential to precision oncology. Total-body imaging, for example, expands surveillance coverage and allows for better delineation of where the disease is located. Hybrid imaging systems improve the spatial information important for tumor heterogeneity assessment and permit the acquisition of multiplexed data at the anatomic, physiologic, and molecular levels, allowing stratification. Emerging new image-guided approaches add to the armamentarium of precision oncology and enable additional treatments, often with better patient acceptance than standard therapies.

Fig. 2: Examples of precision imaging during the treatment history of cancer.

Fig. 2:

A. Case history of a patient with cancer. The lesion size is plotted as a function of time and for primary tumor, locoregional invasion, and distant metastases. Note the frequent imaging. B. Role of precision imaging during different cancer stages. The grey boxes with checkmarks represent the main applications. Note: low-dose imaging is mostly for lung cancer (CT) or breast cancer screening (mammography). IO imaging: intraoperative imaging; radiopharmaceutical therapy (RPT).

Many of the advances in imaging technologies over the last 5 years have been “under the hood” but have vastly improved our current ability to perform anatomic/physiologic mapping, staging, and response monitoring (Fig. 2). Beyond hardware advances, the major driver has been computational advances in artificial intelligence (AI) that have provided new ways for algorithms to reduce scan time, increase image quality, reduce the patient radiation dose (e.g., in computed tomography, CT), automatically quantify radiographic patterns in images, and offer many other advantages. Today, faster imaging has major benefits, including artifact reduction, higher throughput, better coverage, better patient tolerance, and greater affordability. Better spatial resolution has allowed for seamless reconstructions in different planes, the ability to perform CT and MRI angiography, and better staging. Dose reduction helps with screening and the ability to perform more frequent follow-up imaging and pediatric studies. Targeted molecular imaging opened new frontiers in therapy selection and treatment monitoring. Some of the salient advances are summarized below.

Total-body PET

Over the last five years, long-axial field-of-view positron emission tomography (PET) scanners have emerged as an exciting new tool for improving image quality and reducing scan time1. This development helps to address a key question in precision oncology: where is the disease located (Fig. 1)? The new-generation scanners (e.g., μEXPLORER, PennPET, Siemens Biograph Vision Quadra) can acquire images of the main body organs simultaneously or image the total body with a single bed position2 (Fig. 3). The improved sensitivity and spatial resolution of the scanners mean that a PET study can be acquired in a much shorter time (e.g., 2–4 minutes), and with markedly better image quality, than is possible with standard digital PET-CT scanners3. Therefore, the time a patient spends in the scanner is much reduced, leading to enhanced tolerability. In addition, it is possible to significantly reduce the dose of radiopharmaceutical given for a scan without compromising image quality, hence reducing the radiation dose received by the patient1,2. The ability to perform whole-body dynamic imaging also has immediate relevance for the evaluation of the kinetics and biodistribution of novel radiopharmaceuticals3. The cost of the new total-body PET-CT scanners is higher than that of standard PET-CT systems. However, given that they enable higher patient throughput and decreased radiation doses while providing incremental diagnostic information, they will likely be implemented widely in clinical care and research in the next few years.

Fig. 3: Total-body PET Imaging.

Fig. 3:

Examples of studies enabled by total-body PET scanners that image the entire body at once with high detection sensitivity. A. Maximum-intensity projection (MIP) showing total-body parametric image for blood flow using 11C-butanol and quantified in absolute units of mL blood/min/cm3 tissue calculated with kinetic modeling78 (adapted, with permission, from79). B. MIP of the total-body distribution of [89Zr]Zr-crefmirlimab, an antibody fragment that binds to CD8+ T-cells (adapted from80). Uptake is observed in the spleen and bone marrow, with exquisite delineation of lymph nodes throughout the body. Image obtained 48 hours post-injection; the injected dose was only 18 MBq to enable repeat imaging.

Hybrid imaging systems

Hybrid molecular imaging refers to the acquisition and integration of information from functional and anatomic imaging, such as with PET-CT, single photon emission computed tomography (SPECT-CT), or PET-MRI (magnetic resonance imaging). Such imaging has two main advantages: i) display of molecular information on top of anatomic maps (the latter of which are often used for image-guided intervention) and ii) improvement of image reconstruction algorithms for PET or SPECT through the use of CT or MRI. The clinical adoption of PET-MRI in precision imaging has been relatively slow, although MRI by itself is used extensively in breast, prostate, liver, and brain cancers. Recent studies have shown that the combined information from PET and MRI is particularly useful in predicting outcomes in lymphoma after CAR-T-cell therapy4, identifying intraprostatic lesions5, and predicting overall survival in glioma6. In addition to having diagnostic advantages, PET-MRI is preferred over PET-CT in pediatric patients and in patients whose diagnostic work-up requires information from both PET and MRI (e.g., patients with advanced-stage cervical cancer or prostate cancer).

Molecular imaging agents

Over the last decade, many new molecular imaging agents have been tested preclinically7 or through first-in-human microdose exploratory investigational new drug (IND) applications or new drug applications (NDAs), and a smaller number have entered the market8. The latter include [68Ga]Ga-dotatoc (2019), [64Cu]Cu-dotatate (2020), [18F]Fluoroestradiol (2020), and various prostate-specific membrane antigen (PSMA)-targeting PET probes ([68Ga]Ga-PSMA-11, [68Ga]Ga-gozetotide, [18F]F-piflufolastat)9. Additional examples in the pipeline include agents for fibroblast activation protein (FAP; Fig. 4)10 and a large number of labeled antibodies for receptor imaging and cell tracking (ImmunoPET)11. The availability of these new probes improves the physiologic, pharmacologic, and molecular profiling of cancers and enables the assessment of their heterogeneity (Fig. 2 and 4). Novel molecular imaging agents could also be co-developed as companion diagnostics for next-generation therapies.

Fig. 4: Whole-body PET imaging with new fibroblast activation protein inhibitor (FAPI) tracer.

Fig. 4:

Thirty-eight-year-old female patient with a solitary fibrous tumor of the right abdominal wall presenting with lung, peritoneal and bone metastases. A. The left two images represent [18F]FDG PET maximum projection images before and after 4 cycles of treatment. B. As the patient exhausted all treatment options, a [68Ga]Ga-FAPI-46 PET-CT was performed (second from right), displaying high FAP uptake in all [18F]FDG-avid lesions. After four cycles of 90Y-FAPI-46 RLT, restaging revealed partial response according to RECIST criteria. Figure courtesy of Helena Lanzafame, Rainer Hamacher and Wolfgang Fendler (Universitätsmedizin Essen, Germany).

Companion diagnostic imaging

The integration of imaging agents as “companion diagnostics” is another frontier in imaging for precision oncology. The goal is to delineate a patient’s disease landscape, guiding clinicians with unparalleled clarity toward the most effective therapeutic avenues. The convergence of imaging technology with standard-of-care and new therapies presents an unmatched opportunity to improve outcomes. Several examples stand out. For example, [89Zr]Zr-DFO-SC16.56 is being used for the non-invasive in vivo imaging of delta-like ligand 3 (DLL3)-expressing malignancies12, which has been the focus of emerging therapies such as antibody-drug conjugates, T-cell engager molecules, CAR-Ts for small cell lung cancer13 and other neuroendocrine neoplasms14. Another example is the use of 16β−18F-fluoro-5α-dihydrotestosterone ([18F]FDHT), a PET tracer for detecting androgen receptor expression in prostate and breast cancers, which is used to study new therapeutic agents15,16.

Metabolic imaging

The success of [1⁸F]Fluorodeoxyglucose ([18F]FDG) PET has firmly established metabolic imaging in oncology. However, [18F]FDG PET cannot reliably distinguish between glucose uptake in normal cells (oxidative metabolism) and uptake in cancer cells (glycolytic metabolism with excess lactate production). Recently, two MR spectroscopy-based tests that probe dynamic changes in tissue metabolism have emerged: hyperpolarized MRI (HP-MRI) and deuterium metabolic imaging (DMI)17. Hyperpolarization, most commonly of 13C-pyruvate, increases the in vivo MR signal and, thereby, HP-MR image contrast. This allows for rapid acquisition of spatial metabolic maps within seconds of infusion. While technically, all downstream metabolites of HP 13C-pyruvate can be assessed, the conversion of HP 13C-pyruvate to HP 13C-lactate is the most relevant HP-MRI parameter to assess cancer metabolism. There are several other HP probes in preclinical research, including 13C urea, 13C fumarate, and 13C dehydroascorbate (DHA). The shortcoming of HP-MRI is the loss of hyperpolarization and associated signal decay that occurs within minutes after intravenous injection. DMI enables metabolic imaging for >1 hour after oral administration of deuterated (2H-) glucose, and assessment of the conversion of 2H-glucose to 2H-lactate. However, DMI is limited in the spatial resolution and sensitivity that can be achieved for a target organ. Both HP-MRI and DMI hold clinical potential for in vivo interrogation of tumor biology and early assessment of treatment response. As MRI scanners are far more abundant than PET scanners, HP-MRI and DMI – although presently still experimental– could potentially improve patient management and speed up treatment decisions.

Smarter biopsies

Precision oncology has increased the need for accurate tissue sampling (Figs. 1, 2). Advanced imaging has especially contributed to identifying specific sites for targeted biopsies. Tissue sampling is necessary to better understand the heterogeneity of cancer, measure temporal changes in expression profiles and somatic mutations, analyze immune cell infiltration as a response to treatment, perform spatial biology for biomarker discovery, and supply tissue aliquots to drug trial sponsors as part of their reporting requirements. While core biopsies (16–18G) have many advantages for tissue acquisition, they also have some downsides, including procedural complication risks in certain types of biopsies, long turnaround times, and a substantial fraction of “non-diagnostic specimens” in part because of conventional analyses that require large sample sizes. For all of these reasons, new, “smarter” types of biopsies are being explored to improve information content, reduce complication rates, and improve throughput18,19.

Fine-needle aspirates (FNA) are performed with 21–25G needles and typically yield cells rather than core tissue. FNA sampling has been firmly established for conventional cytopathology and flow cytometry analysis in the case of lymphoma. More recently, highly multiplexed analyses of scant specimens have been developed by using DNA origami20 and bioorthogonal chemistries21 for staining and which do not require the use of harsh chemicals that would destroy cells during cycling. Point-of-care analytical systems are being explored for multiplex sample processing, especially for resource-constrained locations and countries22.

Fine-needle biopsies (FNB) utilize small-gauge (20–22G) specialty needles that are designed to obtain microcores with preserved tissue architecture23. The microcores are commonly procured by endoscopic ultrasound, but fine-needle biopsies are also being adapted to percutaneous procedures.

Percutaneous fiberoptic sampling is yet another method that allows precision biopsy. A new development in the interventional community has been fiberoptic-guided biopsies (Spyglass) with forceps24. This emerging field is particularly suited for otherwise inaccessible locations, such as peripheral intraductal cholangiocarcinoma. A second and complementary development is the adaptation of this miniaturized cholangioscope to multiplexed fluorescence imaging using probes, an approach similar to fluorescence-guided surgery. These probes can be designed to “highlight” cancer features, which can be either diagnostic by themselves or biopsied for further analysis.

INTEGRATED DIAGNOSTICS AND THE COMPLEMENTARY ROLES OF LIQUID BIOPSIES AND IMAGING

Integrated diagnostics is an emerging field that involves the use of complementary imaging, laboratory biomarkers, pathology, and patient demographic data augmented with information technology. Integrated diagnostics will: i) offer greater diagnostic accuracy than single tests by identifying complementary biomarkers, ii) shorten the time from diagnosis to delivery of molecularly informed therapy, and iii) improve the longitudinal monitoring of outcomes through alternating use of redundant biomarkers25. Including diagnostic imaging in AI-based integrated diagnostic algorithms is essential for developing the strongest possible predictive and prognostic biomarkers to direct precision oncology.

Liquid biopsy26 refers to the sampling of body fluids such as peripheral blood for analysis of analytes that include cell-free DNA (cfDNA), circulating tumor DNA (ctDNA), circulating tumor cells (CTC), extracellular vesicles and proteins. Similarly to imaging, the field of ctDNA analysis is changing rapidly. Recent introductions of fragmentation analyses27, mutational signatures28 and analysis of repeat elements29 have made ctDNA analysis much more accurate including for early stage disease. It is generally believed that liquid biopsy complements advanced imaging and allows up or downstaging of intermediary imaging findings. However, future studies are required to compare methodologies head to head for specific cancers and clinical indications. Thus, AI-informed integrated diagnostics will likely play a growing role in the future. Applications for liquid biopsies in oncology, and their relationships to imaging, are described below.

Early detection.

Several large-scale clinical trials, such as those using the multi-cancer detection Galleri® test (ISRCTN91431511; based on cancer-specific DNA methylation patterns), reported high specificity (99–100%) but only moderate sensitivity30,31. Sensitivity depends heavily on the tumor stage – not all patients with localized early tumors will have sufficient quantities of circulating tumor DNA32. In one recent study, ctDNA of KRAS mutation alone could only diagnose 25% of stage 1 pancreatic cancer33. Similar results were also observed in a large cohort of gynecological, lung, or GI tract cancers (sensitivity for detecting stage 1 cancer: 24.2%)34. For certain cancers such as lung cancers, imaging tests can be superior to liquid biopsies. For example, a recent study on lung cancer screening showed the sensitivity of CT to be >80%, with a negative predictive value (NPV) of 97.7–100%, but a positive predictive value (PPV) of only 3.3–43.5%35.

Finding the primary.

In patients with metastatic disease at initial presentation, liquid protein biomarkers may be helpful in identifying the primary tumor, which can then be confirmed with anatomic or molecular imaging followed by image-guided biopsies.

Treatment monitoring.

Liquid biopsy and imaging biomarkers may complement in outcome prediction and prognostication, given that both types of biomarkers by themselves have demonstrated potential in different malignancies and treatment scenarios. To date, data on their integrated use are scarce; however, a recent study in follicular lymphoma reported 88% sensitivity and 100% specificity for 2-year progression-free survival for the combination of [18F]FDG-PET and ctDNA36. Some emerging studies indicate that liquid biopsies may be more sensitive than imaging in certain mutation-positive patients and will likely lead to the generation of new guidelines.

Gauging minimal residual disease (MRD) in solid tumors.

MRD is closely associated with disease recurrence, and ctDNA has been explored as a new biomarker. Emerging data suggests that post-operative ctDNA can be a strong prognostic marker of regression-free survival37. It remains to be studied how imaging and liquid biopsy will improve MRD analysis.

Image-guided therapeutics

Radiopharmaceutical therapy (RPT, “radiotheranostics”). RPT refers to the combined imaging and delivery of precision radiotherapeutics, where “what you treat is what you see” (Figs. 4 and 5). The growth of RPT in oncology has been exponential38,39 in part due to its clinical success, marked by improved outcomes, low off-target toxicities, and better quality-of-life data compared to alternative therapies. While different forms of RPT have been explored experimentally and clinically3841, clinical practice has seen a number of mainstream (FDA approved) applications. These have included i) systemic administration of small molecules (e.g., [223Ra]Ra-dichloride42, [131I]I-sodium iodide43, radioligands (e.g., [177Lu]Lu-PSMA-61744, [177Lu]Lu-DOTATATE45) or antibodies (e.g., [131I]I-tositumomab46); and ii) selective transarterial tumor embolization with 90Y and 166Ho microspheres.

Fig. 5: Therapeutic approaches involving radiotheranostics.

Fig. 5:

Therapeutic effects on cancer cells caused by DNA damage induced by either α-, β- or auger-emitting radionuclides can be enhanced via combination with drugs that either cause direct damage to DNA (such as chemotherapies) or inhibit DNA damage repair directly (such as PARP inhibitors) or through modulation of the associated signaling pathways (e.g., with novel androgen-deprivation therapies). Radiotheranostics can also target the tumor microenvironment (fibroblast activation protein) and kill stromal cells, which can indirectly lead to tumor regression. Bystander effects, owing to the use of β-emitters, on the DNA of cancer cells that do not express radiotheranostic target proteins can nonetheless lead to tumor cell death. Targeted radionuclide therapies might also induce antigen presentation following cancer cell death and, when combined with immune checkpoint inhibitors, lead to enhanced antitumor activity. DDR: DNA damage response38.

As the availability of isotopes for diagnostic and therapeutic purposes increases and the number of delivery platforms becomes more sophisticated, truly personalized treatment paradigms can be realized39. Further, many RPT approaches are being deployed in earlier stages of the disease, and ongoing trials are underway in which they are begun after diagnosis. One example is the NETTER-2 trial establishing [177Lu]Lu-DOTATATE as first line treatment in newly diagnosed patients with advanced grade 2 and grade 3, well-differentiated gastroenteropancreatic neuroendocrine tumors47. Another example is in prostate cancer, where treatments are transitioning from usual late-stage metastatic/advanced disease therapy to earlier-line treatment settings such as metastatic hormone-sensitive disease (PSMAddition) and even prior to prostatectomy (LuTectomy)48. There is also excitement about the concept of using RPT in combination with established systemic therapies, including targeted therapies and immunotherapy (e.g., NCT04343885, NCT05146973, NCT03874884, NCT05109728, and NCT03658447), as well as for accessing previously unexplored targets that were not suitable for traditional systemic therapies (Fig. 4).

The expansion of the field is not without its challenges49, including isotope production and supply50,51, workforce expertise, and regulations39,5254; however, efforts to address these issues have been initiated by governments, industry and major professional organizations, as well as in a current Lancet Oncology Commission which is exploring the global availability of theranostics and recommendations for improving patient access.

Transarterial radioembolization (TARE, also known as selective internal radiation therapy, SIRT) with Yttrium-90 (90Y) microspheres is a liver-directed therapy for primary and metastatic disease, for which indications have blossomed following FDA approval and efficacy trials55. In TARE, hepatic artery branches are accessed with microcatheters, injecting 90Y glass or polymer microspheres. These microspheres lodge in tumor-feeding arteries, and 90Y induces locoregional cellular damage. The current indications for TARE include primary treatment of hepatocellular carcinoma (HCC) in nonsurgical candidates, bridging to transplant in HCC, primary treatment of isolated oligometastatic liver lesions (radiation segmentectomy), radiation lobectomy to induce hypertrophy before resection, and palliation or delay of progression for advanced tumor burdens56. To improve dosimetry, planning angiography with the administration of 99mTc macroaggregated albumin is being performed to exclude patients with lung shunting or aberrant abdominal supplies. Cone-beam CT with intraarterial contrast material injection is often performed to exclude non-target perfusion and measure the perfused treatment volume. Bremsstrahlung SPECT/CT or 90Y PET can also be performed after TARE for dosimetry purposes and patient-centric future planning. More recently, 166Ho made its way into the clinic, providing additional therapeutic options complementary to peptide receptor radionuclide therapy (HEPAR Plus trial57.

Image-guided percutaneous ablation methods have transformed the approaches to many primary and metastatic tumors, especially in non-surgical patients. The tumor ablation methods include microwave, radiofrequency, and cryoablation (among others, such as electroporation, histotripsy, and chemical ablation)58. While the different methods have unique advantages and disadvantages, they generally rely on image-guided precision placement of applicators into cancerous lesions in the liver, kidneys, lung, bone, prostate, thyroid, and soft tissue. Following minutes of precision treatments, patients can be discharged the same day. There are usually short recovery periods, less bleeding, and more preservation of organ parenchyma, thus expanding future treatment options.

Image-guided precision drug delivery allows patient-specific administration of next-generation therapeutics (e.g., viral therapeutics, cell therapeutics, immune modulators) through catheters and needle-based approaches. This allows for high local drug concentrations while minimizing systemic therapies. Precision drug delivery may be of particular value to early drug developers in efforts to drive new applications.

Intraoperative Imaging

Intraoperative imaging with ultrasound, fluoroscopy, or MRI has long been used for conventional surgery (Fig. 6). In recent years, however, there has been a shift to precision surgery, where resections are individualized to patients. This potentially allows for improved resection accuracy, sparing essential structures, and faster convalescence. These trends and public demand have driven the growth of minimally invasive surgery, especially robotic surgery and image-guided surgery technologies. Most applications have been in prostate, breast, colorectal, lung cancer, and glioma resections. For example, the main challenge in radical prostatectomy is complete cancer excision with the preservation of continence and erectile function. Positive margins still occur in up to 35% of cases. In breast-conserving surgery, the primary goal is to prevent local recurrence with acceptable cosmetic outcomes. Rates of residual cancer following initially negative lumpectomy margins have been shown to exceed 40% in some studies59.

Fig. 6: Fiberoptic and intraoperative imaging.

Fig. 6:

Fluorescence-based cellular and molecular imaging is an up-and-coming modality, primarily employed for endoscopic and intraoperative imaging. A: results from fluorescent guided surgical detection of residual tumor in the resection cavity using the recently FDA-approved Lumicell system63,64. B: the use of bioorthogonal chemistry has enabled, in vivo, cyclic imaging for up to 12 different targets81.

Fluorescence-guided surgery (FGS). Near-infrared fluorescence imaging uses a combination of injectable fluorescent imaging agents with specialized detection systems to visualize cancers and their margins more accurately. The emergence of the field dates back nearly 25 years60, and a good many different agents and imaging systems have been developed since then61,62. Despite these efforts, it has been challenging to commercialize systems and then test their efficacy prospectively in large-scale studies. A recently FDA-approved system is Lumicell’s activatable fluorescent imaging agent (pegulicianine)63 combined with a handheld device64. In one recent prospective trial involving 406 patients (NCT03686215), the margin status was assessed with or without pegulicianine fluorescence-guided surgery (PFGS). In 27 of 357 patients undergoing surgery, PFGS for surgical margins removed tumors left behind after standard lumpectomy. PFGS prevented second surgeries in 9 of 62 patients with positive margins65

Alternative methods are being explored to enable intraoperative imaging. Early proof-of-principle studies primarily validate the emerging technologies in resection specimens, but the overall future goal is imaging of the resection cavity. The latest modality to be evaluated for assessment of margins is PET-CT66. Additional methods include Cherenkov, Raman and photoacoustic imaging67.

Artificial intelligence in Precision Imaging

Discussions on artificial intelligence (AI) have become central to precision medicine68. For the last several years, AI has been proposed to increase sensitivity in disease detection, enhance measurement reproducibility, reliably extract quantitative disease markers, recognize new patterns encoded within complex data, and serve as an inference engine after empirical “training” using real-world, large-scale data. Since its earliest days, AI has been integral to the imaging sciences69,70.

Most AI-driven imaging workflows focus on the back end of care management, including tumor detection, segmentation71, and staging, serial monitoring of tumor metrics, defining prognosis, radiation treatment planning72, and determination of tumor heterogeneity73. Vendors have rapidly adopted AI at the front end of imaging care to improve image quality, accelerate imaging times, calculate dosimetric profiles for theranostics, and predict whether a patient’s tumor has a sufficient amount of target to undergo targeted therapy.

Despite the tremendous promise noted above, the use of AI in precision imaging is not without certain bottlenecks and controversies. Among the chief bottlenecks is the lack of adequate amounts of data to train convolutional neural networks (CNNs). To decrease the need for manpower and tedious annotation of images in preparation for training, we would need to focus on AI unsupervised methods. However, that is not always possible, as available datasets are either too small or cannot provide reliable images due to non-standardized acquisition parameters74. That has led to the development of methods for creating synthetic images75, for example, through generative adversarial networks76, transfer learning and collaborative/federated learning77. Trustworthiness has become a mounting challenge to the use of AI, even with large-scale datasets. Algorithms can be biased toward one racial group or gender, produce unpredictably erroneous results, and make predictions that have no basis in physical reality. Most of the data presented in the literature to date are from retrospective studies. Prospective studies comparing AI performance to more traditional decision-making processes are needed to define its true potential.

Additional Challenges and Future Developments

While the advances in imaging technologies over the last decade have been nothing short of astounding and hold exceptional promise, several opportunities present themselves to facilitate more rapid progress in precision imaging for oncology:

  • The co-development of companion imaging diagnostics will ensure the success of next-generation patient-specific therapies. Unfortunately, developing and validating imaging probes is often an afterthought and still underfunded. Unlike therapeutic development, imaging agent development is mostly investigator-driven, relying on lengthy funding cycles and often delaying clinical introduction. With the recent exceptions for [68Ga]Ga-PSMA-11 (Illuccix®, 68Ga gozetotide; 8 years to FDA approval in 2020), [18F]F-PSMA (Pylarify®, 18F piflufolastat; 11 years to FDA approval in 2022) or [177Lu]Lu-PSMA (Pluvicto®, 177Lu vipivotide tetraxetan; 7 years to FDA approval in 202244) it is estimated that the average length of imaging approval is still ~13 years, too long to be practical for precision oncology needs. The development and validation of companion imaging diagnostics should occur in the preclinical stage. Many first-in-human imaging studies could be done rapidly as phase I trials to demonstrate safety and proof-of-concept. Currently, phase III trials are expensive and burdensome if industry is not involved, and regulatory approvals are often held to the same standard as for therapeutic drugs. Therefore, regulatory bodies should develop imaging-agent/modality-specific pathways so that approval can be fast-tracked and phase III trials will not require the large-scale cohorts associated with therapeutic drug trial design.

  • For precision oncology and imaging to be synergistic, there has to be continued dialogue and collaboration between different specialties. Except at specialized cancer centers, oncology and imaging/diagnostic departments often continue to operate differently for historical and financial reasons. While a primary oncologist typically follows a single patient longitudinally, different imaging physicians will perform and interpret scans during a patient’s journey (Fig. 1). Naturally, this can lead to discrepancies and inter-observer variability, complicating longitudinal assessments. It is hoped that AI and multidisciplinary conferences will present venues for minimizing discrepancies in longitudinal image analysis.

  • While imaging centers have been at the forefront of AI developments, tighter integration with digital pathology, molecular diagnostics, and AI in clinical oncology seems logical and well justified. Together, we must find ways to manage the growing IT costs and also invest in the future of integrated diagnostics.

  • Capital costs of imaging systems, infrastructure, and new therapies are considerable and have to be harmonized across academic medical centers. Confounding the issue is that reimbursements are decreasing for imaging, just as they are for almost all specialties. Despite the above challenges, there continues to be enormous enthusiasm for future developments and tighter integration of advanced imaging in clinical trials and care.

Summary

Recent advances in medicine and biotechnology have enabled more personalized cancer therapy approaches. However, all too frequently, only limited biomarker analyses are performed on tumors, often before treatment. Because of the dynamic nature of tumor evolution, combating therapy resistance is a game of catch-up requiring sophisticated tools to measure evolution. Here, we have attempted to summarize the extraordinary breadth of progress in developing diagnostic and therapeutic imaging approaches and how these can aid in precision oncology. Advanced imaging and image-guided treatments will continue to play essential roles in precision oncology and will improve cancer outcomes and survival.

Acknowledgments

We thank academic members of the International Society for Strategic Studies in Radiology (IS3R) for helpful discussions. We would also like to thank the following physicians for their contributions: Drs. Boris Hadaschik, Christopher Darr and Pedro Fragoso Costa Universitätsmedizin Essen, Germany for parts of Fig. 2.; Dr. Simon Cherry, University of California, Davis for Fig. 3; Drs. Helena Lanzafame, Rainer Hamacher, Wolfgang Fendler, Universitätsmedizin Essen, Germany for Fig. 4; Drs Cesar Castro, Herbert Kressel and Sanjeeva Kalva for critical review of the manuscript and Ada Muellner for editing.

Funding

This study was not funded by the US National Institutes of Health (NIH), and none of the authors are employed by NIH. There was no commercial sponsorship nor support for the study.

Declaration of interests

HH serves on the board the Board of Directors, IBA (Ion Beam Applications), the External Advisory Board of the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, the International Advisory Board of the University of Vienna, Austria, the Scientific Committee of the DKFZ (German Cancer Research Center), Germany, the Board of Trustees of the DKFZ; the Board of Directors of iCAD; the Advisory Board of The Lancet Oncology; receives stock options from iCAD. KH receives grants from Novartis and Sofie Biosciences and has consulted for Advanced Accelerator Applications, a Novartis company, Amgen, AstraZeneca, Bain Capital, Bayer, Boston Scientific, Convergent, Curium, Debiopharm, EcoR1, Fusion, GE Healthcare, Immedica, Isotopen Technologien München, Janssen, Merck, Molecular Partners, NVision, POINT Biopharma, Pfizer, Radiopharm Theranostics, Rhine Pharma, Siemens Healthineers, Sofie Biosciences, Telix, and Theragnostics, ymabs; has stock options in Sofie Biosciences, Pharma15, Vision, Convergent, Aktis Oncology, AdvanCell; is an advisory board member of Fusion, GE Healthcare; receives honoraria from Peerview and has received travel support from Jansen. JSL reports research support from Clarity Pharmaceuticals and Avid Radiopharmaceuticals; has acted as an advisor for Alpha-9 Theranostics Inc., Boxer, Clarity Pharmaceuticals, Earli Inc., Curie Therapeutics Inc., Evergreen Theragnostics, West Street Life Sciences, Inhibrx, Inc., Luminance Biosciences Inc., NexTech Venture LTD, Sanofi US Services Inc., Solve Therapeutics, Inc., Suba Therapeutics Inc., TPG Capital, Telix Pharmaceuticals LTD, pHLIP Inc., and Precirix; is a co-inventor on technologies licensed to Diaprost, Elucida Oncology, Theragnostics, CheMatech, Daiichi Sankyo and Samus Therapeutics; is the co-founder of pHLIP; holds equity in Summit Biomedical Imaging, Telix Pharmaceuticals, Clarity Pharmaceuticals, and Evergreen Theragnostics and is supported by National Institutes of Health R35 CA232130. MGP has consulted for CraniUS, UCLA Cancer Center, Ventyx, Einseca, ModeX; receives royalties from Lantheus Holdings, Novartis, Intuitive Surgical and Cyclotek; has 70 patents issued or filed related to imaging or informatics; stock options in D&D Pharmatech, PlenaryAI, Earli Inc and Immunosity. AMS reports trial funding from EMD Serono, ITM, Telix Pharmaceuticals, AVID Radiopharmaceuticals, Fusion Pharmaceuticals, and Cyclotek; research funding from Medimmune, AVID Radiopharmaceuticals, Adalta, Antengene, Humanigen, Telix Pharmaceuticals, and Theramyc; and payment for participation in advisory boards of Imagion and Immunos. RW has consulted for ModeRNA, Boston Scientific, Lumicell, Seer Biosciences, Earli Inc, and Accure Health. The other authors declared no conflicts of interest.

Footnotes

Search strategy and selection criteria

References for this Review were identified through searches of PubMed with the search terms “Imaging”, “AI”, “theranostics”, and “biopsy” from 2000 until April, 2024. Articles were also identified through searches of the authors’ own files. Only papers published in English or German were reviewed. The final reference list was generated on the basis of originality and relevance to the broad scope of this article.

References

  • 1.Daube-Witherspoon ME, Pantel AR, Pryma DA, Karp JS. Total-body PET: a new paradigm for molecular imaging. Br J Radiol 2022; 95:20220357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Cherry SR, Jones T, Karp JS, Qi J, Moses WW, Badawi RD. Total-Body PET: Maximizing Sensitivity to Create New Opportunities for Clinical Research and Patient Care. J Nucl Med 2018; 59:3–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Nadig V, Herrmann K, Mottaghy FM, Schulz V. Hybrid total-body pet scanners-current status and future perspectives. Eur J Nucl Med Mol Imaging 2022; 49:445–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Sjöholm T, Korenyushkin A, Gammelgård G et al. Whole body FDG PET/MR for progression free and overall survival prediction in patients with relapsed/refractory large B-cell lymphomas undergoing CAR T-cell therapy. Cancer Imaging 2022; 22:76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Sandgren K, Strandberg SN, Jonsson JH et al. Histopathology-validated lesion detection rates of clinically significant prostate cancer with mpMRI, [68 Ga]PSMA-11-PET and [11 C]Acetate-PET. Nucl Med Commun 2023; 44:997–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lombardi G, Spimpolo A, Berti S et al. PET/MR in recurrent glioblastoma patients treated with regorafenib: [18F]FET and DWI-ADC for response assessment and survival prediction. Br J Radiol 2022; 95:20211018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Haberkorn U, Mier W, Kopka K, Herold-Mende C, Altmann A, Babich J. Identification of Ligands and Translation to Clinical Applications. J Nucl Med 2017; 58:27S–33S. [DOI] [PubMed] [Google Scholar]
  • 8.Herscovitch P Regulatory Agencies and PET/CT Imaging in the Clinic. Curr Cardiol Rep 2022; 24:1361–1371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hofman MS, Lawrentschuk N, Francis RJ et al. Prostate-specific membrane antigen PET-CT in patients with high-risk prostate cancer before curative-intent surgery or radiotherapy (proPSMA): a prospective, randomised, multicentre study. Lancet 2020; 395:1208–1216. [DOI] [PubMed] [Google Scholar]
  • 10.Mori Y, Dendl K, Cardinale J, Kratochwil C, Giesel FL, Haberkorn U. FAPI PET: Fibroblast Activation Protein Inhibitor Use in Oncologic and Nononcologic Disease. Radiology 2023; 306:e220749. [DOI] [PubMed] [Google Scholar]
  • 11.Arroyo A, Lyashchenko SK, Lewis JS. Methods for the Production of Radiolabeled Bioagents for ImmunoPET. Methods Mol Biol 2024; 2729:117–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Tendler S, Dunphy MP, Agee M et al. First-in-human imaging with [89Zr]Zr-DFO-SC16.56 anti-DLL3 antibody in patients with high-grade neuroendocrine tumors of the lung and prostate. Lancet Oncology 2024; in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rudin CM, Reck M, Johnson ML et al. Emerging therapies targeting the delta-like ligand 3 (DLL3) in small cell lung cancer. J Hematol Oncol 2023; 16:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Yao J, Bergsland E, Aggarwal R et al. DLL3 as an Emerging Target for the Treatment of Neuroendocrine Neoplasms. Oncologist 2022; 27:940–951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Scher HI, Beer TM, Higano CS et al. Antitumour activity of MDV3100 in castration-resistant prostate cancer: a phase 1–2 study. Lancet 2010; 375:1437–1446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Boers J, Venema CM, de Vries EFJ et al. Serial [18F]-FDHT-PET to predict bicalutamide efficacy in patients with androgen receptor positive metastatic breast cancer. Eur J Cancer 2021; 144:151–161. [DOI] [PubMed] [Google Scholar]
  • 17.Kaggie JD, Khan AS, Matys T et al. Deuterium metabolic imaging and hyperpolarized 13C-MRI of the normal human brain at clinical field strength reveals differential cerebral metabolism. Neuroimage 2022; 257:119284. [DOI] [PubMed] [Google Scholar]
  • 18.Frenk NE, Spring L, Muzikansky A et al. High-Content Biopsies Facilitate Molecular Analyses and Do Not Increase Complication Rates in Patients With Advanced Solid Tumors. JCO Precis Oncol 2017; 1:1–9. [DOI] [PubMed] [Google Scholar]
  • 19.Oh J, Carlson JCT, Landeros C et al. Rapid Serial Immunoprofiling of the Tumor Immune Microenvironment by Fine Needle Sampling. Clin Cancer Res 2021; 27:4781–4793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ullal AV, Peterson V, Agasti SS et al. Cancer cell profiling by barcoding allows multiplexed protein analysis in fine-needle aspirates. Sci Transl Med 2014; 6:219ra9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ko J, Wilkovitsch M, Oh J et al. Spatiotemporal multiplexed immunofluorescence imaging of living cells and tissues with bioorthogonal cycling of fluorescent probes. Nat Biotechnol 2022; 40:1654–1662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Peterson HM, Chin LK, Iwamoto Y et al. Integrated Analytical System for Clinical Single-Cell Analysis. Adv Sci (Weinh) 2022; 9:e2200415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kovacevic B, Toxværd A, Klausen P et al. Tissue amount and diagnostic yield of a novel franseen EUS-FNB and a standard EUS-FNA needle-A randomized controlled study in solid pancreatic lesions. Endosc Ultrasound 2023; 12:319–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Baars JE, Keegan M, Bonnichsen MH et al. The ideal technique for processing SpyBite tissue specimens: a prospective, single-blinded, pilot-study of histology and cytology techniques. Endosc Int Open 2019; 7:E1241–E1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Jee J, Lebow ES, Yeh R et al. Overall survival with circulating tumor DNA-guided therapy in advanced non-small-cell lung cancer. Nat Med 2022; 28:2353–2363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mattox AK, Bettegowda C, Zhou S, Papadopoulos N, Kinzler KW, Vogelstein B. Applications of liquid biopsies for cancer. Sci Transl Med 2019; 11 [DOI] [PubMed] [Google Scholar]
  • 27.Cristiano S, Leal A, Phallen J et al. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature 2019; 570:385–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bruhm DC, Mathios D, Foda ZH et al. Single-molecule genome-wide mutation profiles of cell-free DNA for non-invasive detection of cancer. Nat Genet 2023; 55:1301–1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Annapragada AV, Niknafs N, White JR et al. Genome-wide repeat landscapes in cancer and cell-free DNA. Sci Transl Med 2024; 16:eadj9283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Schrag D, Beer TM, McDonnell CH et al. Blood-based tests for multicancer early detection (PATHFINDER): a prospective cohort study. Lancet 2023; 402:1251–1260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Klein EA, Richards D, Cohn A et al. Clinical validation of a targeted methylation-based multi-cancer early detection test using an independent validation set. Ann Oncol 2021; 32:1167–1177. [DOI] [PubMed] [Google Scholar]
  • 32.Lee B, Lipton L, Cohen J et al. Circulating tumor DNA as a potential marker of adjuvant chemotherapy benefit following surgery for localized pancreatic cancer. Ann Oncol 2019; 30:1472–1478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Cohen JD, Javed AA, Thoburn C et al. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc Natl Acad Sci U S A 2017; 114:10202–10207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Nicholson BD, Oke J, Virdee PS et al. Multi-cancer early detection test in symptomatic patients referred for cancer investigation in England and Wales (SYMPLIFY): a large-scale, observational cohort study. Lancet Oncol 2023; 24:733–743. [DOI] [PubMed] [Google Scholar]
  • 35.Jonas DE, Reuland DS, Reddy SM et al. Screening for Lung Cancer With Low-Dose Computed Tomography: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA 2021; 325:971–987. [DOI] [PubMed] [Google Scholar]
  • 36.Jiménez-Ubieto A, Poza M, Martin-Muñoz A et al. Real-life disease monitoring in follicular lymphoma patients using liquid biopsy ultra-deep sequencing and PET/CT. Leukemia 2023; 37:659–669. [DOI] [PubMed] [Google Scholar]
  • 37.Chidharla A, Rapoport E, Agarwal K et al. Circulating Tumor DNA as a Minimal Residual Disease Assessment and Recurrence Risk in Patients Undergoing Curative-Intent Resection with or without Adjuvant Chemotherapy in Colorectal Cancer: A Systematic Review and Meta-Analysis. Int J Mol Sci 2023; 24:10230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bodei L, Herrmann K, Schöder H, Scott AM, Lewis JS. Radiotheranostics in oncology: current challenges and emerging opportunities. Nat Rev Clin Oncol 2022; 19:534–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lapi SE, Scott PJH, Scott AM et al. Recent Advances and Impending Challenges for the Radiopharmaceutical Sciences in Oncology. Lancet Oncol 2024; in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Herrmann K, Schwaiger M, Lewis JS et al. Radiotheranostics: a roadmap for future development. Lancet Oncol 2020; 21:e146–e156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Pomykala KL, Hadaschik BA, Sartor O et al. Next generation radiotheranostics promoting precision medicine. Ann Oncol 2023; 34:507–519. [DOI] [PubMed] [Google Scholar]
  • 42.Parker C, Nilsson S, Heinrich D et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med 2013; 369:213–223. [DOI] [PubMed] [Google Scholar]
  • 43.de Keizer B, Koppeschaar HP, Zelissen PM et al. Efficacy of high therapeutic doses of iodine-131 in patients with differentiated thyroid cancer and detectable serum thyroglobulin. Eur J Nucl Med 2001; 28:198–202. [DOI] [PubMed] [Google Scholar]
  • 44.Sartor O, de Bono J, Chi KN et al. Lutetium-177-PSMA-617 for Metastatic Castration-Resistant Prostate Cancer. N Engl J Med 2021; 385:1091–1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Strosberg JR, Caplin ME, Kunz PL et al. 177Lu-Dotatate plus long-acting octreotide versus high-dose long-acting octreotide in patients with midgut neuroendocrine tumours (NETTER-1): final overall survival and long-term safety results from an open-label, randomised, controlled, phase 3 trial. Lancet Oncol 2021; 22:1752–1763. [DOI] [PubMed] [Google Scholar]
  • 46.Davies AJ, Rohatiner AZ, Howell S et al. Tositumomab and iodine I 131 tositumomab for recurrent indolent and transformed B-cell non-Hodgkin’s lymphoma. J Clin Oncol 2004; 22:1469–1479. [DOI] [PubMed] [Google Scholar]
  • 47.Singh S, Halperin DM, Myrehaug S et al. [177Lu]Lu-DOTA-TATE in newly diagnosed patients with advanced grade 2 and grade 3, well-differentiated gastroenteropancreatic neuroendocrine tumors: Primary analysis of the phase 3 randomized NETTER-2 study. Journal of Clinical Oncology 2024; 42:LBA588–LBA588. [Google Scholar]
  • 48.Eapen RS, Buteau JP, Jackson P et al. Administering [177Lu]Lu-PSMA-617 Prior to Radical Prostatectomy in Men with High-risk Localised Prostate Cancer (LuTectomy): A Single-centre, Single-arm, Phase 1/2 Study. Eur Urol 2024; 85:217–226. [DOI] [PubMed] [Google Scholar]
  • 49.Hricak H, Abdel-Wahab M, Atun R et al. Medical imaging and nuclear medicine: a Lancet Oncology Commission. Lancet Oncol 2021; 22:e136–e172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Agency OECDANE. The Supply of Medical Isotopes: An Economic Diagnosis and Possible Solutions. 2019;
  • 51.Goethals PE, Zimmermann R. Marketed Radiopharmaceuticals. MEDraysintell Nuclear Medicine World Market Report & Directory 2023; [Google Scholar]
  • 52.Korde A, Patt M, Selivanova SV et al. Position paper to facilitate patient access to radiopharmaceuticals: considerations for a suitable pharmaceutical regulatory framework. EJNMMI Radiopharm Chem 2024; 9:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Giammarile F, Paez D, Zimmermann R et al. Production and Regulatory Issues for Theranostics. Lancet Oncol 2024; in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Scott AM, Zeglis BM, Lapi SE et al. Trends in Nuclear Medicine and the Radiopharmaceutical Sciences in Oncology: Workforce Challenges and Training in the Age of Theranostics. Lancet Oncol 2024; in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Salem R, Johnson GE, Kim E et al. Yttrium-90 Radioembolization for the Treatment of Solitary, Unresectable HCC: The LEGACY Study. Hepatology 2021; 74:2342–2352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Saini A, Wallace A, Alzubaidi S et al. History and Evolution of Yttrium-90 Radioembolization for Hepatocellular Carcinoma. Journal of Clinical Medicine 2019; 8:55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Braat AJAT, Bruijnen RCG, van Rooij R et al. Additional holmium-166 radioembolisation after lutetium-177-dotatate in patients with neuroendocrine tumour liver metastases (HEPAR PLuS): a single-centre, single-arm, open-label, phase 2 study. Lancet Oncol 2020; 21:561–570. [DOI] [PubMed] [Google Scholar]
  • 58.Mansur A, Garg T, Shrigiriwar A et al. Image-Guided Percutaneous Ablation for Primary and Metastatic Tumors. Diagnostics (Basel) 2022; 12:1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Scopa CD, Aroukatos P, Tsamandas AC, Aletra C. Evaluation of margin status in lumpectomy specimens and residual breast carcinoma. Breast J 2006; 12:150–153. [DOI] [PubMed] [Google Scholar]
  • 60.Weissleder R, Tung CH, Mahmood U, Bogdanov A. In vivo imaging of tumors with protease-activated near-infrared fluorescent probes. Nat Biotechnol 1999; 17:375–378. [DOI] [PubMed] [Google Scholar]
  • 61.Lauwerends LJ, van Driel PBAA, Baatenburg de Jong RJ et al. Real-time fluorescence imaging in intraoperative decision making for cancer surgery. Lancet Oncol 2021; 22:e186–e195. [DOI] [PubMed] [Google Scholar]
  • 62.Mieog JSD, Achterberg FB, Zlitni A et al. Fundamentals and developments in fluorescence-guided cancer surgery. Nat Rev Clin Oncol 2022; 19:9–22. [DOI] [PubMed] [Google Scholar]
  • 63.Whitley MJ, Cardona DM, Lazarides AL et al. A mouse-human phase 1 co-clinical trial of a protease-activated fluorescent probe for imaging cancer. Science Translational Medicine 2016; 8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hwang ES, Beitsch P, Blumencranz P et al. Clinical Impact of Intraoperative Margin Assessment in Breast-Conserving Surgery With a Novel Pegulicianine Fluorescence–Guided System. A Nonrandomized Controlled Trial. JAMA Surgery 2022; 157:573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Smith BL, Hunt KK, Carr D et al. Intraoperative Fluorescence Guidance for Breast Cancer Lumpectomy Surgery. NEJM Evidence 2023; EVIDoa2200333. [DOI] [PubMed] [Google Scholar]
  • 66.Darr C, Costa PF, Kahl T et al. Intraoperative Molecular Positron Emission Tomography Imaging for Intraoperative Assessment of Radical Prostatectomy Specimens. Eur Urol Open Sci 2023; 54:28–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Andreou C, Weissleder R, Kircher MF. Multiplexed imaging in oncology. Nat Biomed Eng 2022; 6:527–540. [DOI] [PubMed] [Google Scholar]
  • 68.Bhinder B, Gilvary C, Madhukar NS, Elemento O. Artificial Intelligence in Cancer Research and Precision Medicine. Cancer Discov 2021; 11:900–915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Rajpurkar P, Lungren MP. The Current and Future State of AI Interpretation of Medical Images. N Engl J Med 2023; 388:1981–1990. [DOI] [PubMed] [Google Scholar]
  • 70.Mikhael PG, Wohlwend J, Yala A et al. Sybil: A Validated Deep Learning Model to Predict Future Lung Cancer Risk From a Single Low-Dose Chest Computed Tomography. J Clin Oncol 2023; 41:2191–2200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Häggström I, Leithner D, Alvén J et al. Deep learning for [18F]fluorodeoxyglucose-PET-CT classification in patients with lymphoma: a dual-centre retrospective analysis. Lancet Digit Health 2024; 6:e114–e125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Palmer JD, Perlow HK, Lehrer EJ, Wardak Z, Soliman H. Novel radiotherapeutic strategies in the management of brain metastases: Challenging the dogma. Neuro Oncol 2024; 26:S46–S55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Acosta PH, Panwar V, Jarmale V et al. Intratumoral Resolution of Driver Gene Mutation Heterogeneity in Renal Cancer Using Deep Learning. Cancer Res 2022; 82:2792–2806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Whybra P, Zwanenburg A, Andrearczyk V et al. The Image Biomarker Standardization Initiative: Standardized Convolutional Filters for Reproducible Radiomics and Enhanced Clinical Insights. Radiology 2024; 310:e231319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Takita H, Matsumoto T, Tatekawa H et al. AI-based Virtual Synthesis of Methionine PET from Contrast-enhanced MRI: Development and External Validation Study. Radiology 2023; 308:e223016. [DOI] [PubMed] [Google Scholar]
  • 76.Hellwig D, Hellwig NC, Boehner S, Fuchs T, Fischer R, Schmidt D. Artificial Intelligence and Deep Learning for Advancing PET Image Reconstruction: State-of-the-Art and Future Directions. Nuklearmedizin 2023; 62:334–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Yang X, Huang K, Yang D, Zhao W, Zhou X. Biomedical Big Data Technologies, Applications, and Challenges for Precision Medicine: A Review. Glob Chall 2024; 8:2300163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Li EJ, López JE, Spencer BA et al. Total-Body Perfusion Imaging with [11C]-Butanol. J Nucl Med 2023; 64:1831–1838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cherry SR, Diekmann J, Bengel FM. Total-Body Positron Emission Tomography: Adding New Perspectives to Cardiovascular Research. JACC Cardiovasc Imaging 2023; 16:1335–1347. [DOI] [PubMed] [Google Scholar]
  • 80.Omidvari N, Jones T, Price PM et al. First-in-human immunoPET imaging of COVID-19 convalescent patients using dynamic total-body PET and a CD8-targeted minibody. Sci Adv 2023; 9:eadh7968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ko J, Lucas K, Kohler R et al. In Vivo Click Chemistry Enables Multiplexed Intravital Microscopy. Adv Sci (Weinh) 2022; 9:e2200064. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES