Skip to main content
The British Journal of Radiology logoLink to The British Journal of Radiology
. 2015 Apr 16;88(1051):20150042. doi: 10.1259/bjr.20150042

Improving external beam radiotherapy by combination with internal irradiation

A Dietrich 1,2,, L Koi 2,3, K Zöphel 1,2,4, W Sihver 5, J Kotzerke 1,2,4, M Baumann 1,2,3,6, M Krause 1,2,3,6
PMCID: PMC4628530  PMID: 25782328

Abstract

The efficacy of external beam radiotherapy (EBRT) is dose dependent, but the dose that can be applied to solid tumour lesions is limited by the sensitivity of the surrounding tissue. The combination of EBRT with systemically applied radioimmunotherapy (RIT) is a promising approach to increase efficacy of radiotherapy. Toxicities of both treatment modalities of this combination of internal and external radiotherapy (CIERT) are not additive, as different organs at risk are in target. However, advantages of both single treatments are combined, for example, precise high dose delivery to the bulk tumour via standard EBRT, which can be increased by addition of RIT, and potential targeting of micrometastases by RIT. Eventually, theragnostic radionuclide pairs can be used to predict uptake of the radiotherapeutic drug prior to and during therapy and find individual patients who may benefit from this treatment. This review aims to highlight the outcome of pre-clinical studies on CIERT and resultant questions for translation into the clinic. Few clinical data are available until now and reasons as well as challenges for clinical implementation are discussed.


External beam radiotherapy (EBRT) alone and in combination with surgery and/or chemotherapy is one of the main modalities for cancer treatment and has a high potential to permanently cure solid tumours even in locally advanced stages by inactivation of cancer stem cells.1 EBRT can be administered precisely to a target volume during a course of fractionated irradiation. The homogeneous energy dose has a high intensity in solid tumour lesions. For some cancers, survival rates after primary radiotherapy are high [e.g. early stage larynx cancer and early stage non-small-cell lung cancer (NSCLC)], whereas for many other entities they are not (e.g. glioblastomas, sarcomas and advanced NSCLC).2

One way to improve radiotherapy is to increase the inactivation of tumour cells. However, the applicable EBRT dose is limited by the radiosensitivity of the surrounding tissue. While EBRT is directed to the local tumour disease, the use of systemic radioimmunotherapy (RIT) offers the possibility to treat both, localized and diffuse tumours and (micro)metastases.3 Radionuclides are bound to carrier molecules that target tumour cells. Thus, they are distributed according to the properties of the tracer and are continuously effective during a longer period compared with EBRT, although dose rates decrease depending on the half-life of the radionuclide. Some free therapeutic radionuclides are effective for specific indications, e.g. 131I for treatment of thyroid cancer or palliative use of 223Ra against bone metastases. However, these cannot be translated to treatment of other entities. Besides, radioactive-labelled cytostatic drugs and hormone derivatives,4 particularly monoclonal antibodies (mAb) have been radiolabelled and investigated.5 Given a substantial difference in the target receptor expression between the tumour cells and surrounding normal tissues, a dose fall-off between both tissues can be expected. The radiolabelled mAb Zevalin® ibritumomab tiuxetan (Zevalin®, Bayer Healthcare Pharmaceuticals, Berlin, Germany) directed against CD20 is approved by the Food and Drug Administration(FDA) and the European Medicines Agency (EMA) for the treatment of follicular B-cell non-Hodgkin's lymphomas, which are generally considered as radiosensitive. However, mAb are large and are thus taken up slowly into solid tumour tissue followed by a long clearance. Additionally, accumulation in solid tumours depends on vascularization, vessel permeability, tumour size, interstitial pressure and other microenvironmental characteristics.6,7 Furthermore, mAb are rather susceptible when labelling under rough conditions. Thus, the application of molecules such as fragment antigen-binding (Fab),810 nanobodies,11,12 affybodies,13,14 single chain variable fragments (scFvs),15,16 aptamers1719 or peptides20,21 is considered. In addition to the effects on target cell, radionuclides with sufficient radiation path length (e.g. β-emitters) can destroy adjacent tumour cells by the crossfire effect, that is through the range of radiation in tissue, cells can be killed without having bound the radionuclide itself.22 This is regarded as a main advantage of RIT for the treatment of solid tumours as plasticity of tumour cells (e.g. loss of target antigen) and delivery barriers can be overcome by some extent. However, the dose-limiting organ in non-myelo-ablative RIT is the red bone marrow and myelosuppression the main toxicity.22 Therefore, the maximum tolerated activity that was applied in clinical RIT trials (reviewed in Navarro-Teulon et al23) did not result in tumour doses >33 Gy in large tumours, which is not enough to achieve permanent local control of solid tumours.

Combination of internal and external radiotherapy

The combination of internal (incorporated) and external radiotherapy (CIERT) is a novel promising approach in radiation oncology. In this review, CIERT is defined more specifically by an integrated (without interval) application of EBRT and systemically applied RIT. Other approaches such as the combination of external radiotherapy with selective internal radiotherapy, radioembolization, brachytherapy, seed implantation, other intravenously applied radionuclide therapies or sequential application of any of these treatments will not be considered here. Furthermore, the focus will be on solid tumours.

The potential benefit of such a combined irradiation is to increase the energy dose applied to the solid tumour lesion, while respecting the limitations of the surrounding normal tissues and the organs at risk (OARs) that are different for both treatment modalities (see above). Figure 1 summarizes the characteristics and OARs of EBRT and RIT and gives an overview on the advantages of the combinatorial approach. Beyond local treatment intensification, another advantage of CIERT can be the combination of local treatment, directed to the solid tumour, and systemic treatment, directed to the subclinically disseminated disease, that is, microscopic tumour lesions not detectable on imaging.

Figure 1.

Figure 1.

Combination of internal and external radiotherapy (CIERT). Treatment characteristics of external beam radiotherapy (EBRT) and radioimmunotherapy (RIT) are summarized and advantages of the combination strategy (CIERT) are depicted. Local treatment of the solid tumour via precise EBRT is supplemented by a systemically applied radiotherapeutic drug. Thereby, the tumour dose is enhanced without additional toxicity and (micro)metastasis are potentially targeted. Further, usage of theragnostic radionuclide pairs has the potential to predict delivery and dose distribution of RIT before and during treatment. OAR, organ at risk.

Many challenges are to be met prior to the initiation of CIERT. For example, thoughts on the treatment schedule of CIERT and dosimetry considerations are inevitable. The EBRT would usually be applied as standard treatment. Considerations on the RIT part equal usual aspects of RIT, for example, application of cold doses as well as the choice of the carrier molecule (according to the tumour target) and radionuclides. Accordingly, new developments in the field of RIT, for example, pre-targeting strategies, might be applicable for CIERT approaches in the future but have not been used in this context so far. Many of those aspects are intensively researched with regard to single treatments and reviewed elsewhere.3,2329 This work focuses on the presentation of pre-clinical and clinical investigations on CIERT as a promising treatment strategy.

Choice of radionuclides and theragnostic potential of combination of internal and external radiotherapy

The main factor of radiation toxicity is damage of DNA. If the amount and severity of radiation-induced damage exceeds the repair capacity of the cell, death occurs during mitosis. The linear energy transfer (LET) describes the energy released by the radiation over a certain distance and influences relative biological effectiveness (RBE).3,30 X-rays as well as γ- and β-emitters have low LET and thus produce individual DNA lesions mainly by indirect ionization that can easily be repaired. By contrast, high and intermediate LET particle emitters cause clusters of DNA damage that are difficult to repair. Thus, α-emitters (high LET) and Auger electrons (intermediate LET) are more cytotoxic at equivalent absorbed doses. The track path length of α-emitters covers only some cell layers (50–100 µm), and Auger electrons have an even shorter range (<1 µm), which, together with the high RBE, makes them suitable for treatment of small volumes such as micrometastasis.3,31 If larger solid tumours are targeted, microenvironmental factors such as perfusion, vessel permeability and the amount of connective tissue influence the distribution of RIT therapeutics. Thus, the application of β-particles may be most promising for CIERT because their path length of 0.5–12.0 mm enables the crossfire effect.3,30

In contrast to mitotic catastrophe caused by irradiation, apoptosis can be induced by some mAb via blockage of the respective receptor and modification of downstream signalling. Thus, the combination of irradiation and mAb may promote the manifestation of sublethal harm to severe damage, which finally lead to cell death. In case of CIERT, radiation is not only applied via EBRT but also by radionuclides bound to the mAb.

A fundamental requisite for the success of radioactivity delivery into solid tumours is that the radionuclide reaches the target and accumulates for an appropriate period. Thus, the pharmacological half-life of the carrier and half-life of the radioactive decay of the chosen nuclide need to be balanced.3,23 Most pre-clinical and clinical studies on CIERT used large mAb (approximately 150 kDa), which show a slow plasma clearance. Thus, intratumoral accumulation peaks usually several days after injection. Accordingly, most studies used β-emitters or emitters of Auger electrons with half-lives of at least several days. Pickhard et al32 recently showed the benefit of using 213Bi bound to an antibody against the epidermal growth factor receptor (EGFR) in combination with EBRT. They demonstrated that different cell death pathways are triggered by this α-emitter and photon irradiation. However, the short half-life of 213Bi (45 min) may limit its usage for solid tumours in vivo if the nuclide is linked to antibodies, because most doses will be applied before the tracer penetrates into tumour tissue. Thus, 213Bi may only be useful to treat haematological malignancies and therefore is not feasible for CIERT. The concept of pre-targeting is intensively researched in association with RIT as a single treatment. The tumour is pre-targeted with the unlabelled complementary prepared antibody, and the radionuclide is delivered via a small molecule recognizing the antibody by the complementary system in a second step. This may lead to higher tumour uptake with lower normal tissue retention (reviewed in van de Watering et al29). However, a combination with EBRT has never been investigated and substantial research on scheduling would be mandatory.

The concept of theragnostic approaches is applicable for CIERT, as theragnostic radionuclide pairs can be used for the RIT part of the therapy. The goal is to combine a diagnostic tool having an imaging radionuclide (positron or γ-radiation emitter) with a derived individualized therapeutic procedure using a therapeutic radionuclide (particle emitter). The tumour and normal tissue uptake of the respective drug can be evaluated for individual patients via positron emission tomography (PET) or single photon emission CT (SPECT) and give predictive information on a potential treatment benefit. The selection of appropriate radionuclides for imaging with regard to their replacement by a radionuclide for therapeutic purposes that exhibit similar chemical and physical properties is a crucial matter. Thus, it is important to consider different characteristics of radiation according to the requirements, such as decay characteristics, dose range and physical half-life of the radionuclides.30 Imaging with radionuclide-labelled conjugates provides pre-therapeutic information such as biodistribution, hints of a limiting or critical organ or tissue, and maximum tolerated dose. Dosimetry is most challenging, as pre-therapeutic imaging may not be congruent to actual delivered doses.33 However, this field is extensively investigated for peptide receptor radionuclide therapy (PRRT), and results are directly transferable to CIERT approaches. After applying therapeutic nuclide-labelled conjugates, the results of such treatment may again be monitored via imaging. A selection of theragnostic combinations of radionuclides are shown in Table 1. 64Cu is denoted here as PET radionuclide but with a considerable part of β-radiation as well as low-energy Auger electrons (0.6–8.3 keV), it has the potential for radiotherapeutic application. However, the half-life is rather short. Furthermore, 67Cu releases γ-radiation directly following the β-decay; thus, it can be used for SPECT,51 but its production is difficult and expensive.52 The positron emitters 86Y and 124I have been described controversially as PET nuclides since besides high β+-radiation energy they emit multiple high-energy γ photons that cause so-called multiple coincidences disturbing PET imaging quality. However, different correction methods allow improved quantitative imaging.50 Moreover, for the application of 90Y-labelled radiopharmaceuticals, it is suggested to estimate the uptake and dosimetry with the nuclide counterpart 86Y.36 Nevertheless, 86Y-PET is far from clinical routine, at least in the near future. Furthermore, 131I also emits γ-radiation that has been used for imaging, and 111In and 123I have a potential for treatment owing to their released Auger electrons.

Table 1.

Potential theragnostic radionuclidesa

Pair Half-life Radiation (keV) Application examples
Study Imaging Model Entity
64Cu/67Cu 12.7 h/2.6 days β+ 653 (17.5%)/β 562 (100%)b Anderson and Ferdani34/Novak-Hofer and Schubiger35 PET; small animal PET/SPECT; biodistribution Patients hypoxia; mice (tm) mAb/patients mAb; mice mAb Fabs lc, cc; SCC/NHL, colc, bc; nb, colc
86Y/90Y 14.7 h/2 days β+ 2766 (17.5%)c/β 2280 (100%) Lopci et al36/McKinney and Beaven37 Small animal PET mice (tm) mAb/patients mAb (Zevalin®) Different xenografts/NHL
89Zr/90Y 3.3 days/2.7 days β+ 902 (22.7%)c/β 2280 (100%) Osborne et al38/Perk et al39 PET; biodistribution Patients mAb/patient mice (tm) mAb (Zevalin) pc/NHL
86Y/177Lu 14.7 h/6.6 days β+ 2766 (17.5%)c/β 498 (79%) A.e. 4.3–65.3 Lopci et al36/Liu et al40 Small animal PET/small animal SPECT mice (tm) mAb/mice (tm) mAb Different xenografts/HNSCC
89Zr/177Lu 3.3 days/6.6 days β+ 902 (22.7%)c/β 498 (79%) A.e. 4.3–65.3 Osborne et al38 PET Patients mAb pc
99mTc/186Re 6 h/3.7 days γ 140 (99%)/β 1069 (71%) A.e. 4.5–69.5 Nagar et al41 SPECT Patients MIBI Parathyroid adenoma
99mTc/188Re 6 h/17 h γ 3140 (99%)/β 1069 (71%) A.e. 47.7–69.9 Müller et al42 Biodistribution mice (tm) folate nasc
111In/90Y 2.8 days/2.7 days γ 171; 245 (100%)/β 2280 (100%) O'Donnell et al43 SPECT Patients mAb pc
123I/131I 13.2 h/8 days γ 159 (97%)/β 606 (89%) Bravo et al44 SPECT Patients NaI thc
124I/131I 4.2 days/8 days β+ 3673 (23%)c/β 606 (89%) Van Nostrand et al45 PET Patients NaI thc
124I/186Re 4.2 days/3.7 days β+ 3673 (23%)c/β 1069 (71%) A.e. 4.5–69.5 Verel et al46 Biodistribution mice (tm) mAb HNSCC
124I/188Re 4.2 days/17 h β+ 3673 (23%)c/β 2120 (71%) A.e. 47.7–69.9 Verel et al46/Torres et al47 Biodistribution/SPECT mice (tm) mAb/patients mAb HNSCC/glioma

A.e, Auger electrons; bc, bladder cancer; cc, cervical carcinoma; colc, colon carcinoma; Fab, Fragment antigen binding; HNSCC, head and neck squamous cell carcinoma; lc, lung carcinoma; mAb, monoclonal antibodies; MIBI, methoxy isobutyl isonitrile; nasc, nasopharyngeal carcinoma; nb, neuroblastoma; NHL, non-Hodgkin's lymphoma; pc, prostate cancer; PET, positron emission tomography; SCC, squamous cell carcinoma (A431); SPECT, single photon emission CT; thc, thyroid cancer; tm, tumour model.

a

Data from Laboratoire National Henri Becquerel: http://www.nucleide.org/DDEP_WG/DDEPdata.htm.48

c

Data from Lubberink and Herzog.50

PRE-CLINICAL RESULTS: PROMISES AND CHALLENGES OF COMBINATION OF INTERNAL AND EXTERNAL RADIOTHERAPY

Pre-clinical investigations on CIERT have been published for more than 25 years53,54 and are summarized in Table 2. Comparison of different data sets is limited because they differ in almost all aspects: model systems, tumour entities, target antigens, radionuclides, delivered activities and doses, treatment schedules and end points. Pre-clinical investigations are used to gain knowledge on principal radiobiological aspects, associations of treatment schedules, target antigen and tumour entity with outcome as well as on principal efficacy.

Table 2.

Pre-clinical studies on combination of internal and external radiotherapy

Model Entity Nuclide mAb Therapy schedulea EBRT dose RIT dose End point Study
Two dimensional cell cultures Glioblastoma, colorectal 125I Murine anti-EGFR (mAb 425) EBRT, after 2 days RIT 6 Gy 0.37 MBq ml−1 Cell growth Bender et al55
Squamous carcinoma of vulva 131I Humanized murine anti-EGFR (Cetuximab) Imprecise, probably simultaneous 2, 4, 10 Gy 0.1 MBq ml−1 Cell growth Rades et al56
HNSCC, squamous carcinoma of vulva 90Y Humanized murine anti-EGFR (Cetuximab) Simultaneous 2, 4 Gy 1 MBq Clonogenicity Saki et al57
HNSCC, squamous carcinoma of vulva 90Y Humanized murine anti-EGFR (Cetuximab) EBRT, after 4 h RIT 2, 4, 6 Gy 0.5, 1, 1.5 MBq ml−1 Clonogenicity Eke et al58
HNSCC 213Bi Humanized murine anti-EGFR (Matuzumab) Simultaneous 2 Gy 0.037 MBq ml−1 Clonogenicity Pickhard et al32
Three dimensional spheroids HNSCC 90Y Humanized murine anti-EGFR (Cetuximab) Simultaneous 2.5–25 Gy 1.2 MBq ml−1 Spheroid control Ingargiola et al59
Xenograft tumours in mice Breast 111In Murine anti-γH2AX with TAT motive RIT, after 1 h EBRT 10 Gy 60 MBq Clonogenicity, tumour growth delay Cornelissen et al60
Breast, HNSCC 111In Murine anti-γH2AX with NLS and EGFR-targeting properties RIT, after 1 h EBRT 10 Gy 5 MBq Clonogenicity Cornelissen et al61
Colorectal 131I 3 F(ab')2 of murine anti-CEA (mAb 35, CE25-B7, B93) fx EBRT, on last day fx RIT starts 16–32 Gy in 5 fx 2 × 55.5 MBq Tumour growth delay, tumour control Buchegger et al62
Colorectal 131I 4 murine anti-CEA (mAb 35, B7, B93, Bl7) Various 30, 40 Gy in 10 fx 3 × 7.4 MBq Tumour growth delay Sun et al63
Cervical adenocarcinoma 131I Murine anti-PLAP + anti-CK8 (H7, TS1) 3 fx EBRT, after 1 week RIT 15 Gy in 3 fx 20 MBq Tumour growth delay Eriksson et al64
HNSCC 90Y Humanized murine anti-EGFR (Cetuximab) EBRT, after 2–4 days RIT 14–50 Gy 2.8 MBq Tumour control Koi et al65
Liver metastasis in mice Colorectal 131I 4 murine anti-CEA (mAb 35, CE25-B7, B93, Bl7) 5 fx EBRT, RIT, 5 fx EBRT, RIT 20 Gy in 10 fx 2 × 5.55 MBq Survival Vogel et al66

γH2AX, phosphorylated histone H2AX; CEA, carcinoembryonic antigen; CK8, cytokeratin 8; EBRT, external beam radiotherapy; EGFR, epidermal growth factor receptor; fx, fractionated; HNSCC, head and neck squamous cell carcinoma; mAb, monoclonal antibodies; NLS, nuclear localization sequence; PLAP, placental alkaline phosphatase; RIT, radioimmunotherapy; TAT, peptide of the trans-activator of transcription protein of HIV-1 virus.

a

If treatments directly followed each other this was counted as simultaneously.

Pre-clinical model systems and their impact on combination of internal and external radiotherapy research

Selection of the model system for pre-clinical experiments on CIERT is of high relevance and should be decided in light of the research question addressed. Four of the studies summarized in Table 2 were investigated in a German framework for radiation research. Herein, the same therapeutic antibody [cetuximab (Erbitux®, Merck KGaA, Darmstadt, Germany) labelled with 90Y], and the same panel of head and neck squamous cell carcinoma (HNSCC) cell lines were used. This offers the possibility to compare the results obtained in different model systems along the translational path from two-dimensional (2D) cell cultures to pre-clinical in vivo models. Within this consortium, Saki et al57 demonstrated a decrease of the surviving fraction of the HNSCC cell line UT-SCC-5 after combination of EBRT with cold Cetuximab, which was further reduced by CIERT. However, this cell line was a non-responder in a study of Gurtner et al67 who combined EBRT with (cold) Cetuximab treatment in xenograft tumours. Koi et al65 showed 2 years later that UT-SCC-5 also did not respond to CIERT when compared with EBRT alone in a tumour control analysis in xenografts. This discrepancy between 2D cell cultures and in vivo results is likely caused by the pharmacokinetics of the therapeutic antibody in vivo. Indeed, UT-SCC-5 xenograft tumours were shown to be the least perfused tumour of a panel of six HNSCC models.68 Furthermore, a histological evaluation and PET study in Koi et al65 demonstrated the lowest tumour uptake and an uneven distribution of the radiolabelled antibody for UT-SCC-5 xenografts compared with two other models. In the same study, a substantial reduction of the tumour control dose 50 (TCD50, dose of EBRT that cures 50% of the xenograft tumours) with CIERT was demonstrated for FaDu xenografts.65 Of note, tumours of this cell line were well perfused and showed the highest uptake of Cetuximab in the PET analysis.65,68 However, they did not or only marginally responded to the combination of EBRT and cold Cetuximab,65,67,69 which leads to the conclusion that the antibody functioned as a carrier of the radionuclide in the CIERT study but did not contribute by itself to the therapeutic effect. The result demonstrates the promising potential of CIERT if the radiolabelled agent accumulates in the target tumour and is well distributed. This encourages the development of theragnostic approaches that have the potential to identify patients who will benefit from CIERT.5,65

Ingargiola et al59 adopted three-dimensional (3D) spheroid cultures to the in vivo tumour control assay. This approach reflects the 3D morphology of tumour cells and pathophysiological gradients (oxygen, metabolites, pH), which may influence their intrinsic radiosensitivity.70,71 CIERT compared with EBRT alone led to a pronounced enhancement of spheroid control probability in the SAS cells as a model. However, SAS and FaDu were already controlled after RIT treatment alone in spheroids as well as 2D cultures.58,59 The obviously higher RIT efficacy in vitro than in xenografts may be explained by differing RIT doses used in the studies. Regardless of the limitations of efficacy tests, in vitro cell culture models offer important possibilities to study other end points such as cellular binding and uptake55,57,59 as well as downstream signalling.32,57,58 Accordingly, in vitro models should be used to uncover underlying mechanisms and can give indications on efficacy. However, to evaluate the therapeutic benefit of combined treatment strategies, in vivo studies are inevitable for translation into the clinic. In general, only a minority of investigators used tumour control as end point.62,63,65 However, studying curative end points in preclinal trials is a pre-requisite for translation into clinical protocols with curative aim as promising results for tumour growth delay not always lead to enhanced control probability.67,72 Additionally, the investigation of normal tissue toxicity should be performed in CIERT studies, as sparing of normal tissue dose is a major aspect of this treatment strategy. Again, only few studies paid attention to this complex issue, which may require the investigation of orthotopic tumour models. Moreover, if specific human antigens are targeted, animal models with transplanted human tumours fail to predict normal tissue response, as the target is not present on murine normal cells.

Scheduling of combination of internal and external radiotherapy

A crucial question that should be addressed in pre-clinical experiments on CIERT is the treatment schedule. Standard EBRT in cancer treatment is applied in multiple daily fractions over several weeks to enable normal tissue repair. Hence, radiolabelled antibodies could be injected before, during or after finishing radiation or can be applied also fractionated, which again offers innumerable possibilities for combination.

In the in vivo studies included in Table 2, a wide variety of schedules were applied. Many early studies showed enhanced tumour uptake of radiolabelled antibodies after EBRT.53,54,73,74 Suggested reasons were an increase of the vascular permeability after EBRT73 and the shrinkage of tumour mass54,75 with RIT being more effective on smaller target volumes.66,75 The effect was dose dependent and did not lead to a higher uptake in normal tissues if X-rays were used.53,63,74 Interestingly, Gridley et al74 showed that the tumour uptake of an anti-carcinoembryonic antigen (CEA) antibody could even be increased by using protons for EBRT instead of X-rays. However, this was accompanied by higher accumulation in normal tissue and traced back to a stronger antigen secretion after proton irradiation. These early studies focused on the improvement of RIT, and tumour uptake was the main end point measured.

The first systematic pre-clinical study on CIERT as a curative combined approach was investigated in 1995 by Buchegger et al.62 A fractionated schedule and high doses of EBRT were combined with high injected RIT activities [mixture of different anti-CEA F(ab')2 fragments]. Of significance, not only tumour uptake was analysed but also efficacy of treatment. The results show a prominent advantage of CIERT when compared with the single treatments in two tumour models without additional toxicity. The same group investigated if tuning of treatment schedule may further increase the outcome.63 Simultaneous application of EBRT and RIT, both in a fractionated scheme, turned out to be most effective causing the longest growth delay compared with other treatment timings in this trial. The associated biodistribution experiment revealed no pronounced changes of antibody uptake in tumours irradiated with low doses but a reduced uptake if 30 Gy (10 fractions) were applied prior to the antibody injection. Additionally, a clear disadvantage of a long delay between EBRT and RIT was shown. However, uptake was not investigated for the complex fractionated timing with the highest therapeutic potential. This schedule was further tested on orthotopic liver metastases of a lung tumour model.66 Again, a benefit of CIERT over the single treatments was demonstrated, which led to translation of the approach into a clinical feasibility study.76

Ruan et al75 addressed the fact that the efficacy of RIT and the tumour response are influenced by the uniformity of antibody microdistribution in the target tissue. To this end, the authors investigated tumour uptake as well as distribution of a radiolabelled antibody directed against the surface protein A33 on autoradiographic slices for different treatment schemes. The simultaneous application of EBRT and RIT led to the highest uptake, but the radiolabelled antibodies were most evenly distributed in non-irradiated tumours. However, no efficacy analysis of the different treatment schedules was performed in this study, making conclusions on the impact of the radionuclide uptake and distribution on treatment response speculative. Generally, the impact of uneven distribution on treatment success strongly depends on the crossfire effect. The magnitude of this effect relies on the used radionuclide and the tissue penetration depth of emitted radiation and may overcome poor dissemination of the radiotherapeutic drug.3,22

Taken together, the application of RIT simultaneously with or after EBRT was in most cases beneficial to a schedule where RIT was applied prior to external irradiation. It may not make a difference if fractionated EBRT starts some days prior to RIT injection or on the same day, as mAb circulate for several days. However, there are indications that the effect of increased antibody uptake after EBRT disappears at higher doses.62,63 The optimal treatment schedule has to be defined for each individual antigen/antibody pair and may be influenced by the chosen tumour model as well. Additionally, increased connective tissue amounts and cystic composition as well as low numbers of residual viable tumour cells were observed in xenografts treated with CIERT compared with single treatments. Thus, not only increased dose delivery but also other radiobiological processes may be involved in the promising pre-clinical results obtained with CIERT. To this end, further pre-clinical research is required to understand mechanisms and enhance the potential of this promising combined treatment approach.

Noteworthy, all studies involving efficacy testing show a benefit of CIERT over single treatments, and normal tissue was spared if this was investigated, as far as this end point can be judged in mouse models. The addition of other approaches to enhance uptake or specificity may further improve CIERT. For example, Cornelissen et al60 were able to generate DNA double strand breaks (dsb) in tumour cells via EBRT, which were subsequently targeted by RIT via an antibody construct with a nuclear targeting sequence, which was directed against the dsb-binding histone γH2AX. In fact, they could find a massive benefit on tumour growth delay of this CIERT strategy in comparison with single treatments. Subsequently, the construct was further extended by EGFR-targeting properties to avoid uptake in normal cells situated in the radiation field.61 However, this strategy may be unfavourable if metastatic disease is a target, because only cells in the radiation field increase γH2AX after irradiation. Nevertheless, the elegant format of the construct shows how intracellular antigens can be targeted. Furthermore, a high specificity was mediated by the combined targeting of a surface receptor and an induced intracellular antigen.

TRANSLATION OF COMBINATION OF INTERNAL AND EXTERNAL RADIOTHERAPY INTO CLINICAL STUDIES

CIERT is composed of EBRT as a local therapy and RIT as a systemic treatment. This approach strictly requires interdisciplinary teamwork between nuclear medicine and radio-oncology.

External radiotherapy is much better understood and therefore widely used in the majority of oncological treatment options, whereas nuclear medicine treatments, except radioiodine treatment of thyroid cancer, are relatively rare and their mechanisms of action are much less known. But beyond radioiodine, these treatments further include RIT of lymphoma, PRRT (e.g. with somatostatin receptor analogues) or systemic treatment of bone metastases with bone-seeking radiotracers [e.g. 223Ra–Cl2 or 153Sm-ethylenediamine tetra (methylene phosphonic acid) (EDTMP)]. Nevertheless, mAb and their fragments are the most popular experimental carriers for radiolabelling. Therefore, first approaches of diagnostic and/or therapeutic applications have been reported since their first synthesis in the early 1980s.77 Consequently, the base of successful in vivo tumour targeting after intravenous application were antigen–mAb binding (e.g. antiferritin mAb),77 the use of the avidin–biotin binding78 and mAb binding to receptors, mostly EGFR.79 Not surprising, the radioisotopes, e.g. 90Y, 131I and 125I, 111In (for dosimetry), applied for the systemic part of CIERT are still in use today.77,80,81 With regard to the literature, reports about integrated CIERT of solid tumours in males are rare, but several clinical trials addressing different tumour-targeting mechanisms have been conducted for three decades.80 The most treated entities with predominantly promising reports are (recurrent) malignant glioma, meningioma, primary and secondary tumours of the liver, brain metastases (especially from NSCLC), osteoplastic bone metastases from prostate cancer, primary breast cancer, malignant pheochromocytoma and paraganglioma. Table 3 gives an overview on early clinical trials. In agreement with pre-clinical results, Order et al77 observed an increasing radiotracer uptake after percutaneous radiotherapy followed by intravenous applied systemic radiotherapy. This schedule led to higher energy doses in pre-radiated tumour regions driven by the inflammatory process initiated by the percutaneous radiotherapy. They achieved imposing remissions but only in rare cases. However, only few studies conducted a real combination of EBRT and RIT; in most cases, a sequential application of the treatment modalities was performed. Furthermore, by the nature of first-in-men trials, the majority of these studies were performed in patients suffering from advanced tumours with a very bad prognosis.81 The vast majority were carried out as Phase I/II trials to demonstrate safety, feasibility and efficacy. Only four studies were designed as prospective randomized controlled trials. First of all, Order et al91 conducted a Radiation Therapy Oncology Group study comparing full-dose chemotherapy to cyclic, systemic application of 131I-antiferritin after induction therapy consisting of EBRT integrated with doxorubicin and 5-fluorouracil in patients with unresectable hepatocellular carcinoma. Baczyk et al89 compared the combination of intravenously applied 153Sm-EDTMP and percutaneous radiotherapy with 153Sm-EDTMP alone in bone metastases. Furthermore, Porter et al82 and Nilsson et al84 conducted similarly designed studies using the radionuclides 89Sr and 223Ra; noteworthy, the latter one is actually a FDA- and EMA-approved α-emitter. However, treatments for bone metastases always do have a palliative intention,89 thus allowing no conclusion on any improvement of tumour curability. Nevertheless, all four authors could conclude the superiority of CIERT to the monotherapy.

Table 3.

Clinical combination of internal and external radiotherapy trials

Study Patients Entity Radiotracer Intention Mode
Msirikale et al53 105 Hepatoma 131I-antiferritin mAb Phase I/II Sequential
Msirikale et al53 6 Hepatocellular cancer 90Y-antiferritin mAb Phase I Sequential
Msirikale et al53 48 Hepatocellular cancer 131I-antiferritin mAb Phase III comparison with chemotherapy Sequential
Porter et al82 126 Bone metastases (PCA) 89Sr–Cl2 Phase III, comparison with mono EBRT Combined
Buchegger et al76 6 Liver metastases (colorectal cancer) 131I-mAb F(ab')2 Feasibility Combined
Quang and Brady83 180 Malignant glioma 125I-anti-epidermal growth factor receptor-mAb 425 Phase II Sequential
Nilsson et al84 64 Bone metastases (PCA) 223Ra–Cl2 Phase II, comparison with mono EBRT Combined
Li et al79 192 Malignant glioma 125I-mAb Phase II Sequential
Rades et al56 1 Brain metastases (non-small-cell lung cancer) 125I-Cetuximab Feasibility Sequential
Burdick et al85 11 Follicular lymphoma 90Y-ibritumomab tiuxetan Feasibility Sequential
Hobbs et al86 1 Paraspinal tumour 153Sm-EDTMP Treatment planning Sequential
Ferrari et al78 14 Breast cancer 90Y-biotin Feasibility Sequential
Fishbein et al87 5 Paraganglioma 131I-metaiodobenzylguanidine Feasibility Sequential
Kreissl et al88 10 Meningioma 177Lu-DOTA conjugates Feasibility Combined
Baczyk et al89 88 Bone metastases (PCA) 153Sm-EDTMP Phase III, comparison with monotherapy Combined
Verburg et al90 5 Malignant glioma 131I-phenylalanine Feasibility Sequential

DOTA, 1,4,7,10-tetra-azacyclododecane-1,4,7,10-tetra-acetic acid; EBRT, external beam radiotherapy; EDTMP, ethylenediamine tetra(methylene phosphonic acid); mAb, monoclonal antibodies; PCA, prostate cancer.

Open questions for clinical application of combination of internal and external radiotherapy

Alongside the acquisition of knowledge in basic radiobiological mechanisms, the biggest “clinical” step forward was made in the field of 3D dosimetry. This is reflected by a significant number of outstanding publications over the past decade accomplishing both radio-oncological standards as well as those of nuclear medicine.78,9296 Nevertheless, despite more or less efficient teamwork between those in nuclear medicine and the radio-oncologists, dosimetry of an internal irradiation respecting all the important factors as residence time of mAb or its fragments, individual differences in individual patient's organ functions, radiation quality (β-emitter) and inhomogeneity of dose prescription is still challenging. Is a radio-oncological “gray” really similar to that in nuclear medicine? That is the question here, and confidence, despite good practice and scientific knowledge, is required too. Considerably fewer articles were published addressing temporal relationships and design considerations of CIERT.24,76,97,98 Another important question is the order of the combined treatments. A further, still controversially discussed question is the manner of antibody application. Whereas Quang and Brady83 applied “cold”, unlabelled antibody at first, followed by the “hot”, labelled after few days, Rades et al99 conducted an inverse sequence. It is well known from the FDA- and EMA-approved RIT with ibritumomab tiuxetan (Zevalin) that pre-conditioning with the “cold” antibody can have distinct advantages, but this is depending on the individual mAb, which may be the biggest challenge of CIERT in the near future. Specifically for mAb, the distribution in small animals is different from the one in humans (e.g. EGFR-mAb and CEA-mAb), limiting conclusions especially on normal tissue toxicities. Chimeric or humanized mouse models are used today for pre-clinical drug development. For example, liver-humanized mice show similarities to the human liver in drug metabolism and toxicity, which are not reflected in normal mice owing to differences in metabolic enzymes and transporters.100,101 However, genetically engineered mouse models that express the desired human antigen in the same cell types as humans have only been created for lymphoma models so far.102 Mice with humanized-receptor expression in normal tissues would have the potential to better reflect the distribution and toxicity of mAb variants and could also be used to study pharmacodynamics of radiolabelled constructs. However, dosimetric images do not allow discrimination between unspecific uptake of mAb in the human liver from specific binding on the receptor addressed.

Taken together, several issues of CIERT are solved, for instance mAb labelling and imaging, or—at least partly—3D dosimetry of intravenously applied radiotracers. The main effort for future research into this treatment option will be the acquisition of knowledge about specific distributions of mAb-based radiotracer in humans, and, much more important, to evaluate possibilities to influence and to steer it.

CONCLUSION

The advantages of a combination of EBRT and RIT are prominent. The possibility to use theragnostic radionuclide pairs facilitates individualization of this treatment strategy. Although, the view to a 30-year-long history of CIERT determines the question: why were the improvements of CIERT in three decades so few? The reasons are sophisticated. One reason may be the very high legal and administrative hurdles, at least in some countries, where such developments could be translated to the clinics. The principal scientific reason, despite more or less efficient teamwork between those in nuclear medicine and the radio-oncologists, is the complexity of a dosimetry of the internal irradiation respecting all the important factors as residence time of carrier molecules, individual differences in patient's organ functions, radiation quality and inhomogeneity of dose prescription. However, knowledge gained by research on the use of (radiolabelled) mAb can be directly transferred to CIERT. Plenty of different strategies to bypass barriers and to enhance tumour uptake are under investigation23 and can potentially be adapted to combined approaches. From in vivo experiments, there is great evidence that the usage of smaller antibody fragments and alternative carrier molecules (aptamers, peptides) may overcome the problem of poor tumour uptake. In vitro and animal experiments are inevitable to study principal efficacy of CIERT approaches and to evaluate radiobiological processes. However, the prediction for patient treatment is limited owing to the human(ized) origin of mAb, resulting in a different biodistribution in the animal. Overall, it is an obligate pre-condition for CIERT that classical radiobiology needs to be adapted to the use of open radionuclides for a real comparison with external radiotherapy, an approach that needs to be driven by radio-oncologists together with researchers in nuclear medicine.

Acknowledgments

ACKNOWLEDGMENTS

The authors wish to thank Dr Ralf Bergmann for provision of the mouse PET-image and Dr Claere von Neubeck for fruitful discussion.

Contributor Information

A Dietrich, Email: antje.dietrich@uniklinikum-dresden.de.

L Koi, Email: Lydia.Koi@uniklinikum-dresden.de.

K Zöphel, Email: Klaus.Zoephel@uniklinikum-dresden.de.

W Sihver, Email: w.sihver@hzdr.de.

J Kotzerke, Email: joerg.kotzerke@tu-dresden.de.

M Baumann, Email: Michael.Baumann@uniklinikum-dresden.de.

M Krause, Email: Mechthild.Krause@uniklinikum-dresden.de.

FUNDING

LK was supported by DFG (Ba 1433/5).

REFERENCES

  • 1.Baumann M, Krause M, Hill R. Exploring the role of cancer stem cells in radioresistance. Nat Rev Cancer 2008; 8: 545–54. doi: 10.1038/nrc2419 [DOI] [PubMed] [Google Scholar]
  • 2.Begg AC, Stewart FA, Vens C. Strategies to improve radiotherapy with targeted drugs. Nat Rev Cancer 2011; 11: 239–53. doi: 10.1038/nrc3007 [DOI] [PubMed] [Google Scholar]
  • 3.Pouget JP, Navarro-Teulon I, Bardiès M, Chouin N, Cartron G, Pèlegrin A, et al. Clinical radioimmunotherapy—the role of radiobiology. Nat Rev Clin Oncol 2011; 8: 720–34. doi: 10.1038/nrclinonc.2011.160 [DOI] [PubMed] [Google Scholar]
  • 4.Aslan O, Biber Muftuler FZ, Yurt Kilcar A, Ichedef C, Unak P. In vivo biological evaluation of 131I radiolabeled-paclitaxel glucuronide (131I-PAC-G). Radiochimica Acta 2012; 100: 339–45. [Google Scholar]
  • 5.Sihver W, Pietzsch J, Krause M, Baumann M, Steinbach J, Pietzsch HJ. Radiolabeled cetuximab conjugates for EGFR targeted cancer diagnostics and therapy. Pharmaceuticals (Basel) 2014; 7: 311–38. doi: 10.3390/ph7030311 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Chauhan VP, Stylianopoulos T, Boucher Y, Jain RK. Delivery of molecular and nanoscale medicine to tumors: transport barriers and strategies. Annu Rev Chem Biomol Eng 2011; 2: 281–98. doi: 10.1146/annurev-chembioeng-061010-114300 [DOI] [PubMed] [Google Scholar]
  • 7.Jain RK. Delivery of molecular and cellular medicine to solid tumors. Adv Drug Deliv Rev 2001; 46: 149–68. [DOI] [PubMed] [Google Scholar]
  • 8.Ding H, Carlton MM, Povoski SP, Milum K, Kumar K, Kothandaraman S, et al. Site specific discrete PEGylation of (124)I-labeled mCC49 Fab' fragments improves tumor MicroPET/CT imaging in mice. Bioconjug Chem 2013; 24: 1945–54. doi: 10.1021/bc400375f [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Wong KJ, Baidoo KE, Nayak TK, Garmestani K, Brechbiel MW, Milenic DE. In vitro and in vivo pre-clinical analysis of a F(ab')(2) fragment of panitumumab for molecular imaging and therapy of HER1 positive cancers. EJNMMI Res 2011; 1. doi: 10.1186/2191-219X-1-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.van Dijk LK, Hoeben BA, Kaanders JH, Franssen GM, Boerman OC, Bussink J. Imaging of epidermal growth factor receptor expression in head and neck cancer with SPECT/CT and 111In-labeled cetuximab-F(ab')2. J Nucl Med 2013; 54: 2118–24. doi: 10.2967/jnumed.113.123612 [DOI] [PubMed] [Google Scholar]
  • 11.Xavier C, Vaneycken I, D'huyvetter M, Heemskerk J, Keyaerts M, Vincke C, et al. Synthesis, preclinical validation, dosimetry, and toxicity of 68Ga-NOTA-anti-HER2 Nanobodies for iPET imaging of HER2 receptor expression in cancer. J Nucl Med 2013; 54: 776–84. doi: 10.2967/jnumed.112.111021 [DOI] [PubMed] [Google Scholar]
  • 12.Evazalipour M, D'Huyvetter M, Tehrani BS, Abolhassani M, Omidfar K, Abdoli S, et al. Generation and characterization of nanobodies targeting PSMA for molecular imaging of prostate cancer. Contrast Media Mol Imaging 2014; 9: 211–20. doi: 10.1002/cmmi.1558 [DOI] [PubMed] [Google Scholar]
  • 13.Wållberg H, Orlova A, Altai M, Hosseinimehr SJ, Widström C, Malmberg J, et al. Molecular design and optimization of 99mTc-labeled recombinant affibody molecules improves their biodistribution and imaging properties. J Nucl Med 2011; 52: 461–9. doi: 10.2967/jnumed.110.083592 [DOI] [PubMed] [Google Scholar]
  • 14.Su X, Cheng K, Jeon J, Shen B, Venturin GT, Hu X, et al. Comparison of two site-specifically (18)F-labeled affibodies for PET imaging of EGFR positive tumors. Mol Pharm 2014; 11: 3947–56. doi: 10.1021/mp5003043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Schneider DW, Heitner T, Alicke B, Light DR, McLean K, Satozawa N, et al. In vivo biodistribution, PET imaging, and tumor accumulation of 86Y- and 111In-antimindin/RG-1, engineered antibody fragments in LNCaP tumor-bearing nude mice. J Nucl Med 2009; 50: 435–43. doi: 10.2967/jnumed.108.055608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fan Q, Cai H, Yang H, Li L, Yuan C, Lu X, et al. Biological evaluation of 131I- and CF750-labeled Dmab(scFv)-Fc antibodies for xenograft imaging of CD25-positive tumors. Biomed Res Int 2014; 2014: 459676. doi: 10.1155/2014/459676 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gomes Sda R, Azéma L, Allard M, Toulmé JJ. Aptamers as imaging agents. Expert Opin Med Diagn 2010; 4: 511–18. doi: 10.1517/17530059.2010.516248 [DOI] [PubMed] [Google Scholar]
  • 18.Rockey WM, Huang L, Kloepping KC, Baumhover NJ, Giangrande PH, Schultz MK. Synthesis and radiolabeling of chelator-RNA aptamer bioconjugates with copper-64 for targeted molecular imaging. Bioorg Med Chem 2011; 19: 4080–90. doi: 10.1016/j.bmc.2011.05.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Varmira K, Hosseinimehr SJ, Noaparast Z, Abedi SM. An improved radiolabelled RNA aptamer molecule for HER2 imaging in cancers. J Drug Target 2014; 22: 116–22. doi: 10.3109/1061186X.2013.839688 [DOI] [PubMed] [Google Scholar]
  • 20.Nock B, Maina T. Tetraamine-coupled peptides and resulting (99m)Tc-radioligands: an effective route for receptor-targeted diagnostic imaging of human tumors. Curr Top Med Chem 2012; 12: 2655–67. [DOI] [PubMed] [Google Scholar]
  • 21.Morgat C, Mishra AK, Varshney R, Allard M, Fernandez P, Hindié E. Targeting neuropeptide receptors for cancer imaging and therapy: perspectives with bombesin, neurotensin, and neuropeptide-Y receptors. J Nucl Med 2014; 55: 1650–7. doi: 10.2967/jnumed.114.142000 [DOI] [PubMed] [Google Scholar]
  • 22.Börjesson PK, Postema EJ, de Bree R, Roos JC, Leemans CR, Kairemo KJ, et al. Radioimmunodetection and radioimmunotherapy of head and neck cancer. Oral Oncol 2004; 40: 761–72. [DOI] [PubMed] [Google Scholar]
  • 23.Navarro-Teulon I, Lozza C, Pèlegrin A, Vivès E, Pouget JP. General overview of radioimmunotherapy of solid tumors. Immunotherapy 2013; 5: 467–87. doi: 10.2217/imt.13.34 [DOI] [PubMed] [Google Scholar]
  • 24.DeNardo GL, Schlom J, Buchsbaum DJ, Meredith RF, O'Donoghue JA, Sgouros G, et al. Rationales, evidence, and design considerations for fractionated radioimmunotherapy. Cancer 2002; 94(4 Suppl.): 1332–48. [DOI] [PubMed] [Google Scholar]
  • 25.Baumann M, Appold S, Petersen C, Zips D, Herrmann T. Dose and fractionation concepts in the primary radiotherapy of non-small cell lung cancer. Lung Cancer 2001; 33(Suppl. 1): S35–45. [DOI] [PubMed] [Google Scholar]
  • 26.Intensity Modulated Radiation Therapy Collaborative Working Group. Intensity-modulated radiotherapy: current status and issues of interest. Int J Radiat Oncol Biol Phys 2001; 51: 880–914. [DOI] [PubMed] [Google Scholar]
  • 27.Purdy JA. Advances in the planning and delivery of radiotherapy: new expectations, new standards of care. Front Radiat Ther Oncol 2011; 43: 1–28. doi: 10.1159/000322396 [DOI] [PubMed] [Google Scholar]
  • 28.Li G, Citrin D, Camphausen K, Mueller B, Burman C, Mychalczak B, et al. Advances in 4D medical imaging and 4D radiation therapy. Technol Cancer Res Treat 2008; 7: 67–81. [DOI] [PubMed] [Google Scholar]
  • 29.van de Watering FC, Rijpkema M, Robillard M, Oyen WJ, Boerman OC. Pretargeted imaging and radioimmunotherapy of cancer using antibodies and bioorthogonal chemistry. Front Med (Lausanne) 2014; 1: 44. doi: 10.3389/fmed.2014.00044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Milenic DE, Brady ED, Brechbiel MW. Antibody-targeted radiation cancer therapy. Nat Rev Drug Discov 2004; 3: 488–99. [DOI] [PubMed] [Google Scholar]
  • 31.Srivastava S, Dadachova E. Recent advances in radionuclide therapy. Semin Nucl Med 2001; 31: 330–41. [DOI] [PubMed] [Google Scholar]
  • 32.Pickhard A, Piontek G, Seidl C, Kopping S, Blechert B, Mißlbeck M, et al. 213Bi-anti-EGFR radioimmunoconjugates and X-ray irradiation trigger different cell death pathways in squamous cell carcinoma cells. Nucl Med Biol 2014; 41: 68–76. doi: 10.1016/j.nucmedbio.2013.09.010 [DOI] [PubMed] [Google Scholar]
  • 33.Förster GJ, Engelbach MJ, Brockmann JJ, Reber HJ, Buchholz HG, Mäcke HR, et al. Preliminary data on biodistribution and dosimetry for therapy planning of somatostatin receptor positive tumours: comparison of (86)Y-DOTATOC and (111)In-DTPA-octreotide. Eur J Nucl Med 2001; 28: 1743–50. [DOI] [PubMed] [Google Scholar]
  • 34.Anderson CJ, Ferdani R. Copper-64 radiopharmaceuticals for PET imaging of cancer: advances in preclinical and clinical research. Cancer Biother Radiopharm 2009; 24: 379–93. doi: 10.1089/cbr.2009.0674 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Novak-Hofer I, Schubiger PA. Copper-67 as a therapeutic nuclide for radioimmunotherapy. Eur J Nucl Med Mol Imaging 2002; 29: 821–30. [DOI] [PubMed] [Google Scholar]
  • 36.Lopci E, Chiti A, Castellani MR, Pepe G, Antunovic L, Fanti S, et al. Matched pairs dosimetry: 124I/131I metaiodobenzylguanidine and 124I/131I and 86Y/90Y antibodies. Eur J Nucl Med Mol Imaging 2011; 38(Suppl. 1): S28–40. doi: 10.1007/s00259-011-1772-6 [DOI] [PubMed] [Google Scholar]
  • 37.McKinney MS, Beaven AW. Yttrium-90 ibritumomab tiuxetan in the treatment of non-Hodgkin lymphoma. Blood Lymphatic Cancer Targets Ther 2014; 4: 45–59. [Google Scholar]
  • 38.Osborne JR, Green DA, Spratt DE, Lyashchenko S, Fareedy SB, Robinson BD, et al. A prospective pilot study of (89)Zr-J591/prostate specific membrane antigen positron emission tomography in men with localized prostate cancer undergoing radical prostatectomy. J Urol 2014; 191: 1439–45. doi: 10.1016/j.juro.2013.10.041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Perk LR, Visser GW, Vosjan MJ, Stigter-van Walsum M, Tijink BM, Leemans CR, et al. (89)Zr as a PET surrogate radioisotope for scouting biodistribution of the therapeutic radiometals (90)Y and (177)Lu in tumor-bearing nude mice after coupling to the internalizing antibody cetuximab. J Nucl Med 2005; 46: 1898–906. [PubMed] [Google Scholar]
  • 40.Liu Z, Ma T, Liu H, Jin Z, Sun X, Zhao H, et al. 177Lu-labeled antibodies for EGFR-targeted SPECT/CT imaging and radioimmunotherapy in a preclinical head and neck carcinoma model. Mol Pharm 2014; 11: 800–7. doi: 10.1021/mp4005047 [DOI] [PubMed] [Google Scholar]
  • 41.Nagar S, Walker DD, Embia O, Kaplan EL, Grogan RH, Angelos P. A novel technique to improve the diagnostic yield of negative sestamibi scans. Surgery 2014; 156: 584–90. doi: 10.1016/j.surg.2014.05.020 [DOI] [PubMed] [Google Scholar]
  • 42.Müller C, Schubiger PA, Schibli R. Isostructural folate conjugates radiolabeled with the matched pair 99mTc/188Re: a potential strategy for diagnosis and therapy of folate receptor-positive tumors. Nucl Med Biol 2007; 34: 595–601. [DOI] [PubMed] [Google Scholar]
  • 43.O'Donnell RT, DeNardo SJ, Yuan A, Shen S, Richman CM, Lara PN, et al. Radioimmunotherapy with (111)In/(90)Y-2IT-BAD-m170 for metastatic prostate cancer. Clin Cancer Res 2001; 7: 1561–8. [PubMed] [Google Scholar]
  • 44.Bravo PE, Goudarzi B, Rana U, Filho PT, Castillo R, Rababy C, et al. Clinical significance of discordant findings between pre-therapy (123)I and post-therapy (131)I whole body scan in patients with thyroid cancer. Int J Clin Exp Med 2013; 6: 320–33. [PMC free article] [PubMed] [Google Scholar]
  • 45.Van Nostrand D, Khorjekar GR, O'Neil J, Moreau S, Atkins FB, Kharazi P, et al. Recombinant human thyroid-stimulating hormone versus thyroid hormone withdrawal in the identification of metastasis in differentiated thyroid cancer with 131I planar whole-body imaging and 124I PET. J Nucl Med 2012; 53: 359–62. doi: 10.2967/jnumed.111.096016 [DOI] [PubMed] [Google Scholar]
  • 46.Verel I, Visser GW, Vosjan MJ, Finn R, Boellaard R, van Dongen GA. High-quality 124I-labelled monoclonal antibodies for use as PET scouting agents prior to 131I-radioimmunotherapy. Eur J Nucl Med Mol Imaging 2004; 31: 1645–52. [DOI] [PubMed] [Google Scholar]
  • 47.Torres LA, Coca MA, Batista JF, Casaco A, Lopez G, García I, et al. Biodistribution and internal dosimetry of the 188Re-labelled humanized monoclonal antibody anti-epidemal growth factor receptor, nimotuzumab, in the locoregional treatment of malignant gliomas. Nucl Med Commun 2008; 29: 66–75. [DOI] [PubMed] [Google Scholar]
  • 48.Nucleide.org. Arinc A, Bé M-M, Browne E, Castro RM, Chechev VP, Chisté V, et al. Le Laboratoire National Henri Becquerel; [updated 13 February, 2015; cited March 2015]. Available from: http://www.nucleide.org/DDEP_WG/DDEPdata.htm [Google Scholar]
  • 49.Periodictable.com. Gray T, Whitby M, Mann N. Element Collection, Inc.; [updated 8 December 2007; cited March 2015]. Available from: http://periodictable.com/Isotopes/029.67/index3.p.full.dm.prod.html [Google Scholar]
  • 50.Lubberink M, Herzog H. Quantitative imaging of 124I and 86Y with PET. Eur J Nucl Med Mol Imaging 2011; 38(Suppl. 1): S10–18. doi: 10.1007/s00259-011-1768-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Engelhardt EL, Schneider RF, Seeholzer SH, Stobbe CC, Chapman JD. The synthesis and radiolabeling of 2-nitroimidazole derivatives of cyclam and their preclinical evaluation as positive markers of tumor hypoxia. J Nucl Med 2002; 43: 837–50. [PubMed] [Google Scholar]
  • 52.Szymański P, Frączek T, Markowicz M, Mikiciuk-Olasik E. Development of copper based drugs, radiopharmaceuticals and medical materials. Biometals 2012; 25: 1089–112. doi: 10.1007/s10534-012-9578-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Msirikale JS, Klein JL, Schroeder J, Order SE. Radiation enhancement of radiolabelled antibody deposition in tumors. Int J Radiat Oncol Biol Phys 1987; 13: 1839–44. [DOI] [PubMed] [Google Scholar]
  • 54.Wong JY, Williams LE, Hill LR, Paxton RJ, Beatty BG, Shively JE, et al. The effects of tumor mass, tumor age, and external beam radiation on tumor-specific antibody uptake. Int J Radiat Oncol Biol Phys 1989; 16: 715–20. [DOI] [PubMed] [Google Scholar]
  • 55.Bender H, Emrich JG, Eshelman J, Chu MA, Steplewski Z, Biersack HJ, et al. Enhancement of monoclonal antibody efficacy: the effect of external beam radiation. Hybridoma 1995; 14: 129–34. [DOI] [PubMed] [Google Scholar]
  • 56.Rades D, Wolff C, Nadrowitz R, Breunig C, Schild SE, Baehre M, et al. Radioactive EGFR antibody cetuximab in multimodal cancer treatment: stability and synergistic effects with radiotherapy. Int J Radiat Oncol Biol Phys 2009; 75: 1226–31. doi: 10.1016/j.ijrobp.2008.12.029 [DOI] [PubMed] [Google Scholar]
  • 57.Saki M, Toulany M, Sihver W, Zenker M, Heldt JM, Mosch B, et al. Cellular and molecular properties of (90)Y-labeled cetuximab in combination with radiotherapy on human tumor cells in vitro. Strahlenther Onkol 2012; 188: 823–32. [DOI] [PubMed] [Google Scholar]
  • 58.Eke I, Ingargiola M, Förster C, Kunz-Schughart LA, Baumann M, Runge R, et al. Cytotoxic properties of radionuclide-conjugated Cetuximab without and in combination with external irradiation in head and neck cancer cells in vitro. Int J Radiat Biol 2014; 90: 678–86. doi: 10.3109/09553002.2014.899446 [DOI] [PubMed] [Google Scholar]
  • 59.Ingargiola M, Runge R, Heldt JM, Freudenberg R, Steinbach J, Cordes N, et al. Potential of a Cetuximab-based radioimmunotherapy combined with external irradiation manifests in a 3-D cell assay. Int J Cancer 2014; 135: 968–80. doi: 10.1002/ijc.28735 [DOI] [PubMed] [Google Scholar]
  • 60.Cornelissen B, Darbar S, Kersemans V, Allen D, Falzone N, Barbeau J, et al. Amplification of DNA damage by a γH2AX-targeted radiopharmaceutical. Nucl Med Biol 2012; 39: 1142–51. doi: 10.1016/j.nucmedbio.2012.06.001 [DOI] [PubMed] [Google Scholar]
  • 61.Cornelissen B, Waller A, Able S, Vallis KA. Molecular radiotherapy using cleavable radioimmunoconjugates that target EGFR and γH2AX. Mol Cancer Ther 2013; 12: 2472–82. doi: 10.1158/1535-7163.MCT-13-0369 [DOI] [PubMed] [Google Scholar]
  • 62.Buchegger F, Rojas A, Delaloye AB, Vogel CA, Mirimanoff RO, Coucke P, et al. Combined radioimmunotherapy and radiotherapy of human colon carcinoma grafted in nude mice. Cancer Res 1995; 55: 83–9. [PubMed] [Google Scholar]
  • 63.Sun LQ, Vogel CA, Mirimanoff RO, Coucke P, Slosman DO, Mach JP, et al. Timing effects of combined radioimmunotherapy and radiotherapy on a human solid tumor in nude mice. Cancer Res 1997; 57: 1312–19. [PubMed] [Google Scholar]
  • 64.Eriksson D, Joniani HM, Sheikholvaezin A, Löfroth PO, Johansson L, Riklund Ahlström K, et al. Combined low dose radio- and radioimmunotherapy of experimental HeLa Hep 2 tumours. Eur J Nucl Med Mol Imaging 2003; 30: 895–906. [DOI] [PubMed] [Google Scholar]
  • 65.Koi L, Bergmann R, Brüchner K, Pietzsch J, Pietzsch HJ, Krause M, et al. Radiolabeled anti-EGFR-antibody improves local tumor control after external beam radiotherapy and offers theragnostic potential. Radiother Oncol 2014; 110: 362–9. doi: 10.1016/j.radonc.2013.12.001 [DOI] [PubMed] [Google Scholar]
  • 66.Vogel CA, Galmiche MC, Buchegger F. Radioimmunotherapy and fractionated radiotherapy of human colon cancer liver metastases in nude mice. Cancer Res 1997; 57: 447–53. [PubMed] [Google Scholar]
  • 67.Gurtner K, Deuse Y, Bütof R, Schaal K, Eicheler W, Oertel R, et al. Diverse effects of combined radiotherapy and EGFR inhibition with antibodies or TK inhibitors on local tumour control and correlation with EGFR gene expression. Radiother Oncol 2011; 99: 323–30. doi: 10.1016/j.radonc.2011.05.035 [DOI] [PubMed] [Google Scholar]
  • 68.Yaromina A, Kroeber T, Meinzer A, Boeke S, Thames H, Baumann M, et al. Exploratory study of the prognostic value of microenvironmental parameters during fractionated irradiation in human squamous cell carcinoma xenografts. Int J Radiat Oncol Biol Phys 2011; 80: 1205–13. doi: 10.1016/j.ijrobp.2011.02.015 [DOI] [PubMed] [Google Scholar]
  • 69.Krause M, Schütze C, Petersen C, Pimentel N, Hessel F, Harstrick A, et al. Different classes of EGFR inhibitors may have different potential to improve local tumour control after fractionated irradiation: a study on C225 in FaDu hSCC. Radiother Oncol 2005; 74: 109–15. [DOI] [PubMed] [Google Scholar]
  • 70.Storch K, Eke I, Borgmann K, Krause M, Richter C, Becker K, et al. Three-dimensional cell growth confers radioresistance by chromatin density modification. Cancer Res 2010; 70: 3925–34. doi: 10.1158/0008-5472.CAN-09-3848 [DOI] [PubMed] [Google Scholar]
  • 71.Hirschhaeuser F, Menne H, Dittfeld C, West J, Mueller-Klieser W, Kunz-Schughart LA. Multicellular tumor spheroids: an underestimated tool is catching up again. J Biotechnol 2010; 148: 3–15. doi: 10.1016/j.jbiotec.2010.01.012 [DOI] [PubMed] [Google Scholar]
  • 72.Krause M, Zips D, Thames HD, Kummermehr J, Baumann M. Preclinical evaluation of molecular-targeted anticancer agents for radiotherapy. Radiother Oncol 2006; 80: 112–22. [DOI] [PubMed] [Google Scholar]
  • 73.Kalofonos H, Rowlinson G, Epenetos AA. Enhancement of monoclonal antibody uptake in human colon tumor xenografts following irradiation. Cancer Res 1990; 50: 159–63. [PubMed] [Google Scholar]
  • 74.Gridley DS, Mackensen DG, Slater JB, Moyers MF, Slater JM. Effects of proton irradiation on radiolabeled monoclonal antibody uptake in human colon tumor xenografts. J Immunother Emphasis Tumor Immunol 1995; 17: 229–37. [DOI] [PubMed] [Google Scholar]
  • 75.Ruan S, O'Donoghue JA, Larson SM, Finn RD, Jungbluth A, Welt S, et al. Optimizing the sequence of combination therapy with radiolabeled antibodies and fractionated external beam. J Nucl Med 2000; 41: 1905–12. [PubMed] [Google Scholar]
  • 76.Buchegger F, Roth A, Allal A, Dupertuis YM, Slosman DO, Delaloye AB, et al. Radioimmunotherapy of colorectal cancer liver metastases: combination with radiotherapy. Ann N Y Acad Sci 2000; 910: 263–9, discussion 269–70. [DOI] [PubMed] [Google Scholar]
  • 77.Order SE, Klein JL, Leichner PK, Frincke J, Lollo C, Carlo DJ. 90Yttrium antiferritin—a new therapeutic radiolabeled antibody. Int J Radiat Oncol Biol Phys 1986; 12: 277–81. [DOI] [PubMed] [Google Scholar]
  • 78.Ferrari ME, Cremonesi M, Di Dia A, Botta F, De Cicco C, Sarnelli A, et al. 3D dosimetry in patients with early breast cancer undergoing intraoperative avidination for radionuclide therapy (IART) combined with external beam radiation therapy. Eur J Nucl Med Mol Imaging 2012; 39: 1702–11. doi: 10.1007/s00259-012-2197-6 [DOI] [PubMed] [Google Scholar]
  • 79.Li L, Quang TS, Gracely EJ, Kim JH, Emrich JG, Yaeger TE, et al. A phase II study of anti-epidermal growth factor receptor radioimmunotherapy in the treatment of glioblastoma multiforme. J Neurosurg 2010; 113: 192–8. doi: 10.3171/2010.2.JNS091211 [DOI] [PubMed] [Google Scholar]
  • 80.Order SE, Stillwagon GB, Klein JL, Leichner PK, Siegelman SS, Fishman EK, et al. Iodine 131 antiferritin, a new treatment modality in hepatoma: a Radiation Therapy Oncology Group study. J Clin Oncol 1985; 3: 1573–82. [DOI] [PubMed] [Google Scholar]
  • 81.Riva P, Franceschi G, Riva N, Casi M, Santimaria M, Adamo M. Role of nuclear medicine in the treatment of malignant gliomas: the locoregional radioimmunotherapy approach. Eur J Nucl Med 2000; 27: 601–9. [DOI] [PubMed] [Google Scholar]
  • 82.Porter AT, McEwan AJ, Powe JE, Reid R, McGowan DG, Lukka H, et al. Results of a randomized phase-III trial to evaluate the efficacy of strontium-89 adjuvant to local field external beam irradiation in the management of endocrine resistant metastatic prostate cancer. Int J Radiat Oncol Biol Phys 1993; 25: 805–13. [DOI] [PubMed] [Google Scholar]
  • 83.Quang TS, Brady LW. Radioimmunotherapy as a novel treatment regimen: 125I-labeled monoclonal antibody 425 in the treatment of high-grade brain gliomas. Int J Radiat Oncol Biol Phys 2004; 58: 972–5. [DOI] [PubMed] [Google Scholar]
  • 84.Nilsson S, Franzén L, Parker C, Tyrrell C, Blom R, Tennvall J, et al. Bone-targeted radium-223 in symptomatic, hormone-refractory prostate cancer: a randomised, multicentre, placebo-controlled phase II study. Lancet Oncol 2007; 8: 587–94. [DOI] [PubMed] [Google Scholar]
  • 85.Burdick MJ, Neumann D, Pohlman B, Reddy CA, Tendulkar RD, Macklis R. External beam radiotherapy followed by 90Y ibritumomab tiuxetan in relapsed or refractory bulky follicular lymphoma. Int J Radiat Oncol Biol Phys 2011; 79: 1124–30. doi: 10.1016/j.ijrobp.2009.12.030 [DOI] [PubMed] [Google Scholar]
  • 86.Hobbs SK, Monsky WL, Yuan F, Roberts WG, Griffith L, Torchilin VP, et al. Regulation of transport pathways in tumor vessels: role of tumor type and microenvironment. Proc Natl Acad Sci U S A 1998; 95: 4607–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Fishbein L, Bonner L, Torigian DA, Nathanson KL, Cohen DL, Pryma D, et al. External beam radiation therapy (EBRT) for patients with malignant pheochromocytoma and non-head and -neck paraganglioma: combination with 131I-MIBG. Horm Metab Res 2012; 44: 405–10. doi: 10.1055/s-0032-1308992 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kreissl MC, Hanscheid H, Löhr M, Verburg FA, Schiller M, Lassmann M, et al. Combination of peptide receptor radionuclide therapy with fractionated external beam radiotherapy for treatment of advanced symptomatic meningioma. Radiat Oncol 2012; 7: 99. doi: 10.1186/1748-717X-7-99 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Baczyk M, Milecki P, Pisarek M, Gut P, Antczak A, Hrab M. A prospective randomized trial: a comparison of the analgesic effect and toxicity of 153Sm radioisotope treatment in monotherapy and combined therapy including local external beam radiotherapy (EBRT) among metastatic castrate resistance prostate cancer (mCRPC) patients with painful bone metastases. Neoplasma 2013; 60: 328–33. doi: 10.4149/neo_2013_044 [DOI] [PubMed] [Google Scholar]
  • 90.Verburg FA, Sweeney R, Hänscheid H, Dießl S, Israel I, Löhr M, et al. Patients with recurrent glioblastoma multiforme. Initial experience with p-[(131)I]iodo-L-phenylalanine and external beam radiation therapy. Nuklearmedizin 2013; 52: 36–42. doi: 10.3413/Nukmed-0510-12-06 [DOI] [PubMed] [Google Scholar]
  • 91.Order S, Pajak T, Leibel S, Asbell S, Leichner P, Ettinger D, et al. A randomized prospective trial comparing full dose chemotherapy to 131I antiferritin: an RTOG study. Int J Radiat Oncol Biol Phys 1991; 20: 953–63. [DOI] [PubMed] [Google Scholar]
  • 92.Bodey RK, Flux GD, Evans PM. Combining dosimetry for targeted radionuclide and external beam therapies using the biologically effective dose. Cancer Biother Radiopharm 2003; 18: 89–97. [DOI] [PubMed] [Google Scholar]
  • 93.Cremonesi M, Ferrari M, Botta F, Guerriero F, Garibaldi C, Bodei L, et al. Planning combined treatments of external beam radiation therapy and molecular radiotherapy. Cancer Biother Radiopharm 2014; 29: 227–37. doi: 10.1089/cbr.2014.1607 [DOI] [PubMed] [Google Scholar]
  • 94.Kotzerke J, Oehme L. Biological aspects of radiation in nuclear medicine. [In German.] Nuklearmedizin 2010; 49(Suppl. 1): S5–10. [PubMed] [Google Scholar]
  • 95.Postema EJ, Borjesson PK, Buijs WC, Roos JC, Marres HA, Boerman OC, et al. Dosimetric analysis of radioimmunotherapy with 186Re-labeled bivatuzumab in patients with head and neck cancer. J Nucl Med 2003; 44: 1690–9. [PubMed] [Google Scholar]
  • 96.Prideaux AR, Song H, Hobbs RF, He B, Frey EC, Ladenson PW, et al. Three-dimensional radiobiologic dosimetry: application of radiobiologic modeling to patient-specific 3-dimensional imaging-based internal dosimetry. J Nucl Med 2007; 48: 1008–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.DeNardo SJ, Kroger LA, Lamborn KR, Miers LA, O'Donnell RT, Kukis DL, et al. Importance of temporal relationships in combined modality radioimmunotherapy of breast carcinoma. Cancer 1997; 80(12 Suppl.): 2583–90. [DOI] [PubMed] [Google Scholar]
  • 98.Hobbs RF, McNutt T, Baechler S, He B, Esaias CE, Frey EC, et al. A treatment planning method for sequentially combining radiopharmaceutical therapy and external radiation therapy. Int J Radiat Oncol Biol Phys 2011; 80: 1256–62. doi: 10.1016/j.ijrobp.2010.08.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Rades D, Nadrowitz R, Buchmann I, Hunold P, Noack F, Schild SE, et al. Radiolabeled cetuximab plus whole-brain irradiation (WBI) for the treatment of brain metastases from non-small cell lung cancer (NSCLC). Strahlenther Onkol 2010; 186: 458–62. doi: 10.1007/s00066-010-2153-y [DOI] [PubMed] [Google Scholar]
  • 100.Grompe M, Strom S. Mice with human livers. Gastroenterology 2013; 145: 1209–14. doi: 10.1053/j.gastro.2013.09.009 [DOI] [PubMed] [Google Scholar]
  • 101.Yoshizato K, Tateno C, Utoh R. Mice with liver composed of human hepatocytes as an animal model for drug testing. Curr Drug Discov Technol 2012; 9: 63–76. [DOI] [PubMed] [Google Scholar]
  • 102.Golay J, Introna M. Mechanism of action of therapeutic monoclonal antibodies: promises and pitfalls of in vitro and in vivo assays. Arch Biochem Biophys 2012; 526: 146–53. doi: 10.1016/j.abb.2012.02.011 [DOI] [PubMed] [Google Scholar]

Articles from The British Journal of Radiology are provided here courtesy of Oxford University Press

RESOURCES