Sie sind auf Seite 1von 14

Critical Reviews in Oncology / Hematology 123 (2018) 7–20

Contents lists available at ScienceDirect

Critical Reviews in Oncology / Hematology


journal homepage: www.elsevier.com/locate/critrevonc

Global comparison of targeted alpha vs targeted beta therapy for cancer: In T


vitro, in vivo and clinical trials

Loredana Marcua,b, Eva Bezakb,c, Barry J. Allend,
a
Department of Physics, Faculty of Science, 1 Universitatii street, University of Oradea, 410087, Romania
b
Sansom Institute for Health Research and the School of Health Sciences, University of South Australia, GPO Box 247, Adelaide SA 5001, Australia
c
Department of Physics, University of Adelaide, Adelaide, SA 5005, Australia
d
School of Medicine, University of Western Sydney, Locked Bag 1797, Penrith NSW 2751, Australia

A R T I C L E I N F O A B S T R A C T

Keywords: Targeted therapy for cancer is a rapidly expanding and successful approach to the management of many in-
Systemic cancer tractable cancers. However, many immunotherapies fail in the longer term and there continues to be a need for
Targeted alpha therapy improved targeted cancer cell toxicity, which can be achieved by radiolabelling the targeting vector with a
Targeted beta therapy radioisotope.
Clinical trials
Such constructs are successful in using a gamma ray emitter for imaging. However, traditionally, a beta
emitter is used for therapeutic applications. The new approach is to use the short range and highly cytotoxic
alpha radiation from alpha emitters to achieve improved efficacy and therapeutic gain.
This paper sets out to review all experimental and theoretical comparisons of efficacy and therapeutic gain for
alpha and beta emitters labelling the same targeting vector. The overall conclusion is that targeted alpha therapy
is superior to targeted beta therapy, such that the use of alpha therapy in clinical settings should be expanded.

1. Introduction improved targeted cancer cell toxicity, which can be achieved by


radiolabelling the targeting vector with a radioisotope.
Anti-cancer treatment, such as surgery, radiotherapy and che- Monoclonal antibodies (Mabs) labeled with radionuclides form so-
motherapy are considered standard of care in today’s oncology. While called radio-immunoconjugates (RIC), which deliver high dose radia-
the advances in science and technology have led to developments in all tion directly to the cancer cell (Allen 2011). Traditionally, beta emitters
these therapeutic areas, the global cancer burden is expected to nearly (i.e. electrons) have been used for therapeutic applications. FDA ap-
double by 2030 and will continue to be the leading cause of death in provals have been granted for targeted radio-immunotherapy for non-
both developed and less developed countries (American Cancer Society, Hodgkin’s lymphoma using 131I-tositumomab (Bexxar®) and 90Y-ibri-
2015). tumomab tiuxetan (Zevalin®) to target CD20 (Kang et al., 2013). More
New therapies are needed to increase systemic cancer control and to recently, the short-range beta-emitting conjugate 177Lu-J591 has been
diminish normal tissue side effects. These should be more targeted and used with encouraging results in a phase 2 clinical trial for castrate
patient specific. Targeted therapy for cancer is a rapidly expanding and resistant prostate cancer, where it targets metastases in both the bone
successful approach to the management of many intractable cancers. and soft tissues and survival time has almost doubled (Tagawa et al.,
For example, monoclonal antibodies to target specific antigens or re- 2013). These beta-emitting conjugates do enhance cytotoxicity to tu-
ceptors have been used effectively to control the progression of dif- mours. However, the long range of betas is a double edged sword. On
ferent cancers. Immuno-modulators such as Rituximab (MabThera®) for one hand, the long range electrons can travel through several cancer
lymphoma, Trastuzumab (Herceptin®) for breast cancer, Alemtuzumab cells thus increasing the average dose to a tumour. On the other hand,
(Campath-1H®) for chronic lymphocytic leukemia, Cetuximab the long range of electrons also results in a larger dose to surrounding
(Erbitux®) for colorectal cancer and Bevacizumab (Avastin®) for color- healthy tissue. Additionally, the low ionisation capacity of electrons
ectal and lung cancers have been used clinically with significant suc- (i.e. low linear energy transfer (LET)) means that beta radiation cannot
cess. However, cancer specific antibodies alone can be insufficient to deliver a lethal dose to a targeted single cancer cell (Marcu et al., 2012).
control cancer progression and there continues to be a need for The new approach is to use the short range and highly cytotoxic


Corresponding author.
E-mail address: bja1940b@outlook.com (B.J. Allen).

https://doi.org/10.1016/j.critrevonc.2018.01.001
Received 19 July 2017; Received in revised form 11 November 2017; Accepted 9 January 2018
1040-8428/ © 2018 Elsevier B.V. All rights reserved.
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

alpha radiation from alpha emitters to achieve improved efficacy and Table 1
therapeutic gain. This new field of targeted radio therapy is gaining Physical and radiobiological properties of alpha and beta radiation.
clinical interest, with a number of clinical trials around the world either
Properties Alpha radiation Beta radiation
being completed, underway or under development (Allen et al., 2016).
The important advantage of alpha radiation is the high LET to targeted Tissue range (μm) 20–80 2000–11500
cancer cells over a short range of several cell diameters (Mulford et al., Linear energy transfer (keV/ ∼100 ∼0.3
μm)
2005). Furthermore, the formation of complex DNA lesions by alpha
RBE (external beam) (Section 5 1
emitters can be explained by the energy distribution at the nanoscale 3.1)
level, though it is likely that the cell membrane and mitochondria are RBE (internal) (section 3.2) > 100 1
also sensitive targets of alpha irradiation (Pouget et al., 2015). Pharmacokinetics (half-life) 1 h–10 d 7 h–7 d
As such, alpha radiation is better suited to the treatment of micro- Decay behaviour One step and One-step decay to stable
multiple alpha nuclide
scopic or small-volume disease since their short range and high energies
decays
potentially offer more efficient and specific killing of tumour cells,
while sparing distant normal cells (Allen et al., 2007).
Nevertheless, most clinical trials continue to use beta emitters ra- The high LET and short range means that alphas are preferred to kill
ther than alpha emitters. This paper sets out to review all experimental isolated cells. If the AIC diffuses into the nucleus, the toxicity increases
comparisons of cytotoxicity, efficacy and therapeutic gain for alpha and markedly which is not the case for betas. The exception is Auger elec-
beta emitters labelling, where possible, the same targeting vector with tron-emitting radioisotopes, such as 77Br, 111In, 123I and 125I, which are
the aim to identify the therapeutic benefit of both modalities. potential therapeutic agents, given the fact that when combined with
suitable molecular carriers, these isotopes have proven high LET cell-
2. Biophysical and radiobiological comparison of alpha and beta killing efficiency (Adelstein et al., 2003).
radiations The long-range crossfire effect gives beta radiation some advantage
over other targeted therapy candidates as the consequences of hetero-
The several cell categories that must be considered when employing geneous uptake of the beta-immunoconjugate (BIC) in the tumour are
TAT as a curative therapeutic modality are: (1) isolated or in transit reduced. With alpha-emitters, this problem can be overcome with in-
cancer cells; (2) targeted cancer cell clusters and tumours (3) non-tar- tralesional injections with AICs (Allen et al., 2005) or by tumour anti-
geted contiguous cancer cells and (4) cancer stem cells. While TAT has a vascular alpha therapy (TAVAT) (Allen et al., 2007), the latter property
powerful effect on contiguous non-targeted cancer cells, this property is being inexistent with beta-emitters (Table 2).
inexistent with beta-emitters. The bystander effect could contribute to cell kill in both beta and
alpha radioimmunotherapy. While several aspects regarding the by-
2.1. Monte Carlo calculations stander effect are still under investigation, it is known that cells that
have not been traversed by radiation but are located in the vicinity of
The different properties of radiations from alpha and beta emitting irradiated cells, can present signs of radiation damage, similar to cells
radioisotopes are apparent from the results of Monte Carlo calculations. affected by ionising radiation (such as chromosomal damage, muta-
Random event probabilities are calculated by Monte Carlo to build up tions, apoptosis) or even radio-adaptation (Pouget et al., 2015). These
the specific energy spectrum in the targeted cell nucleus (Huang et al., effects occur at low dose and dose rates and in targeted therapy can be
2012). The mean nuclear specific energy (Gy) for different radio- particularly relevant due to the non-uniform distribution of radio-
isotopes is shown in Fig. 1 for multiple alpha emissions, single alpha activity that influences risk estimation and clinical outcome in general.
emissions and beta emissions from the designated radioisotopes on the Abscopal effects, which are distant bystander effects that inhibit distant
surface of a cell. 223Ra and 225Ac have about 4 times the specific energy tumours after local irradiation, are also potential players in cell kill
of the single alpha emitters 211At and 213Bi. However, the specific en- through distant cellular signaling. Alpha radioimmunotherapy com-
ergies for beta emitters are approximately two orders of magnitude bined with adoptive T cell therapy to stimulate the immune system was
lower than for 213Bi and three orders below 225Ac. shown to be a successful treatment for multiple myeloma in mice
models, potentially mediated by abscopal effects (Ménager et al., 2015).
2.2. Physical and radiobiological properties The antigen expression of cancer stem cells remains uncertain.
Failure to kill these stem cells means that the cancer cannot be con-
These specific energies, coupled with the short range of alphas as trolled. However, if these cells are either isolated or in transit, they can
shown in Table 1, means that alphas exhibit a much greater toxicity to only be eliminated by alpha rays if their antigenic expression is known.
targeted cells than betas. This may be the case for beta emitters such as 131I and 90Y, used in
combination with monoclonal antibodies for the treatment of non-
Hodgkin’s lymphoma, 177Lu for prostate cancer and 153Sm and 89Sr for
the palliation of bone metastases. Other possible candidates for clinical
studies are 166Ho, 67Co, 186Re and 188Re. While efficacious, none of
these radioisotopes have been curative.

Table 2
Comparative effects of alpha and beta radiation (Allen, 1999).

Cancer Type Alpha particles Beta particles

Isolated cells Yes No


Contiguous non-targeted cells Yes No
stem cells Yes No
Fig. 1. The mean nuclear specific energy (Gy) for different alpha and beta emitting Antivascular effect Yes No
DNA localization effect Yes No
radioisotopes located on the cell membrane are shown for the multiple and single alpha
Clusters Yes Yes
emitters, in comparison with that for the beta emitting radioisotopes (courtesy (Huang
Tumor crossfire No Yes
et al., 2012)).

8
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

Fig. 3. Comparative melanoma cell log-survival for alpha (Bi213) and beta (Tb152) la-
beled 9.2.27 mAb (Courtesy (Rizvi et al., 2000)).

was labeled with radioisotopes with > 90% labelling efficiency high
stability (< 20% leaching) of the radio-immunoconjugate. The HL-60
Fig. 2. Log-survival curves, obtained by clonogenic assay, for A549 cells and endothelial cells were incubated over 24 h with WM53 Mab being unlabelled or
cells (EC) exposed to X-rays or alpha particles (courtesy (Riquier et al., 2013)). labeled with 153Sm(β), 152Tb(β+) and 213Bi(α) labels (Rizvi et al.,
2002).
3. In vitro cytotoxicity Cytotoxicity relative to unlabelled antibody was determined for
DNA synthesis using tritium labeled thymidine incorporation and cell
There are substantial differences between the radiobiological survival using the MTS method. A correction has not been made for the
properties of radioisotopes. These relate to the linear energy transfer different half-lives of the radioisotopes for the 24 h incubation (Fig. 4).
(LET) of the emitted particles, radiochemistry and elemental properties. For either method, only the 213Bi −WM53 conjugate was effective in
Barendsen et al. (1960) was the first to study the survival of cells under killing targeted cancer cells. Free 213Bi was as ineffective as the targeted
alpha, beta and gamma external radiation beams. beta emitting WM-53 conjugates (Rizvi et al., 2002).

3.1. External X-ray and alpha particle irradiation on a human cancer and 3.2.3. Prostate cancer
endothelial cells In an experiment reported by Li et al. (2002a), cells originating from
prostate cancer were treated with varying concentrations of AIC in-
Barendson compared the effect of external photon beams and alpha cubated over 24 h and cell survival measured by MTS assay and ex-
radiation (Barendsen et al., 1960) in terms of cell survival after single or pressed as a mean percent ± SD of control plates containing a non-
multiple exposures of X and alpha radiation. The relative biological specific antibody only (Li et al., 2002a). The survival curves for prostate
effect for alphas vs X-rays was estimated to be around 3 for 1–10% cancer cells (LN3) incubated with a specific 213Bi −J591 conjugate and
survival. a non-specific 213Bi conjugate are shown in Fig. 5. Each point represents
Another experiment looking at the effect of external photon beams a mean of three experiments with each experimental point having tri-
and alphas on two cell lines was undertaken by Riquier et al. (2013). plicate wells. This result clearly demonstrates that the alphas are only
Survival curves are shown in Fig. 2 for A549 cells and endothelial cells effectively cytotoxic to cancer cells when they are targeted and at-
(EC) exposed to x-rays or alpha particles, which were obtained by tached to those cells. The cytotoxicity of alpha labeled specific targeting
clonogenic assay. The survival curves are fitted with the Linear Quad- vector is very much greater (10–100 times) than the cross fire generated
ratic model. The high LET radiation gave a linear curve on the log plot, at these uCi activity incubations. However, if the vector is truly non-
while the low LET radiation a shoulder curve. The relative biological specific or is unlabeled, then it is not directly cytotoxic to the cancer
effect is seen to be close to 5 for 1–10% survival (Riquier et al., 2013). cells.

3.2. Cancer cell survival studies after in vitro incubation of 4. Preclinical in vivo studies
radioimmunoconjugates
In vivo studies use human cancer xenografts in immune-in-
3.2.1. Melanoma competent mice to test pharmacokinetics, efficacy and the maximum
In a study reported by Rizvi et al., the 9.2.27 monoclonal antibody, tolerance dose of radiolabelled targeting vectors. While single cell
which targets the MCSP antigen expressed by most melanoma cells, was radiobiology is determined in vitro studies, in vivo studies are much
investigated. MM138 cells were incubated in different activities of the more complex, involving physiological properties such as bioavail-
same mAb labeled with either 213Bi (alpha) or 152Tb (positron). The ability, clearance, tumour capillary density and permeability and im-
survival of melanoma cells is shown in Fig. 3 (Rizvi et al., 2000). mune response. The objective is to observe efficacy within the max-
Note that the half-lives are 213Bi = 46 m for vs 18.5 h for 152Tb. imum tolerance dose (MTD), efficacy meaning the ability of the therapy
After a 1 h incubation time, the cells were washed. However, attached to stop tumour formation, retard tumour growth or partial or complete
152
Tb conjugates can remain on or in the cell, causing additional cy- tumour regression after subcutaneous, intraperitoneal or intravenous
totoxicity over the 24 h counting time. A half-life correction has not inoculation of cancer cells. Table 3 is a compilation of in vivo studies
been made. The RBE for 37% survival (Do) is 120, after correction for directly comparing the efficacy and toxicity of α-RIT vs β-RIT.
branching ratios. This first result showed the marked difference in
targeted alpha cytotoxicity compared with the beta label. 4.1. Ovarian ascites

3.2.2. Leukaemia The first attempt to compare the effectiveness of alpha- versus beta-
Acute myeloid leukemia cells (HL-60) express the CD33 antigen, emitting radioisotopes was related to the treatment of malignant
which is targeted by the WM53 monoclonal antibody. The WM53 Mab ovarian ascites in mice (Bloomer et al., 1984). The efficacy of the alpha-

9
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

Fig. 4. Log of DNA uptake and cell survival studies with alpha
versus beta targeted therapy, free radioisotopes and non-specific
radio-labeled conjugates (Courtesy (Rizvi et al., 2002)). Only the
Bi213 labeled WM53 MAb showed high cytotoxicity.

therapeutic window in favour of the alpha-emitting isotope that could


be further pursued in clinical settings.

4.4. Ovarian peritoneal carcinoma

The effect of alpha-RIT was investigated using monoclonal huma-


nised anti-CD138 antibody radiolabeled with 213Bi. The effect of RIT
was compared with the outcome after hyperthermic intraperitoneal
chemotherapy, an accredited treatment with new research protocols
(Derrien et al., 2015). The mice undergoing alpha-RIT outlived those
that were treated with chemotherapy (median survival 37.5 days).
Median survival rates for the RIT group could not be calculated as over
50% of mice were still alive after study completion (at 90 days).

Fig. 5. Comparative prostate cancer cell log-survival for the 213Bi labeled specific 4.5. Breast cancer lung metastases
(square) and non-specific mAbs (triangle) (Courtesy (Li et al., 2002b)).
The efficacy of the 225Ac and 90Y radiolabels were compared in a
emitter 211
At-tellurium colloid was compared with the beta-emitting mouse model of breast cancer metastases (Song et al., 2009). A single
colloids 32P, 165Dy and 90Y. Radiocolloids of beta emitters were not administration of 225Ac (14.8 kBq)–labeled anti-rat HER-2/neu mono-
curative, irrespective of the doses used, whereas the 211At-tellurium clonal antibody (7.16.4) completely eradicated breast cancer lung mi-
colloid was shown to be curative for doses of 925 MBq. The increased crometastases in ∼67% of HER-2/neu transgenic mice and led to long-
therapeutic efficacy of the alpha-emitting radioisotope was ascribed to term survival of these mice for up to 1 year. 225Ac was significantly
its densely ionising character. more effective than the 90Y- (4.44 MBq; median survival of 50 days;
P < .001) as well as untreated control (median survival of 41 days;
P < .0001).
4.2. Colon cancer 225
Ac treated metastases received a total dose of 9.6 Gy, sig-
nificantly higher than 2.0 Gy from 213Bi and 2.4 Gy from 90Y.
Since this pioneering study, the following experimental investiga- Biodistribution studies revealed that 225Ac daughters, 221Fr and 213Bi,
tion in solid tumours that showed a therapeutic advantage of α-RIT accumulated in kidneys and probably contributed to the long-term
after a direct comparison with β-RIT was reported by Behr et al. (1999). renal toxicity observed in surviving mice. These data suggest 225Ac
Using monovalent antibody fragments (Fab’) of the Mab CO17-A1, the labeled anti–HER-2/neu monoclonal antibody could significantly pro-
group has evaluated the tumoricidal and normal tissue effects of RIT long survival in HER-2/neu–positive metastatic breast cancer patients.
with an alpha emitter (213Bi/213Po) and a beta emitter (90Y) in a human
colon cancer model in nude mice. The advantageous physical properties
4.6. Prostate cancer
of 213Bi, such as the high mean LET of 100 keV/μm as compared to only
0.2 keV/μm of 90Y as well as its short range in tissue (0.1 mm vs
A novel 213Bi-labeled peptide, DOTA-PEG4-bombesin (DOTA-PESIN)
3.96 mm) lead to a therapeutically superior outcome with α-RIT,
was compared with 177Lu-labeled DOTA-PESIN in a human androgen-
through a 2–3 fold higher radiobiological effectiveness in the tumour
independent prostate carcinoma xenograft model (PC-3 tumour) (Wild
(Behr et al., 1999).
et al., 2011). Animals were injected with 213Bi or 177Lu-DOTA-PESIN to
determine the MTD, biodistribution and dosimetry of each agent; con-
4.3. Neuroendocrine tumours trols were left untreated or were given nonradioactive 175Lu-DOTA-
PESIN.
Alpha-emitting isotopes have shown improvement over beta emit- The MTD of 213Bi-DOTA-PESIN is 25 MBq (25.16 MBq) and for the
177
ters (Miederer et al., 2008). Over the last two decades, peptide receptor Lu conjugate is 112 MBq (111 MBq). Alpha therapy with 213Bi-
radionuclide therapy has been developed for these tumours. Given their DOTA-PESIN was clearly superior to the treatment with 177Lu-DOTA-
high-level expression of somatostatin receptors, somatostatin analogues PESIN at the MTD, although the tumour uptake was not significantly
having high affinity to these receptors have been studied in the context different between the 2 tracers. 213Bi-DOTA-PESIN treatment resulted
of metastasized neuroendocrine tumours. Most commonly, beta-emit- in a highly significant increase in life span to 30 weeks when compared
ting isotopes have been employed (90Y or 177Lu-DOTATOC), that with nontreated controls (P < .0004; log-rank test) and 177Lu-DOTA-
showed a minimum 50% tumour regression in up to 50% of patients PESIN (P = .043). The median survival of animals treated with
(Kam et al., 2012; Waldherr et al., 2001). As demonstrated by pre- 4 × 28 MBq 177Lu-DOTA-PESIN was 12.9 weeks, which was not sig-
clinical studies, the somatostatin receptor radiotherapy can be im- nificantly longer than that of untreated controls (P = 0.085). Both
177
proved by replacing low LET beta emitting isotopes with high LET alpha Lu and 213Bi conjugates showed minimal to slight kidney damage
emitters, such as 225Ac (Miederer et al., 2008). A comparative study on 20–30 weeks post-treatment. These preclinical data indicate that α-RIT
the effectiveness of 177Lu-DOTATOC and 225Ac-DOTATOC showed a is more efficacious than β-RIT. Furthermore, 213Bi-DOTA-PESIN

10
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

Table 3
in vivo studies directly comparing the efficacy and toxicity of α-RIT vs β-RIT.

Cancer type [ref] α-RIT β-RIT Observations Conclusion

Colon cancer (Behr 213


Bi-labeled CO-1A 90
Y-labeled CO-1A 90
Y more nephrotoxic. α-RIT is more effective than β-
213
et al., 1999) Fab′ (dose from 3.7 Fab′ (dose from 3.7 Bi-Fab′ was therapeutically superior to 90Y-Fab′ at RIT (tumour RBE is 2–3 fold
MBq) MBq) each dose. higher).
Neuro-endocrine 225
Ac-DOTATOC 177
Lu-DOTATOC Activities up to 20 kBq have no toxic effects in mice. α-RIT with 225Ac improves
(Miederer et al., (10–130 kBq) (450 kBq-2MBq) outcome compared with177Lu.
2008)
At 20 kBq, Ac reduced tumour growth more effectively
than Lu.
Multiple myeloma 213
Bi-labeled 9E7.4 Lu-labeled 9E7.4 Median survival with α-RIT compared to control: α-RIT superior to β-RIT in both
(Fichou et al., anti-CD138 mAb mAb (18.5 MBq) 80 days vs 37 days; 45% mice cured. tumour control and survival.
2015) (3.7 MBq)
Median survival with β-RIT com-pared to control:
54 days vs 37 days; no mice cured.
Breast cancer lung 225
Ac-7.16.4 anti-rat 90
Y-7.16.4 anti-rat 225
Ac completely eradicated lung micro-metastases in α-RIT markedly superior to β-
metastases HER-2/neu 14.8 kBq HER-2/neu 4.44 ∼67% of mice; with long-term survival up to 1 year. RIT in both tumour control and
(Song et al., 2009) MBq survival.
Significantly more effective than 90Y with median
survival 50 days; untreated controls had a median
survival of 41 days.
Prostate cancer (Wild 213
Bi-DOTA-PESIN 177
Lu-DOTA-PESIN LD40 at the MTD was ∼30 weeks for 225Ac vs 13 weeks α-RIT superior to β-RIT in
et al., 2011) 25 MBq 112 MBq for 177Lu. survival.
213 177
Peritoneal carcino- Bi-d9MAb 1.85 Lu-d9MAb 1.85 Comparable efficacy. But only 177Lu caused As 213Bi-RIT has no toxicity for
matosis (Seidl MBq Over MBq Over lymphoblastic lymphoma, proliferative the same efficacy, it is the
et al., 2011) 24 h = 2.05 MBq·h 24 h = 42.2 MBq·h glomerulonephritis and hepatocarcinoma. preferred treatment.

represents a new approach for use in peptide-receptor TAT for recurrent therapeutic efficacy without toxicity, it should be preferred for the
prostate cancer. treatment of peritoneal carcinomatosis.
Using 212Pb as an alpha emitter for the management of small vo-
4.7. Peritoneal carcinomatosis lume peritoneal carcinomatosis, a comparison was undertaken between
internalizing anti-HER2 mAbs (trastuzumab) and non-internalizing
Nude mice received intraperitoneal inoculation of HSC45-M2 gas- anti-CEA mAbs (35A7) in alpha-RIT (Boudousq et al., 2013). Athymic
tric cancer cells expressing d9-E-cadherin and were treated in- nude mice bearing 2–3 mm tumour xenografts were intraperitoneally
traperitoneally 1 or 8 days later with different activities of specific injected with 370, 740 and 1480 kBq; 37 MBq/mg 212Pb-labeled 35A7,
177
Lu-d9MAb immunoconjugates targeting d9-E-cadherin or with non- trastuzumab or nonspecific mAbs, or with unlabeled mAbs/NaCl. The
specific 177Lu-d9MAb (Seidl et al., 2011). Therapeutic efficacy was results indicated that 212Pb-mAb RIT warrants further investigations as
evaluated by monitoring survival for up to 250 days. For evaluation of a post-surgery adjuvant therapy in patients with peritoneal carcino-
toxicity, both biodistribution of 177Lu-d9MAb and blood cell counts matosis.
were determined at different time points and organs were examined Promising results with 212Pb in tumour xenografts were obtained
histopathologically. when combining 212Pb-trastuzumab with paclitaxel in order to in-
For similar therapeutic efficacies, the intraperitoneal injection re- vestigate the molecular basis of enhanced cell killing by alpha emit-
quired 825-times more 177Lu-RICs than 213Bi-RICs. Thus, one α-particle ters. Yong et al. have investigated the impact of targeted alpha therapy
was as cytotoxic as approximately 1000 β-particles. The markedly dif- on gene expression in a pre-clinical model for disseminated peritoneal
ferent half-lives cause the cumulative activity over 24 h post- admin- disease and observed that the alpha emitter-drug duo inhibits growth
istration of 1.85 MBq 177Lu to be 42.2 MBq·h and that for 213Bi to be arrest and suppresses proliferation through regulation of genes that are
2.05 MBq·h. involved in DNA damage repair and apoptosis (Yong et al., 2014).
Treatment on day 1 after tumour cell inoculation was more effective Similar molecular mechanisms were identified when 212Pb-trastu-
than treatment on day 8, and specific 177Lu-d9MAb conjugates were zumab was administered in combination with gemcitabine, demon-
superior to nonspecific 177Lu-d8MAb. Treatment with 7.4 MBq of 177Lu- strating a great therapeutic potential in pre-clinical models (Yong
d9MAb was most successful, with 90% of the animals surviving longer et al., 2016).
than 250 days. However, treatment with therapeutically effective ac-
tivities of 177Lu-d9MAb was not free of toxic side effects. In some ani- 5. Clinical studies
mals, lymphoblastic lymphoma, proliferative glomerulonephritis and
hepatocarcinoma were seen but were not observed after treatment with 5.1. Radioisotopes and maximum tolerance dose (MTD)
213
Bi-d9MAb at comparable therapeutic efficacy. The therapeutic effi-
cacy of 177Lu-d9MAb is impaired by toxic side effects. Myelotoxicity Radioisotopes used in clinical trials include, from short to long
was observed after intraperitoneal administration of 177Lu-d9MAb as range betas, are 177Lu, 131I, 188Re, 166Ho and 90Y. 177Lu emits a low
lymphoblastic lymphoma in some animals. However, chronic myelo- energy beta (150 keV) with an effective range of ∼ 300 μm (Gledhill,
toxicity could never be observed following treatment of animals with 1973). Alpha emitting radioisotopes and their respective half-lives are
213
Bi-d9MAb. As therapy with 213Bid9MAb revealed comparable 211
At (7 h), 212Bi (60 min), 213Bi (67 min), 223Ra, 225Ac (10 d).

11
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

The linear energy transfer (LET) to targeted cancer cells from 177Lu for staff/family), alpha-emitting radioisotopes are seen to be more ap-
is only ∼0.5 keV/μm so cancer cell toxicity would also be very low. On propriate for AML therapy.
the other hand, 213Bi is an alpha emitting radioisotope with alpha en- The first clinical trial using targeted alpha therapy was designed for
ergy of ∼8 MeV, a range of 80 μm and LET ∼100 keV/μm. This is 200 acute myelogenous leukemia. Within the trial 18 patients with relapsed
times the LET for 177Lu. This means that if 213Bi is used to label the and refractory acute or chronic myeloid leukemia were accrued and
vector, then cancer cell cytotoxicity will be very much greater than for treated with 213Bi-AIC of an anti-CD33 mAb (Jurcic et al., 2002). The
177 213
Lu. Further, normal tissue exposure will be less because of the much alpha-immunoconjugate Bi-CHX-A”-HuM195 (Bi-lintuzumab),
shorter range. However, the short half-life of 46 min compares with 6.7 showed feasibility, safety and anti-leukemic activity in all patients.
d for Lu177 and argues against 213Bi infusion into established tumours. Dose escalation from 10.36 to 37 MBq/kg of AIC was tested. It was
225
Ac has a 10 day half-life with 4 alpha emissions. A comparative study showed that alpha-immunoconjugate uptake by the leukemic cells oc-
shows that on all aspects, 225Ac is equal or superior to 213Bi (Allen, curs within 5 min of systemic injection. While all 17 evaluable patients
2017). Post-androgen deprivation therapy shows a nadir in PSA value. developed myelosuppression, with 22 days median recovery time, no
At this stage tumour may be absent but cells insensitive to androgen significant extramedullary cytotoxicity was observed. Regarding tu-
deprivation are present. This is the ideal time for targeted short range mour control, 93% of patients showed reductions in circulating blasts
and high LET alpha radiation to delay the onset of lethal castration and 78% had reductions in bone marrow leukemic cells 7–10 days after
resistant prostate cancer. treatment.
While animal models offer some guidance regarding tolerance doses Previous studies by the same group employing beta-emitting radio-
for humans they cannot replace phase I trials. Toxicity may depend immunoconjugates (131I-M195, 131I-HuM195, 90Y-HuM195) allowed a
both on the radioisotope and the character of the targeting vector dosimetric comparison between the alpha- and beta-emitters (Caron
(murine or humanised). MTDs are given in terms of body weight, as- et al., 1994; Jurcic et al., 2000; Schwartz et al., 1993). It was showed
sumed to be 70 kg in humans, when not stated. The MTD value reported that the absorbed dose ratios between the bone marrow, liver, spleen
for 213Bi in humans is 37 MBq/kg (Jurcic et al., 2002). The long half-life and the whole body were about 1000 times higher for 213Bi-HuM195
of 225Ac renders the injected activities to be lower than 213Bi by than for the beta emitters. This result was mainly due to the high target
570 kBq/kg (Kratochwil et al., 2015); 225Ac PSMA617 in humans with dose and the considerably reduced whole body dose for 213Bi as com-
metastatic CRPC: 100 kBq/kg per fraction (Kratochwil et al., 2016a), pared to 90Y and 131I.
and 225Ac lintuzumab in humans with relapsed/refractory AML: While this first clinical trial using Bi-lintuzumab showed great po-
111 kBq/kg (Jurcic et al., 2011). tential in tumour control, the large tumour burden (up to 1012 leukemia
cells) did not allow complete remission. Consequently, a phase I/II trial
5.2. Hematologic malignancies was conducted at Memorial Sloan Kettering Cancer Center using partial
cyto-reduction with cytarabine followed by 18.5 MBq to 46.3 MBq per
The first clinical studies designed to target the CD33 antigen that is kg of AIC. The dose limiting toxicity was myelosuppression, which set
expressed by most myeloid leukemias have used the murine M195 or the maximum tolerated dose to 37 MBq. Clinical response was achieved
the humanised HuM195 antibody as anti-CD33 RI Scheinberg et al. in 24% of patients at doses ≥ 37 MBq/kg (Rosenblat et al., 2010).
(1991) reported the first pilot trial using beta-RIT with 131I-M195 in 10 These results confirmed the ability of 213Bi-lintuzumab to trigger re-
patients with AML (Scheinberg et al., 1991). While the study showed mission in patients with high-risk AML, therefore justifying the use of
rapid tumour targeting and internalization of 131I-M195 by the blasts, alpha-emitting radioisotopes for hematological malignancies (see
no tumour control has been achieved with the trialed doses. Conse- Table 4).
quently, a dose-escalation trial (1.85–7.7 GBq/m2) followed in 24 pa- The major shortcoming of Bi-lintuzumab is the short half-life of
213
tients, showing sufficient depletion of leukemia in 8 patients to permit Bi and the necessity for an on-site Bi generator. Since the efficacy of
bone marrow transplant (Schwartz et al., 1993). alpha-RIT could be increased even more with longer lasting activity a
Radiolabeled anti-CD45 mAb (CD45 antigen is expressed on all second generation RIT was developed represented by 225Ac. In addition
leukocytes) has also been investigated in a number of trials, mainly in to its longer half-life (10 days), 225Ac emits 4 alpha particles with a
combination with 131I. The first phase I trial using 131I-BC8 mAb in cytotoxicity 1000 times greater than the cytotoxic effect of 213Bi. To
combination with cyclophosphamide and 12 Gy total body irradiation investigate its clinical potential, a phase I trial that accrued 18 AML
enrolled 44 patients with refractory AML or ALL, or myelodysplastic patients was conducted (Jurcic et al., 2011). Doses delivered ranged
syndrome (Matthews et al., 1999). Thirty-four patients received a between 18.5–150 kBq/kg of 225Ac-lintuzumab, setting the maximum
therapeutic dose of 131I-BC8 mAb that delivered an estimated absorbed tolerated dose to 110 kBq/kg. Peripheral blasts were eliminated in 63%
dose to the liver (the dose limiting organ) between 3.5 Gy and 12.25 Gy. of patients, while reduction in bone marrow blast was achieved in 67%.
The maximum tolerated dose was 10.5 Gy. The radiation doses deliv- More studies are underway investigating the effect of fractionated
225
ered to marrow were 2.3-fold greater than liver and spleen doses. Ac-lintuzumab with low-dose cytarabine in previously untreated
Disease free survival after a median of 65 months among the 25 AML older patients (Jurcic and Rosenblat, 2014).
patients was 28%, while 3 out of 9 ALL patients presented with disease RIT is a promising treatment for hematological malignancies.
free survival up to 66 months post-transplant (Matthews et al., 1999). Radiolabeled monoclonal antibodies are good candidates and the re-
More recent studies support the evidence whereby 131I-anti-CD45 sults of clinical studies warrant further research. While beta-RIT is
added to conventional treatment for acute AML improve survival suitable for the treatment of large tumour burden, alpha-RIT has the
(Mawad et al., 2014). Furthermore, the promising results due to RIT has advantage of a more focused targeting which makes it ideal for isolated
stimulated the development of alpha-radiolabeled anti-CD45 mAb, with tumour cells and residual disease (Bodet-Milin et al., 2016). The un-
the aim to further intensity treatment and diminish side effects. biased advantage of alpha therapy could only be determined via a well-
188
Re and 90Y are two other beta-emitting radioisotopes tested for designed large randomized clinical trial, comparing alpha-RIT with
RIT in conjugation with anti-CD66, the carcinoembryonic antigen ex- beta-RIT labeled to the same targeting agent.
pressed on granulocytes and epithelial cells. While the two radio-
isotopes have very similar ranges in tissue (2.4 mm vs 2.7 mm), yttrium 5.3. Metastatic melanoma
provides the higher bone marrow dose that results in the higher ther-
apeutic efficacy (Ringhoffer et al., 2005). The first report of a clinical trial using RIT for metastatic melanoma
As already mentioned above, to overcome some of the limitations was published in 1985 (Larson et al., 1985). I-131 labeled Fab frag-
imposed by beta-RIT (such as normal tissue toxicity and radiation risk ments of the high molecular weight antigen (HMWA) (melanoma

12
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

Table 4
A compilation of representative beta-RIT and alpha-RIT trials for AML and ALL.

Trial/study Vector Target Conclusions/observations

Beta emitter
Phase I study for AML (Scheinberg et al., 1991) M195 CD33 antigen I-131 Rapid tumour targeting with 131I-M195 without complete response.
131
Phase I/II study for AML(Schwartz et al., 1993) M195 CD33 antigen I-131 I-M195 could be considered as a treatment alternative prior to
bone marrow transplant
131
Phase I study for AML or ALL (Matthews et al., 1999) BC8 mAb CD45 antigen I-131 I-BC8 mAb for advanced AML/ALL was feasible, safe and
warranted further investigations.
Phase I/II study for AML or ALL (Ringhoffer et al., anti-CD66 CD66 antigen Y-90 Overall survival at 12 months – 70%; at 30 months 17%. Higher
2005) Re-188 relapse rate in the rhenium group.

Alpha emitter
Phase I study for AML (Jurcic et al., 2002) MAb lintuzumab CD33 antigen Bi-213 Bi-lintuzumab was shown to be safe, feasible and efficient therapy.
Phase II study for post-chemotherapy of AML MAb lintuzumab CD33 antigen Bi-213 One quarter of patients achieved clinical response, without
(Rosenblat et al., 2010) significant toxicity.
Phase I study for AML (Jurcic et al., 2011; Jurcic and MAb lintuzumab CD33 antigen Ac-225 Ongoing trial
Rosenblat, 2014) Clinical trial NCT01756677 (2015)

Table 5
A compilation of representative beta-RIT and alpha-RIT trials for metastatic melanoma.

Trial/study Vector Target Conclusions/observations

Beta emitter
131
Phase I trial for metastatic melanoma (Larson anti-HMWA HMWA I-131 I-HMWA showed good tumour localization; larger doses resulted in > 50% tumour
et al., 1985) Fab reduction and disease stabilization for 3 months.
188
Phase I trial for metastatic melanoma (Klein MAb 6D2 melanin Re-188 Re-6D2 treatment for advanced metastatic melanoma patients resulted in median
et al., 2013) overall survival of 13 months with no dose-limiting toxicities.

Alpha
emitter
213
Phase I trial for intralesional melanoma cDTPA-9.2.27 MCSP Bi-213 Bi-cDTPA-9.2.27 is an efficient agent without added normal tissue toxicity. Further
(Allen et al., 2005) studies are warranted.
213
Phase I trial of systemic melanoma (Allen cDTPA-9.2.27 MCSP Bi-213 Bi-cDTPA-9.2.27 showed very good response in a select group of patients. There was
et al., 2011; Raja et al., 2007) no evidence of a dose dependence. Several tumour-related factors might influence
response to alpha-RIT, thus more studies are needed.

−associated proteoglycan) was used in the trial that enrolled 10 pa- human trial investigating this agent enrolled 16 patients with meta-
tients. The administered doses ranged from 185 MBq to 481 MBq. All static skin melanoma that were positive to the monoclonal antibody
patients with lesions greater than 1 cm showed good localization of the 9.2.27 (Allen et al., 2005). Intralesional TAT was found to be efficient at
agent is one or more lesions. The patient receiving the highest dose a dose of 22 MBq and safe up to 50 MBq. As shown by melanoma in-
showed a greater than 50% reduction in tumour size as well as disease hibiting activity, apoptosis and Ki67 proliferation marker tests 213Bi-
stabilization for a three-month time period. Despite these promising cDTPA-9.2.27 is a promising agent in the management of unresectable
results, RIT was not pursued for the treatment of unresectable mela- secondary melanomas.
nomas until more recently (Table 5). The same alpha-immunoconjugate was employed for the treatment
A novel approach to beta-RIT of metastatic melanoma is by means of metastatic melanoma based on a single bolus intravenous TAT in-
of melanin targeting with radiolabeled monoclonal antibodies (such as jection. The open-labeled phase I dose escalation trial aimed to estab-
6D2 mAb). Experiments with tumour-bearing nude mice showed some lish the effective dose of 213Bi-cDTPA-9.2.27 in a group of 38 patients,
therapeutic effect without significant toxicities with 166Ho-6D2 and treated with doses ranging from 46 MBq to 925 MBq (Allen et al., 2011;
188
Re-6D2, but not with 90Y (Dadachova et al., 2008; Thompson et al., Raja et al., 2007). Both superficial and internal lesion regression have
2014). The results of two open-label phase I trials investigating safety been achieved at fairly low activities, from 148 to 259 MBq. Median
and efficacy of 188Re-6D2 showed good tolerance and tumour activity survival time for patients with stable disease or partial response (10% of
(Klein et al., 2013). Beside pharmacokinetics, safety and antitumour patients) was 612 days, significantly higher than the median survival
activity, the first phase I trial (phase Ia, 13 patients) aimed to determine time of 266 days among the overall study population (Allen et al.,
the effect of unlabeled mAb on biodistribution of 188Re-6D2, whereas 2011). Since there were no adverse events, the maximum tolerated dose
phase Ib (7 patients) investigated the effect of dose escalation. The fast was not achieved in this trial. While among the responders the results
clearance and relative short half-life of Re (17 h) resulted in good tol- were above expectations, only a small fraction of patients showed po-
erability with maximum doses delivered (3.7 GBq). Median survival sitive outcome. This might be due to several factors such as tumour size,
was about 13 months, which is longer than the median 8.5 months after variations of the vascular structures, size and frequency of fenestrations
standard melanoma treatment. These results warrant further in- and also the expression of targeted receptors. However, tumour anti-
vestigations with 166Ho-6D2 and 188Re-6D2. vascular alpha therapy (TAVAT) is considered to be the main re-
Targeted alpha therapy for intralesional metastatic melanoma was sponsible factor for the positive results obtained in the small group of
aimed to establish a new systemic therapy for this neoplasm. The alpha- patients (Allen et al., 2007).
immunoconjugate designed for this treatment was 213Bi-cDTPA-9.2.27,
exhibiting 120 times greater cytotoxicity to melanoma cells than beta- 5.4. Bone metastases
emitting radioisotopes (Allen et al., 2001; Rizvi et al., 2000). The 9.2.27
mAb is specific for the 250 kDa cell-surface antigen, human melanoma Bone metastasis is still a clinical challenge in prostate and breast
chondroitin sulphate proteoglycan (MCSP), antigen that is expressed in cancer and has a significant impact on the quality of life. While there is
over 90% of melanoma cells lines (Bumol and Reisfeld, 1982). The first no conclusive therapy to control bone metastasis, bone-seeking

13
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

Table 6
A compilation of representative beta-RIT and alpha-RIT trials for bone metastases.

Trial/study Conclusions/observations

Beta
emitter
Phase II randomized trial in advanced androgen-independent prostate cancer (Tu Sr-89 Patients randomized to receive doxorubicin with/without 89Sr. Median
et al., 2001) survival in the 89Sr group was 27.7 months vs 16.8 months of chemo-only
group.
Phase III trial comparing the effect of 89Sr vs external beam radiotherapy for bone Sr-89 No significant difference in overall survival or pain relief at the index site.
metastasis of hormone-resistant prostate cancer (Porter et al., 1993) Progression of pain as measured by sites of new pain showed better outcome
in the 89Sr arm. Quality of life analysis showed superiority of strontium in
pain relief.
Phase III trial comparing the effect of 89Sr vs external beam radiotherapy in Sr-89 There was no significant difference in median survival. Both treatment
metastatic prostate cancer (Quilty et al., 1994) provided pain relief sustained to 3 months in 63.6% after radiotherapy and
66.1% after 89Sr.
Phase III trial comparing the effect of 89Sr vs external beam radiotherapy for bone Sr-89 89
Sr vs palliative local field radiotherapy showed a borderline statistically
metastasis of hormone-resistant prostate cancer (Oosterhof et al., 2003) significant difference in overall survival for the radiotherapy group. No
difference in progression-free survival or toxicity.
Phase III randomized trial for bone metastasis (Serafini et al., 1998) Sm-153 Double-blind placebo-controlled trial of 153Sm-EDTMP in patients with bone
metastasis. Significant pain relief observed in the Sm group in up to 72%
patients during the first 4 weeks. Mild bone marrow suppression.

Alpha
emitter
Phase I/II trial for bone metastases (Nilsson et al., 2007; Nilsson et al., 2005) Ra-223 Median relative change in bone-ALP was −66% in the 223Ra group vs 9.3% in
the placebo group. Median overall survival was 65 weeks for 223Ra vs 46
weeks for placebo. No significant Ra-induced toxicity.
Phase III trial for prostate cancer and symptomatic bone metastases with or without Ra-223 High efficacy of radium. Median overall survival of patients treated with
223
previous docetaxel (ASYMPCA trial) (Hoskin et al., 2014; Sartor et al., 2014) Ra was 14.9 months as compared with the 11.3 months of the placebo
group.
Phase I: Safety, biodistribution, radiation dosimetry and pharmacokinetics study of Ra-223 Initial bone uptake was 52%, with maximum activity of Ra in the bone within
BAY88-8223 in Japanese patients (Yoshida et al., 2016) 2 h of injection and no signs of activity in other organs.
Phase III: Asian population study in the treatment of CRPC patients with bone Ra-223 Ongoing trial
metastasis (completion 2016) Clinical trial NCT01810770 (2013)
Phase I/II: Re-treatment safety of Ra-223 dichloride in CRPC with bone metastases Ra-223 Ongoing trial
(completion 2017) Clinical trial NCT01934790 (2013)
Randomized open-label phase II study evaluating the efficacy/safety of Ra-223 in Ra-223 Ongoing trial
combination with abiraterone acetate or enzalutamide in patients with CRPC Clinical trial NCT02034552 (2013)
and bone metastases (completion 2022)

radioisotopes, both beta and alpha emitters, are under clinical in- of which 33 were assigned to receive four intravenous injections of
vestigation. Of the beta-emitting radioisotopes, 89Sr and 153Sm have 223
Ra every four weeks, with an activity of 50 kBq/kg. Patients were
been widely investigated and approved for palliative treatment. monitored for survival and long term toxicity over 24 months. The
Nevertheless, the results of phase II/III trials conducted to date are 223
Ra group had significant reductions in all five markers of bone for-
inconclusive, as while some failed to demonstrate improvement in mation and bone balance such as bone-ALP, total-ALP, serum type 1
progression-free or overall survival (Oosterhof et al., 2003; Porter et al., procollagen, collagen fragment CTX-1 and ICTP. Also, substantial dif-
1993; Quilty et al., 1994), others showed higher effectiveness in the ferences were observed with changes in PSA from baseline to 4 weeks,
radionuclide arm (Oosterhof et al., 2003; Porter et al., 1993; Quilty PSA decreasing by 24% in the 223Ra group and increasing by 45% in the
et al., 1994; Tu et al., 2001). Nevertheless, the vast majority of these placebo group. Median time to PSA progression was 26 weeks for 223Ra
trials showed some improvement in the quality of life through pain vs 8 weeks for placebo (P = .04). Treatment with 223Ra was well tol-
palliation (see main conclusions in Table 6). erated and no patients discontinued treatment.
As a calcium mimetic, the bone-seeking alpha emitter, 223Ra was The success of the phase II trial has encouraged the wider use of
shown to have potential for palliating cancer patients affected by bone 223
Ra. Therefore, Radium-223 dichloride (Xofigo®) has recently been
metastases. Nilsson et al. (2005) reported on the first clinical experience approved for the treatment of castration-resistant prostate cancer
with radium-223 within a phase I trial that enrolled 15 hormone re- (CRPC) patients with symptomatic bone metastases, due to its effi-
fractory prostate cancer patients and 10 breast cancer patients, with ciency in bone-targeting and increased patient survival (Nilsson, 2014).
bone metastasis (Nilsson et al., 2005). The aim of the trial was to assess The Alpharadin in Symptomatic Prostate Cancer Patients
the safety and tolerability of 223Ra and also to evaluate pain palliation. (ALSYMPCA) is a large, multicenter, randomized, double-blind, pla-
Radium was well tolerated within the administered range (50–250 kBq/ cebo-controlled trial targeting CRPC patients which enrolled 921 pa-
kg). Treatment efficacy was confirmed by the significant reduction in tients with bone metastases (Hoskin et al., 2014; Parker et al., 2013;
serum alkaline phosphatase (ALP) and improved pain control, with pain Sartor et al., 2014). Prior randomization, patients were stratified as a
relief reported by 52%, 60%, and 56% of patients after 7 days, 4 weeks function of: baseline ALP, previous/no use of docetaxel and current/no
and 8 weeks, respectively. Side effects were acceptable without dose use of a bisphosphonate. Treatment with 223Ra consisted of 6 in-
limiting toxicity. Further investigations followed driven by the positive travenous injections at 4-week intervals delivering a dose of 50 kBq/kg.
results. Thus, a randomized, double blind, placebo controlled, multi- The primary endpoint was overall survival, whereas the main sec-
centre phase II study examined the effect of multiple doses of Ra-223 in ondary endpoints targeted the changes in biomarker levels for PSA and
patients with hormone-refractory prostate cancer (Nilsson et al., 2007). ALP as well as the symptomatic bone-related effects. The results of the
Similar end-points were set as in the previous phase I trial, such as the ALSYMPCA trial confirmed the high efficacy of radium as indicated by
reduction in bone-specific alkaline phosphatase concentration and also previous phase I/II trials. The median overall survival of patients
time to occurrence of skeletal-related events. treated with 223Ra was 14.9 months as compared with the 11.3 months
The trial enrolled 64 patients needing external beam radiotherapy, of the placebo group (HR = 0.70, 95% CI). Additionally, the time delay

14
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

to an increase in ALP level in the 223Ra group was 7.4 months compared tumour-associated glycoprotein 72 and tumour growth with increasing
with the placebo group that had only 3.8 months (HR = 0.17, 95% CI) radioactivity administered.
(Hoskin 2014). Overall radium-223 was found to be effective and safe
in patients with CRPC and symptomatic bone metastases. 5.6. Glioma/glioblastoma (GBM)
The positive clinical outcome of the previous trials led to the design
of several studies with 223Ra. Table 6 is a compilation of the completed A highly targeted treatment approach implemented over the last
and ongoing worldwide trials in patients with CRPC and bone metas- couple of decades in order to increase local tumour control in malignant
tases. brain tumours with poor prognosis is the administration of radiolabeled
monoclonal antibodies into the surgically resected cavity. In this way,
5.5. Ovarian carcinoma local irradiation of the residual cancer cells by means of beta- or alpha-
RIT can potentially increase response rate and survival (Table 8).
Several beta-emitters have been investigated in the past for in- Beta-RIT has been trialed in the late 1990s using 131I or 90Y labeled
traperitoneal administration in ovarian cancer patients, such as 90Y, to anti-tenascin mAb (Bigner et al., 1998; Riva et al., 1999). Tenascin is
131
I, 177Lu, some with promising results (Epenetos et al., 2000; a glycoprotein overexpressed in gliomas and glioblastomas, that in-
Rosenblum et al., 1999). However, a large, multinational randomized creases in level with advancing tumour grade (Leins et al., 2003) thus
phase III trial for epithelial ovarian cancer that aimed to compare Y-90 can serve as a target in malignant brain tumours.
labeled murine HMFG1 administered with standard therapy versus In a phase I trial of previously irradiated patients with recurrent or
standard treatment alone showed neither survival benefit nor extension metastatic brain tumours, 131I-labeled anti-tenascin monoclonal anti-
of time to relapse from the addition of 90Y-based radio-immunotherapy body 81C6 was administered in 34 patients, with doses up to 4.4 GBq
(Verheijen et al., 2006). These results are partly due to the biophysical (Bigner et al., 1998). The maximum tolerated dose was reached at 3.7
properties of beta-emitting isotopes, as their long range in tissue led to GBq. Dose-limiting toxicity occurred in the form of neurologic and
suboptimal irradiation of tumour clusters to eradicate micrometastases, hematologic toxicity. The estimated median survival for glioblastoma
and also due to the half-life which is long enough (64 h) to result in patients was 56 weeks. The same beta-RIT was administered within a
normal tissue damage (red marrow irradiation). Given the failure of phase II trial that accrued 33 previously untreated patients with ma-
beta-RIT in treating residual cell clusters, alpha-emitting radioisotopes lignant glioma (Reardon et al., 2002). Median survival was longer than
are under clinical investigation, due to their promising results in vitro. in the previously reported phase I trial (Bigner et al., 1998) (Bigner
A phase I trial reported by Andersson et al. (2009) employed 211At 1998): 79.4 and 86.7 weeks for GBM and all patients, respectively. On
labeled to MX35 F(ab’)2 for recurrent ovarian carcinoma in 9 patients third of patients remained alive at a median follow-up of 93 weeks.
with the aim to assess pharmacokinetics, dosimetry and tolerance doses Toxicity was mainly reversible, though one patient required reopera-
(Andersson et al., 2009). The highest agent concentration administered tion for radionecrosis (Reardon et al., 2002).
was 100 MBq/L. The median follow-up was 23 months, during which Twenty previously treated patients with high-grade malignant
no late toxicities were recorded. While the bone marrow is the main glioma were administered 90Y-labeled Mab conjugates (BC-4, a te-
dose limiting organ in RIT, the largest absorbed dose measured was nascin-affine murine MAb) within a phase I clinical trial (Riva et al.,
0.01 Gy, thus about 2.5% of the estimated tolerable dose. Three patients 1999). Dose escalation from 185 to 1110 MBq was performed and the
remained in clinical remission during the follow-up period. maximum tolerated dose was determined to be 925 MBq. The agent
Next to 211At, there is a growing interest towards the clinical po- showed a good biodistribution in the tumour area, without any diffu-
tential of 212Pb (Table 7), the parental radionuclide of 212Bi, which has sion into the normal brain or healthy organs. Due to the advanced
a more optimal half-life for RIT application (10.6 h vs 61 min). stages of all patients, there was no clinical response recorded in this
Meredith et al. (2014) reported the results of a Phase I trial that in- trial.
cluded three ovarian cancer patients after failing all prior therapies The first clinical trial using an alpha-immunoconjugate injection
(Meredith et al., 2014). Patients were treated with 212Pb-TCMC-tras- into the surgically resected cavity for recurrent brain malignancies,
tuzumab that accumulated preferentially inside the peritoneal cavity, as employed 211At-human antimouse chimeric anti-tenascin mAb 81C6
showed by imaging studies. Patient follow-up (> 6 months) showed no (Zalutsky et al., 2008). The activities used ranged from 74 to 347 MBq,
treatment-related side effects, which correlates with the good phar- however the maximum tolerated dose was not reached. The median
macokinetic and dosimetric results. The results justified the accrual of a survival for glioblastoma patients was 52 weeks as compared to 23
larger number of patients to be treated with 212Pb-based RIT. A recent weeks for a placebo group. While no patient experienced dose-limiting
report by the same group has shown the successful administration of toxicity, 6 of 17 patients developed grade 2 neurotoxicity. Despite the
212
Pb-TCMC-trastuzumab in 18 patients that were monitored for safety fact that the normal liver and spleen have high levels of tenascin, there
and outcome for a year (Meredith et al., 2016). Six dose levels were was no measurable 211At uptake outside the treated area. Overall, local
studied starting from 7.4 MBq/m up to 27.4 MBq/m which was con- administration of 211At-ch81C6 was found to be safe and efficacious.
sidered the upper safety limit. There were no late renal, liver or cardiac Another alpha-emitting radioisotope that has been recently trialed
toxicities reported. Tumour marker studies showed a reduction in in recurrent GBM patients is 213Bi-213 (Morgenstern et al., 2014).

Table 7
A compilation of representative beta-RIT and alpha-RIT trials for ovarian carcinomas.

Trial/study Vector Target Conclusions/observations

Beta emitter
90
Phase III trial of intraperitoneal therapy with beta-RIT with/ muHMFG1 MUC1 Y-90 Y-muHMFG1 did not extend patient survival
without standard treatment (Verheijen et al., 2006)

Alpha
emitter
Phase I trial of intraperitoneal TAT for ovarian cancer MX35 F(ab’)2 NaPi2b At-211 Administration of 211At-MX35 F(ab’)2 allows good tumour uptake
(Andersson et al., 2009) in order to achieve therapeutic doses without significant toxicities.
Phase I trial of intraperitoneal TAT for ovarian cancer TCMC-trastuzumab Her2 Pb-212 Very good accumulation in tumour. No side effects. Has potential
(Meredith et al., 2014) for increased therapeutic ratio as compared to beta-RIT.

15
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

Table 8
A compilation of representative beta-RIT and alpha-RIT trials for recurrent brain malignancies.

Trial/study Vector Target Conclusions/observations

Beta emitter
131
Phase I trial of recurrent malignant gliomas Mab 81C6 tenascin I-131 I-labeled 81C6 seems safe and feasible for patients refractory to other therapies.
(Bigner et al., 1998) Median survival for GBM was 56 weeks and for all patients was 60 weeks.
131
Phase II trial in newly diagnosed malignant MAb 81C6 tenascin I-131 I-labeled 81C6 administered to previously untreated glioma patients resulted in a
gliomas (Reardon et al., 2002) 86.7 weeks median survival for all patients and 79.4 weeks for GBM patients. One
patient (3%) patients developed radionecrosis.
Phase I trial of high-grade malignant gliomas (Riva MAb tenascin Y-90 Good biodistribution, without any diffusion into normal tissues. No systemic toxicity.
et al., 1999) Clinical response was not recorded due to very advanced disease stage.

Alpha
emitter
211
Phase I trial of recurrent brain malignancies (74% MAb 81C6 tenascin At-211 At-ch81C6 was found to be feasible, safe and with promising antitumour activity. 2
glioblastoma) (Zalutsky et al., 2008) of 14 glioblastoma patients survived for nearly 3 years after alpha-RIT. No
radionecrosis was observed.
Pilot trial of glioma (Cordier et al., 2010) DOTA-SP NK1R Bi-213 Critically located gliomas were treated with 213Bi-DOTA-substance P for which 90Y
was not suitable due to long range. Well localised and tolerated treatment, without
additional neurotoxicity.
Pilot trial of glioblastoma (Morgenstern et al., DOTA-SP NK1R Bi-213 The first clinical study using 213Bi-SP for recurrent GBM. Median survival was 18
2014) months compared to 15 months with standard therapy. Safe treatment.

Patients were treated via intracavitary or intratumoral delivery of 213Bi- 177


Lu (beta range 1.5 mm) and 213Bi (alpha range 80 μm). In a pilot trial
labeled DOTA-substance P (213Bi-SP) with a maximum dose of 11 GBq reported by Cordier et al. (2010), 213Bi has been tested for functionally
delivered in 2 GBq fractions. The peptide carrier substance P targets NK critically located gliomas that are not eligible for standard treatment
1 receptors that are overexpressed on GMB cells, thus they represent a (Cordier et al., 2010). 213Bi-DOTA-SP was locally injected in 5 patients
good therapeutic target. The median survival of the 20 evaluable pa- with grade 2–4 gliomas and, without local or systemic toxicity. The
tients was 18 months as compared to 15 months after standard therapy study showed that alpha-RIT with 213Bi is safe, and might have the
alone. There was no significant normal tissue toxicity. Although the great advantage to allow post-treatment resection for otherwise un-
patient group enrolled in the trial was small to state definite conclu- resectable gliomas.
sions, a possible dose-response effect was observed, since those patients While beta-RIT studies show promising results in recurrent brain
that received multiple doses of 213Bi-SP showed better tumour control. malignancies, alpha-RIT has important physical and radiobiological
The neurokinin type-1 receptor (NK1R) which is overexpressed in advantages. Given that brain malignancies are usually hypoxic, the
primary malignant gliomas could potentially be used as a target for the oxygen enhancement ratio of nearly 1 for alpha-emitters show a
treatment of glial brain tumours. A novel targeting vector was devel- minimal dependence on the oxygenation status which leads to efficient
oped in this sense at the University of Basel, in the form of a diffusible cell kill in a hypoxic environment.
peptidic vector 1,4,7,10-tetraazacyclododecane-1-glutaric acid-4,7,10-
triacetic acid-substance P (DOTAGA) (Kneifel et al., 2006). A clinical
pilot study was designed to assess biodistribution, short and long-term 5.7. Neuroendocrine tumours
toxicity, as well as clinical and radiological responses in 20 patients
with grade 2–4 gliomas. Radio-conjugates were prepared with the high Neuroendocrine tumours express somatostatin receptors that enable
energy beta emitter 90Y, the low energy beta emitter 177Lu and the high targeting with high affinity peptides. Radiopeptides that target soma-
energy alpha emitter 213Bi. Injected activities into the resection cavity tostatin receptors in these tumours have therefore been investigated
ranged from 2.25 to 7.5 GBq for 90Y, 1.125-6.375 GBq for 177Lu and over the last decade. Most commonly, octreotide analogue peptides
375–825 MBq for 213Bi. The outcome in all cases was positive and (DOTATOC, DOTATATE) are labeled with beta emitters such as 90Y or
177
without significant toxicity. For invasive tumour cells beyond visible Lu, and show clinical response in up to 30% of patients
tumour margins alpha-RIT may be best suited (Kneifel et al., 2006). (Kwekkeboom et al., 2008; Waldherr et al., 2001).
One shortcoming of 90Y beta radiation is the relatively large range Based on the promising results obtained in a phase II trial (Waldherr
in tissue (maximum of 12 mm), which could produce collateral damage et al., 2001) (see Table 9), the Swiss group has extended the in-
to the normal brain. This outcome could be overcome by the use of vestigations with 90Y DOTATOC on 116 patients with metastatic neu-
roendocrine tumours. Patients were treated with 5.9–7.4 GBq/m2 body

Table 9
A compilation of representative beta-RIT and alpha-RIT trials for neuroendocrine tumours.

Trial/study Vector Target Conclusions/observations

Beta
emitter
90
Phase II trial in neuroendocrine tumours DOTATOC Somatostatin Y-90 Y-Dotatoc led to 24% overall response rate, with complete remission rate of
(Waldherr et al., 2001) receptor 2%. Well tolerated treatment.
Phase III randomized trial for midgut DOTATATE Somatostatin Lu-177 The estimated rate of progression free survival at 20 months was 65.2% vs
neuroendocrine tumours (Strosberg et al., receptor 10.8% for the trial arm treated with 177Lu-Dotatate vs control group
2017) (octreotide alone). Manageable toxicity.

Alpha
emitter
Phase I trial in neuroendocrine tumours DOTATOC Somatostatin Bi-213 The first study with 213Bi-DOTATOC in patients refractory to 90Y/177Lu-
(Kratochwil et al., 2014) receptor DOTATOC showed good stability and pharmacokinetics leading to high
efficacy. Good treatment tolerability without kidney failure.

16
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

surface. Significant symptom reduction was achieved in 83% of pa- PSMA expression increases progressively in higher-grade cancers,
tients, with complete remission in 4%, partial remission in 23% and metastatic and castration-resistant disease (Bostwick et al., 1998; Israeli
stabilization in 62% (Forrer et al., 2006). et al., 1993, 1994; Sweat et al., 1998; Wright et al., 1996). Although
Kwekkeboom et al. (2008) reported on treatment efficacy and first thought to be prostate-specific (Israeli et al., 1993), PSMA ex-
toxicity with 177Lu-octreotate in 504 patients. Treatment efficacy was pression has been identified in cells of the small intestine, proximal
assessable in 310 patients. Among these, tumour response rate, in- renal tubules and salivary glands (Troyer et al., 1995), albeit at levels
cluding complete, partial and minimal response was 46% 100–1000-fold than in tumours (Sokoloff et al., 2000). PSMA is ex-
(Kwekkeboom et al., 2008). The most common acute adverse events pressed in most prostate tumours and associated neovasculature, but
were nausea (25% patients), vomiting (10%), abdominal pain or dis- not on normal vasculature (Chang et al., 1999). This makes it the ideal
comfort (10%), temporary hair loss (62%). Serious delayed toxicities, target for TAT, where alpha radiation with its short range has far
such as liver or renal toxicity were rare. greater endothelial cell toxicity, compared to beta radiation, within the
An interim report of a phase III randomized trial enrolling 229 pa- dose tolerance restriction (Allen et al., 2007; Huang et al., 2012).
tients with midgut neuroendocrine tumours which employed The humanised J591 antibody demonstrates high-affinity binding to
177
Lu-DOTATATE (7.4 GBq every 8 weeks in 4 cycles) vs octreotide PSMA-expressing cells in culture and is rapidly internalized (Liu et al.,
alone showed significantly higher response rate (18% vs 3%) and longer 1997, 1998). J591 demonstrates excellent specificity for prostate
progression-free survival (65.2% vs 10.8% at 20 months) in the cancer xenografts (LNCaP) in mice and for prostate tumours in human.
177
Lu-DOTATATE arm (Strosberg et al., 2017). While the control arm PSMA is further upregulated in LNCaP xenografts following MDV3100
experienced no hematological toxicities, in the beta-RIT group these androgen deprivation therapy (ADT), making PSMA the ideal target
side effects occurred in about 10% of patients. There was no evidence of post-ADT Moreover, PET (89Zr-J591) can be used effectively for se-
renal toxicity during the reported follow-up period. While these pre- quential monitoring of xenograft growth and regression, using 89Zr-
liminary results clearly show a survival benefit with 177Lu-DOTATATE, J591 before and after androgen deprivation (Holland et al., 2010). The
a confirmation will be required by the final trial report. J591 antibody against PSMA demonstrates excellent specificity in the
A critical aspect that has been demonstrated in regards to the clinic for prostate cancer metastases in humans.
177
therapeutic effects of radiolabelled DOTATOC concerns the improved Lu emits a low energy beta (150 keV) with an effective range of
tumour to non-tumour uptake achieved after intra-arterial administra- ∼300 μm (Gledhill, 1973). The trial achieved major decreases in PSA
tion in liver metastatic neuroendocrine tumours (Kratochwil et al., for 67% of subjects, with survival time almost doubling from 12 to 22
2011). Nevertheless, this approach has a shortcoming, as patients with months at the maximum tolerance dose of 2.6 GBq/m2. Overall, a single
recurrences are usually refractory to further treatment with beta ra- dose of 177Lu-J591 was efficacious and well tolerated with reversible
diation therapy. myelosuppression.
Perhaps one of the greatest advantages of alpha-RIT for neu- These results are at the limits of adverse events and do not allow
roendocrine tumours consist of its success in overcoming beta re- higher activities to be administered. Alpha therapy is expected to be
sistance, as an additional treatment line. more efficacious within the tolerance dose. The alpha emitter 213Bi-
The first human study with alpha-RIT was reported by Kratochwil J591 AIC has been tested successfully in vitro and in vivo (Li et al.,
et al. (2014) and employed 213Bi-DOTATOC in seven patients with 2002b), but not in clinical trial. However, alpha and beta RICs have
progressive advanced neuroendocrine liver metastases, which was re- been compared in CRPC patients using the PSMA617 ligand as the
fractory to treatment with 90Y/177Lu-DOTATOC beta emitters. 213Bi- targeting vector (Table 10).
DOTATOC was given in increasing activities in cycles every 2 months
(between 1 and 4 GBq/cycle). Both chronic kidney toxicity and acute 5.8.1. 68Ga-PSMA-617
hematotoxicity were moderate, and even less pronounced than with the Patients with at least 1 lesion suspected of being a prostate cancer
preceding beta therapies. The duration of tumour control with the tumour were injected with this conjugate (Afshar-Oromieh et al.,
earlier 90Y or 177Lu-DOTATOC therapy was up to 3 years and the 2015). At 3 h post-injection, the kidneys and salivary glands showed the
follow-up in patients receiving 213Bi-DOTATOC was reported to be 2 highest uptake in 14 of 19 patients (74%) of the 53 representative tu-
years (Kratochwil et al., 2014). mour lesions at 3 h post-injection. The mean tumour-to-background
Apart from the physical advantages of alpha-emitting isotopes, the ratio for maximum standardized uptake value was 20 ± 17 (range,
molecular mechanisms of tumour damage linked to TAT seem to be an 2.3–84) at 1 h post-injection and 38 ± 39 (range, 3.6–154) at 3 h post-
added reason for their success. A recent study aimed to compare the injection. 68Ga-PSMA-617 shows lesions with high contrast. Images
alpha-emitting 225Ac- and beta emitter 177Lu-labeled somatostatin obtained between 2 and 3 h after injection seem to be the best option
analogue DOTATOC regarding DNA damage induction and the level of for radiotracer uptake and tumour contrast.
cell death (Graf et al., 2014). The group determined the level of γH2AX
as an indicator of DNA double-strand breaks and apoptosis in a neu- 5.8.2. 225Ac vs 177Lu-PSMA-617
roendocrine cell line. Tumours treated with 225Ac-DOTATOC (40 kBq) The Heidelberg trial began in 2014 for prostate cancer patients
showed a higher fraction of cells with γH2AX foci than those treated using either 225Ac or 177Lu labeled PSMA ligand (PSMA617) with pre &
with 177Lu-DOTATOC (30 MBq). The comparative cytotoxicity analysis post-PSMA imaging with 68Ga. 50 patients were administered 130
between the two agents resulted in a factor of 700, which translates into therapies. This trial is ongoing, but early results show superior efficacy
a radiobiological effectiveness of about 5 (Graf et al., 2014). for 225Ac, for which several patients are achieving complete remission,
some of whom had PSA values > 3000 ng/ml (Kratochwil et al.,
5.8. Radio-immunotherapy for castrate resistant prostate cancer (CRPC) 2016a,b).
Patients are stratified for treatment with those with the worst
An important development in prostate cancer therapy has been the prognosis, e.g. bone marrow involvement, visceral and bone metas-
completion of a phase 2 trial of radio-immunotherapy for CRPC patients tases, being assigned to the 225Ac group, while the better patients pri-
using a single dose of the 177Lu-J591 conjugate (Akhtar et al., 2012; marily received 177Lu-PSMA.
Tagawa et al., 2010; Tagawa et al., 2013). The monoclonal antibody The most significant adverse response was xerostomia. The salivary
J591 (Ananias et al., 2009; Bostwick et al., 1998; Mannweiler et al., glands express this PSMA epitope, with 225Ac causing more damage
2009) targets an epitope on the prostate specific membrane antigen than 177Lu. This is a serious and incurable effect, and tends to rule out
(PSMA), which is an integral cell-surface membrane protein expressed the use of 225Ac for earlier applications, such as pre-hormonal therapy.
by most prostate cancer cells. This glandular damage is reduced with 177Lu, but so is the cytotoxicity

17
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

Table 10
Summary of beta-RIT and alpha-RIT trials for CRPC.

Trial/study Vector Target Conclusions/observations

Beta
emitter
Phase 2 for CRPC (Akhtar et al., 2012; J591 PSMA Lu-177 Major decrease in PSA for 67% of subjects, with survival time increasing from 12 to
Tagawa et al., 2010; Tagawa et al., 22 months at the MTD of 2.6 GBq/m2. Overall, a single dose of 177Lu-J591 was
2013) efficacious and well tolerated with reversible myelosuppression at the limits for
Completed adverse events.
Phase 2 for CRPC (Kratochwil et al., 2016a; PSMA617 PSMA617 Lu-177 Patients selected with better prognosis.
Kratochwil et al., 2016b). ligand
Ongoing Reduced efficacy cf Ac225.
Minimal xerostomia in salivary glands.

Alpha
emitter
Phase 2 for CRPC (Kratochwil et al., 2016a; PSMA617 PSMA617 Ac-225 Patients selected with the worst prognosis, e.g. bone marrow involvement, visceral
Kratochwil et al., 2016b) Ongoing ligand and bone metastases. Several patients are achieving complete remission, some of
whom had PSA values > 3000 ng/ml
Major xerostomia in salivary glands.

of targeted cancer cells (Kratochwil et al., 2016b). The PSMA617 ligand Akhtar, N.H., Pail, O., Saran, A., Tyrell, L., Tagawa, S.T., 2012. Prostate-specific mem-
targets a different epitope and has a different biodistribution compared brane antigen-based therapeutics. Adv. Urol. 2012, 973820.
Allen, B., Rizvi, S., Tian, Z., 2001. Preclinical targeted alpha therapy for subcutaneous
with J591, which spares the salivary glands. melanoma. Melanoma Res. 11 (2), 175–182.
Allen, B.J., Raja, C., Rizvi, S., Li, Y., Tsui, W., Graham, P., Thompson, J., Reisfeld, R.,
6. Conclusions Kearsley, J., Morgenstern, A., 2005. Intralesional targeted alpha therapy for meta-
static melanoma. Cancer Biol. Ther. 4 (12), 1318–1324.
Allen, B.J., Raja, C., Rizvi, S., Song, E.Y., Graham, P., 2007. Tumour anti-vascular alpha
This work is a review of the radiobiology, in vitro and in vivo stu- therapy: a mechanism for the regression of solid tumours in metastatic cancer. Phys.
dies of targeted alpha versus beta therapies. In vitro studies and cal- Med. Biol. 52 (13), L15.
Allen, B.J., Singla, A.A., Rizvi, S.M.A., Graham, P., Bruchertseifer, F., Apostolidis, C.,
culations demonstrate the fundamental principles for alpha cytotoxi- Morgenstern, A., 2011. Analysis of patient survival in a Phase I trial of systemic
city: are high specific energy and short range, meaning that cells must targeted α-therapy for metastatic melanoma. Immunotherapy 3 (9), 1041–1050.
be targeted for effective cancer cell kill. While the RBE for 37% survival Allen, B., Marcu, L., Bezak, E., 2016. Targeted alpha therapy for cancer. In: Godfrey, D.J.,
VanDyk, J., Das, S.K., Curran, B.H., Wolbarst, A.A. (Eds.), Advances in Medical
for external alpha vs beta irradiation is ∼ 5, the RBE for internal tar-
Physics. Medical Physics Publishing.
geted alphas relative to betas and non-targeted alphas is close to 120. Allen, B., 1999. Can alpha-immunotherapy succeed where other systemic modalities have
The difference is given by the fact that the range of alphas is very much failed? Nucl. Med. Commun. 20, 3.
smaller than for betas, such that most targeted cancer cells receive a Allen, B., 2017. A comparative evaluation of Ac225 vs Bi213 as therapeutic radioisotopes
for targeted alpha therapy. Aust. Phys. Eng. Sci. Med. 40 (4). http://dx.doi.org/10.
large fraction of the short-range alpha energy but a very, very small 1007/s13246-017-0534-6.
fraction of the long-range beta energy. American Cancer Society, I., 2015. Global Cancer Facts & Figures, 3rd edition. American
Comparing the biological effects of alpha and beta radio- Cancer Society, Inc., Atlanta.
Ananias, H.J., van den Heuvel, M.C., Helfrich, W., de Jong, I.J., 2009. Expression of the
immunotherapy is not a straightforward task, as various factors such as: gastrin-releasing peptide receptor, the prostate stem cell antigen and the prostate-
dose rate, protracted irradiation, associated emission by daughter specific membrane antigen in lymph node and bone metastases of prostate cancer.
radionuclides, chelator stability, etc. may interfere with the analysis Prostate 69 (10), 1101–1108.
Andersson, H., Cederkrantz, E., Bäck, T., Divgi, C., Elgqvist, J., Himmelman, J., Horvath,
(Brady et al., 2013; Pouget et al., 2015). Nevertheless, the physical and G., Jacobsson, L., Jensen, H., Lindegren, S., 2009. Intraperitoneal α-particle radio-
radiobiological properties of α-RIT, as well as clinical trials demon- immunotherapy of ovarian cancer patients: pharmacokinetics and dosimetry of
strate an advantage of the alpha emitting isotopes: 211At-MX35F (abα) 2′a phase I study. J. Nucl. Med. 50 (7), 1153–1160.
Barendsen, G., Beusker, T., AJ, V., Budke, L., 1960. Effects of different ionizing radiations
on human cells in tissue culture: II. Biological experiments. Rad. Res. 13 (6),
(1) in a hypoxic environment, due to minimal dependence on the 841–849.
oxygenation status of alpha particles, Behr, T.M., Behe, M., Stabin, M.G., Wehrmann, E., Apostolidis, C., Molinet, R., Strutz, F.,
Fayyazi, A., Wieland, E., Gratz, S., Koch, L., Goldenberg, D.M., Becker, W., 1999.
(2) overcoming treatment resistance to beta targeted therapy,
High-linear energy transfer (LET) alpha versus low-LET beta emitters in radio-
(3) of a more focused targeting which makes it ideal for isolated cancer immunotherapy of solid tumors: therapeutic efficacy and dose-limiting toxicity of
cells and residual disease, 213Bi- versus 90Y-labeled CO17-1A Fab' fragments in a human colonic cancer model.
(4) in anti-vascular targeted therapies. Cancer Res. 59 (11), 2635–2643.
Bigner, D.D., Brown, M.T., Friedman, A.H., Coleman, R.E., Akabani, G., Friedman, H.S.,
Thorstad, W.L., McLendon, R.E., Bigner, S.H., Zhao, X.G., Pegram, C.N., Wikstrand,
While the number of preclinical and clinical studies to date is rather C.J., Herndon 2nd, J.E., Vick, N.A., Paleologos, N., Cokgor, I., Provenzale, J.M.,
small, α-RIT or β-RIT studies for the above cancers and tumour sites Zalutsky, M.R., 1998. Iodine-131-labeled antitenascin monoclonal antibody 81C6
treatment of patients with recurrent malignant gliomas: phase I trial results. J. Clin.
show that in all the reviewed cases, α-RIT is superior to β-RIT in terms Oncol. 16 (6), 2202–2212.
of efficacy within the tolerance dose. Bloomer, W.D., McLaughlin, W.H., Lambrecht, R.M., Atcher, R.W., Mirzadeh, S., Madara,
J.L., Milius, R.A., Zalutsky, M.R., Adelstein, S.J., Wolf, A.P., 1984. 211At radiocolloid
therapy: further observations and comparison with radiocolloids of 32P, 165Dy, and
References 90Y. Int. J. Radiat. Oncol. Biol. Phys. 10 (3), 341–348.
Bodet-Milin, C., Kraeber-Bodéré, F., Eugène, T., Guérard, F., Gaschet, J., Bailly, C., 2016.
Adelstein, S., Kassis, A., Bodei, L., Mariani, G., 2003. Radiotoxicity of iodine-125 and Radioimmunoterapy for treatment of acute leukemia. Semin. Nucl. Med. 46 (2),
other Auger-electron-emitting radionuclides: background to therapy. Cancer Biother. 135–146.
Radiopharm. 18, 16. Bostwick, D.G., Pacelli, A., Blute, M., Roche, P., Murphy, G.P., 1998. Prostate specific
Afshar-Oromieh, A., Hetzheim, H., Kratochwil, C., Benesova, M., Eder, M., Neels, O.C., membrane antigen expression in prostatic intraepithelial neoplasia and adenocarci-
Eisenhut, M., Kubler, W., Holland-Letz, T., Giesel, F.L., Mier, W., Kopka, K., noma: a study of 184 cases. Cancer 82 (11), 2256–2261.
Haberkorn, U., 2015. The theranostic PSMA ligand PSMA-617 in the diagnosis of Boudousq, V., Bobyk, L., Busson, M., Garambois, V., Jarlier, M., Charalambatou, P., et al.,
prostate cancer by PET/CT: biodistribution in humans, radiation dosimetry, and first 2013. Comparison between internalizing anti-HER2 mAbs and non-internalizing anti-
evaluation of tumor lesions journal of nuclear medicine: official publication. Soc. CEA mAbs in alpha-radioimmunotherapy of small volume peritoneal carcinomatosis
Nucl. Med. 56 (11), 1697–1705. using 212Pb. PLoS One 8 (7 (Jul 29)), e69613.

18
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

Brady, D., O'Sullivan, J.M., Prise, K.M., 2013. What is the role of the bystander response 10-triacetic acid-substance P. Clin. Cancer Res. 12 (12), 3843–3850.
in radionuclide therapies? Front. Oncol. 3 (Aug 19), 215. Kratochwil, C., López-Benítez, R., Mier, W., Haufe, S., Isermann, B., Kauczor, H.-U.,
Bumol, T., Reisfeld, R., 1982. Unique glycoprotein-proteoglycan complex defined by Choyke, P.L., Haberkorn, U., Giesel, F.L., 2011. Hepatic arterial infusion enhances
monoclonal antibody on human melanoma cells. Proc. Natl. Acad. Sci. 79 (4), DOTATOC radiopeptide therapy in patients with neuroendocrine liver metastases.
1245–1249. Endocr. Relat. Cancer 18 (5), 595–602.
Caron, P., Jurcic, J., Scott, A., Fin, R., Digvi, C., Graham, M., Jureidini, I., Sgouros, G., Kratochwil, C., Giesel, F., Bruchertseifer, F., Mier, W., Apostolidis, C., Boll, R., Murphy,
Tyson, D., Old, L., 1994. A phase IB trial of humanized antibody M195 (anti-CD33) in K., Haberkorn, U., Morgenstern, A., 2014. 213Bi-DOTATOC receptor-targeted alpha-
myeloid leukemia: specific targeting without immunogenicity. Blood 83, 1760–1768. radionuclide therapy induces remission in neuroendocrine tumours refractory to beta
Chang, S.S., Reuter, V.E., Heston, W.D., Bander, N.H., Grauer, L.S., Gaudin, P.B., 1999. radiation: a first-in-human experience. Eur. J. Nucl. Med. Mol. Imaging 41 (11),
Five different anti-prostate-specific membrane antigen (PSMA) antibodies confirm 2106–2119.
PSMA expression in tumor-associated neovasculature. Cancer Res. 59 (13), Kratochwil, C., Bruchertseifer, F., Giesel, F., Apostolidis, C., Haberkorn, U., Morgenstern,
3192–3198. A., 2015. Ac-225-DOTATOC – an empiric dose finding for alpha particle emitter
Cordier, D., Forrer, F., Bruchertseifer, F., Morgenstern, A., Apostolidis, C., Good, S., based radionuclide therapy of neuroendocrine tumors. J. Nucl. Med. 56, 1232.
Müller-Brand, J., Mäcke, H., Reubi, J., Merlo, A., 2010. Targeted alpha-radionuclide Kratochwil, C., Bruchertseifer, F., Giesel, F.L., Weis, M., Verburg, F.A., Mottaghy, F.,
therapy of functionally critically located gliomas with 213Bi-DOTA-[Thi8, Met (O2) Kopka, K., Apostolidis, C., Haberkorn, U., Morgenstern, A., 2016a. 225Ac-PSMA-617
11]-substance P: a pilot trial. Eur. J. Nucl. Med. Mol. Imaging 37 (7), 1335–1344. for PSMA-Targeted alpha-Radiation therapy of metastatic castration-Resistant pros-
Dadachova, E., Revskaya, E., Sesay, M.A., Damania, H., Boucher, R., Sellers, R.S., Howell, tate cancer journal of nuclear medicine: official publication. J. Nucl. Med. Off. Publ.
R.C., Burns, L., Thornton, G.B., Natarajan, A., Mirick, G.R., DeNardo, S.J., DeNardo, Soc. Nucl. Med. 57 (12), 1941–1944.
G.L., Casadevall, A., 2008. Pre-clinical evaluation and efficacy studies of a melanin- Kratochwil, C., Giesel, F., Rius, M., Apostolidis, C., Haberkorn, U., Morgenstern, A.,
binding IgM antibody labeled with 188Re against experimental human metastatic 2016b. Ac-225-PSMA617 – a single center experience of 40 patients receiving PSMA-
melanoma in nude mice. Cancer Biol. Ther. 7 (7), 1116–1127. targeted alpha therapy. Deutsche Gesellschaft Fur Nuklearmedizin V68.
Derrien, A., Gouard, S., Maurel, C., Gaugler, M.H., Bruchertseifer, F., Morgenstern, A., Kwekkeboom, D.J., de Herder, W.W., Kam, B.L., van Eijck, C.H., van Essen, M., Kooij,
Faivre-Chauvet, A., Classe, J.M., Cherel, M., 2015. Therapeutic efficacy of alpha-RIT P.P., Feelders, R.A., van Aken, M.O., Krenning, E.P., 2008. Treatment with the
using a (213)Bi-Anti-hCD138 antibody in a mouse model of ovarian peritoneal car- radiolabeled somatostatin analog [177Lu-DOTA0, Tyr3] octreotate: toxicity, efficacy,
cinomatosis. Front. Med. (Lausanne) 2, 88. and survival. J. Clin. Oncol. 26 (13), 2124–2130.
Epenetos, A.A., Hird, V., Lambert, H., Mason, P., Coulter, C., 2000. Long term survival of Larson, S.M., Carrasquillo, J.A., Mcguffin, R.W., Krohn, K.A., Ferens, J.M., Hill, L.D.,
patients with advanced ovarian cancer treated with intraperitoneal radio- Beaumier, P.L., Reynolds, J.C., Hellstrom, K.E., Hellstrom, I., 1985. Use of I-131 la-
immunotherapy. Int. J. Gynecol. Cancer 10 (S1), 44–46. beled, murine fab against a high molecular-weight antigen of human-melanoma –
Fichou, N., Gouard, S., Maurel, C., Barbet, J., Ferrer, L., Morgenstern, A., Bruchertseifer, preliminary experience. Radiology 155 (2), 487–492.
F., Faivre-Chauvet, A., Bigot-Corbel, E., Davodeau, F., 2015. Single-dose anti-CD138 Leins, A., Riva, P., Lindstedt, R., Davidoff, M.S., Mehraein, P., Weis, S., 2003. Expression
radioimmunotherapy: bismuth-213 is more efficient than lutetium-177 for treatment of tenascin-C in various human brain tumors and its relevance for survival in patients
of multiple myeloma in a preclinical model. Front. Med. 2. with astrocytoma. Cancer 98 (11), 2430–2439.
Forrer, F., Waldherr, C., Maecke, H.R., Mueller-Brand, J., 2006. Targeted radionuclide Li, Y., Rizvi, S., Ranson, M., Allen, B., 2002a. 213Bi-PAI2 conjugate selectively induces
therapy with 90Y-DOTATOC in patients with neuroendocrine tumors. Anticancer apoptosis in PC3 metastatic prostate cancer cell line and shows anti-cancer activity in
Res. 26 (1B), 703–707. a xenograft animal model. Brit. J. Cancer 86 (7), 1197–1203.
Gledhill, J., 1973. The range-energy relation for 0.1-600 keV electrons. J. Phys. A: Math. Li, Y., Tian, Z., Rizvi, S., Bander, N., Allen, B., 2002b. In vitro and preclinical targeted
Nucl. Gen. 6 (9), 1420. alpha therapy of human prostate cancer with Bi-213 labeled J591 antibody against
Graf, F., Fahrer, J., Maus, S., Morgenstern, A., Bruchertseifer, F., Venkatachalam, S., the prostate specific membrane antigen. Pros Ca Pros Dis 5, 36–46.
Fottner, C., Weber, M.M., Huelsenbeck, J., Schreckenberger, M., Kaina, B., Miederer, Liu, H., Moy, P., Kim, S., Xia, Y., Rajasekaran, A., Navarro, V., Knudsen, B., Bander, N.H.,
M., 2014. DNA double strand Breaks as predictor of efficacy of the alpha-particle 1997. Monoclonal antibodies to the extracellular domain of prostate-specific mem-
emitter Ac-225 and the electron emitter Lu-177 for somatostatin receptor targeted brane antigen also react with tumor vascular endothelium. Cancer Res. 57 (17),
radiotherapy. PLoS One 9 (2), e88239. 3629–3634.
Holland, J., Divilov, V., Bander, N., Smith-Jones, P., Larson, S., Lewis, J., 2010. Zr-89-dfo- Liu, H., Rajasekaran, A.K., Moy, P., Xia, Y., Kim, S., Navarro, V., Rahmati, R., Bander,
j591 for immuno-PET of prostate-specific membrane antigen expression in vivo. J. N.H., 1998. Constitutive and antibody-induced internalization of prostate-specific
Nucl. Med. 51 (8), 1193–1230. membrane antigen. Cancer Res. 58 (18), 4055–4060.
Hoskin, P., Sartor, O., O'Sullivan, J.M., Johannessen, D.C., Helle, S.I., Logue, J., Ménager, J., Gorin, J.B., Maurel, C., Drujont, L., Gouard, S., Louvet, C., et al., 2015.
Bottomley, D., Nilsson, S., Vogelzang, N.J., Fang, F., 2014. Efficacy and safety of Combining é-Radioimmunotherapy and adoptive t cell therapy to potentiate tumor
radium-223 dichloride in patients with castration-resistant prostate cancer and destruction. PLoS One 10 (6), e0130249.
symptomatic bone metastases, with or without previous docetaxel use: a prespecified Mannweiler, S., Amersdorfer, P., Trajanoski, S., Terrett, J.A., King, D., Mehes, G., 2009.
subgroup analysis from the randomised, double-blind, phase 3 ALSYMPCA trial. Heterogeneity of prostate-specific membrane antigen (PSMA) expression in prostate
Lancet Oncol. 15 (12), 1397–1406. carcinoma with distant metastasis. Pathol. Oncol. Res. 15 (2), 167–172.
Huang, C.-Y., Oborn, B.M., Guatelli, S., Allen, B.J., 2012. Monte Carlo calculation of the Marcu, L., Bezak, E., Allen, B.J., 2012. Biomedical Physics in Radiotherapy for Cancer.
maximum therapeutic gain of tumor antivascular alpha therapy. Med. Phys. 39 (3), Csiro publishing.
1282–1288. Matthews, D.C., Appelbaum, F.R., Eary, J.F., Fisher, D.R., Durack, L.D., Hui, T.E., Martin,
Israeli, R.S., Powell, C.T., Fair, W.R., Heston, W.D., 1993. Molecular cloning of a com- P.J., Mitchell, D., Press, O.W., Storb, R., Bernstein, I.D., 1999. Phase I study of (131)I-
plementary DNA encoding a prostate-specific membrane antigen. Cancer Res. 53 (2), anti-CD45 antibody plus cyclophosphamide and total body irradiation for advanced
227–230. acute leukemia and myelodysplastic syndrome. Blood 94 (4), 1237–1247.
Israeli, R.S., Powell, C.T., Corr, J.G., Fair, W.R., Heston, W.D., 1994. Expression of the Mawad, R., Gooley, T.A., Rajendran, J.G., Fisher, D.R., Gopal, A.K., Shields, A.T.,
prostate-specific membrane antigen. Cancer Res. 54 (7), 1807–1811. Sandmaier, B.M., Sorror, M.L., Deeg, H.J., Storb, R., Green, D.J., Maloney, D.G.,
Jurcic, J.G., Rosenblat, T.L., 2014. Targeted alpha-particle immunotherapy for acute Appelbaum, F.R., Press, O.W., Pagel, J.M., 2014. Radiolabeled anti-CD45 antibody
myeloid leukemia. American Society of Clinical Oncology educational book/ASCO. with reduced-intensity conditioning and allogeneic transplantation for younger pa-
Am. Soc. Clin. Oncol. Meet. e126–e131. tients with advanced acute myeloid leukemia or myelodysplastic syndrome. Biol.
Jurcic, J., Divgi, C., McDevitt, M., 2000. Potential for myeloablation with yttrium-90-Hu Blood Marrow Transplant. 20 (9), 1363–1368.
(anti-CD33) in myeloid leukemia. Proc. Am. Soc. Clin. Oncol. 19 (8), M195. Meredith, R.F., Torgue, J., Azure, M.T., Shen, S., Saddekni, S., Banaga, E., Carlise, R.,
Jurcic, J.G., Larson, S.M., Sgouros, G., McDevitt, M.R., Finn, R.D., Divgi, C.R., Ballangrud, Bunch, P., Yoder, D., Alvarez, R., 2014. Pharmacokinetics and imaging of 212Pb-
Å.M., Hamacher, K.A., Ma, D., Humm, J.L., 2002. Targeted α particle im- TCMC-trastuzumab after intraperitoneal administration in ovarian cancer patients.
munotherapy for myeloid leukemia. Blood 100 (4), 1233–1239. Cancer Biother. Radiopharm. 29 (1), 12–17.
Jurcic, J., Rosenblat, T., McDevitt, M., 2011. Phase I trial of the targeted alpha-Particle Meredith, R.F., Torgue, J.J., Rozgaja, T.A., Banaga, E.P., Bunch, P.W., Alvarez, R.D.,
nano-Generator actinium-225(225Ac)-Lintuzumab (anti-CD33; HuM195) In acute Straughn Jr., J.M., Dobelbower, M.C., Lowy, A.M., 2016. Safety and outcome mea-
myeloid leukemia (AML). Blood 2011, 1. sures of first-in-Human intraperitoneal alpha radioimmunotherapy with 212Pb-
Kam, B.L., Teunissen, J.J., Krenning, E.P., de Herder, W.W., Khan, S., van Vliet, E.I., TCMC-Trastuzumab. Am. J. Clin. Oncol. http://dx.doi.org/10.1097/COC.
Kwekkeboom, D.J., 2012. Lutetium-labelled peptides for therapy of neuroendocrine 0000000000000353. (epub ahead of print).
tumours. Eur. J. Nucl. Med. Mol. Imaging 39 (Suppl 1), S103–112. Miederer, M., Henriksen, G., Alke, A., Mossbrugger, I., Quintanilla-Martinez, L.,
Kang, H., Lee, S., Byun, B., Kim, K., Lim, I., Choi, C., Suh, C., Kim, W., Nam, S., Lee, S., Senekowitsch-Schmidtke, R., Essler, M., 2008. Preclinical evaluation of the alpha-
Eom, H., Shin, D., Lim, S., 2013. Repeated radioimmunotherapy with (131)I-ritux- particle generator nuclide 225Ac for somatostatin receptor radiotherapy of neu-
imab for patients with low-grade and aggressive relapsed or refractory B cell non- roendocrine tumors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 14 (11),
Hodgkin lymphoma. Cancer Chemother. Pharmacol. 71 (4), 945–953. 3555–3561.
Klein, M., Lotem, M., Peretz, T., Zwas, S.T., Mizrachi, S., Liberman, Y., Chisin, R., Morgenstern, A., Krolicki, L., Kunikowska, J., Koziara, H., Krolicki, B., Jakucinski, M.,
Schachter, J., Ron, I.G., Iosilevsky, G., Kennedy, J.A., Revskaya, E., de Kater, A.W., Apostolidis, C., Bruchertseifer, F., 2014. Targeted alpha therapy of glioblastoma
Banaga, E., Klutzaritz, V., Friedmann, N., Galun, E., Denardo, G.L., Denardo, S.J., multiforme: first clinical experience with 213Bi-substance P. J. Nucl. Med. 55 (Suppl.
Casadevall, A., Dadachova, E., Thornton, G.B., 2013. Safety and efficacy of 188- 1), 390.
rhenium-labeled antibody to melanin in patients with metastatic melanoma. J. Skin Mulford, D., Scheinberg, D., Jurcic, J., 2005. The promise of targeted (American Cancer
Cancer 828329. Society)-particle therapy Journal of nuclear medicine: official publication. Soc. Nucl.
Kneifel, S., Cordier, D., Good, S., Ionescu, M.C., Ghaffari, A., Hofer, S., Kretzschmar, M., Med. 46 (Suppl. 1), 199S–204S.
Tolnay, M., Apostolidis, C., Waser, B., 2006. Local targeting of malignant gliomas by Nilsson, S., Larsen, R.H., Fosså, S.D., Balteskard, L., Borch, K.W., Westlin, J.-E., Salberg,
the diffusible peptidic vector 1, 4, 7, 10-tetraazacyclododecane-1-glutaric acid-4, 7, G., Bruland, Ø.S., 2005. First clinical experience with α-emitting radium-223 in the

19
L. Marcu et al. Critical Reviews in Oncology / Hematology 123 (2018) 7–20

treatment of skeletal metastases. Clin. Cancer Res. 11 (12), 4451–4459. Schmidtke, R., 2011. Lu-177-immunotherapy of experimental peritoneal carcino-
Nilsson, S., Franzén, L., Parker, C., Tyrrell, C., Blom, R., Tennvall, J., Lennernäs, B., matosis shows comparable effectiveness to Bi-213-immunotherapy, but causes toxi-
Petersson, U., Johannessen, D.C., Sokal, M., 2007. Bone-targeted radium-223 in city not observed with Bi-213. Eur. J. Nucl. Med. Mol. 38 (2), 312–322.
symptomatic, hormone-refractory prostate cancer: a randomised, multicentre, pla- Serafini, A.N., Houston, S.J., Resche, I., Quick, D.P., Grund, F.M., Ell, P.J., Bertrand, A.,
cebo-controlled phase II study. Lancet Oncol. 8 (7), 587–594. Ahmann, F.R., Orihuela, E., Reid, R.H., Lerski, R.A., Collier, B.D., McKillop, J.H.,
Nilsson, S., 2014. Alpha-emitter radium-223 in the management of solid tumors: current Purnell, G.L., Pecking, A.P., Thomas, F.D., Harrison, K.A., 1998. Palliation of pain
status and future directions. American Society of Clinical Oncology educational associated with metastatic bone cancer using samarium-153 lexidronam: a double-
book/ASCO. Am. Soc. Clin. Oncol. Meet. e132–e139. blind placebo-controlled clinical trial. J. Clin. Oncol. 16 (4), 1574–1581.
Oosterhof, G.O., Roberts, J.T., de Reijke, T.M., Engelholm, S.A., Horenblas, S., von der Sokoloff, R.L., Norton, K.C., Gasior, C.L., Marker, K.M., Grauer, L.S., 2000. A dual-
Maase, H., Neymark, N., Debois, M., Collette, L., 2003. Strontium(89) chloride versus monoclonal sandwich assay for prostate-specific membrane antigen: levels in tissues,
palliative local field radiotherapy in patients with hormonal escaped prostate cancer: seminal fluid and urine. Prostate 43 (2), 150–157.
a phase III study of the European Organisation for Research and Treatment of Cancer, Song, H., Hobbs, R.F., Vajravelu, R., Huso, D.L., Esaias, C., Apostolidis, C., Morgenstern,
Genitourinary Group. Eur. Urol. 44 (5), 519–526. A., Sgouros, G., 2009. Radioimmunotherapy of breast cancer metastases with alpha-
Parker, C.C., Pascoe, S., Chodacki, A., O'Sullivan, J.M., Germá, J.R., O’Bryan-Tear, C.G., particle emitter 225Ac: comparing efficacy with 213Bi and 90Y. Cancer Res. 69 (23),
Haider, T., Hoskin, P., 2013. A randomized, double-blind, dose-finding, multicenter, 8941–8948.
phase 2 study of radium chloride (Ra 223) in patients with bone metastases and Strosberg, J., El-Haddad, G., Wolin, E., Hendifar, A., Yao, J., Chasen, B., Mittra, E., Kunz,
castration-resistant prostate cancer. Eur. Urol. 63 (2), 189–197. P.L., Kulke, M.H., Jacene, H., Bushnell, D., O'Dorisio, T.M., Baum, R.P., Kulkarni,
Porter, A.T., McEwan, A.J., Powe, J.E., Reid, R., McGowan, D.G., Lukka, H., H.R., Caplin, M., Lebtahi, R., Hobday, T., Delpassand, E., Van Cutsem, E., Benson, A.,
Sathyanarayana, J.R., Yakemchuk, V.N., Thomas, G.M., Erlich, L.E., Crook, J., 1993. Srirajaskanthan, R., Pavel, M., Mora, J., Berlin, J., Grande, E., Reed, N., Seregni, E.,
Results of a randomized phase-III trial to evaluate the efficacy of strontium-89 ad- Oberg, K., Lopera Sierra, M., Santoro, P., Thevenet, T., Erion, J.L., Ruszniewski, P.,
juvant to local field external beam irradiation in the management of endocrine re- Kwekkeboom, D., Krenning, E., Investigators, N.-T., 2017. Phase 3 trial of 177Lu-
sistant metastatic prostate cancer. Int. J. Radiat. Oncol. Biol. Phys. 25 (5), 805–813. Dotatate for midgut neuroendocrine tumors. N. Engl. J. Med. 376 (2), 125–135.
Pouget, J.P., Lozza, C., Deshayes, E., Boudousq, V., Navarro-Teulon, I., 2015. Introduction Sweat, S., Pacelli, A., Murphy, G., Bostwick, D., 1998. Prostate-specific membrane antigen
to radiobiology of targeted radionuclide therapy. Front. Med. (Lausanne) 17 (2), 12. expression is greatest in prostate adenocarcinoma and lymph node metastases.
Quilty, P.M., Kirk, D., Bolger, J.J., Dearnaley, D.P., Lewington, V.J., Mason, M.D., Reed, Urology 52 (4), 637–640.
N.S., Russell, J.M., Yardley, J., 1994. A comparison of the palliative effects of Tagawa, S.T., Beltran, H., Vallabhajosula, S., Goldsmith, S.J., Osborne, J., Matulich, D.,
strontium-89 and external beam radiotherapy in metastatic prostate cancer. Petrillo, K., Parmar, S., Nanus, D.M., Bander, N.H., 2010. Anti-prostate-specific
Radiother. Oncol. 31 (1), 33–40. membrane antigen-based radioimmunotherapy for prostate cancer. Cancer 116 (4
Raja, C., Graham, P., Rizvi, S., Song, E., Goldsmith, H., Thompson, J., Bosserhoff, A., Suppl), 1075–1083.
Morgenstern, A., Apostolidis, C., Kearsley, J., 2007. Interim analysis of toxicity and Tagawa, S.T., Milowsky, M.I., Morris, M., Vallabhajosula, S., Christos, P., Akhtar, N.H.,
response in phase 1 trial of systemic targeted alpha therapy for metastatic melanoma. Osborne, J., Goldsmith, S.J., Larson, S., Taskar, N.P., 2013. Phase II study of lutetium-
Cancer Biol. Ther. 6 (6), 846–852. 177?Labeled anti-Prostate-Specific membrane antigen monoclonal antibody J591 for
Reardon, D., Akabani, G., Coleman, R., Friedman, A., Friedman, H., Herndon, J., 2002. metastatic castration-Resistant prostate cancer. Clin. Cancer Res. 19 (18),
Phase II trial of murine (131)I-labeled antitenascin monoclonal antibody 81C6 ad- 5182–5191.
ministered into surgically created resection cavities of patients with newly diagnosed Thompson, S., Ballard, B., Jiang, Z., Revskaya, E., Sisay, N., Miller, W.H., Cutler, C.S.,
malignant gliomas. J. Clin. Oncol. 20 (5), 1389–1397. Dadachova, E., Francesconi, L.C., 2014. 166Ho and 90Y labeled 6D2 monoclonal
Ringhoffer, M., Blumstein, N., Neumaier, B., Glatting, G., von Harsdorf, S., Buchmann, I., antibody for targeted radiotherapy of melanoma: comparison with 188Re radiolabel.
Wiesneth, M., Kotzerke, J., Zenz, T., Buck, A., Schauwecker, P., Stilgenbauer, S., Nucl. Med. Biol. 41 (3), 276–281.
Döhner, H., Reske, S., Bunjes, D., 2005. 188Re or 90Y-labelled anti-CD66 antibody as Troyer, J.K., Beckett, M.L., Wright Jr., G.L., 1995. Detection and characterization of the
part of a dose-reduced conditioning regimen for patients with acute leukaemia or prostate-specific membrane antigen (PSMA) in tissue extracts and body fluids. Int. J.
myelodysplastic syndrome over the age of 55: results of a phase I-II study. Br. J. Cancer 62 (5), 552–558.
Haematol. 130 (4), 604–613. Tu, S.M., Millikan, R.E., Mengistu, B., Delpassand, E.S., Amato, R.J., Pagliaro, L.C.,
Riquier, H., Wera, A., Heuskin, A., Feron, O., Lucas, S., Michiels, C., 2013. Comparison of Daliani, D., Papandreou, C.N., Smith, T.L., Kim, J., Podoloff, D.A., Logothetis, C.J.,
X-ray and alpha particle effects on a human cancer and endothelial cells: survival 2001. Bone-targeted therapy for advanced androgen-independent carcinoma of the
curves and gene expression profiles. Radiother. Oncol. 106, 397–403. prostate: a randomised phase II trial. Lancet 357 (9253), 336–341.
Riva, P., Franceschi, G., Frattarelli, M., 1999. Loco-regional radioimmunotherapy of high- Verheijen, R.H., Massuger, L.F., Benigno, B.B., Epenetos, A.A., Lopes, A., Soper, J.T.,
grade malignant gliomas using specific monoclonal antibodies labeled with 90Y: a Markowska, J., Vyzula, R., Jobling, T., Stamp, G., Spiegel, G., Thurston, D., Falke, T.,
phase I study. Clin. Cancer Res. 5 (Suppl), 3275s–3280s. Lambert, J., Seiden, M.V., 2006. Phase III trial of intraperitoneal therapy with yt-
Rizvi, S., Sarkar, S., Goozee, G., Allen, B., 2000. Radioimmunoconjugates for targeted trium-90-labeled HMFG1 murine monoclonal antibody in patients with epithelial
[alpha] therapy of malignant melanoma. Melanoma Res. 10 (3), 281–289. ovarian cancer after a surgically defined complete remission. J. Clin. Oncol. 24 (4),
Rizvi, S., Henniker, A., Goozee, G., Allen, B., 2002. In vitro testing of the leukaemia 571–578.
monoclonal antibody WM-53 labeled with alpha and beta emitting radioisotopes. Waldherr, C., Pless, M., Maecke, H., Haldemann, A., Mueller-Brand, J., 2001. The clinical
Leuk. Res. 26 (1), 37–43. value of [90Y-DOTA]-D-Phe1-Tyr3-octreotide (90Y-DOTATOC) in the treatment of
Rosenblat, T.L., McDevitt, M.R., Mulford, D.A., Pandit-Taskar, N., Divgi, C.R., Panageas, neuroendocrine tumours: a clinical phase II study. Ann. Oncol. 12 (7), 941–945.
K.S., Heaney, M.L., Chanel, S., Morgenstern, A., Sgouros, G., Larson, S.M., Wild, D., Frischknecht, M., Zhang, H., Morgenstern, A., Bruchertseifer, F., Boisclair, J.,
Scheinberg, D.A., Jurcic, J.G., 2010. Sequential cytarabine and alpha-particle im- Provencher-Bolliger, A., Reubi, J.C., Maecke, H.R., 2011. Alpha- versus beta-particle
munotherapy with bismuth-213-lintuzumab (HuM195) for acute myeloid leukemia. radiopeptide therapy in a human prostate cancer model (213Bi-DOTA-PESIN and
Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 16 (21), 5303–5311. 213Bi-AMBA versus 177Lu-DOTA-PESIN). Cancer Res. 71 (3), 1009–1018.
Rosenblum, M.G., Verschraegen, C.F., Murray, J.L., Kudelka, A.P., Gano, J., Cheung, L., Wright Jr., G.L., Grob, B.M., Haley, C., Grossman, K., Newhall, K., Petrylak, D., Troyer, J.,
Kavanagh, J.J., 1999. Phase I study of 90Y-labeled B72. 3 intraperitoneal adminis- Konchuba, A., Schellhammer, P.F., Moriarty, R., 1996. Upregulation of prostate-
tration in patients with ovarian cancer: effect of dose and EDTA coadministration on specific membrane antigen after androgen-deprivation therapy. Urology 48 (2),
pharmacokinetics and toxicity. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 5 (5), 326–334.
953–961. Yong, K.J., Milenic, D.E., Baidoo, K.E., Brechbiel, M.W., 2014. Impact of α-targeted ra-
Sartor, O., Coleman, R., Nilsson, S., Heinrich, D., Helle, S.I., O'Sullivan, J.M., Fosså, S.D., diation therapy on gene expression in a pre-clinical model for disseminated perito-
Chodacki, A., Wiechno, P., Logue, J., 2014. Effect of radium-223 dichloride on neal disease when combined with paclitaxel. PLoS One 9 (Sep 30), e108511.
symptomatic skeletal events in patients with castration-resistant prostate cancer and Yong, K.J., Milenic, D.E., Baidoo, K.E., Brechbiel, M.W., 2016. Cell killing mechanisms
bone metastases: results from a phase 3, double-blind, randomised trial. Lancet and impact on gene expression by gemcitabine and 212Pb-Trastuzumab treatment in
Oncol. 15 (7), 738–746. a disseminated i.p. tumor model. PLoS One 11 (7 (Jul 28)), e0159904.
Scheinberg, D.A., Lovett, D., Divgi, C.R., Graham, M.C., Berman, E., Pentlow, K., Feirt, N., Yoshida, K., Kaneta, T., Takano, S., Sugiura, M., Kawano, T., Hino, A., Yamamoto, T.,
Finn, R.D., Clarkson, B.D., Gee, T.S., et al., 1991. A phase I trial of monoclonal an- Shizukuishi, K., Kaneko, M., Zurth, C., Inoue, T., 2016. Pharmacokinetics of single
tibody M195 in acute myelogenous leukemia: specific bone marrow targeting and dose radium-223 dichloride (BAY 88–8223) in Japanese patients with castration-re-
internalization of radionuclide. J. Clin. Oncol. 9 (3), 478–490. sistant prostate cancer and bone metastases. Ann. Nucl. Med. 30 (7), 453–460.
Schwartz, M.A., Lovett, D.R., Redner, A., Finn, R.D., Graham, M.C., Divgi, C.R., Dantis, L., Zalutsky, M.R., Reardon, D.A., Akabani, G., Coleman, R.E., Friedman, A.H., Friedman,
Gee, T.S., Andreeff, M., Old, L.J., et al., 1993. Dose-escalation trial of M195 labeled H.S., McLendon, R.E., Wong, T.Z., Bigner, D.D., 2008. Clinical experience with α-
with iodine 131 for cytoreduction and marrow ablation in relapsed or refractory particle?emitting 211At: treatment of recurrent brain tumor patients with 211At-la-
myeloid leukemias. J. Clin. Oncol. 11 (2), 294–303. beled chimeric antitenascin monoclonal antibody 81C6. J. Nucl. Med. 49 (1), 30–38.
Seidl, C., Zockler, C., Beck, R., Quintanilla-Martinez, L., Bruchertseifer, F., Senekowitsch-

20

Das könnte Ihnen auch gefallen