• Keine Ergebnisse gefunden

Let’s embrace optical imaging: a growing branch on the clinical molecular imaging tree

N/A
N/A
Protected

Academic year: 2022

Aktie "Let’s embrace optical imaging: a growing branch on the clinical molecular imaging tree"

Copied!
9
0
0

Wird geladen.... (Jetzt Volltext ansehen)

Volltext

(1)

https://doi.org/10.1007/s00259-021-05476-z EDITORIAL

Let’s embrace optical imaging: a growing branch on the clinical molecular imaging tree

Milou E. Noltes1,2 · Gooitzen M. van Dam1,3 · Wouter B. Nagengast4 · Pieter J. van der Zaag1,5 · Riemer H. J. A. Slart1,6 · Wiktor Szymanski7,8 · Schelto Kruijff1,2 · Rudi A. J. O. Dierckx1,8

© The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature 2021

The history of nuclear medicine (and molecular imaging)

Nuclear medicine first became recognised as a potential medical speciality in the early 1950s of the last century when Seidlin reported on the success of radioactive iodine (I-131) in treating a patient with advanced thyroid cancer [1, 2]. The use of the rectilinear scanner contributed to the widespread clinical use of diagnostic nuclear medicine and resulted in planar, somewhat spotty images which constituted of plotted dots [3]. Importantly, this methodology already acted on the tracer principle, i.e. the use of a radioactive compound (radi- opharmaceutical) to visualise in vivo processes and to reveal physiological and pathophysiological changes. In those days, nuclear medicine often served as an alternative approach or an add-on tool to the radiological morphological imaging techniques [4]. Demonstrating, for example, a blood–brain barrier disruption using brain diethylenetriaminepentaacetic acid (DTPA) scans as a sign for a possible brain tumour was certainly — at the time — less invasive when compared

to the radiological pneumoventriculography, which used air injected in the ventricular system as an indirect contrast for adjacent brain tumours. 99mTc colloid liver scans, for example, added to the detection of hepatic defects in case of primary liver tumours or liver metastases [5]. Due to the clinical breakthroughs over the following decades in radiol- ogy, in computed tomography (CT) and in magnetic reso- nance imaging (MRI) with increasing resolution and accu- racy, the clinical dissemination of these imaging techniques was rapid. In addition, this development resulted in a reduc- tion of the nuclear medicine scans of the aforementioned type. In parallel, nuclear medicine itself as an imaging field saw the development of single-photon emission computed tomography (SPECT) [6] and positron emission tomogra- phy (PET) [7, 8] imaging modalities, benefitting from the ever-increasing computation power and continuous improve- ments in hardware detection systems, such as new crystals and digital detectors [6, 9, 10]. The clinical deployment of SPECT was boosted by the availability and development of 99mTc generators and pharmaceutical kits [11], allowing more efficient and cost-effective in-house institutional radi- opharmaceutical labelling. Similarly, clinical PET imaging

This article is part of the Topical Collection on Editorial

* Rudi A. J. O. Dierckx r.a.dierckx@umcg.nl

1 Medical Imaging Center,

Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands

2 Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands

3 AxelaRx/TRACER B.V, Groningen, the Netherlands

4 Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands

5 Molecular Biophysics, Zernike Institute, University of Groningen, Nijenborgh 4, 9747 AG Groningen, the Netherlands

6 Faculty of Science and Technology, Department of Biomedical Photonic Imaging, University of Twente, Enschede, Netherlands

7 Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 7, 9747AG Groningen, the Netherlands

8 Medical Imaging Center, Department of Radiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands

Published online: 31 August 2021

(2)

disseminated rapidly in the hospital environment with the breakthrough and growing availability of fluorodeoxyglu- cose (18F-FDG) [12] for oncological indications. Due to fast and accurate computational capacity, nuclear medicine was able to make quantum leaps, made possible by, for example, iterative image reconstruction rather than filtered back pro- jection, combined with corrections for time of flight, depth of interaction and movement [7, 13]. As a next step, the nuclear clinical imaging systems converged with radiologi- cal imaging into hybrid systems, as SPECT-CT, PET-CT or PET-MRI, adding the advantages of radiological systems to the nuclear imaging techniques, with the latest addition being total body PET-CT [14]. Today, developments are continuously pushing the existing frontiers, including, for example, artificial intelligence, nanotechnology and secured giant cloud-based data servers for global shared data ware- houses combining imaging data with clinical, histopatho- logical and patient outcome datasets.

The rise of molecular imaging

Molecular imaging, developed by ‘the father of molecular imaging’, the late Dr. Sanjiv Gambhir, is defined as ‘the visualization, characterization, and measurement of biologi- cal processes at the molecular and cellular levels in humans and other living systems’ [15]. Molecular imaging typically includes two- or three-dimension imaging as well as quan- tification over time [15] and differs from other more tradi- tional imaging methods in that tracers are used to image molecular targets and pathways occurring within an area of interest, in contrast to imaging that differentiates quali- ties, such as density (CT), reflectance (ultrasound) or water content/properties (MRI). The pioneering role of PET in the field of molecular imaging relied heavily on its inher- ent strengths, which include a larger range of radiopharma- ceuticals, moreover, introducing radionuclides of biological atoms, higher sensitivity and resolution of the detection sys- tem and more accurate quantification as compared to SPECT imaging. The early clinical applications of PET emerged in the 1980s, especially in the field of neurology, followed by cardiology and later on in oncology in the 1990s [16, 17]. In neurology, PET was initially used to characterise cerebral perfusion and assess brain tumours non-invasively [18–20]. In cardiology, the development of [O-15]-H2O, a radioactive variation of regular water, led to the clinical and currently gold standard application of quantitative evaluation of myocardial blood flow using PET [21]. Additionally, PET became more widely accepted as a clinical application due to its role in oncology for staging and treatment monitoring [22].

While the nineteenth century in medicine was domi- nated by the focus on infectious diseases, in high-income

countries in the last century, healthcare focused especially on oncology as detailed, for instance, in the Pulitzer-winning book The Emperor of All Maladies: A Biography of Cancer written by S. Mukherjee [23]. The discoveries in tumour biology and detailed insights into oncological cellular pathways, as illustrated in the seminal Hallmark of Cancer article by Hanahan and Weinberg in 2011 [24], shifted the focus towards precision medicine and the need for molecular imaging, providing an additional boost for the domain of nuclear medicine.

FDG-PET emerged as a game changer in expanding the scope of PET imaging, especially in clinical oncology, and its appearance was heralded in a landmark article by the late Henry Wagner in 1991: ‘Clinical PET, its time has come’ [25].

Presently, a broad spectrum of PET tracers are being devel- oped to visualise molecular biomarkers and used in clinical or research settings in many clinical domains, resulting in novel avenues, for example, immuno-PET (e.g. 89Zr-immuno-PET) to measure target engagement of therapeutic antibodies and tailoring of therapy with monoclonal antibodies [26] or amy- loid PET (e.g. 11C-Pittsburgh Compound-B PET) to detect amyloid depositions in the brain [27]. Other mostly radiologi- cal molecular imaging tools, such as MR spectroscopy, hyper- polarized MRI or MR chemical exchange saturation transfer (CEST) imaging, may be considered smaller players in terms of the scope of future applications and still not close to becom- ing clinical routine.

Finally, iodine radionuclides and related diagnostics and therapy demonstrated from the early start of nuclear medi- cine the potential of radionuclide therapies and theranostics (i.e. diagnostic combined with therapeutic capacity) [28].

Theranostics, nowadays, is a rapidly growing field in clinical nuclear medicine using, for example, alpha emitters such as

213Bi-DOTATOC receptor-targeted alpha-radionuclide therapy for neuroendocrine tumours [29] and prostate-specific mem- brane antigen (PSMA)-directed theranostics in prostate cancer [30]. All the assets of nuclear medicine and the era of targeted (molecular) medicine proved to be a match made in heaven and contributed significantly to the rapid worldwide dissemination of nuclear medicine, initially in many high-income countries but presently on a global scale.

In 2002, the European Journal of Nuclear Medicine (EJNM) changed its branding into the European Journal of Nuclear Medicine and Molecular Imaging (EJNMMI), claim- ing the representation and embodiment of the unique selling point proposition of molecular imaging as ‘molecular imaging has its roots in nuclear medicine and in many ways is a direct extension of our existing discipline’ [31]. In 2012, the Society of Nuclear Medicine recognised the growing diversity in the field by adding ‘molecular imaging’ to its title [32]. At preclin- ical conferences, especially such as the European Society for Molecular Imaging (ESMI) and similar societies, this embodi- ment started to happen on a smaller scale and continued within

(3)

the EANM as reflected by the initiation of a committee on Translational Molecular Imaging and Therapy in 2007.

Nuclear medicine and molecular imaging in Groningen as an illustration of history

The aforementioned overview of a global history of molecu- lar imaging was also reflected in the local developments in Groningen, the Netherlands. Groningen had pioneered PET since the early 1970s. In 2005, a new department was con- stituted in Groningen, called ‘the Department of Nuclear Medicine and Molecular Imaging’, arguably one of the first nuclear medicine departments to adopt the molecu- lar imaging label. This new department emerged from the fusion of the previous clinical department of conventional nuclear medicine and the PET research facility. Given the need for collaboration within a single group, which united the strengths of one speciality within a single centre while performing the four classical university hospital tasks of patient care, research, teaching and training, a new name was needed. This new name symbolised a shared future while giving credit to feelings, achievements and assets of the previous entities. ‘’Nuclear medicine’’ needed to stay in the title, as it reflected the name used for the speciality and was internationally recognised by authorities, patients and referring physicians. Hence, the PET entity was repre- sented by adding ‘’molecular imaging’’, reflecting the con- tinuous effort at the molecular forefront of its radiochemistry to translate growing insights in molecular pathways at the benefit of in vivo imaging. This was especially demonstrated by the immuno-PET work driven by medical oncology, in collaboration with pharmacists and nuclear medicine col- laborators, presenting, for example, results from a first- in-human study assessing the feasibility of imaging with

89Zr-labeled atezolizumab (anti-PD-L1) to predict response to PD-L1 blockade in several tumour types [33]. After the configuration of a joint medical imaging centre, consist- ing of the Department of Nuclear Medicine and Molecular Imaging and the Department of Radiology, the ‘TracerLab’

in Groningen was founded as a synergistic initiative between these departments, together with the Stratingh Institute for Chemistry from the Faculty of Sciences and Engineering.

The TracerLab aims at developing and applying innovative chemistry for molecular imaging in general, including PET, MRI and, last but not least, optical imaging. Multidiscipli- nary collaboration further allows to translate the imaging tracers in the chain from bench to bedside.

The rise of optical molecular imaging

Driven by the need for clinical answers in the new era of precision medicine, the development of molecular imag- ing tools beyond PET and SPECT is rapidly expanding in terms of both targets and accessibility. The first intraopera- tive fluorescence optical imaging procedure was carried out in 1948 when intravenous fluorescein was used to visualise intracranial neoplasms during neurosurgery [34]. Efforts to increase specificity led to the first-in-human study assess- ing the feasibility of intra-operative tumour-targeted fluores- cence molecular imaging real-time during surgery in 2011, where folate conjugated to fluorescein isothiocyanate was used as a targeted optical imaging agent to detect ovarian cancer and possible peritoneal metastases [35]. As put for- ward in this editorial and related review, optical imaging is no longer restricted to preclinical studies and is already translated into routine clinical care by both surgery- and endoscopy-focused groups in phase I–III clinical trials.

Illustrating the rapid rise of optical imaging research until now, the review in this same issue of the journal had to use a metanarrative approach including artificial intelligence to filter more than 6000 presently available studies, to finally address 58 publications on clinical studies relevant within the scope of the review.

Similar to the interest of medical oncologists in PET- CT and the development and validation of novel radiophar- maceuticals, surgical oncologists and gastroenterologists, amongst other clinicians such as ophthalmologists, are par- ticularly keen on optical imaging as an additional method to address their specific challenges in diagnosis before and during surgical or endoscopic treatment. In Groningen, for example, the strength of optical molecular imaging was translated into preclinical and clinical work by surgeons and gastroenterologists, in close collaboration with disci- plines like pathology and hospital pharmacy, amongst others [35–38]. Due to the rapid deployment of their research and clinical translation into routine daily clinical care, these are now in need of a partner to embrace the clinical support, development of optical tracers, standardisation, validation and thus methodological development, also towards regula- tory agencies like the FDA and EMA. Consequently, in Gro- ningen, the aforementioned separate activities and experts in the field of molecular imaging are now functionally merging for more sustainability, innovative synergism and economies of scale towards this ambition.

While developing this innovative yet challenging imag- ing field, optical-imaging-adopting clinicians need partners who can support them with a proven platform and long- standing experience on how to adopt a sound and reliable methodological development in clinical adaptation [39]. Recent articles in Lancet Oncology demonstrate the socioeconomic

(4)

need for a concerted effort of imaging and oncologists on a global scale [40, 41]. Optical imaging may benefit from classical imaging stakeholders to represent and propagate its methodology towards regulators and reimbursement poli- cies. As an experienced multidisciplinary speciality within the same field of molecular imaging, nuclear medicine is well suited to help address these needs, having gone through the same ordeal. The beating heart of nuclear medicine in terms of ongoing innovation is radiochemistry, in essence providing biologically active molecules labelled with a radionuclide, while in optical imaging, the label is a fluo- rophore, creating the common ground for merging speciali- ties. Radiochemists, trained to develop labelling methods for complex bioactive molecules, are instrumental in creating molecular avenues for optical tracer developments. Radi- opharmacists have the expertise to take this role as imag- ing modality-spanning pharmacists to implement in-house production of optical tracers. Medical physics, as the back- bone of nuclear medicine, has a longstanding experience and focus on quantification and standardisation, which still presents a major challenge in the upcoming field of optical imaging. Finally, nuclear medicine physicians are used to thinking and discussing with referring physicians in terms of molecular pathway. Along the same lines, biologists in our preclinical facilities may help accommodate the translation to their colleagues in surgery, gastroenterology and other fields to come. As a result, the above provides the instant glue between optical and nuclear medicine, a bond to be cherished.

Similarities, differences

and complementarity between optical imaging and nuclear medicine

and molecular imaging

Clinical

While PET imaging allows for a standardised, non-invasive and highly sensitive full-body scan, optical imaging provides real-time, wide-field imaging with high spatial resolution (Fig. 1). Although the sensitivity and specificity of the avail- able nuclear imaging modalities as SPECT-CT and PET- CT are superior to those of any other current clinical imag- ing modality, optical imaging has the potential to visualise biomarkers at the cellular and molecular level in a real-life environment without the need for ionising radiation and its inherent risks. It thereby facilitates the improvement of early detection and staging of disease in a multitude of imaging sessions. While PET is primarily used preoperatively for, for example, diagnostic or therapy response purposes, optical imaging is mainly used as a supportive imaging tool before and during interventional procedures. In optical imaging, the imaging data flows back and forth in real-time, i.e. it is experienced by the operator while performing a procedure or within minutes of imaging, for example, a specimen on the back-table. In contrast, in PET imaging, the informa- tion is obtained during the scanning procedure and not used in a real-time setting in the operating theatre or during an endoscopic procedure. Optical imaging has already shown

Fig. 1 The advantages (green) and drawbacks (red) of human PET imaging (left) and optical imaging (right)

(5)

to increase surgical efficacy in head and neck cancer [42], breast cancer [43] and colorectal cancer [44] and to improve (early) disease detection by endoscopy in, for example, oesophageal cancer [38] by using fluorescent labels linked to compounds that target disease-specific biomarkers. Fur- thermore, it has been applied into the field of infectious dis- ease [45–47] and cardiovascular medicine to detect bacteria in implants and vulnerable atherosclerotic plaques [48–50].

Optical imaging has also been employed for decades to obtain detailed images from within the eye to assess retinal perfusion with fluorescein or indocyanine green (ICG) by using a dedicated split lamp and more recently to assess the retinal layers using optical coherence tomography (OCT), a cross-sectional tomographic imaging technique that meas- ures light back-scattered or back-reflected within the tissue [51]. Finally, there is a crucial role for ‘closed-field’ ex vivo imaging during optical guided procedures, where the sur- gical specimen is imaged on the back-table in the operat- ing room, or in the pathology examination room, within a black-box, thereby allowing for pathological assessment of the excised tissue immediately after resection, with greater control over optical parameters such as illumination inten- sity, field homogeneity and geometry, camera distance and the lack of interference of ambient background light [52]. Closed-field imaging will enable more accurate and standardised operating procedures for the visualization of excised tissue and thereby provides a more reliable and accu- rate comparison of imaging data obtained by the observers, individual centres and study results, which is of need for regulatory approval. These preliminary clinical results show the potential of optical imaging to guide interventions and improve (early) disease detection in real-time. Furthermore, in vivo fluorescence imaging can be used to predict thera- peutic outcomes, as shown for cancer and Crohn’s disease [53, 55, 56].

Optical imaging is also well-suited for theranostic pur- poses since it can facilitate the detection of disease, deter- mine the target location and dosimetry and monitor thera- peutic effects in situ [53]. Exemplary is a self-reporting and self-adapting theranostic nanoparticle based on poly(lactic- co-glycolic acid) designed to encapsulate camptothecin as an anticancer drug and a caspase-3 activatable fluorescent peptide [54]. The fluorescence is turned off due to the fluo- rescence resonance energy transfer (FRET) effect and can be turned on upon reaction with caspase-3, an important pro- tease in apoptosis, to achieve visualised therapeutic report- ing for precise tumour treatment [54]. Of special interest is also the potential of a theranostic combination of optical imaging and photopharmacology, an emerging field in which light is used to activate drugs selectively and reversibly in real-time [57].

Chemistry

A fluorescent tracer’s design must consider its pharmacolog- ical properties, toxicity profile, chemical/metabolic stability and in vivo imaging performance. In general, the chemical challenges in nuclear and optical imaging are similar; while introducing the imaging label, maintaining the functional- ity and favourable pharmacokinetics of the targeting moiety is vital [58]. In the earlier evolutionary stages of nuclear molecular imaging, radiochemists have developed valuable expertise in this respect. However, it must be noted that there is a key structural difference between the labels used in nuclear and optical imaging: while radionuclides are often typically single atoms (11C, 18F) already present in many chemical structures, and their introduction to the targeting molecule can therefore be often achieved in a near-seamless way, the fluorophores used in optical imaging are typically large, planar, lipophilic molecules, whose incorporation into the targeting molecule has the potential to change its proper- ties significantly. A limitation of PET imaging is that it relies solely on the accumulation of tracers, and the signal in this modality is continuously detected and cannot be activated, e.g. through the catalytic activation of an imaging probe by a specific enzyme resulting in the dequenching of the fluorescence signal. Conversely, in optical imaging, this can be achieved when a quenched fluorophore conjugated on a cleavable backbone encounters a molecular target, such as a proteolytic enzyme with a specific cleavage site activity [59, 60] for the backbone, so the quenched fluorophore becomes activated ones the enzyme is presented at the site of inter- est, e.g. proteases such as matrix metalloproteases [61]. In a similar off/on strategy, fluorescent tracers can be designed to be quenched (non-activatable) in the off-state and activated, for example, by an acidic microenvironment using an ultra- pH sensitive amphiphilic polymer [36].

Clearly, it remains challenging to find a single imag- ing method that can satisfy the increasing need for diag- nostic information in all instances and clinical condition.

Therefore, chemists can at times convert or even expand existing imaging agents to another modality as, for exam- ple, an antibody can be labelled with a radionuclide such as 89-zirconium (89Zr) or a near-infrared fluorescent dye such as IRDye-800CW. Exemplary for dual-modality imaging is the vascular endothelial growth factor (VEGF)-targeting antibody bevacizumab labelled with IRDye800CW or 89Zr- bevacizumab [62]. A hybrid tracer, for example, is [111In]

In-DTPA-trastuzumab-IRDye800CW, a monoclonal anti- body against the human epidermal growth factor 2 (HER2) receptor and dual-labelled trastuzumab with both a fluoro- phore (IRDye800CW) and a radioactive label (111In) used for multimodal imaging of HER2-positive breast cancer [63–65]. The design of such hybrid tracers can combine desirable features of two different imaging modalities and

(6)

can potentially improve quantification and understanding of the underlying biological targets through the complementary information provided [66].

Physics

Optical imaging uses excitation light varying from the vis- ible light (380–750 nm) (e.g. for fluorescein- or Cyanine 5.5- based tracers) to the near-infrared spectrum (750–1400 nm) (e.g. tracers employing ICG or IRDye-800CW as fluoro- phores) to excite fluorescent tracers from a ground state to an excited state, from which decay to a lower energy state occurs, through the emission of fluorescence at a lower energy, i.e. higher wavelength. On the other hand, PET meas- ures two high-energy annihilation gamma photons produced after positron emission from a radionuclide-tagged tracer molecule. In optical imaging, the tissue penetration of the fluorescent signal is limited to the range of several centime- tres at best, while in PET, there is a virtually unlimited pen- etration depth. However, spatial resolution of (pre)clinical PET is in the range of 1–4 mm [67], whereas optical imaging can reach few to hundreds of micrometres depending on the optical imaging modality used [53, 68]. Additionally, in con- trast to PET, optical imaging uses non-ionising radiation and is easy to use and relatively inexpensive in terms of camera costs and tracer synthesis, although the cost-effectiveness still has to be confirmed by health economic studies (Fig. 1).

While optical imaging outperforms PET in spatial resolution of subsurface disease, PET is superior to optical imaging in its non-invasive quantitative assessment of larger volumes.

The latter is possible through such techniques as attenuation and scatter corrections, providing uniform images that allow for high reading confidence and accurate quantitative assess- ments. In optical imaging, such corrections resulting in accurate quantitative assessments are already implemented in preclinical small imaging optical fluorescence imaging systems but still need to be developed for clinical applica- tions. As the fluorescence signal detected is influenced by optical tissue properties, such as absorption and scattering properties, wide-field fluorescence imaging alone does not necessarily reflect true tracer distribution. Several methods have been proposed to correct for the aforementioned opti- cal properties, such as multi-diameter single-fibre reflection/

single-ibre fluorescence (MDSFR/SFF) spectroscopy [69, 70]. However, such a quantification method has yet to be translated to wide-field fluorescence imaging.

Standardisation and implementation

In optical imaging, standardisation of imaging procedures is currently lacking, mainly due to a lack of uniform guide- lines. The variety of camera systems, illumination sources,

data processing methods and imaging procedures in use makes an adequate comparison between study results and observers challenging. The professional environment that was built for imaging techniques, such as PET, allowed the field of nuclear medicine to flourish in a clinical setting towards standard of care. To date, this is still lacking in the optical imaging field. In this respect, the optical molecular imaging community should benefit from the knowledge gained in clinical PET standardisation and quantifica- tion. If optical imaging were to be implemented into the standard of care, it needs an established uniform transla- tional pathway. As nuclear imaging techniques, such as PET, had to tackle similar issues, we must join forces as a clinical molecular imaging community and share our les- sons learned, irrespective of the applied imaging modal- ity. The pathway developed to bring nuclear medicine and molecular imaging into the standard of care may provide the needed roadmap for advanced clinical translation of optical imaging and more recent innovations, such as clini- cal multispectral optoacoustic tomographic imaging. We, therefore, advocate more intensified multidisciplinary and international collaborations to facilitate the implementa- tion of optical and optoacoustic imaging as the next step after X-ray CT, PET, SPECT, MRI and ultrasound imag- ing. Clinical questions need to be answered to improve patient care; the choice of the methodology is secondary, i.e. the methodology should be selected based on the clini- cal or research question.

Take‑home message: cross‑fertilization within nuclear medicine and molecular imaging

A unique opportunity exists to develop a stronger bond between the multidisciplinary nuclear medicine community and pioneering optical imaging clinicians, being major play- ers in hospitals in need of accurate, reliable and reproducible imaging data similar for neurology, cardiology and medical oncology. The history of PET imaging development and the hybrid PET-imaging technologies like PET/CT and PET/

MRI, combined with the more recent theranostic clinical implementations, provides inspiration and a navigator for a bright future of optical and optoacoustic imaging. As has been the case with PET, fundamental challenges lay ahead for optical imaging, but applying and implementing proven strategies from the field of nuclear imaging will contribute to overcome those hurdles. The optical field should benefit from the knowledge gained in PET chemistry, standardisa- tion and quantification and the established pathway to bring imaging techniques into the standard of care. This collabo- ration may present us with an opportunity to build on the

(7)

molecular imaging part of ‘nuclear medicine and molecular imaging’ and provide us with a technique that complements our current molecular imaging modalities, such as real-time intraoperative guidance and early-stage cancer detection, to name a few.

The authors of this editorial have a background in nuclear medicine, surgery, gastroenterology, chemistry, optical engi- neering and physics and support each other in this endeavour at a local, regional level. We appeal to colleagues in our com- munities to strengthen these synergistic bonds on a broader (inter-)national level at a larger scale and not to miss out on this unique chance for a historical rendezvous of molecular imaging modalities. In short, let us all embrace optical imag- ing as a growing branch on the clinical molecular imaging tree and a global opportunity to enrich our molecular arma- mentarium for the benefit of the patient.

Acknowledgements Prof. Dr. P. H. Elsinga, Prof. Dr. A. A. Lam- mertsma and Prof. Dr. J. Pruim are gratefully acknowledged for their critical review of this editorial.

Declarations

Ethics approval Institutional review board approval was not required because the paper is an editorial.

Informed consent Not applicable.

Conflict of interest GMvD is CEO, founder and shareholder of TRAC- ER Europe BV / AxelaRx. The other authors declare no competing interests.

References

1. Seidlin SM, Marinelli LD, Oshry E. Radioactive iodine therapy;

effect on functioning metastases of adenocarcinoma of the thyroid.

J Am Med Assoc. 1946;132:838–47.

2. Keston AS, Ball RP, Frantz VK, Palmer WW. Storage of radio- active iodine in a metastasis from thyroid carcinoma. Science.

1942;95:362–3.

3. Blahd WH. Ben Cassen and the development of the rectilinear scanner. Semin Nucl Med. 1996;26:165–70.

4. Richmond C. Sir Godfrey Hounsfield BMJ. 2004;329:687.

5. Cosgrove DO, McCready VR. Diagnosis of liver metastases using ultrasound and isotope scanning techniques. J R Soc Med.

1978;71:652–7.

6. Hutton BF. The origins of SPECT and SPECT/CT. Eur J Nucl Med Mol Imaging. 2014;41(Suppl 1):S3-16.

7. Jones T, Townsend D. History and future technical innovation in positron emission tomography. J Med Imaging (Bellingham) 2017;4:011013.

8. Wagner HN Jr. A brief history of positron emission tomography (PET). Semin Nucl Med. 1998;28:213–20.

9. Slomka PJ, Pan T, Berman DS, Germano G. Advances in SPECT and PET hardware. Prog Cardiovasc Dis. 2015;57:566–78.

10. van Sluis J, de Jong J, Schaar J, Noordzij W, van Snick P, Dierckx R, Borra R, Willemsen A, Boellaard R. Performance

characteristics of the digital biograph vision PET/CT system. J Nucl Med. 2019;60:1031–6.

11. Richards P, Tucker WD, Srivastava SC. Technetium-99m: an his- torical perspective. Int J Appl Radiat Isot. 1982;33:793–9.

12. Petroni D, Menichetti L, Poli M. Historical and radiopharmaceuti- cal relevance of [18F]FDG. J Radioanal Nucl. 2020;323:1017–31.

13. Hutton BF, Erlandsson K, Thielemans K. Advances in clini- cal molecular imaging instrumentation. Clin Trans Imaging.

2018;6:31–45.

14. Cherry SR, Jones T, Karp JS, Qi J, Moses WW, Badawi RD. Total- body PET: maximizing sensitivity to create new opportunities for clinical research and patient care. J Nucl Med. 2018;59:3–12.

15. Pomper,M, VanBrocklin,H, Anderson,C. What is molecular imag- ing?. J Nucl Med 2008:163.

16. Anand SS, Singh H, Dash AK. Clinical applications of PET and PET-CT. Med J Armed Forces India. 2009;65:353–8.

17. Coleman RE, Delbeke D, Guiberteau MJ, Conti PS, Royal HD, Weinreb JC, Siegel BA, Federle MF, Townsend DW, Berland LL, American College of Radiology, Society of Nuclear Medicine, Society of Computed Body Tomography and Magnetic Reso- nance. Concurrent PET/CT with an integrated imaging system:

intersociety dialogue from the joint working group of the Ameri- can College of Radiology, the Society of Nuclear Medicine, and the Society of Computed Body Tomography and Magnetic Reso- nance. J Nucl Med 2005;46:1225–39.

18. Brownell G, Sweet W. Localization of brain tumors with positron emitters. Nucleonics. 1953;11:40–5.

19. Sweet WH, Brownell GL. Localization of intracranial lesions by scanning with positron-emitting arsenic. J Am Med Assoc.

1955;157:1183–8.

20. Davis KM, Ryan JL, Aaron VD, Sims JB. PET and SPECT imag- ing of the brain: history, technical considerations, applications, and radiotracers. Semin Ultrasound CT MR. 2020;41:521–9.

21. Pelletier-Galarneau M, Martineau P, El Fakhri G. Quanti- fication of PET myocardial blood flow. Curr Cardiol Rep 2019;21:11,019–1096-x.

22. Basu S, Alavi A. PET-based personalized management in clini- cal oncology: an unavoidable path for the foreseeable future.

PET Clin. 2016;11:203–7.

23. Mukherjee S. The emperor of all maladies: a biography of can- cer. Scribner; 2010.

24. Hanahan D, Weinberg RA. Hallmarks of cancer: the next gen- eration. Cell. 2011;144:646–74.

25. Wagner HN Jr. Clinical PET: its time has come. J Nucl Med.

1991;32:561–4.

26. Cancer MR. Imaging with antibodies. Nature. 2017;543:743–6.

27. Klunk WE, Engler H, Nordberg A, Wang Y, Blomqvist G, Holt DP, Bergström M, Savitcheva I, Huang GF, Estrada S, Ausén B, Debnath ML, Barletta J, Price JC, Sandell J, Lopresti BJ, Wall A, Koivisto P, Antoni G, Mathis CA, Långström B.

Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B. Ann Neurol. 2004;55:306–19.

28. Ehrhardt JD Jr, Güleç S. A review of the history of radioactive iodine theranostics: the origin of nuclear ontology. Mol Imaging Radionucl Ther. 2020;29:88–97.

29. Kratochwil C, Giesel FL, Bruchertseifer F, Mier W, Apos- tolidis C, Boll R, Murphy K, Haberkorn U, Morgenstern A.

213Bi-DOTATOC receptor-targeted alpha-radionuclide therapy induces remission in neuroendocrine tumours refractory to beta radiation: a first-in-human experience. Eur J Nucl Med Mol Imaging. 2014;41:2106–19.

30. Virgolini I, Decristoforo C, Haug A, Fanti S, Uprimny C. Cur- rent status of theranostics in prostate cancer. Eur J Nucl Med Mol Imaging. 2018;45:471–95.

(8)

31. Gambhir SS, Ell PJ. Welcome to the European journal of nuclear medicine and molecular imaging. Eur J Nucl Med Mol Imaging.

2002;29:1–2.

32. Society of Nuclear Medicine and Molecular Imaging. SNM changing name to society of nuclear medicine and molecular imaging. Oncol Times 2012;34.

33. Bensch F, van der Veen EL, Lub-de Hooge MN, Jorritsma-Smit A, Boellaard R, Kok IC, Oosting SF, Schröder CP, Hiltermann TJN, van der Wekken AJ, Groen HJM, Kwee TC, Elias SG, Gie- tema JA, Bohorquez SS, de Crespigny A, Williams SP, Mancao C, Brouwers AH, Fine BM, de Vries EGE. (89)Zr-atezolizumab imaging as a non-invasive approach to assess clinical response to PD-L1 blockade in cancer. Nat Med. 2018;24:1852–8.

34. Moore GE, Peyton WT. The clinical use of fluorescein in neurosurgery; the localization of brain tumors. J Neurosurg.

1948;5:392–8.

35. van Dam GM, Themelis G, Crane LM, Harlaar NJ, Pleijhuis RG, Kelder W, Sarantopoulos A, de Jong JS, Arts HJ, van der Zee AG, Bart J, Low PS, Ntziachristos V. Intraoperative tumor-spe- cific fluorescence imaging in ovarian cancer by folate receptor-α targeting: first in-human results. Nat Med. 2011;17:1315–9.

36. Voskuil FJ, Steinkamp PJ, Zhao T, van der Vegt B, Koller M, Doff JJ, Jayalakshmi Y, Hartung JP, Gao J, Sumer BD, Witjes MJH, van Dam GM, SHINE study group. Exploiting metabolic acidosis in solid cancers using a tumor-agnostic pH-activata- ble nanoprobe for fluorescence-guided surgery. Nat Commun 2020;11:3257,020–16814–4.

37. Voskuil FJ, de Jongh SJ, Hooghiemstra WTR, Linssen MD, Steinkamp PJ, de Visscher SAHJ, Schepman KP, Elias SG, Meersma GJ, Jonker PKC, Doff JJ, Jorritsma-Smit A, Nagen- gast WB, van der Vegt B, Robinson DJ, van Dam GM, Witjes MJH. Fluorescence-guided imaging for resection margin evaluation in head and neck cancer patients using cetuximab- 800CW: a quantitative dose-escalation study. Theranostics.

2020;10:3994–4005.

38. Nagengast WB, Hartmans E, Garcia-Allende PB, Peters FTM, Linssen MD, Koch M, Koller M, Tjalma JJJ, Karrenbeld A, Jorritsma-Smit A, Kleibeuker JH, van Dam GM, Ntziachristos V. Near-infrared fluorescence molecular endoscopy detects dys- plastic oesophageal lesions using topical and systemic tracer of vascular endothelial growth factor A. Gut. 2019;68:7–10.

39. Koller M, Qiu SQ, Linssen MD, Jansen L, Kelder W, de Vries J, Kruithof I, Zhang GJ, Robinson DJ, Nagengast WB, Jorritsma- Smit A, van der Vegt B, van Dam GM. Implementation and benchmarking of a novel analytical framework to clinically evalu- ate tumor-specific fluorescent tracers. Nat Commun 2018;9:3739, 018–05727-y.

40. Ward ZJ, Scott AM, Hricak H, Atun R. Global costs, health ben- efits, and economic benefits of scaling up treatment and imaging modalities for survival of 11 cancers: a simulation-based analysis.

Lancet Oncol. 2021;22:341–50.

41. Ward ZJ, Scott AM, Hricak H, Abdel-Wahab M, Paez D, Lette MM, Vargas HA, Kingham TP, Atun R. Estimating the impact of treatment and imaging modalities on 5-year net survival of 11 cancers in 200 countries: a simulation-based analysis. Lancet Oncol. 2020;21:1077–88.

42. Rosenthal EL, Warram JM, de Boer E, Chung TK, Korb ML, Brandwein-Gensler M, Strong TV, Schmalbach CE, Morlandt AB, Agarwal G, Hartman YE, Carroll WR, Richman JS, Clemons LK, Nabell LM, Zinn KR. Safety and tumor specificity of cetuximab- IRDye800 for surgical navigation in head and neck cancer. Clin Cancer Res. 2015;21:3658–66.

43. Lamberts LE, Koch M, de Jong JS, Adams ALL, Glatz J, Kranen- donk MEG, Terwisscha van Scheltinga AGT, Jansen L, de Vries J, Lub-de Hooge MN, Schröder CP, Jorritsma-Smit A, Linssen MD, de Boer E, van der Vegt B, Nagengast WB, Elias SG, Oliveira S,

Witkamp AJ, Mali WPTM, Van der Wall E, van Diest PJ, de Vries EGE, Ntziachristos V, van Dam GM. Tumor-specific uptake of fluorescent bevacizumab-IRDye800CW microdosing in patients with primary breast cancer: a phase I feasibility study. Clin Cancer Res 2017;23:2730–41.

44. Boogerd LSF, Hoogstins CES, Schaap DP, Kusters M, Handgraaf HJM, van der Valk MJM, Hilling DE, Holman FA, Peeters KCMJ, Mieog JSD, van de Velde CJH, Farina-Sarasqueta A, van Lijns- choten I, Framery B, Pèlegrin A, Gutowski M, Nienhuijs SW, de Hingh IHJT, Nieuwenhuijzen GAP, Rutten HJT, Cailler F, Burg- graaf J, Vahrmeijer AL. Safety and effectiveness of SGM-101, a fluorescent antibody targeting carcinoembryonic antigen, for intraoperative detection of colorectal cancer: a dose-escalation pilot study. Lancet Gastroenterol Hepatol. 2018;3:181–91.

45. Ning X, Lee S, Wang Z, Kim D, Stubblefield B, Gilbert E, Murthy N. Maltodextrin-based imaging probes detect bacteria in vivo with high sensitivity and specificity. Nat Mater. 2011;10:602–7.

46. Panizzi P, Nahrendorf M, Figueiredo JL, Panizzi J, Marinelli B, Iwamoto Y, Keliher E, Maddur AA, Waterman P, Kroh HK, Leuschner F, Aikawa E, Swirski FK, Pittet MJ, Hackeng TM, Fuentes-Prior P, Schneewind O, Bock PE, Weissleder R. In vivo detection of Staphylococcus aureus endocarditis by targeting path- ogen-specific prothrombin activation. Nat Med. 2011;17:1142–6.

47. van Oosten M, Schäfer T, Gazendam JA, Ohlsen K, Tsompanidou E, de Goffau MC, Harmsen HJ, Crane LM, Lim E, Francis KP, Cheung L, Olive M, Ntziachristos V, van Dijl JM, van Dam GM.

Real-time in vivo imaging of invasive- and biomaterial-associated bacterial infections using fluorescently labelled vancomycin. Nat Commun. 2013;4:2584.

48. Dweck MR, Doris MK, Motwani M, Adamson PD, Slomka P, Dey D, Fayad ZA, Newby DE, Berman D. Imaging of coronary atherosclerosis - evolution towards new treatment strategies. Nat Rev Cardiol. 2016;13:533–48.

49. Hara T, Ughi GJ, McCarthy JR, Erdem SS, Mauskapf A, Lyon SC, Fard AM, Edelman ER, Tearney GJ, Jaffer FA. Intravascu- lar fibrin molecular imaging improves the detection of unhealed stents assessed by optical coherence tomography in vivo. Eur Heart J. 2017;38:447–55.

50. Jaffer FA, Calfon MA, Rosenthal A, Mallas G, Razansky RN, Mauskapf A, Weissleder R, Libby P, Ntziachristos V. Two-dimen- sional intravascular near-infrared fluorescence molecular imag- ing of inflammation in atherosclerosis and stent-induced vascular injury. J Am Coll Cardiol. 2011;57:2516–26.

51. Schmidt-Erfurth U, Klimscha S, Waldstein SM, Bogunović H. A view of the current and future role of optical coherence tomogra- phy in the management of age-related macular degeneration. Eye (Lond). 2017;31:26–44.

52. Liu JTC, Sanai N. Trends and challenges for the clinical adoption of fluorescence-guided surgery. J Nucl Med. 2019;60:756–7.

53. Yun SH, Kwok SJJ. Light in diagnosis, therapy and surgery. Nat Biomed Eng 2017;1:https:// doi. org/ 10. 1038/ s4155 1,016- 0008.

54. Luo Y, Huang L, Yang Y, Zhuang X, Hu S, Ju H, Yu BY, Tian J.

A programmed nanoparticle with self-adapting for accurate can- cer cell eradication and therapeutic self-reporting. Theranostics.

2017;7:1245–56.

55. Ntziachristos V, Schellenberger EA, Ripoll J, Yessayan D, Graves E, Bogdanov A,Jr, Josephson L, Weissleder R. Visualization of antitumor treatment by means of fluorescence molecular tomog- raphy with an annexin V-Cy5.5 conjugate. Proc Natl Acad Sci U S A 2004;101:12294–9.

56. Atreya R, Neumann H, Neufert C, Waldner MJ, Billmeier U, Zopf Y, Willma M, App C, Münster T, Kessler H, Maas S, Gebhardt B, Heimke-Brinck R, Reuter E, Dörje F, Rau TT, Uter W, Wang TD, Kiesslich R, Vieth M, Hannappel E, Neurath MF. In vivo imag- ing using fluorescent antibodies to tumor necrosis factor predicts therapeutic response in Crohn’s disease. Nat Med. 2014;20:313–8.

(9)

57. Lerch MM, Hansen MJ, van Dam GM, Szymanski W, Feringa BL. Emerging targets in photopharmacology. Angew Chem Int Ed Engl. 2016;55:10978–99.

58. van Leeuwen FW, de Jong M, EANM Translational Molecular Imaging Committee. Molecular imaging: the emerging role of optical imaging in nuclear medicine. Eur J Nucl Med Mol Imaging 2014;41:2150–3.

59. Bremer C, Tung CH, Weissleder R. In vivo molecular target assessment of matrix metalloproteinase inhibition. Nat Med.

2001;7:743–8.

60. Jager NA, Wallis de Vries BM, Hillebrands JL, Harlaar NJ, Tio RA, Slart RH, van Dam GM, Boersma HH, Zeebregts CJ, Westra J. Distribution of matrix metalloproteinases in human atheroscle- rotic carotid plaques and their production by smooth muscle cells and macrophage subsets. Mol Imaging Biol 2016;18:283–91.

61. Savariar EN, Felsen CN, Nashi N, Jiang T, Ellies LG, Steinbach P, Tsien RY, Nguyen QT. Real-time in vivo molecular detection of primary tumors and metastases with ratiometric activatable cell-penetrating peptides. Cancer Res. 2013;73:855–64.

62. Terwisscha van Scheltinga AG, van Dam GM, Nagengast WB, Ntziachristos V, Hollema H, Herek JL, Schröder CP, Kosterink JG, Lub-de Hoog MN, de Vries EG. Intraoperative near-infrared fluorescence tumor imaging with vascular endothelial growth factor and human epidermal growth factor receptor 2 targeting antibodies. J Nucl Med 2011;52:1778–85.

63. Deken MM, Bos DL, Tummers WSFJ, March TL, van de Velde CJH, Rijpkema M, Vahrmeijer AL. Multimodal image-guided surgery of HER2-positive breast cancer using [(111)In]In-DTPA- trastuzumab-IRDye800CW in an orthotopic breast tumor model.

EJNMMI Res 2019;9:98,019–0564-z.

64. Hekman MC, Rijpkema M, Muselaers CH, Oosterwijk E, Huls- bergen-Van de Kaa CA, Boerman OC, Oyen WJ, Langenhuijsen JF, Mulders PF. Tumor-targeted dual-modality imaging to improve

intraoperative visualization of clear cell renal cell carcinoma: a first in man study. Theranostics 2018;8:2161–70.

65. Li D, Zhang J, Chi C, Xiao X, Wang J, Lang L, Ali I, Niu G, Zhang L, Tian J, Ji N, Zhu Z, Chen X. First-in-human study of PET and optical dual-modality image-guided surgery in glio- blastoma using (68)Ga-IRDye800CW-BBN. Theranostics.

2018;8:2508–20.

66. Li C, Yang Y, Mitchell GS, Cherry SR. Simultaneous PET and multispectral 3-dimensional fluorescence optical tomography imaging system. J Nucl Med. 2011;52:1268–75.

67. Moses WW. Fundamental limits of spatial resolution in PET. Nucl Instrum Methods Phys Res A. 2011;648(Supplement 1):S236–40.

68. DSouza AV, Lin H, Henderson ER, Samkoe KS, Pogue BW.

Review of fluorescence guided surgery systems: identification of key performance capabilities beyond indocyanine green imaging.

J Biomed Opt 2016;21:80901.

69. Middelburg TA, Hoy CL, Neumann HA, Amelink A, Robinson DJ. Correction for tissue optical properties enables quantitative skin fluorescence measurements using multi-diameter single fiber reflectance spectroscopy. J Dermatol Sci. 2015;79:64–73.

70. Tjalma JJJ, Koller M, Linssen MD, Hartmans E, de Jongh S, Jorritsma-Smit A, Karrenbeld A, de Vries EG, Kleibeuker JH, Pennings JP, Havenga K, Hemmer PH, Hospers GA, van Etten B, Ntziachristos V, van Dam GM, Robinson DJ, Nagengast WB.

Quantitative fluorescence endoscopy: an innovative endoscopy approach to evaluate neoadjuvant treatment response in locally advanced rectal cancer. Gut. 2020;69:406–10.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Referenzen

ÄHNLICHE DOKUMENTE

Our results demonstrate that the amount of free air is larger in upper GI and distal lower GI perforations than in other sites of the gut, and upper GI perforations have a greater

We then detail recent progress on imaging technology, including advancements in tracer design and injection methods, that have allowed visualization of lymphatic vessels with

The MR imaging study (Fig. 1) showed a soft tissue process of the index finger with two components: a fluid- filled region with contrast-enhancing rim corresponding to an abscess

SPECT imaging of three numerical phantoms (Original) perturbed by abrupt rigid motion and reconstructed with the EM algorithm (Reconstruction), the DDMC and the CRMC

In the present study we investigated whether in vivo ultrasound molecular imaging of the expression of VCAM-1 could be used to assess the effects of statins on vascular

Christian; Quantitative PET/CT Scanner Performance Characterization Based Upon the Society of Nuclear Medicine and Molecular Imaging Clinical Trials Network Oncology Clinical

Continuously Moving Robotic freehand SPECT using Gamma Cameras 91 The reconstruction quality in all four cases produced satisfactory results for all the nodules.. This is

We addressed this issue and found that ground state depletion (GSD) microscopy can resolve heterostructured nanowires with a 5-fold resolution enhancement over confocal microscopy.