Skip to main content
Erschienen in: European Journal of Nuclear Medicine and Molecular Imaging 11/2021

Open Access 29.04.2021 | Original Article

EANM position paper on the role of radiobiology in nuclear medicine

verfasst von: An Aerts, Uta Eberlein, Sören Holm, Roland Hustinx, Mark Konijnenberg, Lidia Strigari, Fijs W.B. van Leeuwen, Gerhard Glatting, Michael Lassmann

Erschienen in: European Journal of Nuclear Medicine and Molecular Imaging | Ausgabe 11/2021

Executive Summary

With an increasing variety of radiopharmaceuticals for diagnostic or therapeutic nuclear medicine as valuable diagnostic or treatment option, radiobiology plays an important role in supporting optimizations. This comprises particularly safety and efficacy of radionuclide therapies, specifically tailored to each patient. As absorbed dose rates and absorbed dose distributions in space and time are very different between external irradiation and systemic radionuclide exposure, distinct radiation-induced biological responses are expected in nuclear medicine, which need to be explored. This calls for a dedicated nuclear medicine radiobiology. Radiobiology findings and absorbed dose measurements will enable an improved estimation and prediction of efficacy and adverse effects. Moreover, a better understanding on the fundamental biological mechanisms underlying tumor and normal tissue responses will help to identify predictive and prognostic biomarkers as well as biomarkers for treatment follow-up. In addition, radiobiology can form the basis for the development of radiosensitizing strategies and radioprotectant agents. Thus, EANM believes that, beyond in vitro and preclinical evaluations, radiobiology will bring important added value to clinical studies and to clinical teams. Therefore, EANM strongly supports active collaboration between radiochemists, radiopharmacists, radiobiologists, medical physicists, and physicians to foster research toward precision nuclear medicine.
Hinweise
G.G. and M.L. shared senior authorship.
This article is part of the Topical Collection on Radiation biology

Publisher’s note

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

Introduction

In recent years, the number of radiopharmaceuticals for diagnostic and therapeutic applications has increased considerably. In addition, theranostics are being developed that use the same molecular targeting platform for both imaging and treatment. This has led to an increased medical use in both malignant as well as benign conditions. Consequently, an improved understanding of the biological processes, with special regard to the effects of ionizing radiation to normal tissues and tumors, is required. This is to determine the absorbed dose-effect relationship more precisely, as a prerequisite for achieving an optimal diagnostic or therapeutic outcome. In general, absorbed doses are low (< 20 mGy) for most organs in diagnostic procedures [1]. However, when multiple diagnostic examinations or therapeutic applications are undertaken, this is no longer the case. Repeated diagnostic irradiations can result in cumulative absorbed doses in normal organs and tissues up to a few hundred milligray [2]. For therapies, absorbed doses can exceed previously suggested absorbed dose limits (e.g., 23 Gy for the kidneys in peptide receptor radionuclide therapies [3]). For absorbed doses of < 1 Gy, stochastic effects of ionizing radiation may be observed, whereas for therapies, a mixture of stochastic and deterministic effects is expected. Radiation-related adverse effects strongly depend on both the individual absorbed doses [4] and the individual radiation sensitivity [58]. Thus, without an individualized approach in radionuclide therapy, a group of patients may be over-treated, jeopardizing patient safety. Conversely, patients may be undertreated, leading to suboptimal treatment efficacy.
Systemic radiation delivery via radiopharmaceuticals is inherently different from irradiation by external radiation sources. As a consequence, distinct radiation-induced biological responses are expected for radiopharmaceuticals posing considerable challenges for in vitro, preclinical and clinical studies investigating radionuclide applications. Several research topics are suggested that should be addressed with regard to radiobiology in relation to the systemic use of radiopharmaceuticals and which have not yet been rigorously investigated [911]. Presently, there are limited studies related to the use of radiobiology in nuclear medicine. Typical examples of such studies are provided in Table 1.
Table 1
Non-exhaustive list of typical examples of nuclear medicine radiobiology studies including a description of the method or biomarker applied, targets, radionuclides, and activity/absorbed dose range
Topics investigated
Method/biomarker
Target
Radionuclide
Model
Activity/dose range
Remark
References
DNA damage
ɣ-H2AX, 53BP1 foci in PMBCs
Blood
177Lu, 131I, 223Ra
Patients
< 100 mGy
Ex vivo and in vivo data
[7, 1216]
DNA damage
ɣ-H2AX, 53BP1 foci in tumor cells
Neuroendocrine tumor, (SST2), prostate tumor (PSMA)
177Lu, 213Bi
Cell culture, mouse
< 2.5 MBq 177Lu in vitro, 30 MBq 177Lu in vivo, 0.3 MBq 213Bi in vitro, < 6.6 MBq 213Bi in vivo
SST2 agonist vs. antagonist
[17, 18]
DNA damage Imaging
[111In]In-anti-γH2AX-TAT, [89Zr]Zr-DFO-anti-γH2AX-TAT
Neuroendocrine tumor, pancreatic carcinoma
177Lu, 225Ac
Mouse
< 20 MBq 177Lu, 37 kBq 225Ac
DNA damage monitoring after [177Lu]Lu-DOTA-TATE therapy
[19, 20]
Preclinical therapeutic value, cell survival, cell cycle progression
Tumor volume, cell survival, cell cycle analysis
Non-Hodgkin lymphoma (CD37)
177Lu
Cell culture, mouse, patient samples
< 6 MBq/mL 177Lu in vitro, < 500 MBq/kg 177Lu in vivo
Radioimmunotherapy
[21]
In vitro cytotoxicity
Cell-free plasmid DNA damage, DNA damage, cell survival, cell viability, microautoradiography cell distribution assay
Breast cancer (HER2), prostate tumor
67Ga, 111In
Cell culture
< 0.3 MBq/mL 67Ga in vitro; 1.1 Bq/cell (15 MBq/mL) 67Ga or 111In in vitro, 0.1 MBq/mL 67Ga or 111In cell free
Auger electrons
[22, 23]
Combination with other agents: radiosensitizing agents
53BP1, micronuclei in cell cultures, cell survival, cell viability, cell cycle progression, DNA damage response, gene expression, tumor perfusion, tumor, tumor radioactivity uptake, tumor volume
Neuroendocrine tumor (SST2), small cell lung cancer (SST2), prostate tumor (PSMA), neuroblastoma
PARP, protein folding, DNA and DNA synthesis, Hedgehog signaling, nicotinamide phosphoribosyltransferase, topoisomerase I, proteasome, P53-MDM2 interaction nutlin-3, and the copper-chelated form of the oxidizing agent disulfiram, G2/M cell cycle arrest
177Lu, 131I
Cell culture, spheroids, mouse, patients
< 6 MBq/mL 177Lu in vitro, < 30 MBq 177Lu in vivo, < 6 MBq 177Lu ex vivo patients, 4 × 7.8 GBq patients, 0.37 MBq/mL 131I in vitro, 20 MBq 131I in vivo
Olaparib, 1,5-dihydroxyisoquinoline, PJ-34, veliparib, talazoparib, Hsp90 inhibitor, androgen receptor inhibitor, capecitabine, temozolomide, sonidegib, NAMPT inhibitor, topotecan, bortezomib, the inhibitor of the P53-MDM2 interaction nutlin-3 and the copper-chelated form of the oxidizing agent disulfiram (DSF:Cu), EBRT
[21, 2437]
Combination with other agents: upregulation of the therapeutic target
Transcriptional, translational, and functional analysis, tracer uptake
Neuroendocrine tumor (SST2)
n.a.
Cell culture
n.a.
[38, 39]
Combination with other agents: chemotherapeutic drugs
Cell viability, biodistribution, tumor volume
Breast cancer
131I
Cell culture, mouse
< 7.4 MBq/mL in vitro, 7.4 MBq in vivo
Human serum albumin–paclitaxel nanoparticles
[40]
Combination with other agents: radioprotectant agents
Biodistribution, tumor response
Kidneys
177Lu
Mouse
30 MBq
Kidney-preserving agent
[41]
Tumor radionuclide/receptor distribution
[111In]In-EGF and [111In]In-labeled trastuzumab imaging, autoradiography, immunofluorescence microscopy
Breast cancer (EGFR, HER2), head and neck cancer (EGFR), neuroendocrine tumor (SST2)
111In, 177Lu
Spheroids, cell culture, mouse, patients (ex vivo)
1 MBq/mL 177Lu in vitro, 30 MBq 177Lu in vivo
[42, 43]
Molecular profiling
Blood NET transcript analysis
Neuroendocrine tumors (SST2)
177Lu
Patients
[177Lu]Lu-DOTA-TATE-based PRRT
NETest, PPQ: PRRT predictive quotient (PPQ)
[4446]
Molecular profiling
Whole genome microarray analysis
Neuroendocrine tumor (SST2), thyroid gland, various normal tissues, Kidney
177Lu, 131I, 211At
Mouse, rat
< 15 MBq 177Lu, < 4.7 MBq 131I, < 42 kBq 211At
[4750]
Molecular profiling
Targeted next-generation sequencing of DNA damage-repair associated genes
Prostate cancer
225Ac
Biopsies
[225Ac]Ac-PSMA-617 therapy
[8]
Relative biological effectiveness
Cell survival
Neuroendocrine tumor (SST2)
177Lu, 213Bi
Cell culture
< 10 Gy (177Lu), < 5.2 MBq (7 Gy) 213Bi
RBE = 6
[51]
Radiation quality effects
Cell survival, cell viability, gene expression, DNA damage, in vivo therapy studies
 
131I, 161Tb, 177Lu
Cell culture, mouse
< 9.25 MBq 131I in vitro
[52, 53]
Cell membrane-mediated non-targeted effects
Cell membrane lipid raft analysis, underlying signaling pathways, cell survival, DNA damage tumor volume
Colon cancer (CEA), vulvar squamous carcinoma (A431 HER2 + CEA), ovarian carcinoma (SKOV3 MISRII), endometrial carcinoma (AN3CA MISRII)
125I, 212Pb/212Bi, 213Bi
Cell culture, mouse
< 0.5 MBq/ml 212Pb in vitro, 0.5 MBq/mL 213Bi in vitro, < 4 MBq 125I in vitro, 1.48 MBq 212Pb in vivo, 37 MBq 125I in vivo
[54, 55]
Single cell and micrometastases dosimetry
Calculation
Neuroendocrine tumor (SST2)
177Lu, 161Tb
Cell culture, computed cell model
2.5 MBq/mL 177Lu in vitro
[56, 57]
Radiobiology, generic dose models
Calculation
Kidneys, tumor
   
Development of the linear-quadratic model for nuclear medicine
[5864]
Thyroid dose-toxicity model
Retrospective calculations of TCP, NTCP
Thyroid treatment
131I
Patients
< 560 MBq [65]
Retrospective analysis
[65, 66]
Hepatocellular carcinoma tumor response
Prospective study based on [99mTc]Tc macro-aggregated albumin dosimetry
Liver treatment glass microspheres
90Y
Patients
> 205 Gy
Prospective study
[67]
Hepatic dose-toxicity model
BED, TD, EUD
Liver treatment glass and resin microspheres
90Y
< 250 Gy BED50
Dose-toxicity model
[68]
Kidney dose-toxicity model
BED, TCP, NTCP
SST2 agonists, treatment of neuroendocrine tumors
90Y, 177Lu
Patients
40 Gy BED
Clinical trial
[69]
Kidneys and red bone marrow toxicity model
BED
Neuroendocrine tumor
177Lu
Virtual patients
40 Gy2.5 kidneys BED, 2 Gy15 red bone marrow BED
In silico clinical trial
[70]
mIBG treatment
Retrospective calculations
Neuroblastoma mIBG treatment
131I
Patients
30 GBq
Two fractions
[71, 72]
Predicting tumor response
BED
Prostate carcinoma
177Lu
Patients
7.3 ± 0.3 GBq
Prediction of tumor volume shrinking using PBPK/PD modeling
[73]
BED biologically effective dose, EBRT external beam radiation therapy, EUD equivalent uniform dose, NAMPT nicotinamide phosphoribosyltransferase, NTCP normal-tissue complication probability, PMBC peripheral mononuclear blood cells, RBE relative biological effectiveness, SST2 somatostatin receptor subtype 2, TCP tumor control probability, TD tolerable dose, PARP poly-[ADP-ribose]-polymerase 1, PRRT peptide receptor radionuclide therapy
In addition, in a recent review it was stated that, for radiation oncology, incomplete physics and dosimetry reporting limits the progress of radiobiology [74]. The authors concluded that there is not only a crucial deficiency on experimental details but also a lack of interaction between medical physicists and radiobiologists. The reporting of results pertaining to radiobiology in nuclear medicine is often provided in activities rather than absorbed doses (Table 1). Therefore, future radiobiology studies in nuclear medicine will benefit from including good practice of dosimetry reporting [75]. Conversely, dosimetry calculations should be based on available experimental biological data.
Consequently, in nuclear medicine, patient care optimization principles, which include not only absorbed dose or dose rate parameters but also radiobiological parameters, should be integrated. To this end, close interaction and collaboration between radiochemists, radiopharmacists, radiobiologists, medical physicists, and physicians will be needed.

Radiobiology

Radiobiology (also known as radiation biology) is a branch of biology concerned with the biological effects of ionizing radiation on living organisms. Radiobiology studies the interactions of ionizing radiation on atomic and molecular structures and consequently their induced effects on cells, tissues, and organs, both normal and diseased. As such, radiobiology enhances the understanding of biological outcome (harm or benefit) from ionizing radiation exposure.
When ionizing radiation impinges living matter, it deposits energy along its path leading to atomic ionization, thereby damaging biological molecular structures (Fig. 1). In the common paradigm, DNA is considered the critical target for radiation damage [76]. However, not only DNA, but also proteins, lipids and metabolites may be modified by ionizing radiation [77, 78]. In direct action, absorption of ionizing radiation will happen at the site of the atoms of the cellular molecules. Subsequent ionization events may cause breakage of chemical bonds. It may also convert atoms and molecules into free radicals with very reactive unpaired electrons that can further react with neighboring molecules after which a chain of damaging reactions may occur. The indirect effect from the absorption of ionizing radiation is the production of free hydroxyl and other highly reactive radicals, due to the hydrolysis of water molecules. Despite their short existence, they can still diffuse to and damage other cellular molecules. Moreover, oxygen can create reactive oxygen or nitrogen radical species with greater stability, longer lifetimes, and thus wider diffusion possibilities [76, 78]. The abundance of these oxygen radicals generates a condition known as oxidative stress which can further impact cellular signaling and alter metabolic pathways resulting in, among others, cell death mechanisms, senescence, and inflammation [78, 79]. Furthermore, ionizing radiation can harm supramolecular structures like cellular membranes, mitochondria, the endoplasmic reticulum, the Golgi apparatus, the lysosomal system, and the cytoskeleton [80]. Finally, also aspects beyond cellular boundaries are increasingly being considered in radiobiology, like the tumor microenvironment, intercellular communication, immune responses, and the abscopal effect [81, 82].
Research in radiobiology can rely on the newest techniques and insights in biology in general and is exploiting (epi)genomics, proteomics, metabolomics, high-throughput screening, and exploring new models like stem cells, organoids, in vivo orthotopic and subcutaneous patient-derived xenograft models, or siRNA- or CRISPR/Cas9-derived models. These are anticipated to lead to new hypotheses to understand the effects generated from ionizing radiation on biological systems and to improve therapies based on ionizing radiation [78, 83].
Today, improved insights into the dose-response effects caused by ionizing radiation on tumor cell killing as well as on acute and long-term normal tissue collateral damage are impacting greatly treatment planning in external beam radiation therapy (EBRT) [83, 84]. Of several models, the linear-quadratic (LQ) model has been best validated by experimental and clinical data to describe cell survival fractions. The selection of accurate LQ parameters, α, β, and α/β, is pivotal for a reliable estimate of radiation response. Clinically, the LQ model is mainly used to estimate equivalent radiotherapy schedules (e.g., calculate the equivalent dose in 2-Gy fractions), but increasingly also to predict tumor control probability (TCP) and normal tissue complication probability (NTCP) using logistic models [76, 85]. In addition, radiobiological discoveries are guiding clinical trials that test EBRT combined with inhibitors of the DNA damage response and immune or cell cycle checkpoint inhibitors. To have maximum impact for individual patients, predictive biomarkers should be identified that enable the rational selection of treatments to combine with EBRT. Further research into the radiobiology of tumor metabolism, cancer stem cells, and the tumor microenvironment has the potential to translate current knowledge and future gains to the clinic [83].
The question arises whether there is a need for a dedicated nuclear medicine radiobiology or whether we can rely on radiobiological models derived for EBRT or brachytherapy.
The response of a living system to an irradiation strongly depends on the distribution of absorbed doses across space and time. As these dose distributions in EBRT and brachytherapy are very different to those in radionuclide therapy, extrapolation of EBRT or brachytherapy radiobiology to radionuclide therapy is not straightforward. Indeed, the specific physical characteristics of radionuclide therapy (mixed radiation qualities, time-varying and protracted exposure, low absorbed dose rates, and inhomogeneous dose distributions) differ from those of conventional EBRT (short exposure time, high absorbed dose rate, and mostly homogeneous irradiation fields) and brachytherapy (even in the low dose rate case there is a well-defined source distribution). As a result, the responses of irradiated tissues and of the human/patient are expected to be different for radionuclide therapy [10, 86, 87]. For example, due to the time-varying and comparatively low dose rates in radionuclide therapies, the DNA damage induction and repair may strongly differ compared to EBRT [7, 12, 13, 75]. Consequently, there is a need for the generation and application of more radiobiological knowledge specific for nuclear medicine diagnostic and therapeutic procedures.
Efforts to gather more evidence in radiobiology regarding systemic exposure to ionizing radiation in nuclear medicine applications have been increasing recently; this is illustrated in this paragraph at the example of [177Lu]Lu-DOTA-[Tyr3]octreotate ([177Lu]Lu-DOTA-TATE) radionuclide therapy in patients with advanced, progressive, somatostatin receptor subtype (SST2)-positive midgut neuroendocrine tumors (NETs) that was studied in the NETTER-1 phase III trial [88]. Even though [177Lu]Lu-DOTA-TATE is clearly successful in terms of survival benefits, current figures could be further improved. In addition, treatment is limited by potential adverse effects on the kidneys and the bone marrow, hindering the use to its full potential. This emphasizes the need to further optimize [177Lu]Lu-DOTA-TATE radionuclide therapy to further improve efficacy while reducing toxicity. This includes improved dosimetry hand in hand with a deep biological evaluation of superior radionuclides, improved SST2 ligands, increased SST2 levels, the role of tumor microenvironment, and combinations with immunotherapy, targeted therapy or DNA modulating agents, as well as predictive markers for improved patient selection and treatment follow-up [24, 44, 45, 8991].

Position of the EANM

Radiobiology is particularly relevant for nuclear medicine therapies, as these treatments differ substantially from irradiation by external radiation sources. This is highlighted in the common strategic research agenda for radiation protection in medicine [92] developed by the five medical societies involved in the medical application of ionizing radiation, which later founded the European Alliance for Medical Radiation Protection Research (EURAMED) (https://​www.​euramed.​eu/​)1. An improved understanding of the biological processes with special regard to the effects of ionizing radiation to normal tissues and tumors is needed to individualize the use of existing and future developed radiopharmaceuticals. Therefore, the radiobiological knowledge concerning the specific needs of nuclear medicine (e.g., patient-specific and tumor-specific radiation sensitivity, dose-effect relationships, spatio-temporal properties, therapy response, normal tissue effects, role of microenvironment and systemic reactions, combination therapies) must be obtained and considered together with physical and medical parameters in the development of nuclear medicine procedures. This will also foster the principles stated in the EC Directive 2013/59/Euratom, article 56, that exposures of target volumes in nuclear medicine treatments shall be individually planned and their delivery appropriately verified [93]. How to interpret the EC directive for nuclear medicine therapies is further detailed in the recently published EANM position paper on article 56 of the Council Directive 2013/59/Euratom [94].
Absorbed dose measurements can be combined with radiobiological parameters to enable an improved estimation and prediction of efficacy and adverse effects, which can further support treatment planning [94]. This additional input is presently, however, very rarely used, as only limited studies related to therapeutic use of radiopharmaceuticals and including radiobiological parameters are available. Of note, the precision dosimetry approach to describe the dose on the cellular and subcellular level in targeted radionuclide therapy is under development [56, 57].
In diagnostic nuclear medicine applications, especially in longitudinal procedures, the role of radiobiology and the long-term consequences of radiobiology-related findings, such as reported in studies on DNA damage and repair, still has to be defined [95101]. Currently, these studies provide no evidence that diagnostic nuclear medicine procedures are not safe.
Consequently, EANM believes that, to further optimize nuclear medicine procedures for each individual patient, efforts should be undertaken to promote the integration of radiobiology within nuclear medicine by endorsing further research and teaching activities. The knowledge of different disciplines, such as biology, chemistry, medicine, pharmacy, and physics, can then be combined for providing reproducible results, which are, ideally, traceable to metrological standards.

Essential radiobiological studies

The nature of radiation exposure resulting from nuclear medicine procedures is diverse and comprises different radiation qualities, absorbed doses, dose rates, and temporal and spatial dose distributions [102, 103]. Low doses are encountered in diagnostic procedures as well as from out-of-target therapeutic exposures. High doses are expected within the tumor and in the close proximity of the tumor during radionuclide therapy. The determination of the absorbed dose to the tissue and on a (sub-)cellular scale are a prerequisite for defining dose-effect relationships, both in estimating (pre-)clinical therapy outcome and normal tissue toxicity as well as in assessing the cellular and molecular mechanisms, including repair capacity (Fig. 2).
With the ambition to maximize the benefits of radiopharmaceutical products that are effective and safe for each individual patient, preclinical and translational science undertakes dedicated research to understand the biological characteristics of tumor and normal tissue intrinsic radiosensitivities and the fundamental biological mechanisms underlying the therapeutic and short- and long-term cytotoxic effects of radiopharmaceutical products, as well as determining the dose-effect relationship herein (Fig. 2) [911]. Essential aspects to investigate related to patient- and tumor-specific radiosensitivities include genetic background and underlying anomalies that impact radiosensitivity, target distribution, tumor heterogeneity, tumor micro-environment, and tumor growth and metastatic spread. Important endpoints to be assessed in tumor and healthy tissue responses are DNA damage, damage to other cell elements, death and survival pathways, oxidative stress and inflammation, effect on the immune and systemic reactions, and repair capacities. To this end, high-end molecular and cellular biology tools, omics data analyses (proteomic, transcriptomic, genomic, radiomics) as well as bioimaging (microscopy, PET/CT or SPECT/CT, autoradiography) are available. As such, radiobiological data may reveal patient-specific radiation sensitivity traits useful as predictive biomarkers of response for a personalized radionuclide therapy regimen (e.g., genomic traits, target level and distribution, anomalies in signaling pathways altering radiosensitivity) as well as biomarkers useful for therapy response monitoring, both on a therapeutic aspect as well as for normal tissue damage. Radiobiological findings may also be used as input for the development of radiosensitizers or radioprotectant agents. Moreover, the radiobiology of fractionation schemes (how many/how much activity per fraction) as well as the radiobiology of combination therapies (combinations of radionuclide therapy with chemotherapy, tyrosine kinase inhibitors, immunotherapy, hormone therapy, or radiosensitizers) [9, 25] is not well explored and could be helpful in defining optimal treatment strategies. Finally, standardization of biological study protocols as well as depositing study data in repositories is required to allow comparison and combining of cohorts.
Some publications, regarding both external and internal irradiation, indicate that there is a very low dose range (< 10 mGy) which shows a different dose response compared to higher doses [99, 104107]. Therefore, extrapolating from higher absorbed doses and dose rates to very low doses and dose rates is not straightforward and needs further research. It is likely that other, different, biological responses exist after low-dose and high-dose ionizing radiation exposure using radiopharmaceuticals for both tumor and normal tissues.

Investigating low-dose radiation effects in nuclear medicine

The linear-no-threshold (LNT) model based on the extrapolation of epidemiological data at high absorbed doses is currently used to estimate the risk at low doses [108], although this is a matter of debate [109, 110]. An important aspect of the justification of using this model is that radiation carcinogenesis has been assumed to be primarily driven by the damage to the DNA and subsequent mutation of growth-regulating genes in target cells. Yet, a number of other potential mechanisms contributing to and modulating radiation carcinogenesis have been proposed, including epigenetic mechanisms of gene regulation such as DNA methylation and miRNA expression, transmissible genomic instability, bystander effects, and adaptive response. The extent to which these modulating effects and non-mutational mechanisms challenge the validity of the LNT risk extrapolation model needs to be determined. For this purpose, the use of well-validated animal and human cellular/tissue models of radiation carcinogenesis (both solid cancers and leukemias) is required. In addition, also non-cancer effects (e.g., cardiovascular and neurocognitive) should be considered and studied [111].
The key question here is whether the LNT model is valid for internal radiation exposure such as that encountered from nuclear medicine procedures with typically low dose rates, heterogeneous dose distributions, and a protracted nature of exposure. Therefore, the determination of corresponding low-dose-effect relationships can be a basis for risk assessment also in radiation protection (ALARA, LNT hypothesis, second cancer risk), e.g., in medical imaging or staff exposure.
To describe and monitor such effects, studies are needed to identify biomarkers for assessing short-term or medium-/long-term stochastic radiation risks (cancer and non-cancer) that (1) are sufficiently sensitive in the low-dose range (< 100 mGy), (2) are strongly linked to medium-/long-term side effects of ionizing radiation, and (3) possess definite dose/dose rate/dose fractionation/radiation quality dependencies.

Investigating therapeutic radiation effects in nuclear medicine

Currently, dose-effect relations are not fully utilized in most radionuclide therapies, as these therapies are given at a minimal activity that is deemed safe in all patients and effective to some extent. This often results in suboptimal therapy delivery. Radioembolization therapies arguably form the exception to this rule, as both normal liver thresholds and tumor target absorbed doses are considered as input to the treatment planning of these therapies.
More studies should be undertaken to determine TCP and NTCP curves for specific radionuclide therapies. In this context, radiobiological data (e.g., LQ α/β parameters and repair kinetics) can serve as input for better dose-effect modeling [58, 59, 65, 70] taking into account the radiation quality, dose rate, dose fractionation, and dose distribution on the tissue as well as on the (sub)cellular scale [60]. Finally, results from comparison studies with external beam radiation therapy could inform on better treatment strategies in nuclear medicine.
Radiobiology and dosimetry should be integrated in all stages of the development of individualized radionuclide therapy drugs. Preclinical experiments should deliver the radiobiological data through standardized and controlled settings with multiple cancer models to study response variability. Radiobiological concepts should ideally form the basis for the design of clinical trial protocols. Phase I studies focus on safety and thus should consider both absorbed dose and individual patient radiosensitivity. Phase II studies should ideally be based on absorbed dose thresholds and individual radiosensitivity. In many cases, the choice is made for a phase II trial with fixed activity at the maximum tolerable activity from phase I in order to simplify the clinical protocol. However, without absorbed doses available, it is impossible to build knowledge on dose-effect relationships and prospective clinical trials based on individually absorbed doses are crucial [67].

Discussion

Radiobiological knowledge is not yet used in many nuclear medicine applications, or it is used only in a basic phenomenological manner, such as that integrated in the model of biologically effective dose (BED). This can be attributed to two main reasons:
1.
Detailed radiobiological knowledge is currently not readily available, because of
 
a.
The specifics of nuclear medicine procedures and their multiple parameters involved (dose, dose rate, individual DNA repair capacity, …) as well as the heterogeneity of the conditions being treated.
 
b.
The complexity of its integration into the clinical procedures for example due to technical constraints (e.g., microscopes, bone marrow biopsies) or missing know-how.
 
c.
The associated patient load (time per patient per measurement).
 
d.
The resources and costs associated (personnel, supplies, and others).
 
2.
The phenomenological parameters (LQ α/β parameters, DNA repair) known from EBRT are incorrectly thought to suffice fully for the needs of all nuclear medicine applications.
 
All of the above points need to be addressed for continued improvement of nuclear medicine procedures, including the adequate integration of radiobiology. To solve item 1.a requires intensified research as previously discussed, while item 1.b needs education and training of all involved, scientists and physicians. Both call for the need of standardized procedures to produce reliable data. Lastly, items 1.c and 1.d are an issue for cost-benefit analysis which is mandatory for all medical procedures. This applies also to item 2 whose applicability and justification should be applied adhering to the requirements of best available science and cost-benefit analysis.
Thus, to develop an optimal nuclear medicine procedure one needs to acquire and include all knowledge and appropriate commitment from all involved in the process. This will inevitably include time investment from the patient for more measurements. Such collection of data and inclusion of a priori knowledge is the required input for a rigorous cost-benefit analysis. Implemented therapies will then be well founded, both scientifically and in terms of cost and effort.

Conclusion

While the role of radiobiology for diagnostics remains to be clarified, there is a clear role for radiobiology in optimizing the benefits of therapeutic radiopharmaceuticals to ensure that they are effective and safe for each individual patient. Just as radiobiology data are routinely used in EBRT treatment planning, nuclear medicine could also benefit from a deeper integration of such data. Therefore, there is a need to better define the dose-effect relationships of systemic ionizing radiation for tumors as well as for normal tissue. As absorbed dose rates and absorbed dose distributions in space and time are very different between external irradiation and systemic radionuclide exposure, distinct radiation-induced biological responses are expected in nuclear medicine and need to be explored. It is expected that a better understanding of radiobiological parameters can contribute to fully exploit the capabilities of new and existing nuclear medicine applications to be effective and safe for each individual patient. To this end, a strong link between radiochemists, radiopharmacists, radiobiologists, medical physicists, and physicians is warranted to design sound study designs. In particular, the inclusion of radiobiologists in the clinical team will be advantageous.

Acknowledgments

We thank all EANM committee members, who responded to the nuclear medicine radiobiology questionnaire by the EANM Radiobiology Working Group, for their valuable input.

Declarations

Conflict of interest

M.L. has received research grants by IPSEN Pharma and Nordic Nanovector. A.A., G.G., M.K., S.H., R.H, and F.v.L. have no conflicts of interest to declare that are relevant to the content of this article. U.E., L.S. and R.H. have no conflicts of interest to declare. M.L. and R.H. are members of the EANM board. The other authors are members of the following EANM committees: Radiation Protection (G.G., S.H.); Dosimetry (U.E., M.K., L.S.); Translational Molecular Imaging & Therapy (A.A., F.v.L.).

Ethical approval

This article does not contain any studies with human participants or animals performed by any of the authors.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Radiologie

Kombi-Abonnement

Mit e.Med Radiologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Radiologie, den Premium-Inhalten der radiologischen Fachzeitschriften, inklusive einer gedruckten Radiologie-Zeitschrift Ihrer Wahl.

Fußnoten
1
EURAMED represents the European Association of Nuclear Medicine (EANM), the European Federation of Organizations for Medical Physics (EFOMP), the European Federation of Radiographer Societies (EFRS), the European Society of Radiology (ESR), and the European Society for Radiotherapy and Oncology (ESTRO).
 
Literatur
11.
Zurück zum Zitat Verburg FA, Nonnekens J, Konijnenberg MW, de Jong M. To go where no one has gone before: the necessity of radiobiology studies for exploration beyond the limits of the “holy Gray” in radionuclide therapy. Eur J Nucl Med Mol Imaging. 2021. https://doi.org/10.1007/s00259-020-05147-5. Verburg FA, Nonnekens J, Konijnenberg MW, de Jong M. To go where no one has gone before: the necessity of radiobiology studies for exploration beyond the limits of the “holy Gray” in radionuclide therapy. Eur J Nucl Med Mol Imaging. 2021. https://​doi.​org/​10.​1007/​s00259-020-05147-5.
16.
Zurück zum Zitat Schumann S, Eberlein U, Lapa C, Müller J, Serfling S, Lassmann M, et al. α-Particle-induced DNA damage tracks in peripheral blood mononuclear cells of [223Ra]RaCl2-treated prostate cancer patients. Eur J Nucl Med Mol Imaging. 2021. https://doi.org/10.1007/s00259-020-05170-6. Schumann S, Eberlein U, Lapa C, Müller J, Serfling S, Lassmann M, et al. α-Particle-induced DNA damage tracks in peripheral blood mononuclear cells of [223Ra]RaCl2-treated prostate cancer patients. Eur J Nucl Med Mol Imaging. 2021. https://​doi.​org/​10.​1007/​s00259-020-05170-6.
18.
20.
Zurück zum Zitat Poty S, Mandleywala K, O'Neill E, Knight JC, Cornelissen B, Lewis JS. 89Zr-PET imaging of DNA double-strand breaks for the early monitoring of response following α- and β-particle radioimmunotherapy in a mouse model of pancreatic ductal adenocarcinoma. Theranostics. 2020;10:5802–14. https://doi.org/10.7150/thno.44772. Poty S, Mandleywala K, O'Neill E, Knight JC, Cornelissen B, Lewis JS. 89Zr-PET imaging of DNA double-strand breaks for the early monitoring of response following α- and β-particle radioimmunotherapy in a mouse model of pancreatic ductal adenocarcinoma. Theranostics. 2020;10:5802–14. https://​doi.​org/​10.​7150/​thno.​44772.
21.
Zurück zum Zitat Pichard A, Marcatili S, Karam J, Constanzo J, Ladjohounlou R, Courteau A, et al. The therapeutic effectiveness of 177Lu-lilotomab in B-cell non-Hodgkin lymphoma involves modulation of G2/M cell cycle arrest. Leukemia. 2020;34:1315–28. https://doi.org/10.1038/s41375-019-0677-4. Pichard A, Marcatili S, Karam J, Constanzo J, Ladjohounlou R, Courteau A, et al. The therapeutic effectiveness of 177Lu-lilotomab in B-cell non-Hodgkin lymphoma involves modulation of G2/M cell cycle arrest. Leukemia. 2020;34:1315–28. https://​doi.​org/​10.​1038/​s41375-019-0677-4.
27.
28.
Zurück zum Zitat Cullinane C, Waldeck K, Kirby L, Rogers BE, Eu P, Tothill RW, et al. Enhancing the anti-tumour activity of 177Lu-DOTA-octreotate radionuclide therapy in somatostatin receptor-2 expressing tumour models by targeting PARP. Sci Rep. 2020;10:10196. https://doi.org/10.1038/s41598-020-67199-9. Cullinane C, Waldeck K, Kirby L, Rogers BE, Eu P, Tothill RW, et al. Enhancing the anti-tumour activity of 177Lu-DOTA-octreotate radionuclide therapy in somatostatin receptor-2 expressing tumour models by targeting PARP. Sci Rep. 2020;10:10196. https://​doi.​org/​10.​1038/​s41598-020-67199-9.
36.
Zurück zum Zitat Tesson M, Rae C, Nixon C, Babich JW, Mairs RJ. Preliminary evaluation of prostate-targeted radiotherapy using 131I-MIP-1095 in combination with radiosensitising chemotherapeutic drugs. J Pharm Pharmacol. 2016;68:912–21. https://doi.org/10.1111/jphp.12558. Tesson M, Rae C, Nixon C, Babich JW, Mairs RJ. Preliminary evaluation of prostate-targeted radiotherapy using 131I-MIP-1095 in combination with radiosensitising chemotherapeutic drugs. J Pharm Pharmacol. 2016;68:912–21. https://​doi.​org/​10.​1111/​jphp.​12558.
37.
41.
43.
Zurück zum Zitat Feijtel D, Doeswijk GN, Verkaik NS, Haeck JC, Chicco D, Angotti C, et al. Inter and intra-tumor somatostatin receptor 2 heterogeneity influences peptide receptor radionuclide therapy response. Theranostics. 2021;11. Feijtel D, Doeswijk GN, Verkaik NS, Haeck JC, Chicco D, Angotti C, et al. Inter and intra-tumor somatostatin receptor 2 heterogeneity influences peptide receptor radionuclide therapy response. Theranostics. 2021;11.
54.
Zurück zum Zitat Paillas S, Ladjohounlou R, Lozza C, Pichard A, Boudousq V, Jarlier M, et al. Localized irradiation of Cell Membrane by Auger Electrons Is Cytotoxic Through Oxidative Stress-Mediated Nontargeted Effects. Antioxid Redox Signal. 2016;25:467–84. https://doi.org/10.1089/ars.2015.6309. Paillas S, Ladjohounlou R, Lozza C, Pichard A, Boudousq V, Jarlier M, et al. Localized irradiation of Cell Membrane by Auger Electrons Is Cytotoxic Through Oxidative Stress-Mediated Nontargeted Effects. Antioxid Redox Signal. 2016;25:467–84. https://​doi.​org/​10.​1089/​ars.​2015.​6309.
58.
Zurück zum Zitat Strigari L, Benassi M, Chiesa C, Cremonesi M, Bodei L, D'Andrea M. Dosimetry in nuclear medicine therapy: radiobiology application and results. Q J Nucl Med Mol Imaging. 2011;55:205–21.PubMed Strigari L, Benassi M, Chiesa C, Cremonesi M, Bodei L, D'Andrea M. Dosimetry in nuclear medicine therapy: radiobiology application and results. Q J Nucl Med Mol Imaging. 2011;55:205–21.PubMed
67.
Zurück zum Zitat Garin E, Tselikas L, Guiu B, Chalaye J, Edeline J, de Baere T, et al. Personalised versus standard dosimetry approach of selective internal radiation therapy in patients with locally advanced hepatocellular carcinoma (DOSISPHERE-01): a randomised, multicentre, open-label phase 2 trial. Lancet Gastroenterol Hepatol. 2020. https://doi.org/10.1016/s2468-1253(20)30290-9. Garin E, Tselikas L, Guiu B, Chalaye J, Edeline J, de Baere T, et al. Personalised versus standard dosimetry approach of selective internal radiation therapy in patients with locally advanced hepatocellular carcinoma (DOSISPHERE-01): a randomised, multicentre, open-label phase 2 trial. Lancet Gastroenterol Hepatol. 2020. https://​doi.​org/​10.​1016/​s2468-1253(20)30290-9.
72.
Zurück zum Zitat Flux GD, Chittenden SJ, Saran F, Gaze MN. Clinical applications of dosimetry for mIBG therapy. Q J Nucl Med Mol Imaging. 2011;55:116–25.PubMed Flux GD, Chittenden SJ, Saran F, Gaze MN. Clinical applications of dosimetry for mIBG therapy. Q J Nucl Med Mol Imaging. 2011;55:116–25.PubMed
76.
Zurück zum Zitat Castellucci P, Deandreis D, Krizsán A, Mirzaei S, Prior J, Sattler B, et al. European nuclear medicine guide. A joint publication by EANM and UEMS/EBNM 2020. Castellucci P, Deandreis D, Krizsán A, Mirzaei S, Prior J, Sattler B, et al. European nuclear medicine guide. A joint publication by EANM and UEMS/EBNM 2020.
92.
Zurück zum Zitat European Association of Nuclear Medicine (EANM), European Federation of Organizations for Medical Physics (EFOMP), European Federation of Radiographer Societies (ESR), European Society of Radiology (ESR), European Society for Radiotherapy and Oncology (ESTRO). Common strategic research agenda for radiation protection in medicine. Insights Imaging. 2017;8:183–97. https://doi.org/10.1007/s13244-016-0538-x. European Association of Nuclear Medicine (EANM), European Federation of Organizations for Medical Physics (EFOMP), European Federation of Radiographer Societies (ESR), European Society of Radiology (ESR), European Society for Radiotherapy and Oncology (ESTRO). Common strategic research agenda for radiation protection in medicine. Insights Imaging. 2017;8:183–97. https://​doi.​org/​10.​1007/​s13244-016-0538-x.
93.
Zurück zum Zitat Council of the European Union. European Council Directive 2013/59/Euratom on basic safety standards for protection against the dangers arising from exposure to ionising radiation and repealing directives 89/618/Euratom, 90/641/Euratom, 96/29/Euratom, 97/43/Euratom and 2003/122/Euratom. Off J EU. 2014;L13:1–73. Council of the European Union. European Council Directive 2013/59/Euratom on basic safety standards for protection against the dangers arising from exposure to ionising radiation and repealing directives 89/618/Euratom, 90/641/Euratom, 96/29/Euratom, 97/43/Euratom and 2003/122/Euratom. Off J EU. 2014;L13:1–73.
94.
Zurück zum Zitat Konijnenberg M, Herrmann K, Kobe C, Verburg F, Hindorf C, Hustinx R, et al. EANM position paper on article 56 of the Council Directive 2013/59/Euratom (basic safety standards) for nuclear medicine therapy. Eur J Nucl Med Mol Imaging. 2020. https://doi.org/10.1007/s00259-020-05038-9. Konijnenberg M, Herrmann K, Kobe C, Verburg F, Hindorf C, Hustinx R, et al. EANM position paper on article 56 of the Council Directive 2013/59/Euratom (basic safety standards) for nuclear medicine therapy. Eur J Nucl Med Mol Imaging. 2020. https://​doi.​org/​10.​1007/​s00259-020-05038-9.
102.
Zurück zum Zitat European Association of Nuclear Medicine, Flux G, Stokke C, (Editors). Internal Dosimetry Task Force Report. Treatment planning for molecular radiotherapy: potential and prospects. 2017. European Association of Nuclear Medicine, Flux G, Stokke C, (Editors). Internal Dosimetry Task Force Report. Treatment planning for molecular radiotherapy: potential and prospects. 2017.
108.
Zurück zum Zitat ICRP. Publication 103: The 2007 recommendations of the International Commission of Radiological Protection. Ann ICRP. 2007;37(2–4). ICRP. Publication 103: The 2007 recommendations of the International Commission of Radiological Protection. Ann ICRP. 2007;37(2–4).
111.
Zurück zum Zitat Bouffler S, Auvinen A, Cardis E, Durante M, Jourdain JR, Harms-Ringdahl M, et al. Strategic Research Agenda of the Multidisciplinary European Low Dose Initiative (MELODI). 2019 Bouffler S, Auvinen A, Cardis E, Durante M, Jourdain JR, Harms-Ringdahl M, et al. Strategic Research Agenda of the Multidisciplinary European Low Dose Initiative (MELODI). 2019
Metadaten
Titel
EANM position paper on the role of radiobiology in nuclear medicine
verfasst von
An Aerts
Uta Eberlein
Sören Holm
Roland Hustinx
Mark Konijnenberg
Lidia Strigari
Fijs W.B. van Leeuwen
Gerhard Glatting
Michael Lassmann
Publikationsdatum
29.04.2021
Verlag
Springer Berlin Heidelberg
Erschienen in
European Journal of Nuclear Medicine and Molecular Imaging / Ausgabe 11/2021
Print ISSN: 1619-7070
Elektronische ISSN: 1619-7089
DOI
https://doi.org/10.1007/s00259-021-05345-9

Weitere Artikel der Ausgabe 11/2021

European Journal of Nuclear Medicine and Molecular Imaging 11/2021 Zur Ausgabe