4,264
Views
60
CrossRef citations to date
0
Altmetric
Review

Recent advances in nuclear and hybrid detection modalities for image-guided surgery

, , , , &
Pages 711-734 | Received 19 Apr 2019, Accepted 08 Jul 2019, Published online: 26 Jul 2019

ABSTRACT

Introduction: Radioguided surgery is an ever-evolving part of nuclear medicine. In fact, this nuclear medicine sub-discipline actively bridges non-invasive molecular imaging with surgical care. Next to relying on the availability of radio- and bimodal-tracers, the success of radioguided surgery is for a large part dependent on the imaging modalities and imaging concepts available for the surgical setting. With this review, we have aimed to provide a comprehensive update of the most recent advances in the field.

Areas covered: We have made an attempt to cover all aspects of radioguided surgery: 1) the use of radioisotopes that emit γ, β+, and/or β radiation, 2) hardware developments ranging from probes to 2D cameras and even the use of advanced 3D interventional imaging solutions, and 3) multiplexing solutions such as dual-isotope detection or combined radionuclear and optical detection.

Expert opinion: Technical refinements in the field of radioguided surgery should continue to focus on supporting its implementation in the increasingly complex minimally invasive surgical setting, e.g. by accommodating robot-assisted laparoscopic surgery. In addition, hybrid concepts that integrate the use of radioisotopes with other image-guided surgery modalities such as fluorescence or ultrasound are likely to expand in the future.

1. Introduction

Image-guided surgery is a key component in the realization of precision surgery, especially through the ability to intraoperatively identify disease based on its molecular properties. Radioguided surgery is one of the earliest and still most used forms of interventional molecular imaging. By intraoperatively exploring the use of radiopharmaceuticals that are embedded within the field of nuclear medicine, guidance can be facilitated towards the location and extent of the disease [1].

The concept of radioguided surgery originated about 6 decades ago. The first radiation detection probe system was evaluated by Selverstone et al. who reported in 1949 on the use of a handheld gaseous ionization detector (i.e. a Geiger-Müller tube) to localize suspected brain tumors that contained β emitting 32P isotope [Citation2]. In 1956, Harris et al. reported on the first use of probe based 131I isotope detection in a patient who underwent thyroidectomy [Citation3]. From that moment onward, applications and new innovations for radioguided surgery have evolved substantially and many studies have reported on the introduction of new detection modalities, radiopharmaceuticals and clinical applications [Citation4].

To date, radioguided surgery has been successfully applied in a plurality of clinical indications, mostly oncological (see ). When examining this list, one soon realizes that the indications are highly dependent on the radiopharmaceutical used. In some cases, for example, in sentinel node imaging, radiopharmaceuticals are used specifically dedicated to surgical guidance. Here low dose to the patient and surgical staff and compatibility with scintigraphy or SPECT/CT are considered selection criteria. However, there are also multiple examples where radioguidance procedures can make use of radiopharmaceuticals that were originally designed to have a role in diagnostic PET/CT such as 18F-FDG. This ability to build on the well-established discipline of nuclear medicine and the plurality of clinical radiopharmaceuticals that have come available through decades of radiochemical developments occurring across the globe, drives the field of radioguided surgery and gives it a unique edge over other image-guided surgery approaches.

Table 1. Clinical application of nuclear detection modalities in radioguided surgery. (In vivo human use only.)

Next to its reliance on the availability of specific radiopharmaceuticals, the implementation of radioguided surgery is highly dependent on the availability of dedicated (intraoperative) tracing and imaging modalities. In the current review, we evaluate advances made in the engineering of detection modalities that have been specifically designed for the purpose of radioguided surgery. In doing so, we have made an attempt at placing new innovations within the perspective of existing technologies and clinical applications.

2. Overview radioguidance modalities

2.1. 1D nuclear detection probes

Handheld nuclear detection probes are 1D detector modalities designed to measure the number of incident γ photons or β particles within a small FOV and a specific time frame (see ). When small in size, such probes can be used to trace radioactivity even in small confined surgical spaces. Typically, audible and numerical feedback, provided in counts/s, describe the intensity of the radioactive signal that was measured. As a result, these devices have proven to be useful tools for the intraoperative localization of tissues with radiopharmaceutical uptake (e.g. cancerous tissue). Surgical nuclear detection probes are developed for either: (1) low-to-mid energy γ ray detection (≤400 keV), often simply called ‘gamma probes’, (2) high-energy γ ray detection (>400 keV), often called ‘high-energy gamma probes’, (3) β+ particle detection (i.e. positively charged electrons or positrons), often called ‘beta+ probes’, and (4) β particle detection (i.e. negatively charged electrons or negatrons), often called ‘beta- probes’.

Figure 1. 1D nuclear detection probes. (a) Open surgery low-to-mid energy gamma probe, with example during ROLL procedure with ICG-99mTc-nanocolloid for recurrent melanoma (from [Citation219]). (b) Laparoscopic surgery low-to-mid energy gamma probe, with example during SN procedure with ICG-99mTc-nanocolloid for prostate cancer. (c) DROP-IN gamma probe for robot-assisted laparoscopic surgery, for example, during lymphatic salvage procedures in recurrent prostate cancer using 99mTc-PSMA. (d) High energy gamma probe for open surgery, with example in head and neck squamous cell carcinoma showing 18F-FDG avid lymph node with metastases (from [Citation127]). (e) Open surgery beta- probe, with example of ex vivo tumor with 90Y-DOTATOC in meningioma (from [Citation144]).

Figure 1. 1D nuclear detection probes. (a) Open surgery low-to-mid energy gamma probe, with example during ROLL procedure with ICG-99mTc-nanocolloid for recurrent melanoma (from [Citation219]). (b) Laparoscopic surgery low-to-mid energy gamma probe, with example during SN procedure with ICG-99mTc-nanocolloid for prostate cancer. (c) DROP-IN gamma probe for robot-assisted laparoscopic surgery, for example, during lymphatic salvage procedures in recurrent prostate cancer using 99mTc-PSMA. (d) High energy gamma probe for open surgery, with example in head and neck squamous cell carcinoma showing 18F-FDG avid lymph node with metastases (from [Citation127]). (e) Open surgery beta- probe, with example of ex vivo tumor with 90Y-DOTATOC in meningioma (from [Citation144]).

2.1.1. Gamma (γ) probes (≤400 kev)

Typical application of low-to-mid energy γ probes is tailored towards the use of ‘SPECT isotopes’ (see ) and form factors are available for open, laparoscopic and even robot-assisted laparoscopic surgery (see )) [Citation99]. Compared to β emissions, γ rays have a high penetration depth of >10 cm. Today the γ probe is one of the most widely applied modalities for image-guided surgery. As can be seen from , the most common surgical applications are: sentinel node (SN) procedures using 99mTc-labelled radiocolloids (e.g. melanoma, head and neck oncology, breast cancer, penile cancer, prostate cancer), radioguided occult lesion localization (ROLL) procedures using 99mTc-labelled macro-aggregates (e.g. breast cancer, lung cancer, thyroid cancer) and radioguided seed localization (RSL) procedures using 125I-seeds (e.g. breast cancer) [Citation100Citation103]. Building on many proof-of-concept studies, receptor targeted tumor resections are also increasingly being performed with the use of γ probes (e.g. prostate specific membrane antigen (99mTc or 111In)) [Citation104]. Below, we provide a short summary of the working principles for this well-established technology. However, a more detailed description can be found in more topic-specific reviews such as those of Heller et al. or Povoski et al. [Citation105,Citation106].

Table 2. Characteristics of radionuclides interesting for radioguided surgery.

2.1.1.1. Background and recent advances

Two main detector setups are in use for γ probes: (1) a scintillation detector, which consists of a scintillation crystal coupled (directly or via optical fiber [Citation107]) to a photomultiplier tube (PMT) or photodiode, or (2) an ionization detector, which consists of a semiconductor crystal structure. Next to having real-time sample times (typically <1 s), γ probes have been shown to allow for the identification of lesions as small as 2 mm [Citation108]. To allow for spatial resolution, some form of collimation is needed around the detector setup to only allow radiation originating from the front-side of the γ probe to be detected. To achieve collimation, shielding of the detector with a certain thickness of a high-atom-number material is required. Lead and tungsten, or some sort of alloy of these materials, are the most generally used materials in collimators [Citation106]. Given the relative stopping powers, however, gold and platinum might even be better alternatives; relative stopping powers of 1 (lead), 1.2 (tungsten (8% copper)), 1.6 (gold) and 1.7 (platinum (iridium alloy)) [Citation109]. The collimator material also reflects on the size and weight of the collimator, where a typical collimator diameter for low-energy γ probes ranges between 10 and 12 mm. Even with the collimator in place, lesions with low radiopharmaceutical uptake can still be hard to distinguish when located in close proximity to high-uptake background tissues, typically denoted as the ‘shine-through’ problem. To this end, some groups have experimented with blocking background radiation (e.g. injection site) with the intraoperative use of additional lead-shielding [Citation110,Citation111].

In the evolution of surgical techniques, more and more minimally invasive alternatives are being pursued, of which (robot-assisted) laparoscopic surgery is a prime example [Citation112]. To enable the use of radioguidance during laparoscopic surgery, laparoscopic versions of the low-to-mid energy γ probe have been introduced. From an engineering perspective, this basically means the shaft of an open surgery probe has to be elongating so that it can be used through a trocar [Citation113]. Unfortunately, with the trocar functioning as fixed rotation point, the maneuverability is drastically restricted, ultimately limiting the accuracy of lesion localization in anatomically difficult to reach locations [Citation114,Citation115]. In case of shine-through issues, this restricted maneuverability can especially be limiting when (low-uptake) lesions are located near high-uptake background tissues [Citation116Citation118]. To help focus the probe FOV, alternative laparoscopic γ probe designs have incorporated a detection window at, e.g. 45° or 90° angles [Citation119]. Because the angle of the detector would accommodate detection in specific anatomical locations, this could mean that different probe versions would be needed during a single procedure. When robot-assisted procedures are performed, next to the reduction in rotational freedom, the operating surgeon is also deprived of his/her ability to guide probe placement: based on verbal feedback provided by the surgeon, the bedside assistant takes care of the probe. Obviously, such placement issues become increasingly complicated when laparoscopic probes are used with angled detectors. To provide a solution for the challenges encountered during traditional laparoscopic radioguided surgery, and to optimally facilitate robot-assisted procedures, a small-sized tethered DROP-IN γ probe concept was introduced in 2016 (see )) [Citation99]. This probe, which essentially has a flexible shaft, was designed to allow the surgeon to independently manipulate the probe position using the surgical instruments (e.g. the wristed PROGRASP tool of the da Vinci robot). Gripping was designed so that it facilitates the identification of lesions in areas with a high background activity. Following technological evaluation in pigs [Citation99], it was successfully translated to in-human use (n = 12 patients) during robot-assisted prostate cancer SN procedures (with ICG-99mTc-nanocolloid [Citation120]) and robot-assisted salvage resection of lymphatic metastases in recurrent prostate cancer (with 99mTc-PSMA I&S [Citation121]).

2.1.2. High-energy gamma (γ) probes (>400 kev)

High-energy γ probes are generally designed to detect the indirect 511 keV annihilation γ-emissions of PET isotopes (see ), facilitating the surgical translation of diagnostic PET radiopharmaceuticals (see )). Therefore, they are sometimes also called ‘PET probes’. From a technical point, these probes are very similar to traditional low-energy γ probes, but employ different materials and designs to facilitate the detection and collimation of high-energy γ rays (i.e. >400 keV). As seen from , the most common surgical application of these probes is during open surgical localization of 18F-FDG avid lesions (e.g. colorectal cancer, breast cancer, lymphoma, ovarian cancer) [Citation122]. Topic-specific reviews are provided by Daghighian et al. and Povoski et al. [Citation123,Citation124].

2.1.2.1. Background and recent advances

Since ionization detectors typically are less sensitive for high-energy γ rays, scintillation-based detectors appear to be the preferred choice for high-energy γ probes [Citation106]. In general, to provide enough stopping power for these γ rays, scintillation crystals are longer than those used in low-to-mid energy γ probes (typically >2 cm [Citation124] and <1 cm [Citation99], respectively).

Largely dependent on the probe collimator, the smallest lesion size as detected with a high-energy γ probe, was reported to be 5 mm [Citation124]. High-energy γ probe collimators comprise of the same materials as chosen for low-to-mid energy γ probes, but since the 511 keV γ rays require more stopping power, the collimation surrounding the detector has to be substantially thicker. This also results in a substantially enlarged diameter of the end products: typically a probe-head diameter between 25–35 mm [Citation124,Citation125]. This also negatively influences the weight, where a high-energy γ probe is generally much heavier than a low-to-mid energy γ probe [Citation126Citation129]. Therefore, both probe diameter and weight restrict the intraoperative utility of such probes [Citation130]. Since minimal invasive laparoscopic surgery is slowly moving towards smaller trocar diameters (<12 mm), the typical 25–35 mm high-energy γ probe is simply not compatible. Still, a 15 mm diameter laparoscopic high-energy γ probe is reported in literature: a single case study (ovarian cancer) showed its success in 2005 [Citation131], however, follow-up hasn’t been reported since and further preliminary studies (colorectal and lung cancer) suggest that collimation in this small design is inadequate for successful use [Citation124].

Further refinements on intraoperative tracing of 511 keV emissions will most likely focus towards the design of innovative detector-collimator designs that allow for the engineering of more user-friendly probes [Citation123]. One innovative detector design, already applied in-human trials, comprises an arrangement of multiple detectors, able to perform electronical collimation of the γ signal without the need of a heavy collimator [Citation126,Citation128,Citation130,Citation132]. Unfortunately, this multi-detector setup still renders the device rather big (~30 mm diameter probe-head). It has been suggested that use of K-alpha x-ray fluorescence could be used to fabricate much lighter, and possibly smaller, detectors [Citation133,Citation134].

2.1.3. Beta+ (β+) and beta- (β-) probes

Instead of detecting γ-rays, β particles (including β+ and β), can be detected using β probes (see )). Unlike γ-rays, such emissions have a very limited tissue penetration of a few mm rendering this a superficial detection technology [Citation124,Citation135]. The upside of this limitation is that β tracing is less likely to suffer from radiation ‘shine-through’ (see above), originating from high-uptake background tissues. Hence, the technology could help realize detailed examination of the resection planes with respect to clear tumor margins [Citation136,Citation137] and could potentially support lesion detection in high background areas [Citation130]. Although most dedicated β probe systems can detect both β+ and β particles, there is a clear distinction; the systems are focused on isotopes with different emission characteristics. Similar to high-energy γ probes, β+ probes facilitate the intraoperative identification of PET-radiopharmaceuticals. Instead of detecting the secondary annihilation γ-rays, rather they directly detect the β+ particles emitted by these PET isotopes (see ). Apart from ex vivo evaluation in breast cancer [Citation124], melanoma [Citation138] and cervical cancer [Citation139], small-sized in vivo evaluations have been reported using 18F-FDG (colorectal cancer, melanoma, adenocarcinoma, thyroid cancer and breast cancer [Citation140Citation255]). Studies have also been performed using 124I-Hu-A33 (colorectal cancer [Citation142]) and 124I-cG250 (renal cancer [Citation142]) (see ).

Radioguidance based on β detection is based on the use of isotopes that have therapeutic properties (see ). With this emission type, the high background of 511 keV annihilation γ rays are avoided. Although the use of β emitting isotopes in radioguided surgery was already explored in 1949 (32P in brain tumor surgery) [Citation2], instrumentation was not sensitive for routine implementation [Citation136]. With the advance of modern technology, there has been a renewed interest for this surgical methodology. This has resulted in the first ex vivo experiments of modern β radioguided surgery using 90Y-DOTATOC in meningioma [Citation143]. Topic-specific literature is provided by Heller et al., Daghighian et al. and Camillocci et al. [Citation106,Citation124,Citation136].

2.1.3.1. Background and recent advances

The most widely applied detector composition for β probes comprises a scintillation setup. Because of the short-range attenuation of β particles, minimal collimation is needed and probe designs can be relatively small and lightweight. The materials that are often applied are stainless steel [Citation144], PVC [Citation136] and ABS [Citation143]. It should be noted that the sample times of β probes depend on the energy of the β particle, but values are often not clearly stated with respect to in vivo measurements.

Although the β probe plastic scintillators are relatively insensitive for γ rays, γ background signals can still render β tracing problematic. This is because both β+ and β particles can induce additional γ ray backgrounds within the human tissue. The first form is bremsstrahlung, produced by both β+ and β particles as they are decelerated within the tissue (emission probability of only 0.1%) [Citation136]. According to Russomando et al., using a p-terphenyl based detector, the sensitivity to such bremsstrahlung can be reduced to a negligible <1% [Citation145]. Contrary to pure β emitting isotopes, β+ emitting isotopes always come with high-energy γ rays due to the positron annihilation process. This high background of γ rays has been shown to be especially problematic when tracing low-energy β+ emissions such as that of 18F (635 keV) [Citation256]. To solve this issue, most β+ detection probes implement a dual-detector design: a first detector for both β and γ radiation and a second detector for γ radiation only [Citation124]. These detectors can either be placed next to or even behind each other. In this design the ‘γ only’ detector is shielded from β radiation by the first detector or some form of β collimation [Citation124,Citation144,Citation146]. By correcting the ‘combined β and γ signal’ (first detector) with the ‘γ only signal’ (second detector), a background rejection can be implemented to drastically improve the signal-to-noise ratio. Since background subtraction is not needed for pure β isotopes, these probes do not feature a dual-detector setup.

Current β+ and β probes have mainly focused towards open surgical applications. However, a technical description of a laparoscopic β+ probe version [Citation124] and a single case study in lung cancer using 18F-FDG have been reported [Citation147]. Interestingly, due to the small sizes possible, β+ detectors have also been suggested for intravascular detection of 18F-FDG avid atheroma [Citation148]. Next to experiments on laparoscopic versions of these probe technologies, future developments will most likely focus towards the realization of the first in vivo evaluations of the modern β probes.

2.2. 2D portable nuclear detection cameras

Nuclear detection cameras combine the output of a set of 1D detectors to form a 2D image of the radiopharmaceutical distribution within a specific FOV (see ). With that, they provide a logical evolution of the above-mentioned detection probe concepts. The image output of portable cameras can be considered to be more intuitive than the numerical and acoustic readout of probes. Furthermore, it may reduce localization time by allowing the mapping of a larger field of view. By using longer exposure times, the statistical noise can be reduced, but this comes at a cost for the ability to provide real-time feedback. Although full 3D insight is missing, the possibility to make an image from different directions can already help increase the diagnostic accuracy when lesions are located relatively deep within the anatomy or multiple lesions are overlapping. Unfortunately, the size of the bulkier detector can limit intraoperative maneuverability and mentally connecting the images displayed on a screen with the actual patient can be a demanding task. Furthermore, probe detection modalities are often found to be more sensitive than camera detection modalities [Citation106]. Providing a map for the radiopharmaceutical distribution within a small surgical area (e.g. SN mapping), portable nuclear detection cameras have proven to be a useful tool, with clinical systems available for low-to-mid energy γ ray detection and experimental systems available for β+ particle detection.

Figure 2. 2D portable nuclear detection cameras. (a–b) Portable gamma camera imaging during SN procedure for head and neck melanoma with ICG-99mTc-nanocolloid (from [Citation253Citation254]). (c–e) Augmented reality overlay of gamma camera findings within the anatomical context of the patient during head and neck cancer SN procedure (from [Citation255]). (f) Setup of a portable beta+ camera for radioguided surgery, with evaluation in a tongue tumor rabbit model showing uptake in primary tumor (g) and lymph nodes (h) using 18F-FDG (from [Citation162]).

Figure 2. 2D portable nuclear detection cameras. (a–b) Portable gamma camera imaging during SN procedure for head and neck melanoma with ICG-99mTc-nanocolloid (from [Citation253Citation254]). (c–e) Augmented reality overlay of gamma camera findings within the anatomical context of the patient during head and neck cancer SN procedure (from [Citation255]). (f) Setup of a portable beta+ camera for radioguided surgery, with evaluation in a tongue tumor rabbit model showing uptake in primary tumor (g) and lymph nodes (h) using 18F-FDG (from [Citation162]).

2.2.1. Portable gamma (γ) cameras

Imaging the emitted γ rays, portable γ cameras are designed to image radiopharmaceutical uptake in a small FOV (typically around 50 × 50 mm2) during surgery (see ). Most systems are designed to image γ ray energies between roughly 50–300 keV, making them compatible with traditional SPECT isotopes (e.g. 99mTc) (see ). Therefore, as seen from , portable γ cameras are mostly applied for open surgery SN procedures using 99mTc-labelled radiocolloids (e.g. in head and neck cancer, breast cancer, melanoma or penile cancer) and ROLL procedures using 99mTc-labelled macro-aggregates (e.g. breast cancer, thyroid cancer), but also in SNOLL procedures using 99mTc-nanocolloid and 99mTc-MAA (i.e. breast cancer), resection of neuroendocrine tumors using 123I-MIBG (e.g. paraganglioma) and resection for thyroid disorders 99mTc-MIBI [Citation101,Citation149]. To our knowledge, intraoperative usage with PET radiopharmaceuticals has not been shown in literature yet. Specific reviews reporting on portable γ cameras in more detail are provided by Tsuchimochi et al. and Hellingman et al. [Citation150,Citation151].

2.2.1.1. Background and recent advances

Similar to the γ probe technology, most γ camera detectors are either based on a scintillator system or an ionization system [Citation150,Citation152]. Effectively, camera exposure times used during surgery range between 5 and 60 s [Citation106]. Typical system resolutions of a portable γ camera, at distances <5 cm to the targeted tissue, are found between roughly 1.5–10 mm [Citation150,Citation151]. Typical collimator designs are the parallel-hole and the pinhole collimators, fabricated out of lead or tungsten [Citation153]. In a parallel-hole collimator, parallel septa of collimation material are used to prevent overflow of γ signal between individual pixels of the camera, only allowing what is in front of the specific pixel to be detected. Consequently, this fixes the size of the camera FOV. These parallel-hole collimators generally have a square or hexagonal pattern in front of the detector and often come in low energy high resolution or low energy high sensitivity configurations; for example, longer septa increase camera resolution, but decrease camera sensitivity. In a pinhole collimation configuration, a single focusing aperture is created with the collimation in front of the detector, very similar to photographic cameras used in daily life. When an object is close to the camera, this allows for a magnified image on the detector (i.e. higher resolution, smaller FOV) [Citation154]. However, when an object is far from the camera, the image on the detector is miniaturized allowing for a larger FOV with lower resolution. Just as with a parallel-hole setup, the pinhole setup also allows for an interchangeable collimator configuration, where a smaller aperture allows for higher resolution, but lower sensitivity [Citation155]. Literature is somewhat contradicting on this point, most likely due to differences in working distances, detector technologies, and collimator geometries used, parallel-hole is generally thought to provide higher sensitivity (especially at large distances), while resolution is higher with pinhole (especially at close distances) [Citation106,Citation151].

Portable γ cameras can be used as a handheld device (e.g. CrystalCam) or one that is mounted on an articulating arm (e.g. Sentinella) (see )). The first model is more mobile and can be applied for investigations under difficult angles, but the second is more ergonomic over longer periods of time (typical camera weight 0.7–2.7 kg [Citation150]) and can provide a sharper image when long exposure times are needed. A downside of using γ cameras is that these images traditionally only display the distribution of radioactive counts on a screen in the OR, and thus miss a connection to the anatomical context of the patient. To provide such a connection and make use of the system more intuitive, camera developments focus on integrating γ images with optical images of the area of interest via an augmented reality image overlay. This directly places the nuclear findings within the anatomical context of the patient (see )) [Citation156Citation159]. In the clinic, portable γ cameras are only used as open surgery systems (i.e. outside of the patient), however, experimental setups are devised allowing for the acquisition of γ images via a laparoscopic device as well [Citation107].

2.2.2. Beta (β) cameras

Although intraoperative imaging of PET radiopharmaceuticals with a portable γ camera has not been shown yet, some research groups are pioneering this use with handheld β+ cameras (see )) [Citation124,Citation160Citation164].

2.2.2.1. Background and recent advances

Similar as what was reported for β probes, background as a result of the high-energy annihilation γ’s limits the implementation of this camera technology [Citation163]. Hence, like the probes, most β+ camera systems incorporate a dual-detector design. However, different from typical β+ probes reported, these β+ cameras do not use two separate detectors to determine the contribution of the individual signals, but rather use a single detector with two stacked scintillation materials in front: the first converting both β+ and γ signals to visible light and the second only converting the deeper penetrating γ rays to visible light. These light pulses can subsequently be separated based on shape [Citation163]. Similar as with β+ probes, subtracting the γ-only signal from the signal of the first scintillator (i.e. γ and β+) allows for a γ rejection to be applied. Typically, the first scintillator is a relatively thin plastic scintillator (e.g. p-terphenyl), while the second scintillator is made of a thicker inorganic scintillator (e.g. LYSO or BGO). This scintillation stack is generally coupled to either a position-sensitive PMT, silicon photomultiplier (SiPM) detector arrays or an EMCCD. Current prototype systems hardly use any collimation, though it is suggested that shielding on the sides and back would be a requirement in vivo, preventing detector noise from lesion surrounding anatomies [Citation160,Citation162].

Most experimental systems are now targeted towards 18F-FDG; preliminary ex vivo (18F-FDG avid existed breast tumor [Citation124]) or in vivo animal evaluations (18F-FDG avid rabbit tongue tumor [Citation161,Citation162]) indicate possible translation of this technology (see )). As far as we know, portable β cameras have not been reported. That said, CMOS sensors have been suggested as relative simple detector for β particle imaging, without the need for a scintillation material [Citation165].

2.3. 3D nuclear detection modalities

To provide full in-depth insight regarding the radiopharmaceutical distribution within the patient, some form of 3D imaging is required. For this purpose, SPECT/CT and PET/CT have become the golden standard for diagnostic molecular imaging. For interventional purposes, these scans can be used to provide a reference roadmap for planning of the procedure. Furthermore, similar as the use of nuclear detection probes (1D) and cameras (2D), SPECT- or PET-based imaging (3D) can also be of value during radioguided interventions, including interventional radiology and surgery (see ). Especially for surgical application, however, it is often not logistically and financially feasible to use a full-sized SPECT or PET system in the OR itself. Therefore, pre-interventional scans are used in a navigation workflow, whereby an augmented/mixed reality overlay on to the patient is shown or a virtual reality navigation environment is provided, directing the interventional instruments towards the lesions with high radiopharmaceutical uptake [Citation166]. Such navigation procedures have been pursued during PET-guided biopsy and PET- and SPECT-guided intraoperative lesion localization. As result of innovative use of a surgical navigation setup, intraoperatively acquired freehand SPECT has also been introduced. This technology provides a method to acquire a patient roadmap in the operating room. The biggest challenge with navigated approaches in general are tissue-induced deformations, which degrade the registration between the patient and the PET- or SPECT-based patient map [Citation167]. Real-time confirmation with an intraoperative detection device can help correct for such inaccuracies.

Figure 3. 3D nuclear detection modalities for radioguided interventions. (a–b) Setup for navigated 18F-FDG PET-biopsy using a registered robotic biopsy arm (from [Citation257]). (c–d) Example of trajectory planning in patient with suspected lymphoma. (e) Setup for SPECT/CT-based surgical navigation in head and neck cancer SN procedure, showing near-infrared (NIR) tracking of patient and instrument positions for registration with the patient scans. (f) Augmented reality overlay of SPECT/CT scan in the operating room for a penile cancer SN procedure (from [Citation180]). (g) Virtual reality navigation towards SN in breast cancer using the SPECT/CT patient data (from [Citation101]). (h) Intraoperative creation of 3D freehand SPECT images using a tracked portable gamma camera, including subsequent augmented reality overlay (i) and virtual reality navigation (j) during a head and neck cancer SN procedure.

Figure 3. 3D nuclear detection modalities for radioguided interventions. (a–b) Setup for navigated 18F-FDG PET-biopsy using a registered robotic biopsy arm (from [Citation257]). (c–d) Example of trajectory planning in patient with suspected lymphoma. (e) Setup for SPECT/CT-based surgical navigation in head and neck cancer SN procedure, showing near-infrared (NIR) tracking of patient and instrument positions for registration with the patient scans. (f) Augmented reality overlay of SPECT/CT scan in the operating room for a penile cancer SN procedure (from [Citation180]). (g) Virtual reality navigation towards SN in breast cancer using the SPECT/CT patient data (from [Citation101]). (h) Intraoperative creation of 3D freehand SPECT images using a tracked portable gamma camera, including subsequent augmented reality overlay (i) and virtual reality navigation (j) during a head and neck cancer SN procedure.

2.3.1. Navigated PET- or SPECT-guided biopsy

Image-guided percutaneous biopsy followed by pathology plays an essential role in the management of oncological patients. Biopsies provide detailed molecular and genetic information of the disease and are critical for both diagnosis and staging. A non-successful biopsy, which occurs in 5–31% of the oncological cases, demands a repeating biopsy or even a conversion to open (surgical) procedures, which can postpone diagnosis and lead to additional costs, patient pain and anxiety [Citation168,Citation169]. Image-guided percutaneous biopsies are commonly performed using morphological imaging (e.g. fluoroscopy, US, CT, and MR imaging). In some cases, morphological changes are not predictive for the disease progression, which can lead to inconclusive findings [Citation170]. Here targeting of molecular features as identified on, e.g. SPECT/CT or PET/CT can help decrease the number of inconclusive biopsies. One of the main advantages of such a radioguided biopsy approach is the ability to specifically target the lesion(s) with highest radiopharmaceutical uptake (see ).

2.3.1.1. Background and recent advances

Intra-procedural 18F-FDG PET/CT-guided biopsy using an automated robot arm has been reported [Citation171,Citation172]. Here, positioning of the biopsy needle and tissue sampling was fully automated using a registered robot arm based on lesion coordinates defined in the PET/CT images (see )). Fixation of the patient with a vacuum-immobilization device helped preventing inaccuracies due to patient movement. Radhakrishnan et al. reported this led to 100% successful diagnostic biopsies in 25 patients with intrathoracic lesions [Citation172] and Kumar et al. reported a diagnostic yield of 98.6% in 73 patients with bone lesions [Citation171]. A down-side of such procedures is that they are time-consuming and can increase the radiation burden to the patient and medical staff [Citation171]. SPECT/CT-guided biopsies have also been suggested, using, for example, 99mTc-MIBI [Citation173]. Navigated approaches of this method were not found in literature yet, but the manual method already provided a 100% diagnostic yield (n = 12) in intrathoracic lesions.

2.3.2. Navigated PET- or SPECT-guided surgery

Similar as to navigated biopsies, 3D PET and SPECT images have also proven their ability to guide surgical lesion localization via a surgical navigation workflow in vivo (see )).

2.3.2.1. Background and recent advances

18F-FDG PET/CT navigation of a laparoscope was demonstrated during lymph node salvage surgery for breast cancer and recurrent lymphoma [Citation174]. SPECT/CT navigation has been applied more often, with navigation of a γ probe during open surgery SN procedures using (ICG-)99mTc-nanocolloid (e.g. penile cancer, melanoma, breast cancer, thyroid cancer and prostate cancer [Citation101,Citation175Citation177]), the resection of neuroendocrine tumors using 123I-MIBG (e.g. paraganglioma [Citation101]) and parathyroid adenoma using 99mTc-MIBI [Citation178]. All intraoperative studies so far were based on the same underlying technology: an intraoperative navigation system with near-infrared optical tracking and optical tracking fiducials attached to both the patient and the navigated instruments (see )). This tracking setup allowed for a registration between the preoperative PET- or SPECT-based patient maps and the intraoperative instruments. During surgery, this allowed for more accurate positioning of the surgical instruments (see )). However, these studies also indicated that navigation to soft tissues based on preoperative images can suffer from inaccuracies due to tissue deformations. By navigating a γ probe or fluorescence camera, the real-time feedback provided could partially correct for such inaccuracies [Citation175,Citation176,Citation179Citation181].

2.3.3. 3D intraoperative freehand imaging and subsequent navigation

Rather than rely on preoperatively acquired 3D scans, also intraoperative 3D scans can be obtained using a so-called freehand imaging technology. This technology makes innovative use of nuclear detection modalities already available in the OR to create 3D images of the radiopharmaceutical distribution within the patient. Freehand SPECT (fhSPECT) is the most common form of nuclear freehand 3D imaging available in the OR [Citation182,Citation183] (see and is the only form evaluated in vivo during radioguided surgery so far.

2.3.3.1. Background and recent advances

Using a surgical navigation system, the position and orientation of a γ probe or camera can be followed during γ tracing in de OR. While freely moving the γ probe or camera around the tissue of interest, this navigation system tracks the detector position relative to the patient, and at the same time, the read-out of the detector in each different position is recorded. Subsequently, dedicated signal processing software is able to create a 3D scan of the tracer distribution as related to the tracked patient [Citation182,Citation184,Citation185]. The quality of this fhSPECT scan depends on the radiopharmaceutical uptake (i.e. signal intensity) and the number of positions and orientations (i.e. angular coverage) scanned around the tissue of interest [Citation156,Citation183]. Acquisition times can vary, but in general total scanning time should be kept roughly below 5 min [Citation156,Citation182,Citation186].

Since the clinical introduction in 2010, fhSPECT has shown to be useful for several nuclear detection modalities, including open surgery γ probes [Citation187Citation189], open surgery portable γ cameras [Citation156,Citation186,Citation190], laparoscopic γ probes [Citation179,Citation191,Citation192] and even the recently introduced DROP-IN γ probe [Citation193]. As a result, this technology has already been evaluated during many different radioguided surgery procedures, such as SN in breast cancer [Citation156,Citation182,Citation187], RSL in breast cancer [Citation189], SN in head and neck cancers [Citation186,Citation188,Citation194Citation197], SN in melanoma [Citation186,Citation198,Citation199], SN in prostate cancer [Citation179], SN in penile cancer [Citation179], SN in gynecology [Citation191], ROLL for pulmonary lesions [Citation192], PSMA-targeted prostate salvage procedures [Citation108], parathyroid adenoma [Citation178], NET tumors [Citation200] and various bone lesions [Citation201].

Apart from using γ detection modalities, some have experimented with using β+ probes to create freehand β+ surface mapping [Citation137,Citation202]. Though proven successful in the laboratory, this concept has not been performed in vivo yet.

2.4. Multiplexing modalities

Despite the strong advantages provided for radioguided surgery, it is obvious to all that each individual detection signal (i.e. γ or β emissions) also has weaknesses. One way to overcome such weaknesses is to include the use of complementing guidance technologies during the same procedure. This multiplexing of the complementary information of different signatures in to a single procedure yields so-called ‘hybrid’, ‘dual-modality’ or ‘multi-modal’ procedures. The exploration of multiplexing concepts within the framework of radioguided surgery has substantially increased during the last 10 years. Multiplexing concepts have made use of dual-isotope detection (i.e. γ emissions with different energies or a combination of γ and β emissions), combined nuclear and optical detection (i.e. a combination of γ and fluorescence emissions or γ and Cerenkov emissions), combined use of β detection and optical coherence tomography and combined use of γ detection and ultrasound imaging. While procedures that rely on two signatures are more challenging from a technical perspective, from a clinical point-of-view, guidance towards targeted lesions may be optimized and logistics can be simplified.

2.4.1. Dual-isotope detection

Where the use of multiple radiopharmaceuticals for a single surgical guidance procedure is rare, dual-isotope multiplexing has been applied for the use of 125I-seeds and 99mTc-radiocolloid pharmaceuticals. Both low-to-mid energy γ probes [Citation102] and γ cameras [Citation116] have shown to be capable of such multiplexing.

2.4.1.1. Background and recent advances

Every radioactive isotope comes with a characteristic emission spectrum based on emission type (i.e. γ photon or β particle) and emission energy (see ). Such a defined emission spectrum can consist of a single type and energy, but often consists of a combination of multiple types and/or energies. These defined emissions allow for the differentiation between multiple radiopharmaceuticals, each harboring a complementing radioactive isotope [Citation203,Citation204].

γ detectors typically describe detection events as an electrical pulse, where height of the pulse is a reflection of the γ ray energy. However, energy resolution of the detector determines how accurate this energy estimation is. By optimally classifying detection events within chosen energy ‘windows’, a differentiation can be made between different isotopes, provided that both are individually detectable with the device. The more unique the emission spectra of the radioactive isotopes are (i.e. no overlap of energy peaks), the more trivial such differentiation becomes.

To this extend, low-to-mid energy γ probes have been used to support combined use of the SN procedure (using 99mTc) and the RSL procedure (using 125I) in open breast cancer surgery [Citation205]. Individual isotope tracing was achieved using a single γ probe that allows for switching between a 99mTc and 125I energy window (i.e. a window either around 140 or 27 keV, respectively). Similar energy discrimination even allows simultaneous imaging of a 99mTc radiopharmaceutical distribution with respect to the 125I-seed location. For portable γ cameras, the dual-isotope concept has been used to realize more convenient γ probe placement during laparoscopic SN procedures [Citation114,Citation116,Citation206]. This concept was realized by placing a 125I seed on the tip of the laparoscopic γ probe able to detect the radiopharmaceutical 99mTc-nanocolloid, while monitoring the location of this tip with respect to the targeted lesion. However, some groups report a certain level of additional background is present during 125I detection with both γ probe and camera, due to the 99mTc originating scattered γ rays [Citation102].

Next to multiplexing several γ ray energies, it has also been suggested to use a combination of β+ and γ detection during surgery [Citation141,Citation207]. Such joint emissions are common for all PET isotopes (e.g. 18F), producing both β+ particles and annihilation γ rays (see ). In this setup, the γ-signal could be used for initial localization of the targeted lesion, while the β+ signal enables examination of, for example, tumor margins. As discussed in the β+ probe section, these nuclear emissions can be discriminated using a dual-detector setup: one sensitive to both β+ and γ and another sensitive to γ only. Despite being technically possible, to the best of our knowledge, hybrid γ and β+ detection has only been pursued in a single in vivo study for recurrent melanoma using 18F-FDG yet [Citation255]. This study describes the use of a nuclear detection probe in five patients, capable of switching between β+ and γ detection via a footswitch pedal.

2.4.2. Optical emissions – fluorescence

To provide the surgeon with the best guidance as currently possible, many recent advances focus on the use of optical and nuclear detection modalities within a single surgical workflow [Citation153,Citation208,Citation209]. Within the field of image-guided surgery, fluorescence imaging is rapidly gaining popularity, often providing detailed real-time visualization of targeted tissues within their anatomical context [Citation210]. Unfortunately, despite the intuitive use of this technology, it remains a superficial technique (<1 cm) [Citation211]. Therefore, in the case of targeted resections, a combination with nuclear medicine approaches is preferred. The hybrid (i.e. fluorescent and radioactive) radiopharmaceutical indocyanine green (ICG)-99mTc-nanolloid has been successfully applied during, among others, SN procedures performed across different indications [Citation212] including breast cancer [Citation213], cervical cancer [Citation214], penile cancer [Citation215], prostate cancer [Citation216] and head and neck cancers [Citation217,Citation218]. The same hybrid concept also has demonstrated value during ROLL procedures performed in melanoma and lymphoma patients [Citation219].

2.4.2.1. Background and recent advances

Fluorescence imaging is an optical method that exploits the nature of fluorescent dyes: under excitation with a dye specific wavelength of light (typically between 400 and 800 nm), these substances emit light with a lower energy themselves (typically between 500 and 900 nm). Due to this shift in energy, optical filters can be used to separate excitation from emission light, allowing for precise visual localization of the fluorescent pharmaceutical. Of all the hybrid detection systems described for radioguided surgery, the combination of γ and fluorescence is the one most addressed in clinical literature. The optonuclear probe is a hybrid version of the traditional γ probe (see )), providing both low-to-mid energy γ tracing and ICG fluorescence tracing in a single device [Citation220Citation222]. This functionality is achieved by extending a γ probe design with two optical fibers, one coupled to an ICG excitation laser and the other coupled to an optically filtered (>810 nm) PMT. It has been used during both open and laparoscopic surgery for SN procedures in head and neck cancers, penile cancer, breast cancer, melanoma and cervical cancer [Citation220Citation222]. Recently, it was shown that an integration of this hybrid detection probe with a surgical navigation system was even capable of producing 3D fluorescence tomography reconstructions (i.e. freehand Fluorescence scans) in addition to the above described fhSPECT modality [Citation258]. This hybrid imaging and navigation technology was evaluated in ex vivo prostate cancer samples using ICG-99mTc-nanocolloid. KleinJan et al. investigated the use of image-based hybrid modalities in vivo (i.e. penile cancer SN procedure), by using a tailored bracket to physically connect either a γ probe or portable γ camera to a fluorescence camera system [Citation154]. Here, the in-depth information of radioguidance provided the ability to more accurately position the fluorescence camera. A portable and fully integrated hybrid γ and fluorescence camera has only been studied in preclinical evaluations [Citation223]. In this system, γ and fluorescent emissions are both collected at the front of the device, but in the camera are separated through an angled mirror (45°) to allow for separate detection. With an innovative wavelength division-based multiplexing method, Kang et al. propose an integrated hybrid laparoscope for simultaneous γ and fluorescence imaging in a laparoscopic setting [Citation107]. In this system, γ, white light and fluorescent emissions are all collected through the same tungsten pinhole collimator. Subsequently, all emissions are passed through a gadolinium oxyorthosilicate (GSO) scintillator, converting only the γ rays to visible blue light (~400–500 nm). Hereafter all emissions are transported via de laparoscope imaging fiber bundle and collected with three different cameras based on wavelength. However, usability of this system still has to be determined.

Figure 4. Multiplexing modalities for radioguided surgery combining gamma and fluorescence detection. (a) Optonuclear probe design with detection elements for both low-to-mid energy gamma tracing and ICG fluorescence tracing (from [Citation220]). Application during open surgery breast cancer (b–c) and laparoscopic surgery cervix cancer (d) SN procedure using ICG-99mTc-nanocolloid (from [Citation221]). (e) Position tracking and navigation of an open surgery fluorescence camera during a penile cancer SN procedure using ICG-99mTc-nanocolloid. (f) Similar navigated fluorescence camera setup for a laparoscopic prostate cancer SN procedure (from [Citation179]). (g–h) The fluorescence laparoscope is guided towards the lesion locations via augmented reality overlays of the freehand SPECT scans. (i) Fluorescence confirmation of the lesion of interest.

Figure 4. Multiplexing modalities for radioguided surgery combining gamma and fluorescence detection. (a) Optonuclear probe design with detection elements for both low-to-mid energy gamma tracing and ICG fluorescence tracing (from [Citation220]). Application during open surgery breast cancer (b–c) and laparoscopic surgery cervix cancer (d) SN procedure using ICG-99mTc-nanocolloid (from [Citation221]). (e) Position tracking and navigation of an open surgery fluorescence camera during a penile cancer SN procedure using ICG-99mTc-nanocolloid. (f) Similar navigated fluorescence camera setup for a laparoscopic prostate cancer SN procedure (from [Citation179]). (g–h) The fluorescence laparoscope is guided towards the lesion locations via augmented reality overlays of the freehand SPECT scans. (i) Fluorescence confirmation of the lesion of interest.

Next to the use of fully integrated hybrid detection modalities, hybrid surgical navigation concepts have been used to successfully integrate nuclear guidance (i.e. SPECT/CT or fhSPECT) with fluorescence guidance in vivo [Citation175,Citation179Citation181]. In this approach, the surgical targets (e.g. SNs) are defined based on the 3D preoperative SPECT/CT or intraoperative fhSPECT scans. Hereafter, the tracked fluorescence camera is navigated towards these targets, using virtual or augmented reality displays of the SPECT findings in the fluorescence video display (see )). In this way, fluorescence guidance is extended to deeper lying lesions (>1 cm), while <1 cm inaccuracies of the navigation workflow are corrected via the real-time fluorescence feedback. Promising evaluations of this approach were shown in open surgery penile SN and laparoscopic prostate SN procedures.

2.4.3. Optical emissions – cerenkov

An alternative means to combine nuclear with optical imaging is provided by Cerenkov imaging (see ). Ciarrocchi et al., Das et al. and Spinelli et al. provide comprehensive overviews regarding the current status of clinical Cerenkov imaging [Citation224Citation226].

2.4.3.1. Background and recent advances

Cerenkov radiation is a phenomena first described and explained in literature by Pavel Cerenkov in 1934 [Citation227]. It is an optical emission that occurs when charged particles, such as β+ and β particles, travel through a dielectric medium with a phase velocity higher than the speed of light in that medium [Citation225,Citation228]. By passing through the medium at such velocities, the charged particles create an asymmetric polarization of the medium along their traveled path, which promotes the emission of (ultra)violet (~250–450 nm) light and to a lower extend visible light (<25% emission >450 nm) [Citation229,Citation230]. The amount of light coming from these types of emissions increases with the particle energy and amount of particles (i.e. radiation dose). The total light yield at conventional clinical 18F-FDG conditions is unfortunately relatively low, being roughly an order of 3 to 4 less in intensity as compared to typical fluorescence-guided surgery [Citation231]. To increase intensity of the faint signal, a higher amount of the β emitting radiopharmaceutical could be injected, though increasing the dose to the patients and possibly the surgical staff. Although 18F-FDG appears to be the generally used tracer, this isotope is actually rated as a low to moderate Cerenkov emitter, where isotopes such as the β emitting 90Y provide a much higher Cerenkov intensity (factor 23) due to the higher β particle energy [Citation232].

Theoretically, instrumentation to detect Cerenkov radiations is rather straightforward, where only an optical camera is needed to capture the light emitted in the tissue as a result of the injected radiopharmaceutical. Practically, due to the low signal intensity, a controlled light-tight environment is needed (i.e. blocking the ambient light) to accommodate detection by a highly sensitive (cooled) (EM)CCD camera with typically long exposure times (1–5 min). These practical features limit current Cerenkov-guided surgery, making it understandable that most work with this technology is performed in a preclinical setting so far [Citation233]. Nevertheless, there have been some nice examples of in-human use (see )): non-surgical imaging of 18F-FDG avid lymph nodes in the axilla [Citation234], non-surgical imaging of thyroid radiation therapy using 131I [Citation235], non-surgical imaging of external beam radiation therapy in breast cancer [Citation236] and intraoperative imaging using 18F-FDG in rectal cancer endoscopic surgery [Citation237]. Furthermore, ex vivo analysis of surgical specimens within the OR has also been suggested for direct margin evaluation [Citation224]. Although no research is reported on this format yet, one could imagine a hybrid application where γ rays are used for lesion localization and Cerenkov imaging is used for margin evaluation [Citation208].

Figure 5. Multiplexing modalities for radioguided monitoring and interventions using various detection signals. (a) Setup for a SN biopsy based on the fhSPECT-ultrasound fusion (from [Citation208]). (b–c) fhSPECT-US fusion images for breast cancer SN localization (from [Citation248]). (d–f) Combined use of beta+ and OCT for ex vivo localization of 18F-FDG avid ovarian cancer lesions, showing increased radiopharmaceutical uptake with a beta+ distribution map (d), anatomical microstructure details using OCT (e) and histopathological evaluation (f) (from [Citation240]). (g) 18F-FDG avid axilla lymph nodes as shown with PET/CT can successfully be imaged with Cerenkov imaging (h) as well (from [Citation236]). (i) Similar Cerenkov imaging during thyroid therapy using 131I (from [Citation237]). (j) Intraoperative Cerenkov imaging of 18F-FDG in endoscopic surgery of rectal cancer (from [Citation239]). (k–l) Augmented reality overlay of Cerenkov images with an example of tumor tissue (k) and normal tissue (l).

Figure 5. Multiplexing modalities for radioguided monitoring and interventions using various detection signals. (a) Setup for a SN biopsy based on the fhSPECT-ultrasound fusion (from [Citation208]). (b–c) fhSPECT-US fusion images for breast cancer SN localization (from [Citation248]). (d–f) Combined use of beta+ and OCT for ex vivo localization of 18F-FDG avid ovarian cancer lesions, showing increased radiopharmaceutical uptake with a beta+ distribution map (d), anatomical microstructure details using OCT (e) and histopathological evaluation (f) (from [Citation240]). (g) 18F-FDG avid axilla lymph nodes as shown with PET/CT can successfully be imaged with Cerenkov imaging (h) as well (from [Citation236]). (i) Similar Cerenkov imaging during thyroid therapy using 131I (from [Citation237]). (j) Intraoperative Cerenkov imaging of 18F-FDG in endoscopic surgery of rectal cancer (from [Citation239]). (k–l) Augmented reality overlay of Cerenkov images with an example of tumor tissue (k) and normal tissue (l).

2.4.4. Optical coherence tomography

β+ imaging has also been proposed in combination with optical coherence tomography (OCT) [Citation238]. OCT is a technique often compared to ultrasound. However, instead of sound pulses emitted and received by the ultrasound probe, an OCT probe typically emits near-infrared light pulses in to the tissue and detects the back-reflected light [Citation259]. Spatial resolution of this technique is in the order of μm’s. Similar as β+ detection, OCT is a superficial technique with a penetration depth of only a few mm’s. In a combined setup, 2D β+ imaging is suggested to be used for localization of the lesions with high radiopharmaceutical uptake, while OCT provides anatomical imaging to investigate the tissue microstructures [Citation239]. So far, only a single prototype system is reported, where the detection head consists of a single OCT fiber, surrounded by multiple β scintillators. Preliminary ex vivo results illustrate the potential of this technique (see )); ex vivo ovarian cancer samples (using 18F-FDG) showed detailed morphological features with OCT that could potentially be used for detection of early malignant cancer [Citation238].

2.4.5. Ultrasound

Interventional use of nuclear medicine technologies with ultrasound (US) imaging has also been pursued. In such a setup, nuclear detection modalities such as γ cameras and fhSPECT identify lesions with high radiopharmaceutical uptake, while ultrasound can be used to provide anatomical detail of the surrounding tissue.

2.4.5.1. Background and recent advances

Pani et al. describe a handheld ultrasound probe with an integrated small FOV γ camera [Citation240]. By placing a piezoelectric US crystal in front of, and partially overlapping with, a custom γ collimator, this device can simultaneously image ultrasound and γ information, which can be displayed as a fused image. Partial overlap of the detectors combined with a custom software algorithm allows for <1 mm error in image registration. Using a slit model collimator design, combined with the NaI(Tl) scintillator and position sensitive PMTs, the resulting high γ counting efficiency allows for real-time acquisition of γ images. This in turn ensures that the fused γ/US images can be displayed in a real-time fashion. Unfortunately, a hard acquisition time is not mentioned. The reported resolution (<2 mm) was acceptable for a γ camera but is quite poor for an US device. In vivo evaluation has still to be shown.

Integration of US and γ images are also shown via a surgical navigation setup [Citation241]. In this setup, the 3D molecular imaging capabilities of fhSPECT are acquired and subsequently fused with the real-time US information by tracking the position of both the patient and the US probe (see )). Registration of this US-fhSPECT fusion has shown to be accurate for various (para)thyroid diseases [Citation242,Citation243] using 99mTc-MIBI or 99mTcO4. Mapping of sentinel lymph nodes was also shown for breast cancer, melanoma, cutaneous lesions, vulva cancer and head and neck cancers [Citation244Citation246]. In these cases, the potential of US-fhSPECT fusion was further supported, facilitating sentinel lymph node identification if lymph nodes were not well visible with US or when various lymph nodes were located in close proximity to each other. However, to allow for an accurate fhSPECT-US registration (i.e. order of mm’s) in these procedures, specific attention was needed to minimize patient repositioning and tissue deformations.

3. Expert opinion

In this article, we’ve provided a comprehensive overview of nuclear detection modalities for image-guided surgery, specifically placing new developments within the framework of existing technology. During the last years, dedicated detection modalities have become available for specific medical applications such as the tethered DROP-IN γ probe for robot-assisted laparoscopic surgery. Also, use of a wide range of nuclear emissions has been explored: γ radiation (low-to-mid or high energy), β radiation (β+ and β). displays a simplified and generalized summary of all modalities discussed.

Table 3. Simplified general overview of detection modalities available for radioguided surgery, including human use only.

From a clinical perspective, the main advantages of using low-to-mid energy γ detection are the ease of use (e.g. low sampling time), the great availability of (hybrid) detection devices for different surgical techniques and the relative low dose to patient and surgical staff. Based on this, one can argue there is a clinical demand for the development of more receptor-specific SPECT radiopharmaceuticals. Contradictory to this, the main advantages of using either high energy γ or β+ detection is the availability of a plurality of PET radiopharmaceuticals. For the exploration of available PET radiopharmaceuticals, further engineering of high energy γ and β+ detection modalities is required to improve ease of handling and promote integration in laparoscopic surgery. Considering that PET isotopes come with high-energy γ rays, decrease of radiopharmaceutical dosing would also be desired, decreasing radiation dose to patient and surgical staff. Since detection of pure β isotopes demands the use of therapeutic radiopharmaceuticals, more detailed evaluations are needed to decide on the potential role of this approach. Specific challenges lie with preoperative lesion mapping since β particles have low penetration depth, and radiation dose to the patient.

Though the use of radioactive isotopes in medicine is only expected to grow, it does not mean there aren’t shortcomings and disadvantages to its use in surgery. The limitations of conventional radioguided surgery approaches have resulted in an increasing tendency to apply radioguidance in combination with other imaging signatures (e.g. fluorescence, Cerenkov, OTC, and US) or together with software-based guidance solutions (e.g. surgical navigation and augmented/mixed/virtual reality displays). With all the research available on investigating hybrid surgical devices [Citation153,Citation208,Citation209], especially the link between radioguidance and fluorescence guidance appears to be important for surgical interventions. Here, the complementary advantages of both techniques will provide a valuable roadmap for surgery, were nuclear information is used for diagnostics (e.g. SPECT or PET), surgical planning (e.g. SPECT/CT) and intraoperative translation and coarse localization (e.g. γ probe or portable camera), while fluorescence information provides high-resolution visual confirmation and delineation of the targeted lesions during excision [Citation260]. With the many successful evaluations of ICG-99mTc-nanocolloid [Citation212], we are under the opinion that this field will rapidly expand when more hybrid radiopharmaceuticals will make their way in to the clinic.

3.1. Five-year view

Radioguided surgery is already standard for image-guided surgery procedures such as the SN, ROLL and RSL and it is likely that the clinical impact of radioguided surgery will increase through the expanding clinical availability of disease-specific radiopharmaceuticals. The recent example of lymphatic salvage procedures in recurrent prostate cancer that make use of PSMA I&S (99mTc or 111In) provides a great example how innovations in the area of radiopharmaceutical design impact the field of radioguided surgery [Citation247]. Here it should be noted that efforts towards the realization of hybrid radiopharmaceuticals are likely to promote the use of multiplexing approaches. Examples of potential future hybrid procedures are: prostate cancer (PSMA I&F) [Citation248], neuroendocrine tumors (Cy5-111In-DTPA-Tyr3-octreotate) [Citation249], breast cancer ([111In]-DTPA-trastuzumab-IRDye800) [Citation250] and renal cancer (111In-girentuximab-IRDye800CW) [Citation251].

From a modality perspective, significant improvements are to be expected for high-energy γ detection modalities (e.g. improved ergonomics) and β detection modalities (e.g. first in vivo evaluations of the role of modern β detection). Nevertheless, we think the main focus of radioactive isotopes for surgery will probably remain confined to the more traditional low- and mid-energy γ detection (i.e. 99mTc and 111In). A combination of factors still makes this kind of nuclear detection dominant during surgery: (1) a clear connection to preoperative total body molecular imaging (i.e. SPECT/CT) available, (2) in-depth information offered for lesion localization and confirmation, (3) ease of use due to low sample times (<1 s) and plurality of form factors possible with relative lightweight and small dimensions, and (4) a relative low dose to patient and surgical staff [Citation254].

In the pursue of minimal invasive interventions, ensuring radical excisions while decreasing surgical complications, thereby improving patient outcome and quality of life, an ever more amount of surgical interventions will be pushed towards the realm of robot-assisted (laparoscopic) surgery. This will push the development of dedicated technologies such as the tethered DROP-IN γ probe [Citation99,Citation120,Citation121]. At the same time, the technological evolution of the field is expected to provide a platform for integration of augmented-, mixed- or virtual-reality displays and navigation in routine procedures [Citation167].

Article highlights

  • One-dimensional (1D) γ-detection probes provide the technical fundament of radioguided surgery.

  • Additionally, nuclear imaging during interventions has been evolving towards the detection of alternative signals (e.g. β radiation) or signal combinations (e.g. γ and fluorescence) as well. These efforts not only expand toward two-dimensional (2D) and three-dimensional (3D) imaging, but even include augmented- and virtual-reality approaches.

  • Key issues of performance for such image guidance modalities are:

    1. Sensitivity, which determines the ability to detect low radiopharmaceutical uptake and allows for short sample times. To facilitate surgical implementation, sample times have to be pushed towards real-time (<1 s) detections.

    2. Spatial resolution, which describes the ability to distinguish between lesions in close proximity to each other.

    3. Specificity, which defines the ability to discriminate the target tissue from background, sometimes called contrast.

    4. Ease of handling, which is determined by the ability of the surgeon to maneuver the nuclear detection device in an ergonomic and intuitive fashion.

    5. Rotational freedom during lesion localization.

    6. Signal representation, which determines how the surgeon needs to interpret the findings during lesion localization (e.g. audible, numerical or via image display).

  • These key issues of performance are not defined by a single aspect of the nuclear detection modality, but rather are a result of the complete design, applied working distance and integration within the surgical methodology. The latter underlines that the surgical procedure will influence the choice of device.

  • Clinically, a balance has to be created between the radiation dose that comes with a specific procedure (risk) and the benefit it creates (outcome). From the perspective of the patient, clinical outcome should outweigh the risk of accumulating dose.

  • For the medical staff, legally allowed annual dose exposure boundaries may limit the number of procedures that can be performed by individual surgeons. Most important dose factors are energy of the emitted γ or β particles (i.e. lower energy gives lower dose) and the amount of radioactivity left at the procedure (which is a combination of injected activity, isotope half-life and time after injection).

  • Uniquely, provided proper handling, use of β radiation hardly results in any dose risk to the surgical staff; β radiation has a limited penetration of only a few mm’s. Unfortunately, radiation dose to the patient does increase.

Declaration of interest

FWB van Leeuwen is a consultant for Hamamatsu photonics and is Chief Innovation Officer at the ORSI Academy surgical training center. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was funded by an NWO-TTW-VICI grant (grant no. TTW 16141).

References

  • Herrmann K, Nieweg OE, Povoski SP. Radioguided surgery. Switzerland: Springer International; 2016. DOI: 10.1007/978-3-319-26051-8.
  • Selverstone B, Sweet WH, Robinson CV. The clinical use of radioactive phosphorus in the surgery of brain tumors. Ann Surg. 1949;130(4):643.
  • Harris C, Bigelow R, Francis J, et al. A CsI (Tl)-crystal surgical scintillation probe. Nucleonics. New York City: McGraw-Hill Publishing Co., Inc., 1956;14:102–108.
  • Povoski SP. The history of radioguided surgery: early historical milestones and the development of later innovative clinical applications. In: Herrmann K, Nieweg OE, Povoski SP, eds. Radioguided surgery. Switzerland: Springer International; 2016. p. 3–12.
  • Alex JC. The application of sentinel node radiolocalization to solid tumors of the head and neck: a 10-year experience. Laryngoscope. 2004 Jan;114(1):2–19.
  • Chondrogiannis S, Ferretti A, Facci E, et al. Intraoperative hand-held imaging γ-camera for sentinel node detection in patients with breast cancer: feasibility evaluation and preliminary experience on 16 patients. Clin Nucl Med. 2013;38(3):e132–e136.
  • Duch J. Portable gamma cameras: the real value of an additional view in the operating theatre. Eur J Nucl Med Mol Imaging. 2011 April 01;38(4):633–635.
  • Hoogendam JP, Hobbelink MG, Veldhuis WB, et al. Preoperative sentinel node mapping with (99m)Tc-nanocolloid SPECT-CT significantly reduces the intraoperative sentinel node retrieval time in robot assisted laparoscopic cervical cancer surgery. Gynecol Oncol. 2013 May;129(2):389–394.
  • Jeschke S, Lusuardi L, Myatt A, et al. Visualisation of the lymph node pathway in real time by laparoscopic radioisotope- and fluorescence-guided sentinel lymph node dissection in prostate cancer staging. Urology. 2012 Nov;80(5):1080–1086.
  • Leong SP, Kim J, Ross M, et al. A phase 2 study of (99m)Tc-tilmanocept in the detection of sentinel lymph nodes in melanoma and breast cancer. Ann Surg Oncol. 2011 Apr;18(4):961–969.
  • Marcinow AM, Hall N, Byrum E, et al. Use of a novel receptor-targeted (CD206) radiotracer, 99mTc-tilmanocept, and SPECT/CT for sentinel lymph node detection in oral cavity squamous cell carcinoma: initial institutional report in an ongoing phase 3 study. JAMA Otolaryngol Head Neck Surg. 2013 Sep;139(9):895–902.
  • Mariani G, Moresco L, Viale G, et al. Radioguided sentinel lymph node biopsy in breast cancer surgery. J Nucl Med. 2001 Aug;42(8):1198–1215.
  • Murawa D, Nowaczyk P, Wiegering A. Radioguided sentinel lymph node mapping and biopsy in colorectal cancer. In: Herrmann K, Nieweg OE, Povoski SP, eds. Radioguided surgery. Switzerland: Springer International; 2016. p. 279–297.
  • Rietbergen DD, Van Den Berg NS, Van Leeuwen FW, et al. Hybrid techniques for intraoperative sentinel lymph node imaging: early experiences and future prospects. Imaging Med. 2013;5(2):147–159.
  • Saikawa Y, Otani Y, Kitagawa Y, et al. Interim results of sentinel node biopsy during laparoscopic gastrectomy: possible role in function-preserving surgery for early cancer. World J Surg. 2006 Nov;30(11):1962–1968.
  • Satoh M, Ito A, Kaiho Y, et al. Intraoperative, radio-guided sentinel lymph node mapping in laparoscopic lymph node dissection for Stage I testicular carcinoma. Cancer. 2005 May 15;103(10):2067–2072.
  • Sondak VK, King DW, Zager JS, et al. Combined analysis of phase III trials evaluating [(9)(9)mTc]tilmanocept and vital blue dye for identification of sentinel lymph nodes in clinically node-negative cutaneous melanoma. Ann Surg Oncol. 2013 Feb;20(2):680–688.
  • Surasi DS, O’Malley J, Bhambhvani P. 99mTc-Tilmanocept: a novel molecular agent for lymphatic mapping and sentinel lymph node localization. J Nucl Med Technol. 2015 Jun;43(2):87–91.
  • Takeuchi H, Goto O, Yahagi N, et al. Function-preserving gastrectomy based on the sentinel node concept in early gastric cancer. Gastric Cancer. 2017 Mar;20(Suppl 1):53–59.
  • Tokin CA, Cope FO, Metz WL, et al. The efficacy of Tilmanocept in sentinel lymph mode mapping and identification in breast cancer patients: a comparative review and meta-analysis of the (9)(9)mTc-labeled nanocolloid human serum albumin standard of care. Clin Exp Metastasis. 2012 Oct;29(7):681–686.
  • Valdes Olmos RA, Rietbergen DD, Vidal-Sicart S, et al. Contribution of SPECT/CT imaging to radioguided sentinel lymph node biopsy in breast cancer, melanoma, and other solid cancers: from “open and see” to “see and open”. Q J Nucl Med Mol Imaging. 2014 Jun;58(2):127–139.
  • van Den Berg NS, Buckle T, KleinJan GH, et al. Multispectral fluorescence imaging during robot-assisted laparoscopic sentinel node biopsy: a first step towards a fluorescence-based anatomic roadmap. Eur Urol. 2017 July 01;72(1):110–117.
  • Wallace AM, Han LK, Povoski SP, et al. Comparative evaluation of [(99m)tc]tilmanocept for sentinel lymph node mapping in breast cancer patients: results of two phase 3 trials. Ann Surg Oncol. 2013 Aug;20(8):2590–2599.
  • Weckermann D, Thalgott M, Holl G, et al. Radioguided surgery in urological malignancies. Indian J Urol. 2008 Jan;24(1):4–9.
  • Cabanas RM. An approach for the treatment of penile carcinoma. Cancer. 1977 Feb;39(2):456–466.
  • Betancourt JH, Vera CD, Martinez-Sarmiento M, et al. Application of the radio-guided occult lesion localization technique for renal lumpectomy: from the laboratory to the patient. Clin Nucl Med. 2017;42(11):e467–e468.
  • Cerit ET, Yalcin MM, zkan C, et al. Guided intraoperative scintigraphic tumor targeting of metastatic cervical lymph nodes in patients with differentiated thyroid cancer: a single-center report. Arch Endocrinol Metab. 2018 Oct;62(5):495–500.
  • Galetta D, Bellomi M, Grana C, et al. Radio-guided localization and resection of small or ill-defined pulmonary lesions. Ann Thorac Surg. 2015 Oct;100(4):1175–1180.
  • Lavoue V, Nos C, Clough KB, et al. Simplified technique of radioguided occult lesion localization (ROLL) plus sentinel lymph node biopsy (SNOLL) in breast carcinoma. Ann Surg Oncol. 2008 Sep;15(9):2556–2561.
  • Manca G, Mazzarri S, Rubello D, et al. Radioguided occult lesion localization: technical procedures and clinical applications. Clin Nucl Med. 2017 Dec;42(12):e498–e503.
  • Monti S, Galimberti V, Trifiro G, et al. Occult breast lesion localization plus sentinel node biopsy (SNOLL): experience with 959 patients at the European Institute of Oncology. Ann Surg Oncol. 2007 Oct;14(10):2928–2931.
  • Paredes P, Vidal-Sicart S, Zanón G, et al. Radioguided occult lesion localisation in breast cancer using an intraoperative portable gamma camera: first results. Eur J Nucl Med Mol Imaging. 2008;35(2):230–235.
  • Vitral G, Salgado H, Rangel J. Use of radioguided surgery in abdominal wall endometriosis: an innovative approach [case report]. World J Nucl Med. 2018 July 1;17(3):204–206.
  • Zurrida S, Galimberti V, Monti S, et al. Radioguided localization of occult breast lesions. Breast. 1998;7(1):11–13.
  • Donker M, Straver ME, Wesseling J, et al. Marking axillary lymph nodes with radioactive iodine seeds for axillary staging after neoadjuvant systemic treatment in breast cancer patients: the MARI procedure. Ann Surg. 2015;261(2):378–382.
  • van der Noordaa ME, Pengel KE, Groen E, et al. The use of radioactive iodine-125 seed localization in patients with non-palpable breast cancer: a comparison with the radioguided occult lesion localization with 99m technetium. Eur J Surg Oncol. 2015 Apr;41(4):553–558.
  • Essner R, Hsueh EC, Haigh PI, et al. Application of an [18F] fluorodeoxyglucose-sensitive probe for the intraoperative detection of malignancy. J Surg Res. 2001;96(1):120–126.
  • Kim WW, Kim JS, Hur SM, et al. Radioguided surgery using an intraoperative PET probe for tumor localization and verification of complete resection in differentiated thyroid cancer: a pilot study. Surgery. 2011 Mar;149(3):416–424.
  • Kraeber-Bodéré F, Cariou B, Curtet C, et al. Feasibility and benefit of fluorine 18–fluoro-2-deoxyglucose–guided surgery in the management of radioiodine-negative differentiated thyroid carcinoma metastases. Surgery. 2005;138(6):1176–1182.
  • Lee GO, Costouros NG, Groome T, et al. The use of intraoperative PET probe to resect metastatic melanoma. BMJ Case Rep. 2010;2010:bcr1220092593.
  • Molina MA, Goodwin WJ, Moffat FL, et al. Intra-operative use of PET probe for localization of FDG avid lesions. Cancer Imaging. 2009 Sep;29(9):59–62.
  • Povoski SP, Hall NC, Murrey DA, et al. Feasibility of a multimodal 18 F-FDG-directed lymph node surgical excisional biopsy approach for appropriate diagnostic tissue sampling in patients with suspected lymphoma. BMC Cancer. 2015;15(1):378.
  • Vos C, Hartemink K, Muller S, et al. Clinical applications of FDG-probe guided surgery. Acta Chir Belg. 2012;112(6):414–418.
  • Arbizu J, Rodriguez-Fraile M, Dominguez-Prado I, et al. Whole body 18fluoro-l-dopa PET–CT: a useful tool for location and surgical guidance in primary carcinoid tumours. Eur J Nucl Med Mol Imaging. 2008;35(8):1577.
  • Aras G, Gültekin SS, Küçük NÖ, et al. Intraoperative gamma probe guidance with 99m Tc-pertechnetate in the completion thyroidectomy. Ann Nucl Med. 2009;23(5):421–426.
  • Ubhi C, Hardy J, Pegg C. Mediastinal parathyroid adenoma: a new method of localization. Br J Surg. 1984;71(11):859–860.
  • Adams S, Acker P, Lorenz M, et al. Radioisotope‐guided surgery in patients with pheochromocytoma and recurrent medullary thyroid carcinoma: a comparison of preoperative and intraoperative tumor localization with histopathologic findings. Cancer: Interdiscip Int J Am Cancer Soc. 2001;92(2):263–270.
  • Al-Saeedi F. Role of 99mTc-(V) DMSA in detecting tumor cell proliferation. Anal Chem Insights. 2007;2:117739010700200001.
  • Axelsson C, Nielsen B, Graff J. Radioisotope-guided surgical biopsy of costal metastases in breast cancer patients. Scand J Surg. 2002;91(4):333–335.
  • Isgoren S, Demir H, Daglioz-Gorur G, et al. Gamma probe guided surgery for osteoid osteoma: is there any additive value of quantitative bone scintigraphy? Revista Española de Medicina Nuclear e Imagen Molecular (English Edition). 2013 July 01;32(4):234–239.
  • Krag D, Ford P, Patel M, et al. A simplified technique to resect abnormal bony radiolocalizations using a gamma counter. Surg Oncol. 1992;1(5):371–377.
  • Robinson LA, Muro-Cacho C, Hubbell DS. Intraoperative gamma probe-directed biopsy of asymptomatic suspected bone metastases. Ann Thorac Surg. 1998;65(5):1426–1432.
  • Von Meyenfeldt E, Siebenga J, Van Der Pol H, et al. Radionuclide-guided biopsy of bone lesions in cancer patients; a reliable, well-tolerated technique. Eur J Surg Oncol. 2014;40(2):193–196.
  • Duarte G, Cabello C, Torresan R, et al. Radioguided intraoperative margins evaluation (RIME): preliminary results of a new technique to aid breast cancer resection. Eur J Surg Oncol. 2007;33(10):1150–1157.
  • García-Talavera P, González-Selma ML, Ruiz M, et al. The value of early SPECT/CT and hand-held γ-camera in radio-guided surgery: a case of a hard-to-locate parathyroid adenoma. Clin Nucl Med. 2014;39(11):1009–1011.
  • Ikeda Y, Takayama J, Takami H. Minimally invasive radioguided parathyroidectomy for hyperparathyroidism. Ann Nucl Med. 2010;24(4):233–240.
  • Mariani G, Gulec SA, Rubello D, et al. Preoperative localization and radioguided parathyroid surgery. J Nucl Med. 2003;44(9):1443–1458.
  • Martinez DA, King DR, Romshe C, et al. Intraoperative identification of parathyroid gland pathology: a new approach. J Pediatr Surg. 1995;30(9):1306–1309.
  • Placzkowski K, Christian R, Chen H. Radioguided parathyroidectomy for recurrent parathyroid cancer. Clin Nucl Med. 2007;32(5):358–360.
  • Vilela Filho O, Carneiro Filho O. Gamma probe-assisted brain tumor microsurgical resection: a new technique. Arq Neuropsiquiatr. 2002;60(4):1042–1047.
  • Schattner A, Cohen A, Wolfson L, et al. Unexplained systemic symptoms and Gallium-67–guided decisions. Am J Med Sci. 2001;321(3):198–200.
  • Anderson RS, Eifert B, Tartt S, et al. Radioimmunoguided surgery using indium-111 capromab pendetide (PROSTASCINT) to diagnose supraclavicular metastasis from prostate cancer. Urology. 2000;56(4):669.
  • Badalament RA, Burgers JK, Petty LR, et al. Radioimmunoguided radical prostatectomy and lymphadenectomy. Cancer. 1993;71(7):2268–2275.
  • Rauscher I, Düwel C, Wirtz M, et al. Value of 111In‐prostate‐specific membrane antigen (PSMA)‐radioguided surgery for salvage lymphadenectomy in recurrent prostate cancer: correlation with histopathology and clinical follow‐up. BJU Int. 2017;120(1):40–47.
  • Rauscher I, Horn T, Eiber M, et al. Novel technology of molecular radio-guidance for lymph node dissection in recurrent prostate cancer by PSMA-ligands. World J Urol. 2018 Apr;36(4):603–608.
  • Povoski SP, Hall NC, Murrey DA Jr, et al. Multimodal imaging and detection strategy with 124 I-labeled chimeric monoclonal antibody cG250 for accurate localization and confirmation of extent of disease during laparoscopic and open surgical resection of clear cell renal cell carcinoma. Surg Innov. 2013;20(1):59–69.
  • Dawson P, Blair S, Begent R, et al. The value of radioimmunoguided surgery in first and second look laparotomy for colorectal cancer. Dis Colon Rectum. 1991;34(3):217–222.
  • Gu J, Zhao J, Li Z, et al. Clinical application of radioimmunoguided surgery in colorectal cancer using 125 I-labeled carcinoembryonic antigen-specific monoclonal antibody submucosally. Dis Colon Rectum. 2003;46(12):1659–1666.
  • Hladik P, Vizda J, Bedrna J, et al. Immunoscintigraphy and intra‐operative radioimmunodetection in the treatment of colorectal carcinoma. Colorectal Dis. 2001;3(6):380–386.
  • Krag DN, Haseman MK, Ford P, et al. Gamma probe location of 111 indium‐labeled B72. 3: an extension of immunoscintigraphy. J Surg Oncol. 1992;51(4):226–230.
  • Martin JE, Tuttle S, Rousseau M, et al. Radioimmunoguided surgery: intraoperative use of monoclonal antibody 17-1A in colorectal cancer. Hybridoma. 1986;5:S97–108.
  • Muxi A, Pons F, Vidal-Sicart S, et al. Radioimmunoguided surgery of colorectal carcinoma with an 111In-labelled anti-TAG72 monoclonal antibody. Nucl Med Commun. 1999;20(2):123–130.
  • Nieroda C, Mojzisik C, Hinkle G, et al. Radioimmunoguided surgery (RIGS) in recurrent colorectal cancer. Cancer Detect Prev. 1991;15(3):225–229.
  • O’Dwyer PJ, Mojzisik CM, Hinkle GH, et al. Intraoperative probe-directed immunodetection using a monoclonal antibody. Arch Surg. 1986;121(12):1391–1394.
  • Xu G, Zhang M, Liu B, et al., editors. Radioimmunoguided surgery in gastric cancer using 131‐I labeled monoclonal antibody 3H11. Semin Surg Oncol. New York: John Wiley & Sons, Inc. 1994;10:88-94. DOI: 10.1002/ssu.2980100205.
  • Wang C, Wang Y, Su X, et al. Iodine-125 labeled monoclonal antibody 3H11: in radioimmunoguided surgery for primary gastric cancer. Zhonghua Wai Ke Za Zhi. 2000;38(7):507–509.
  • Ind T, Granowska M, Britton K, et al. Peroperative radioimmunodetection of ovarian carcinoma using a hand-held gamma detection probe. Br J Cancer. 1994;70(6):1263.
  • Adams S, Baum RP, Hertel A, et al. Intraoperative gamma probe detection of neuroendocrine tumors. J Nucl Med. 1998;39(7):1155.
  • Cuccurullo V, Di Stasio GD, Mansi L. Radioguided surgery with radiolabeled somatostatin analogs: not only in GEP-NETs. Nucl Med Rev Cent East Eur. 2017;20(1):49–56.
  • Cuntz MC, Levine EA, O’Dorisio TM, et al. Intraoperative gamma detection of 125 I-lanreotide in women with primary breast cancer. Ann Surg Oncol. 1999;6(4):367–372.
  • Einspieler I, Novotny A, Okur A, et al. First experience with image-guided resection of paraganglioma. Clin Nucl Med. 2014;39(8):e379–81.
  • Gay E, Vuillez JP, Palombi O, et al. Intraoperative and postoperative gamma detection of somatostatin receptors in bone-invasive en plaque meningiomas. Operative Neurosurg. 2005;57(suppl_1):107–113.
  • Grossrubatscher E, Vignati F, Dalino P, et al. Use of radioguided surgery with [111 In]-pentetreotide in the management of an ACTH-secreting bronchial carcinoid causing ectopic Cushing’s syndrome. J Endocrinol Invest. 2005;28(3):72–78.
  • Gulec SA, Baum R. Radio-guided surgery in neuroendocrine tumors. J Surg Oncol. 2007 Sep 15;96(4):309–315.
  • Hubalewska-Dydejczyk A, Kulig J, Szybinski P, et al. Radio-guided surgery with the use of [99m Tc-EDDA/HYNIC] octreotate in intra-operative detection of neuroendocrine tumours of the gastrointestinal tract. Eur J Nucl Med Mol Imaging. 2007;34(10):1545–1555.
  • Kaemmerer D, Prasad V, Daffner W, et al. Radioguided surgery in neuroendocrine tumors using Ga-68-labeled somatostatin analogs: a pilot study. Clin Nucl Med. 2012;37(2):142–147.
  • Mansi L, Rambaldi PF, Panza N, et al. Diagnosis and radioguided surgery with 111In-pentetreotide in a patient with paraneoplastic Cushing’s syndrome due to a bronchial carcinoid. Eur J Endocrinol. 1997;137(6):688–690.
  • Martelli H, Ricard M, Larroquet M, et al. Intraoperative localization of neuroblastoma in children with 123I-or 125I-radiolabeled metaiodobenzylguanidine. Surgery. 1998;123(1):51–57.
  • Öhrvall U, Westlin JE, Nilsson S, et al. Intraoperative gamma detection reveals abdominal endocrine tumors more efficiently than somatostatin receptor scintigraphy. Cancer: Interdiscip Int J Am Cancer Soc. 1997;80(S12):2490–2494.
  • Panareo S, Carcoforo P, Lanzara S, et al. Radiolabelled somatostatin analogs for diagnosis and radio-guided surgery of neuroendocrine breast cancer undetectable with conventional imaging procedures. Breast. 2008;17(1):111–114.
  • Ricard M, Tenenbaum F, Schlumberger M, et al. Intraoperative detection of pheochromocytoma with iodine-125 labelled meta-iodobenzylguanidine: a feasibility study. Eur J Nucl Med. 1993;20(5):426–430.
  • Sadowski SM, Millo C, Neychev V, et al. Feasibility of radio-guided surgery with 68 Gallium-DOTATATE in patients with gastro-entero-pancreatic neuroendocrine tumors. Ann Surg Oncol. 2015;22(3):676–682.
  • Sánchez N, Tapias A, Bowles H, et al. Multimodal approach in radioguided surgery in a case of multiple paraganglioma. Revista Española de Medicina Nuclear e Imagen Molecular (English Edition). 2018 Jan 01;37(1):41–45.
  • Schirmer WJ, O’Dorisio TM, Schirmer TP, et al. Intraoperative localization of neuroendocrine tumors with 125I-TYR (3)-octreotide and a hand-held gamma-detecting probe. Surgery. 1993;114(4):745–752.
  • van Hulsteijn LT, Corssmit EP, van der Hiel B, et al. Is there a role for radioguided surgery with iodine-labeled metaiodobenzylguanidine in resection of neuroendocrine tumors? Clin Nucl Med. 2012;37(11):1083–1088.
  • Woltering EA, Barrie R, O’Dorisio TM, et al. Detection of occult gastrinomas with iodine 125-labeled lanreotide and intraoperative gamma detection. Surgery. 1994;116(6):1139–1147.
  • Ashkenazy M, Davis L, Martin J. An evaluation of the technic and results of the radioactive di-iodo-fluorescein test for the localization of intracranial lesions. J Neurosurg. 1951;8(3):300–314.
  • Berland TL, Smith SL, Metzger PP, et al. Intraoperative gamma probe localization of the ureters: a novel concept. J Am Coll Surg. 2007 Oct 01;205(4):608–611.
  • van Oosterom MN, Simon H, Mengus L, et al. Revolutionizing (robot-assisted) laparoscopic gamma tracing using a drop-in gamma probe technology. Am J Nucl Med Mol Imaging. 2016;6(1):1–17.
  • Valdes Olmos RA, Vidal-Sicart S, Manca G, et al. Advances in radioguided surgery in oncology. Q J Nucl Med Mol Imaging. 2017 Sep;61(3):247–270.
  • Bowles H, Sánchez N, Tapias A, et al. Radioguided surgery and the GOSTT concept: from pre-operative image and intraoperative navigation to image-assisted excision. Revista Española de Medicina Nuclear e Imagen Molecular (English Edition). 2017 May 01;36(3):175–184.
  • Pouw B, de Wit‐van der Veen LJ, Stokkel MP, et al. Heading toward radioactive seed localization in non‐palpable breast cancer surgery? A meta‐analysis. J Surg Oncol. 2015;111(2):185–191.
  • Valdes Olmos RA, Vidal-Sicart S, Giammarile F, et al. The GOSTT concept and hybrid mixed/virtual/augmented reality environment radioguided surgery. Q J Nucl Med Mol Imaging. 2014 Jun;58(2):207–215.
  • Maurer T, Robu S, Schottelius M, et al. 99mTechnetium-based prostate-specific membrane antigen–radioguided surgery in recurrent prostate cancer. Eur Urol. 2019;75(4):659-666. DOI: 10.1016/j.eururo.2018.03.013.
  • Povoski SP, Neff RL, Mojzisik CM, et al. A comprehensive overview of radioguided surgery using gamma detection probe technology. World J Surg Oncol. 2009 Jan;27(7):11.
  • Heller S, Zanzonico P. Nuclear probes and intraoperative gamma cameras [review]. Semin Nucl Med. 2011 May;41(3):166–181.
  • Kang HG, Song SH, Han YB, et al. Proof-of-concept of a multimodal laparoscope for simultaneous NIR/gamma/visible imaging using wavelength division multiplexing. Optics express. 2018;26(7):8325-8339.
  • Maurer T, Weirich G, Schottelius M, et al. Prostate-specific membrane antigen–radioguided surgery for metastatic lymph nodes in prostate cancer. Eur Urol. 2015;68(3):530–534.
  • Woolfenden J, Barber HB. Design and use of radiation detector probes for intraoperative tumor detection using tumor-seeking radiotracers. in Freeman LM (ed): Nuclear Medicine Annual. Philadelphia, Lippincott-Raven Publishers, 1990, pp 151-173.
  • Vidal-Sicart S, Vermeeren L, Solà O, et al. The use of a portable gamma camera for preoperative lymphatic mapping: a comparison with a conventional gamma camera. Eur J Nucl Med Mol Imaging. 2011;38(4):636–641.
  • Warncke SH, Mattei A, Fuechsel FG, et al. Detection rate and operating time required for γ probe-guided sentinel lymph node resection after injection of technetium-99m nanocolloid into the prostate with and without preoperative imaging. Eur Urol. 2007;52(1):126–133.
  • KleinJan GH, van Den Berg NS, de Jong J, et al. Multimodal hybrid imaging agents for sentinel node mapping as a means to (re) connect nuclear medicine to advances made in robot-assisted surgery. Eur J Nucl Med Mol Imaging. 2016;43(7):1278–1287.
  • Ohyama C, Chiba Y, Yamazaki T, et al. Lymphatic mapping and gamma probe guided laparoscopic biopsy of sentinel lymph node in patients with clinical stage I testicular tumor. J Urol. 2002 Oct;168(4 Pt 1):1390–1395.
  • van der Poel HG, Buckle T, Brouwer OR, et al. Intraoperative laparoscopic fluorescence guidance to the sentinel lymph node in prostate cancer patients: clinical proof of concept of an integrated functional imaging approach using a multimodal tracer. Eur Urol. 2011 Oct;60(4):826–833.
  • Jeschke S, Beri A, Grüll M, et al. Laparoscopic radioisotope-guided sentinel lymph node dissection in staging of prostate cancer. Eur Urol. 2008;53(1):126–133.
  • Vermeeren L, Olmos RV, Meinhardt W, et al. Intraoperative radioguidance with a portable gamma camera: a novel technique for laparoscopic sentinel node localisation in urological malignancies. Eur J Nucl Med Mol Imaging. 2009;36(7):1029–1036.
  • Kitagawa Y, Kitajima M. Gastrointestinal cancer and sentinel node navigation surgery. J Surg Oncol. 2002;79(3):188–193.
  • Kitagawa Y, Kitano S, Kubota T, et al. Minimally invasive surgery for gastric cancer—toward a confluence of two major streams: a review. Gastric Cancer. 2005;8(2):103–110.
  • Acar C, Kleinjan GH, van Den Berg NS, et al. Advances in sentinel node dissection in prostate cancer from a technical perspective. Int J Urol. 2015 Oct;22(10):898–909.
  • Meershoek P, van Oosterom MN, Simon H, et al. Robot-assisted laparoscopic surgery using DROP-IN radioguidance: first-in-human translation. Eur J Nucl Med Mol Imaging. 2019 Jan; 46(1): 49–53.
  • Leeuwen F, Oosterom M, Meershoek P, et al. Minimal-invasive robot-assisted image-guided resection of PSMA-positive lymph nodes in recurrent prostate cancer. Clin Nucl Med. 2019;44(7):580–581.
  • Povoski SP, Hall NC, Murrey DA, et al. Multimodal imaging and detection approach to 18 F-FDG-directed surgery for patients with known or suspected malignancies: a comprehensive description of the specific methodology utilized in a single-institution cumulative retrospective experience. World J Surg Oncol. 2011;9(1):152.
  • Povoski SP, Murrey DA, Hall NC. 18 F-FDG-directed surgery and 18 F-FDG-directed interventional procedures. In: Herrmann K, Nieweg OE, Povoski SP, eds. Radioguided Surgery. Switzerland: Springer International; 2016. p. 419–445.
  • Daghighian F, Fong Y. Detectors for intraoperative molecular imaging: from probes to scanners. In: Fong Y., Giulianotti P., Lewis J, et al, eds. Imaging and visualization in the modern operating room. New York, NY: Springer; 2015. p. 55–67.
  • Gulec SA, Daghighian F, Essner R. PET-Probe: evaluation of technical performance and clinical utility of a handheld high-energy gamma probe in oncologic surgery. Ann Surg Oncol. 2016;23(5):9020–9027.
  • García JR, Fraile M, Soler M, et al. PET/CT-guided salvage surgery protocol. Results with ROLL technique and PET probe. Revista Española De Medicina Nuclear (english Edition). 2011 July 01;30(4):217–222.
  • Gulec SA, Hoenie E, Hostetter R, et al. PET probe-guided surgery: applications and clinical protocol. World J Surg Oncol. 2007 Jun;7(5):65.
  • Freesmeyer M, Wurst C, Überrueck T, et al. Intraoperative identification of a neuro endocrine tumour diagnosed by 68Ga-DOTATOC PET but undetectable by surgical palpation or conventional imaging. Nuklearmedizin. 2009;48(05):N50–N51.
  • González SJ, González L, Wong J, et al. An analysis of the utility of handheld PET probes for the intraoperative localization of malignant tissue. J Gastrointestinal Surg. 2011;15(2):358–366.
  • Heuveling DA, Karagozoglu KH, Van Lingen A, et al. Feasibility of intraoperative detection of sentinel lymph nodes with 89-zirconium-labelled nanocolloidal albumin PET-CT and a handheld high-energy gamma probe. EJNMMI Res. 2018;8(1):15.
  • Barranger E, Kerrou K, Petegnief Y, et al. Laparoscopic resection of occult metastasis using the combination of FDG-positron emission tomography/computed tomography image fusion with intraoperative probe guidance in a woman with recurrent ovarian cancer. Gynecol Oncol. 2005;96(1):241–244.
  • De Jong J, Van Ginkel R, Slart R, et al. FDG-PET probe-guided surgery for recurrent retroperitoneal testicular tumor recurrences. Eur J Surg Oncol. 2010;36(11):1092–1095.
  • Martin EW, Chapman GJ, Subramaniam VV, et al. Intraoperative detection of gamma emissions using K-alpha X-ray fluorescence. Expert Rev Med Devices. 2010;7(4):431–434.
  • Povoski SP, Chapman GJ, Murrey DA, et al. Intraoperative detection of 18 F-FDG-avid tissue sites using the increased probe counting efficiency of the K-alpha probe design and variance-based statistical analysis with the three-sigma criteria. BMC Cancer. 2013;13(1):98.
  • Collamati F, Bocci V, Castellucci P, et al. Radioguided surgery with β radiation: a novel application with Ga 68. Sci Rep. 2018;8(1):16171.
  • Camillocci ES, Baroni G, Bellini F, et al. A novel radioguided surgery technique exploiting β− decays. Sci Rep. 2014;4:4401.
  • Monge F, Shakir DI, Lejeune F, et al. Acquisition models in intraoperative positron surface imaging. Int J Comput Assist Radiol Surg. 2017;12(4):691–703.
  • Essner R, Daghighian F, Giuliano AE. Advances in FDG PET probes in surgical oncology. Cancer J. 2002;8(2):100–108.
  • Yen T-C, See L-C, Lai C-H, et al. 18F-FDG uptake in squamous cell carcinoma of the cervix is correlated with glucose transporter 1 expression. J Nucl Med. 2004;45(1):22–29.
  • Zervos EE, Desai DC, DePalatis LR, et al. 18F-labeled fluorodeoxyglucose positron emission tomography-guided surgery for recurrent colorectal cancer: a feasibility study. J Surg Res. 2001;97(1):9–13.
  • Piert M, Burian M, Meisetschläger G, et al. Positron detection for the intraoperative localisation of cancer deposits. Eur J Nucl Med Mol Imaging. 2007;34(10):1534–1544.
  • Strong VE, Humm J, Russo P, et al. A novel method to localize antibody-targeted cancer deposits intraoperatively using handheld PET beta and gamma probes. Surg Endosc. 2008 Feb;22(2):386–391.
  • Camillocci ES, Schiariti M, Bocci V, et al. First ex vivo validation of a radioguided surgery technique with β-radiation. Phys Med. 2016;32(9):1139–1144.
  • Daghighian F, Mazziotta JC, Hoffman EJ, et al. Intraoperative beta probe: a device for detecting tissue labeled with positron or electron emitting isotopes during surgery. Med Phys. 1994;21(1):153–157.
  • Russomando A, Bellini F, Bocci V, et al. An intraoperative beta-detecting probe for radio-guided surgery in tumour resection. IEEE Trans Nucl Sci. 2016;63(5):2533–2539.
  • Yamamoto S, Matsumoto K, Sakamoto S, et al. An intra-operative positron probe with background rejection capability for FDG-guided surgery. Ann Nucl Med. 2005;19(1):23.
  • Mester C, Bruschini C, Magro P, et al., editors. A handheld β+ probe for intra-operative detection of radiotracers. Limerick, Ireland: SENSORS, 2011. IEEE. DOI: 10.1109/ICSENS.2011.612714.
  • Strauss HW, Mari C, Patt BE, et al. Intravascular radiation detectors for the detection of vulnerable atheroma. J Am Coll Cardiol. 2006;47(8 Supplement):C97–C100.
  • Lombardi A, Nigri G, Scopinaro F, et al. High-resolution, handheld camera use for occult breast lesion localization plus sentinel node biopsy (SNOLL): a single-institution experience with 186 patients. Surgeon. 2015 Apr;13(2):69–72.
  • Tsuchimochi M, Hayama K. Intraoperative gamma cameras for radioguided surgery: technical characteristics, performance parameters, and clinical applications. Phys Med. 2013 Mar 01;29(2):126–138.
  • Hellingman D, Vidal-Sicart S. The use of intraoperative small and large field of view gamma cameras for radioguided surgery. In: Herrmann K, Nieweg OE, Povoski SP, eds. Radioguided Surgery. Switzerland: Springer International Publishing; 2016. p. 35–56.
  • Bugby SL, Lees JE, Bhatia B, et al. Characterisation of a high resolution small field of view portable gamma camera. Phys Med. 2014;30(3):331–339.
  • Sajedi S, Sabet H, Choi HS. Intraoperative biophotonic imaging systems for image-guided interventions. Nanophotonics. 2018;8(1):99–116.
  • KleinJan GH, Hellingman D, van Den Berg NS, et al. Hybrid surgical guidance: does hardware integration of γ-and fluorescence imaging modalities make sense? J Nucl Med. 2017;58(4):646–650.
  • Hellingman D, Klop WMC, Olmos RAV. Detecting near-the-injection-site sentinel nodes in head and neck melanomas with a high-resolution portable gamma camera. Clin Nucl Med. 2015;40(1):e11–e16.
  • Engelen T, Winkel BM, Rietbergen DD, et al. The next evolution in radioguided surgery: breast cancer related sentinel node localization using a freehandSPECT-mobile gamma camera combination. Am J Nucl Med Mol Imaging. 2015;5(3):233–245.
  • Hellingman D, Vidal-Sicart S, Paredes P, et al. A new portable hybrid camera for fused optical and scintigraphic imaging: first clinical experiences. Clin Nucl Med. 2016;41(1):e39–e43.
  • Lees JE, Bugby SL, Bark A, et al. A hybrid camera for locating sources of gamma radiation in the environment. J Instrum. 2013;8(10):P10021.
  • Bugby S, Lees J, Ng A, et al. Investigation of an SFOV hybrid gamma camera for thyroid imaging. Phys Med. 2016;32(1):290–296.
  • Yamamoto S, Higashi T, Matsumoto K, et al. Development of a positron-imaging detector with background rejection capability. Ann Nucl Med. 2006;20(10):655.
  • Singh B, Stack BC, Thacker S, et al. A hand-held beta imaging probe for FDG. Ann Nucl Med. 2013;27(3):203–208.
  • Sabet H, Stack BC, Nagarkar VV. A hand-held, intra-operative positron imaging probe for surgical applications. IEEE Trans Nucl Sci. 2015;62(5):1927–1934.
  • Verdier M-A, Spadola S, Pinot L, et al. Gamma-background rejection method for a dual scintillator positron probe dedicated to radio-guided surgery. Nucl Instrum Methods Phys Res A. 2018;912:315–319.
  • Spadola S, Verdier M-A, Pinot L, et al. Design optimization and performances of an intraoperative positron imaging probe for radioguided cancer surgery. J Instrum. 2016;11(12):P12019.
  • Solestizi LA, Biasini M, Bocci V, et al. Use of a CMOS image sensor for beta-emitting radionuclide measurements. J Instrum. 2018;13(07):P07003.
  • Waelkens P, van Oosterom MN, van Den Berg NS, et al. Surgical navigation: an overview of the state-of-the-art clinical applications. In: Herrmann K, Nieweg OE, Povoski SP, editors. Radioguided Surgery. Switzerland: Springer International Publishing; 2016. p. 57–73.
  • van Oosterom MN, van der Poel HG, Navab N, et al. Computer-assisted surgery: virtual-and augmented-reality displays for navigation during urological interventions. Curr Opin Urol. 2018;28(2):205–213.
  • Didolkar MM, Anderson ME, Hochman MG, et al. Image guided core needle biopsy of musculoskeletal lesions: are nondiagnostic results clinically useful? Clin Orthop Relat Res. 2013;471(11):3601–3609.
  • Maciel MJS, Tyng CJ, Barbosa PNVP, et al. Computed tomography-guided percutaneous biopsy of bone lesions: rate of diagnostic success and complications. Radiol Bras. 2014;47(5):269–274.
  • El-Haddad G. PET-based percutaneous needle biopsy. PET Clin. 2016 Jul;11(3):333–349.
  • Kumar R, Mittal BR, Bhattacharya A, et al. 18F-FDG PET/CT-guided real-time automated robotic arm–assisted needle navigation for percutaneous biopsy of hypermetabolic bone lesions: diagnostic performance and clinical impact. Am J Roentgenol. 2019;212(1):W10–W18.
  • Radhakrishnan RK, Mittal BR, Gorla AKR, et al. Real-time intraprocedural 18F-FDG PET/CT-guided biopsy using automated robopsy arm (ARA) in the diagnostic evaluation of thoracic lesions with prior inconclusive biopsy results: initial experience from a tertiary health care centre. Br J Radiol. 2017;90(1080):20170258.
  • Hua Q, Zhu X, Zhang L, et al. Initial experience with real-time hybrid single-photon emission computed tomography/computed tomography-guided percutaneous transthoracic needle biopsy. Nucl Med Commun. 2017;38(6):556–560.
  • Wiegmann AL, Broucek JR, Fletcher RN, et al. Image-guided navigation in lymph node biopsy. JSLS. 2018;22(2).
  • Brouwer OR, Buckle T, Bunschoten A, et al. Image navigation as a means to expand the boundaries of fluorescence-guided surgery. Phys Med Biol. 2012;57(10):3123.
  • Brouwer OR, van Den Berg NS, Mathéron HM, et al. Feasibility of intraoperative navigation to the sentinel node in the groin using preoperatively acquired single photon emission computerized tomography data: transferring functional imaging to the operating room. J Urol. 2014;192(6):1810–1816.
  • van Den Berg NS, Engelen T, Brouwer OR, et al. A pilot study of SPECT/CT-based mixed-reality navigation towards the sentinel node in patients with melanoma or merkel cell carcinoma of a lower extremity. Nucl Med Commun. 2016;37(8):812–817.
  • Rahbar K, Colombo-Benkmann M, Haane C, et al. Intraoperative 3-D mapping of parathyroid adenoma using freehand SPECT. EJNMMI Res. 2012;2(1):51.
  • van Oosterom MN, Meershoek P, KleinJan GH, et al. Navigation of fluorescence cameras during soft tissue surgery—is it possible to use a single navigation setup for various open and laparoscopic urological surgery applications?. J Urol. 2018;199(4):1061–1068.
  • KleinJan GH, van Den Berg NS, van Oosterom MN, et al. Toward (hybrid) navigation of a fluorescence camera in an open surgery setting. J Nucl Med. 2016;57(10):1650–1653.
  • van Oosterom MN, Engelen MA, van Den Berg NS, et al. Navigation of a robot-integrated fluorescence laparoscope in preoperative SPECT/CT and intraoperative freehand SPECT imaging data: a phantom study. J Biomed Opt. 2016;21(8):086008.
  • Wendler T, Herrmann K, Schnelzer A, et al. First demonstration of 3-D lymphatic mapping in breast cancer using freehand SPECT. Eur J Nucl Med Mol Imaging. 2010;37(8):1452–1461.
  • Bluemel C, Matthies P, Herrmann K, et al. 3D scintigraphic imaging and navigation in radioguided surgery: freehand SPECT technology and its clinical applications. Expert Rev Med Devices. 2016;13(4):339–351.
  • Hartl A, Shakir DI, Kojchev R, et al. Freehand SPECT reconstructions using look up tables. Medical Imaging 2012: Image-Guided Procedures, Robotic Interventions, and Modeling. 2012. International Society for Optics and Photonics.
  • Hartl A, Shakir DI, Lasser T, et al. Detection models for freehand spect reconstruction. Phys Med Biol. 2015;60(3):1031.
  • KleinJan GH, Karakullukcu B, Klop WMC, et al. Introducing navigation during melanoma-related sentinel lymph node procedures in the head-and-neck region. EJNMMI Res. 2017 Aug 17;7(1):65.
  • Bluemel C, Schnelzer A, Okur A, et al. Freehand SPECT for image-guided sentinel lymph node biopsy in breast cancer. Eur J Nucl Med Mol Imaging. 2013;40(11):1656–1661.
  • Heuveling DA, van Weert S, Karagozoglu KH, et al. Evaluation of the use of freehand SPECT for sentinel node biopsy in early stage oral carcinoma. Oral Oncol. 2015;51(3):287–290.
  • Pouw B, de Wit-van der Veen LJ, van Duijnhoven F, et al. Intraoperative 3D navigation for single or multiple 125i-seed localization in breast-preserving cancer surgery. Clin Nucl Med. 2016 May;41(5):e216–20.
  • Opfermann T, Winkens T, Freesmeyer M, et al. Radiation exposure of the investigator’s hand during fusion imaging of the thyroid with 99mTcO4-free-hand SPECT and ultrasound. Radiat Prot Dosimetry. 2015;168(4):531–536.
  • Markus A, Ray ASC, Bolla D, et al. Sentinel lymph node biopsy in endometrial and cervical cancers using freehand SPECT—first experiences. J Gynecological Surg. 2016;13(4):499–506.
  • Müller J, Putora PM, Schneider T, et al. Handheld single photon emission computed tomography (handheld SPECT) navigated video-assisted thoracoscopic surgery of computer tomography-guided radioactively marked pulmonary lesions. Interact Cardiovasc Thorac Surg. 2016;23(3):345–350.
  • Fuerst B, Sprung J, Pinto F, et al. First robotic SPECT for minimally invasive sentinel lymph node mapping. IEEE Trans Med Imaging. 2016;35(3):830–838.
  • Bluemel C, Herrmann K, Kübler A, et al. Intraoperative 3-D imaging improves sentinel lymph node biopsy in oral cancer. Eur J Nucl Med Mol Imaging. 2014;41(12):2257–2264.
  • Bluemel C, Herrmann K, Müller–Richter U, et al. Freehand SPECT‐guided sentinel lymph node biopsy in early oral squamous cell carcinoma. Head Neck. 2014;36(11):E112–E116.
  • Mandapathil M, Teymoortash A, Heinis J, et al. Freehand SPECT for sentinel lymph node detection in patients with head and neck cancer: first experiences. Acta Otolaryngol. 2014 Jan;134(1):100–104.
  • Schilling C, Gnansegaran G, Thavaraj S, et al. Intraoperative sentinel node imaging versus SPECT/CT in oral cancer–A blinded comparison. Eur J Surg Oncol. 2018;44(12):1901–1907.
  • Mihaljevic A, Rieger A, Belloni B, et al. Transferring innovative freehand SPECT to the operating room: first experiences with sentinel lymph node biopsy in malignant melanoma. Eur J Surg Oncol. 2014;40(1):42–48.
  • Sulzbacher L, Klinger M, Scheurecker C, et al. Clinical usefulness of a novel freehand 3D imaging device for radio-guided intraoperative sentinel lymph node detection in malignant melanoma. Clin Nucl Med. 2015 Sep;40(9):e436–40.
  • Castillo V, Rioja ME, Diaz-Laugart E, et al. Intraoperative demonstration of 3D tumor localization using freehand SPECT in a duodenum NET patient. J Nucl Med. 2012;53(supplement 1):1252.
  • Rietbergen D, van der Hage J, Meershoek P, et al. FreehandSPECT with 99mTc-HDP can be used to guide percutaneous biopsy of skeletal lesions detected on bone scintigraphy. Eur J Surg Oncol. 2019;45(2):e87.
  • Wendler T, Traub J, Ziegler SI, et al., editors. Navigated three dimensional beta probe for optimal cancer resection. International Conference on Medical Image Computing and Computer-Assisted Intervention; 2006: Copenhagen, Denmark: Springer.
  • Welling MM, Spa SJ, van Willigen DM, et al. In vivo stability of supramolecular host–guest complexes monitored by dual-isotope multiplexing in a pre-targeting model of experimental liver radioembolization. J Control Release. 2019;293:126–134.
  • Goorden MC, van der Have F, Kreuger R, et al. VECTor: a preclinical imaging system for simultaneous submillimeter SPECT and PET. J Nucl Med. 2013;54(2):306–312.
  • Dauer LT, Thornton C, Miodownik D, et al. Radioactive seed localization with 125I for nonpalpable lesions prior to breast lumpectomy and/or excisional biopsy: methodology, safety, and experience of initial year. Health Phys. 2013;105(4):356–365.
  • Meinhardt W, van der Poel H, Valdés Olmos R, et al. Laparoscopic sentinel lymph node biopsy for prostate cancer: the relevance of locations outside the extended dissection area. In: Prostate cancer. 2012. Doi: 10.1155/2012/751753
  • Tipnis S, Nagarkar V, Shestakova I, et al. Feasibility of a beta-gamma digital imaging probe for radioguided surgery. IEEE Trans Nucl Sci. 2004;51(1):110–116.
  • Bugby SL, Lees JE, Perkins AC. Hybrid intraoperative imaging techniques in radioguided surgery: present clinical applications and future outlook. Clin Transl Imaging. 2017;5(4):323–341.
  • Gambini JP, Quinn TP. Hybrid tracers and devices for intraoperative imaging: the future for radioguided surgery? Clin Transl Imaging. 2016;4(5):343–351.
  • van Leeuwen FW, Hardwick JC, van Erkel AR. Luminescence-based imaging approaches in the field of interventional molecular imaging. Radiology. 2015;276(1):12–29.
  • Stoffels I, Dissemond J, Pöppel T, et al. Intraoperative fluorescence imaging for sentinel lymph node detection: prospective clinical trial to compare the usefulness of indocyanine green vs technetium Tc 99m for identification of sentinel lymph nodes. JAMA Surg. 2015;150(7):617–623.
  • KleinJan GH, van Werkhoven E, van Den Berg NS, et al. The best of both worlds: a hybrid approach for optimal pre- and intraoperative identification of sentinel lymph nodes. Eur J Nucl Med Mol Imag. 2018 Apr 25;45(11):1915–1925.
  • Schaafsma BE, Verbeek FP, Rietbergen DD, et al. Clinical trial of combined radio‐and fluorescence‐guided sentinel lymph node biopsy in breast cancer. Br J Surg. 2013;100(8):1037–1044.
  • Paredes P, Vidal-Sicart S, Campos F, et al. Role of ICG-99m Tc-nanocolloid for sentinel lymph node detection in cervical cancer: a pilot study. Eur J Nucl Med Mol Imaging. 2017;44(11):1853–1861.
  • Brouwer OR, van Den Berg NS, Matheron HM, et al. A hybrid radioactive and fluorescent tracer for sentinel node biopsy in penile carcinoma as a potential replacement for blue dye. Eur Urol. 2014;65(3):600–609.
  • KleinJan GH, van Den Berg NS, Brouwer OR, et al. Optimisation of fluorescence guidance during robot-assisted laparoscopic sentinel node biopsy for prostate cancer. Eur Urol. 2014 Dec;66(6):991–998.
  • Stoffels I, Leyh J, Pöppel T, et al. Evaluation of a radioactive and fluorescent hybrid tracer for sentinel lymph node biopsy in head and neck malignancies: prospective randomized clinical trial to compare ICG-99m Tc-nanocolloid hybrid tracer versus 99m Tc-nanocolloid. Eur J Nucl Med Mol Imaging. 2015;42(11):1631–1638.
  • Christensen A, Juhl K, Charabi B, et al. Feasibility of real-time near-infrared fluorescence tracer imaging in sentinel node biopsy for oral cavity cancer patients. Ann Surg Oncol. 2016;23(2):565–572.
  • KleinJan G, Brouwer O, Mathéron HM, et al. Hybrid radioguided occult lesion localization (hybrid ROLL) of 18F-FDG-avid lesions using the hybrid tracer indocyanine green-99mTc-nanocolloid. Revista Española de Medicina Nuclear e Imagen Molecular (English Edition). 2016;35(5):292–297.
  • van Den Berg NS, Simon H, Kleinjan GH, et al. First-in-human evaluation of a hybrid modality that allows combined radio- and (near-infrared) fluorescence tracing during surgery [Article]. Eur J Nucl Med Mol Imaging. 2015 Oct;42(11):1639–1647.
  • Vidal-Sicart S, Seva A, Campos F, et al. Clinical use of an opto-nuclear probe for hybrid sentinel node biopsy guidance: first results. Int J Comput Assist Radiol Surg. 2019 Feb;14(2):409-416. DOI: 10.1007/s11548-018-1816-5.
  • Poumellec M, Dejode M, Figl A, et al. Sentinel node detection using optonuclear probe (gamma and fluorescence) after green indocyanine and radio-isotope injections. Gynecol Obstet Fertil. 2016;44(4):207–210.
  • Lees J, Bugby S, Alqahtani M, et al. A multimodality hybrid gamma-optical camera for intraoperative imaging. Sensors. 2017;17(3):554.
  • Ciarrocchi E, Belcari N. Cerenkov luminescence imaging: physics principles and potential applications in biomedical sciences. EJNMMI Phys. 2017;4(1):14.
  • Das S, Thorek DL, Grimm J. Cerenkov imaging. Adv Cancer Res. 2014;124:213–234. Elsevier.
  • Spinelli AE, Boschi F. Novel biomedical applications of Cerenkov radiation and radioluminescence imaging. Phys Med. 2015;31(2):120–129.
  • Cherenkov PA. Visible emission of clean liquids by action of γ radiation. Dokl Akad Nauk SSSR. 1934;2:451.
  • Jelley J. Cerenkov radiation and its applications. Br J Appl Phys. 1955;6(7):227.
  • Glaser AK, Zhang R, Andreozzi JM, et al. Cherenkov radiation fluence estimates in tissue for molecular imaging and therapy applications. Phys Med Biol. 2015;60(17):6701.
  • Thorek DL, Robertson R, Bacchus WA, et al. Cerenkov imaging-a new modality for molecular imaging. Am J Nucl Med Mol Imaging. 2012;2(2):163.
  • Chin PT, Welling MM, Meskers SC, et al. Optical imaging as an expansion of nuclear medicine: cerenkov-based luminescence vs fluorescence-based luminescence. Eur J Nucl Med Mol Imaging. 2013;40(8):1283–1291.
  • Gill RK, Mitchell GS, Cherry SR. Computed Cerenkov luminescence yields for radionuclides used in biology and medicine. Phys Med Biol. 2015;60(11):4263.
  • Boschi F, E Spinelli A. Cerenkov luminescence imaging at a glance. Current Mol Imaging. 2014;3(2):106–117.
  • Thorek DL, Riedl CC, Grimm J. Clinical Cerenkov luminescence imaging of 18F-FDG. J Nucl Med. 2014;55(1):95–98.
  • Spinelli AE, Ferdeghini M, Cavedon C, et al. First human cerenkography. J Biomed Opt. 2013;18(2):020502.
  • Jarvis LA, Zhang R, Gladstone DJ, et al. Cherenkov video imaging allows for the first visualization of radiation therapy in real time. Int J Radiat Oncol* Biol* Phys. 2014;89(3):615–622.
  • Hu H, Cao X, Kang F, et al. Feasibility study of novel endoscopic Cerenkov luminescence imaging system in detecting and quantifying gastrointestinal disease: first human results. Eur Radiol. 2015;25(6):1814–1822.
  • Yang Y, Biswal NC, Wang T, et al. Potential role of a hybrid intraoperative probe based on OCT and positron detection for ovarian cancer detection and characterization. Biomed Opt Express. 2011;2(7):1918–1930.
  • Gamelin J, Yang Y, Biswal N, et al. A prototype hybrid intraoperative probe for ovarian cancer detection. Opt Express. 2009;17(9):7245–7258.
  • Pani R, Pellegrini R, Cinti M, et al. Integrated ultrasound and gamma imaging probe for medical diagnosis. J Instrum. 2016;11(03):C03037.
  • Okur A, Hennersperger C, Runyan B, et al., editors. FhSPECT-US guided needle biopsy of sentinel lymph nodes in the axilla: is it feasible? International Conference on Medical Image Computing and Computer-Assisted Intervention; 2014: Boston, MA, USA: Springer.
  • Freesmeyer M, Opfermann T, Winkens T. Hybrid integration of real-time US and freehand SPECT: proof of concept in patients with thyroid diseases. Radiology. 2014;271(3):856–861.
  • Bluemel C, Kirchner P, Kajdi GW, et al. Localization of parathyroid adenoma with real-time ultrasound: freehand SPECT fusion. Clin Nucl Med. 2016;41(3):e141–2.
  • de Bree R, Pouw B, Heuveling DA, et al. Fusion of freehand SPECT and ultrasound to perform ultrasound-guided fine-needle aspiration cytology of sentinel nodes in head and neck cancer. AJNR Am J Neuroradiol. 2015 Nov;36(11):2153–2158.
  • Freesmeyer M, Winkens T, Opfermann T, et al. Real-time ultrasound and freehand-SPECT. Nuklearmed Nucl Med. 2014;53(06):259–264.
  • Bluemel C, Safak G, Cramer A, et al. Fusion of freehand SPECT and ultrasound: first experience in preoperative localization of sentinel lymph nodes. Eur J Nucl Med Mol Imaging. 2016;43(13):2304–2312.
  • Horn T, Krönke M, Rauscher I, et al. Single lesion on prostate-specific membrane antigen-ligand positron emission tomography and low prostate-specific antigen are prognostic factors for a favorable biochemical response to prostate-specific membrane antigen-targeted radioguided surgery in recurrent prostate cancer. Eur Urol. 2019 Apr 12. pii: S0302-2838(19)30278-7. doi: 10.1016/j.eururo.2019.03.045.
  • Schottelius M, Wurzer A, Wissmiller K, et al. Synthesis and preclinical characterization of the PSMA-targeted hybrid tracer PSMA-I&F for nuclear and fluorescence imaging of prostate cancer. J Nucl Med. 2019;60(1):71–78.
  • Santini C, Kuil J, Bunschoten A, et al. Evaluation of a fluorescent and radiolabeled hybrid somatostatin analog in vitro and in mice bearing H69 neuroendocrine xenografts. J Nucl Med. 2016;57(8):1289–1295.
  • Wang X, Aldrich MB, Marshall MV, et al. Preclinical characterization and validation of a dual-labeled trastuzumab-based imaging agent for diagnosing breast cancer. Chin J Cancer Res. 2015;27(1):74.
  • Hekman MC, Boerman OC, de Weijert M, et al. Targeted dual-modality imaging in renal cell carcinoma: an ex vivo kidney perfusion study. Clin Cancer Res. 2016;22(18):4634–4642.
  • Brouwer OR, Klop WMC, Buckle T, et al. Feasibility of sentinel node biopsy in head and neck melanoma using a hybrid radioactive and fluorescent tracer. Ann Surg Oncol. 2012;19(6):1988–1994.
  • Vidal-Sicart S, Orsini F, Giammarile F, et al. Radioguided surgery for malignant melanoma. In: Strauss H., Mariani G., Volterrani D, eds. Nuclear oncology: from pathophysiology to clinical applications. Switzerland: Springer 2016. p. 1–33.
  • Bunschoten A, van Den Berg NS, Olmos RAV, et al. Tracers applied in radioguided surgery. In: Herrmann K, Nieweg OE, Povoski SP, eds. Radioguided Surgery. Switzerland: Springer International Publishing; 2016. p. 75–101. DOI: 10.1007/978-3-319-26051-8_5.
  • Franc BL, Mari C, Johnson D, et al. The role of a positron-and high-energy gamma photon probe in intraoperative localization of recurrent melanoma. Clin Nucl Med. 2005;30(12):787–791.
  • Mancini-Terracciano C, Donnarumma R, Bencivenga G, et al. Feasibility of beta-particle radioguided surgery for a variety of “nuclear medicine” radionuclides. Phys Med. 2017;43:127–133.
  • Kumar R, Mittal BR, Bhattacharya A, et al. Diagnostic performance of real-time robotic arm-assisted 18 f-fdg pet/ct-guided percutaneous biopsy in metabolically active abdominal and pelvic lesions. European Journal Of Nuclear Medicine and Molecular Imaging. 2019;46(4):838-847.
  • Van Oosterom MN, Meershoek P, Welling MM, et al. Extending the hybrid surgical guidance concept with freehand fluorescence tomography. Ieee Transactions on Medical Imaging 2019. DOI: 10.1109/TMI.2019.2924254
  • van Manen L, Dijkstra J, Boccara C, et al. The clinical usefulness of optical coherence tomography during cancer interventions. J Cancer Res Clin Oncol. 2018;144(10):1967–1990.
  • van Leeuwen FW, Valdés-Olmos R, Buckle T, et al. Hybrid surgical guidance based on the integration of radionuclear and optical technologies. Br J Radiol. 2016;89(1062):20150797.