Next Article in Journal
A Systematic Pan-Cancer Analysis of Genetic Heterogeneity Reveals Associations with Epigenetic Modifiers
Next Article in Special Issue
Interventional Techniques for the Management of Cancer-Related Pain: Clinical and Critical Aspects
Previous Article in Journal
Immunohistochemical Expression of Autophagy-Related Proteins in Advanced Tubular Gastric Adenocarcinomas and Its Implications
Previous Article in Special Issue
Cancer-Related Neuropathic Pain
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Personalized Radiation Therapy in Cancer Pain Management

1
Department of Oncology, Medical University of Bialystok, 15-027 Białystok, Poland
2
Department of Radiation Therapy, Comprehensive Cancer Center of Białystok, 15-027 Bialystok, Poland
3
Student Scientific Association Affiliated with Department of Oncology, Medical University of Bialystok, 15-027 Bialystok, Poland
*
Author to whom correspondence should be addressed.
Cancers 2019, 11(3), 390; https://doi.org/10.3390/cancers11030390
Submission received: 3 February 2019 / Revised: 5 March 2019 / Accepted: 11 March 2019 / Published: 19 March 2019
(This article belongs to the Special Issue Cancer Pains)

Abstract

:
The majority of advanced cancer patients suffer from pain, which severely deteriorates their quality of life. Apart from analgesics, bisphosphonates, and invasive methods of analgesic treatment (e.g., intraspinal and epidural analgesics or neurolytic blockades), radiation therapy plays an important role in pain alleviation. It is delivered to a growing primary tumour, lymph nodes, or distant metastatic sites, producing pain of various intensity. Currently, different regiments of radiation therapy methods and techniques and various radiation dose fractionations are incorporated into the clinical practice. These include palliative radiation therapy, conventional external beam radiation therapy, as well as modern techniques of intensity modulated radiation therapy, volumetrically modulated arch therapy, stereotactic radiosurgery or stereotactic body radiation therapy, and brachytherapy or radionuclide treatment (e.g., radium-223, strontium-89 for multiple painful osseous metastases). The review describes the possibilities and effectiveness of individual patient-tailored conventional and innovative radiation therapy approaches aiming at pain relief in cancer patients.

1. Introduction

The majority of cancer patients suffer from pain during course of their disease [1]. Pain may be a result of a growing primary tumour, metastases in lymph nodes, but predominately it occurs in patients experiencing distant metastases, particularly to the skeletal system. Pain management can significantly improve patients’ quality of life [2]. Antineoplastic treatment, e.g., radiation therapy, may also cause transient pain, which has to be managed pharmacologically [3]. Radiation therapy (RT) per se, however, significantly contributes to pain alleviation in cancer patients in multiple clinical scenarios [4,5]. Pain relief after radiation therapy may be achieved in as many as 60–80% of patients [2]. Recently, a strong need of substantial engagement of radiation oncologists in pain relief (among other symptoms produced by advanced disease) in cancer patients was a subject of the American Society for Radiation Oncology (ASTRO) members’ statement [6].

2. Painful Bone Metastases

About 50% of all cancer patients will develop bone metastases [7]. The frequency of bone metastases’ occurrence depends on tumour type, with breast, prostate, and lung cancers accounting for 85% of the cases [8,9]. Other primaries which frequently produce bone metastases include urinary bladder, renal, endometrial, and thyroid cancer as well as melanoma [9]. Bone pain may be caused by a local invasion of a metastatic tumour, which leads to remodelling of the microenvironment (changing the equilibrium between the activity of osteoblasts and osteoclast and bone structural degradation), direct nerve root invasion, or an increased release of inflammatory mediators, which stimulate nerve fibres [10,11,12]. Furthermore, a spasm of surrounding muscles may cause discomfort to the patient. Radiotherapy is the most effective treatment for bone metastases [13].

2.1. Pathomechanism of Analgesic Effect of Radiation

The exact mechanism of radiation-induced pain relief is unknown. The analgesic effect of radiation is achieved by stimulating ossification, diminishing osteoclasts’ activity in bone microenvironment, and killing cancer cells along with reduced osteolysis, which results in decreasing tumour burden [12,14]. Rapid pain relief (even after 24 h), observed in some patients, indicates a decrease of inflammatory cell activity as well as chemical pain mediators concentration in the radiation field, which play a role in the analgesic effect of radiation [15]. The RT fractionation scheme in patients with bone metastasis influences the level of mineralization, bone density, and recalcification of the irradiated site [16,17], which is associated with pain response. The recalcification rate was lower in the single fraction group (120%) compared to patients who received fractionated RT (173%, p < 0.0001) with a slight trend favouring 10 × 3 Gy RT scheme [17]. An association was observed between radiotherapy-driven bone pain relief and low osteoclast activity markers’ (pyridinoline—PYD, and deoxypyridinoline—DPD) concentrations in the urine before and after RT [18]. Re-irradiation for painful bone metastases also influences osteoclast activity visualized by urinary markers—PYD and DPD [19]. Significant differences at the baseline markers’ urinary levels were detected between 40 responders and 69 non-responders to re-irradiation (p = 0.03 for PYD and p = 0.04 for DPD) [19].

2.2. External Beam Radiation Therapy for Painful Bone Metastases

Radiation therapy with external beams may be delivered using different radiation techniques, the most common of which, for the purpose of irradiation of painful bone metastases, are the following: Two-dimensional radiation therapy (2D-RT) (Figure 1), still used especially in patients with expected short survival, and three-dimensional conformal radiation therapy (3D-CRT) (Figure 2). Modern modifications of the 3D-CRT radiation techniques include intensity modulated radiation therapy (IMRT), volumetrically modulated arch therapy (VMAT), or tomotherapy [20], which allow for better tumour coverage along with sparing surrounding normal tissue, thus decreasing potential toxicity of the treatment (Figure 2). Nowadays, sophisticated stereotactic radiosurgery (SRS) or stereotactic body radiation therapy (SBRT), allowing for delivery of a very high biological dose of radiation for very precisely delineated tumour masses, is increasingly used in selected groups of patients (Figure 2). Qualification of a particular patient for certain techniques and fractionation schedules in clinical practice depends on multiple factors (Table 1).

2.2.1. Conformal Radiation Therapy

The optimal choice of fractionation schedule for the treatment of painful bone metastases is still an unresolved issue. Most skeletal metastases are multiple in nature and only 10% are diagnosed as solitary ones [21].
During the last three decades, many randomized and observational trials focused on the optimal choice of the dose and fractionation regimen for pain relief [9,17,22,23,24,25,26,27,28]. They demonstrated an equivalent analgesic effect and durability of a single 8 Gray (Gy) fraction with multiple fractionation schedules, such as 30 Gy delivered in 10 fractions, 24 Gy given in 6 fractions, or 20 Gy in 5 fractions (Table 2) [9,17,21,22,23,24,25,26,27,28].
A meta-analysis of 25 randomized clinical trials revealed that up to 80% of patients with uncomplicated bone metastases experience analgesic response to external beam radiotherapy and 25–30% of patients achieve complete pain relief within 3–4 weeks after radiation therapy [2]. Uncomplicated bone metastases include metastatic tumour masses without massive infiltration towards soft tissue, characterized by a low risk of imminent pathological fracture and no evidence of spinal cord compression or cauda equina compression, and which were not previously irradiated [15]. Pain control persisting 3–6 months after radiation therapy, however, is experienced by 35% of patients only [2]. Of note, the re-treatment rate to in-field pain recurrence was higher in those patients who received single-fraction regimens than in those treated with multiple fraction schemes (20% vs. 8%, p < 0.00001) [2]. Yet, it is unclear if this was due to lower durability of pain control or to the physician’s comfort with re-treating after a lower initial radiation dose. It should be emphasized that single fraction treatment was associated with lower acute toxicity (nausea and vomiting, diarrhoea, fatigue, or skin toxicity). On the other hand, single fraction treatment was related to a higher rate of spinal cord compression and pathological fractures, but the difference did not reach statistical significance. The limitation of this meta-analysis are nonuniform primary end-points in different studies [2].
Despite the same efficacy of a single 8 Gy fraction, compared with multiple-fraction regimens and available published guidelines [11,15], to date this single fraction schedule is underused in the clinical practice to treat painful bone metastases [33]. Fischer-Valuck et al. [34], however, report increased usage of short course radiation therapy (1 × 8 Gy or 5 × 4 Gy) over long course treatment in 2014, compared to 2000, particularly in academic centres, in elderly prostate cancer patients living more than 15 miles from the treatment facility. In fact, single radiation fraction treatment allows patients to receive the planning procedure and radiation therapy within the same day, which is of particular importance for those with poor performance status [35,36].
The decision making should be based on treatment-related costs as well. In this context, some study results are of interest. A Dutch randomized controlled trial enrolling 1157 patients revealed that the cost of radiation therapy, including re-treatments and other nonmedical costs, was significantly lower for a single fraction regimen compared to a multiple fraction schedule ($2438 vs. $3311, p < 0.001) [37]. Similar financial results were obtained by the Radiation Therapy Oncology Group (RTOG, trial 9714) [38]. The New Zealand study revealed that single fraction radiotherapy costs per patient amounted to NZ$1344 and were lower than the costs of multiple fractionation regimens for prostate cancer patients [39]. Furthermore, SEER (The Surveillance, Epidemiology and End Results) analysis found a difference of $3094 between single fraction and multiple fractionations (10 fractions or more) [40]. It has been estimated that an absolute increase of 10% of single fraction radiation therapy administration for painful bone metastases in prostate cancer patients’ population could generate more than $70 million per year in health cost savings [41,42,43]. Of interest, in India the average distance travelled per day by a patient for radiation treatment is about 100 km [27], which means that patients undergoing single fraction treatment will travel 100 km for a complete radiation regimen, whereas patients undergoing 10-fraction radiation therapy will travel approx. 1000 km. It increases the costs from Rs1010 to Rs9700 per patient [27,37,38,39,40,41,42,43].
The neuropathic pain component represents a special issue. It occurs in 19–39% of cancer patients [44,45,46]. Pain management with pharmacotherapy may be troublesome, although it is the mainstay of the treatment [46,47]. Of interest, in a prospective observational study, patients with tumour-related neuropathic pain components (93 out of 302 patients) were more likely to achieve a pain response after radiation therapy administration than those without such component [48]. In this scenario, multifraction radiation therapy may be preferred to a single fraction, as it leads to longer durability of pain control [31]. It is associated with a higher remineralization rate of irradiated vertebrae, in comparison to a single fraction [17].
In case of spine metastases, combining percutaneous vertebroplasty (decreasing pain through restoring mechanical stability of vertebrae) with radiation therapy (exhibiting antitumor effect) provides better pain relief than radiotherapy alone [49] (Figure 3).
In a scenario of spinal cord or cauda equina compression caused by a metastatic mass, direct decompressive surgery followed by radiation therapy seems to be superior to radiotherapy alone [50]. The usual dose is 20 Gy delivered in 5 days or 30 Gy given in 10 days [15]. In patients who are unfit for surgical management, radiation therapy is indicated to alleviate the pain and decrease neurologic complications [15], although the optimal dose and radiation regimen are not established yet. Taking into account medical emergency in spinal cord compression, a shorter radiation schedule is desirable. Results of a randomized controlled trial, performed in a group of 276 patients with life expectancies of fewer than 6 months, indicate that both regimens (8 Gy × 2 days and multifraction split regimen: 5 × 3 Gy and 5 Gy × 3) are equally efficient (pain relief in 56% vs. 59% patients, motor capacity in 68% vs. 71% patients), thus the short course should be recommended as the treatment of choice in the group of patients [51]. In patients with bone fractures, surgical management along with radiation therapy should be performed (Figure 2).
It should also be depicted that the histology of a metastatic tumour influences its radiosensitivity and the analgesic effect of radiation therapy. Arcangeli et al. [52] demonstrated that non-small cell lung cancer metastases appeared to be the least responsive among all primary tumours, with 46% complete pain relief in comparison to bone metastases from breast and prostate cancers (65%, p = 0.04 and 83%, p-0.002, respectively).
In summary, the decision on RT fractionation should be supported by the American Society for Radiation Oncology (ASTRO) Evidence-Based Guideline [53]. The panel states there exists pain equivalency following a single 8 Gy fraction, 20 Gy in 5 fractions, 24 Gy in 6 fractions, and 30 Gy in 10 fractions for patients with previously unirradiated painful bone metastases. Patients should be made aware that single fraction RT is associated with a higher rate of re-treatment to the same painful site than in fractionated treatment. A single fraction treatment may be particularly convenient and sensible for patients with limited life expectancy. Patients who experience persistent or recurrent pain more than one month following external beam radiation therapy (EBRT) for symptomatic peripheral bone metastases or in spine lesions should be considered for re-treatment while adhering to normal tissue dosing constraints described in the available literature [53].

2.2.2. Hemibody Irradiation (HBI)

In patients with multiple painful bone metastases, hemibody irradiation using external beams may be a treatment option. The upper, middle or, lower half of the skeleton may be irradiated. Both photon beams and electron beams may be used [54,55]. Delivering 6–8 Gy in single fraction results in pain response in 70% of patients after 24–48 h [54]. The intensity of pain is reduced significantly, from 8 to 1, according to the visual-analogue scale, and there is a decrease in morphine consumption [54,55]. It is a very convenient treatment type for advanced cancer patients since it involves a short hospital stay and acceptable side effects [54].

2.2.3. Stereotactic Surgery/Stereotactic Body Radiation Therapy

As was mentioned earlier, since conventionally delivered radiation therapy leads to pain relief in approximately 80% of patients, but complete pain alleviation is only experienced by 25–30% of them, another concept of radiation therapy has emerged. Furthermore, the oligometastatic state in clinical practice has been defined when 1–5 distant metastases are found in a cancer patient [56]. Advances in radiation therapy planning software, custom patients immobilization, and sophisticated radiation delivery equipment utilizing multiple conformal beams or arc therapy with intensity modulation and image guidance, with each treatment allowing for accuracy within millimetres, facilitate obtaining a very precisely covered radiation target with extremely high biological doses of radiation delivered in 1 to few fractions [57] (Figure 2C). Single fraction radiation therapy is called “radiosurgery” whereas delivering multiple fractions—stereotactic body radiation therapy (SBRT). The latter is alternatively called stereotactic ablative radiotherapy since a huge biological dose of radiation is delivered to the treated target. Indications for SBRT include one to three vertebral metastases, less than 5 cm in diameter [52]. Symptomatic spinal cord compression may be a contraindication for the treatment, as the myelopathy after SBRT may increase [57]. Several doses per fraction are used, from single 15–24 Gy fractions to 18–36 Gy delivered in 1–5 fractions [57,58,59,60,61,62]. Of note, data concerning efficacy and safety of the treatment come from retrospective analyses [46,47,48,49,50,51]. Nevertheless, SRS/SBRT is widely used in most radiation therapy centres. About 40% of radiation oncologists in the US report that they use spine SBRT in their radiation departments and the single fraction is preferable due to greatest patient comfort and outpatient treatment convenience [63]. Unfortunately, many studies assessing the efficacy of spine SBRT report excellent local control (LC approximately 90%), while they do not provide information on pain control [57].
Most studies report that pain control continues for 1-year after spine SBRT in as many as 84–90% of patients [58,59,60,61,62,64] and about 50% of patients experience complete pain relief at 6 months [61,65].
To date, the optimal dose and fractionation schedule of SRS/SBRT is unclear. Chang et al. [58] reported pain control in 89.2% of patients suffering from different types of tumours. Ryu et al. [59], in a phase II dose escalation trial, demonstrated that a dose above 14 Gy in a single fraction was associated with increased pain control (one-year actuarial pain control was 84%, whereas 46% of patients experienced complete pain relief, 18.9%—partial relief, and 16.2% reported stable pain intensity). Currently, a RTOG 0631 phase 2/3 randomized study compares pain relief between 8 Gy in a single fraction given with conventional radiotherapy versus 16 Gy in a single fraction delivered with stereotactic techniques [66]. Sprave et al. [60], in a single-institution randomized explorative trial on a group of 55 patients suffering from painful spinal metastases, documented that after single-fraction SRS (24 Gy), patients achieved quicker and improved pain responses, in comparison to 3DCRT (30 Gy in 10 fractions). McGee et al. [67], in a retrospective analysis of 96 patients undergoing SRS to the spine metastases from primary tumours of radioresistant histology (hepatocellular cancer, cholangiocarcinoma, renal cell carcinoma, melanoma, or leiomyosarcoma), found high rates of pain relief (93%), but pointed out that hepatocellular carcinoma was associated with an inferior response to radiation therapy. The concept of single fraction SRS has some limitations. The most important one is the risk of postradiation myelopathy since vertebral masses are in close proximity to the spinal cord, which is characterized by a limited dose of tolerance [62,68]. In cases of radiosurgical treatment, the epidural tumour component is the most frequent site of treatment failure, since the spine and epidural tumour mass was spared from a high dose of radiation [58,59,60,62,66,67,68]. Thus, a distance of more than 3 mm from the tumour mass and spinal cord is desired [66].
Fractionated SBRT to spine metastases (27 Gy in 3 fractions) resulted in pain relief in 52% of renal cancer patients after one year [61]. An 84% improvement in symptomatic patients was also observed in a study performed by Gibbs et al. [62]. In a retrospective, international, and multicentre study based on 387 spinal metastases treated with SBRT (median total dose—24 Gy in 3 fractions) it was demonstrated that worse outcomes of radiation therapy were associated with an interval between primary tumour diagnosis and SBRT smaller than 30 months and the presence of histology of primary cancer, such as non-small cell lung cancer, renal cell cancer, and/or melanoma [69]. Prior to the treatment, patients were pain-free or reported pain of mild/moderate or severe intensity in 18.2%, 64.9%, and 16.9%, respectively. The patients remained pain-free at the time of the last clinical assessment (median follow-up of 11.5 months) in 76.8%, 56.3%, and 43.8%, respectively [64,69]. According to ASTRO guidelines, advanced radiation techniques, such as SBRT as the primary treatment for painful spine bone lesions, should be considered in the setting of a clinical trial or with data collected in a registry, given that insufficient data is available to routinely use this treatment [53].

2.2.4. Re-Irradiation

Palliative radiation therapy of bone metastasis for recurrent pain after previous palliative radiotherapy may be administered. It depends, however, on the location, prior radiation dose, fractionation schedule, and the time between radiation treatments [70]. Results obtained by Chow et al. [36], in a randomized trial, demonstrated that retreatment with a single fraction for painful bone metastases produces an effect equal to a multifraction conventional radiation therapy. The second conventional radiation treatment should be given no earlier than after 1 month [36]. Response to re-irradiation with conventional radiation therapy, however, was modest, with overall response rates of 45–52% and complete pain relief experienced by only 11–14% of patients [36]. These indicate a need for more effective re-treatment, and SBRT offers such opportunity [71,72]. A systematic review of studies concerning SBRT in re-irradiation of spine masses proved safety and good results in terms of local control and pain relief, although data is of low-quality or limited [72]. Only some studies reported results concerning pain control [58,73,74,75,76,77]. (Table 3).
In the case of previously irradiated, but progressing spinal metastases, re-irradiation should be ordered with caution, since the spinal cord is a radiosensitive structure [78,79]. The risk of myelopathy after re-irradiation is presumably low if the following conditions are met: The cumulative dose is less 135.5 Gy2, the interval between treatments is more than 6 months, and no course of radiation therapy exceeds the dose of 98 Gy2 [70,80] Sahgal et al. [64] found that an interval between conventional palliative radiation therapy and SBRT re-irradiation longer than 5 months, when maximum point dose to the thecal sac is limited to nBED 20–25 Gy (2/2), appears to be safe. Furthermore, the cumulative point max to the thecal sac should not exceed 70 Gy (2/2) and the SBRT thecal sac point max dose should not be above 50% of the total cumulative dose [64]. According to ASTRO guidelines, advanced radiation techniques, such as SBRT re-treatment for recurrent pain in spine bone lesions, may be feasible, effective, and safe, but this approach should be limited to clinical trial participation or on a registry given limited data supporting routine use [53].
This review is concentrated on radiation influence on pain control in cancer patients, but one has to realise that the final decision on qualification of patients to the SRS/SBRT should be taken at a multidisciplinary tumour board, assisted by the oncologic, neurologic, mechanical, and systemic framework defined by Laufer et al. [81], which allows for personalization of therapy.

3. Brachytherapy for the Treatment of Painful Bone Metastases

Brachytherapy consists in the application of radioactive sources inside the patient’s body (directly to the tumour burden or to the postoperative tumour bed) temporarily or permanently, which is meant to damage cancer cells’ DNA and destroy their ability to divide and grow. It allows for the use of a higher total dose of radiation to treat a smaller area in less time than the conventional external beam radiation therapy. Brachytherapy is a rarely used treatment option for bone metastases. Recently, a systematic review was published, which summarized the role of the treatment modality in painful spinal metastases [82]. Seven studies (which analysed treatment efficacy on pain control) reported a decrease of pain intensity after brachytherapy [83,84,85,86,87,88,89] (Table 4).

Pain Flare Syndrome

Pain relief contributes to improved quality of life of cancer patients, which is currently one of the most important goals of the treatment. Radiation therapy is widely used for decreasing pain in the population, however, in some patients “pain flare” can occur after this treatment. It is observed in 2–40% of patients [90,91] and is defined as a temporary increase of bone pain at the treated site during radiation therapy or early after its cessation [92]. Although the precise mechanism of this phenomenon is not recognized yet, biochemical mediators of inflammation, which are released upon the radiation therapy or transient oedema compressing nerves at the site of treatment, are suggested to contribute to this toxicity [93]. Steroids decrease pain flare intensity [28]. Furthermore, anti-inflammatory medications may prevent or reduce the risk of toxicity.

4. Radioactive Isotopes for the Treatment of Painful Bone Metastases

In cancer patients suffering from multiple bone metastases, bone-seeking radiopharmaceuticals have proven to be an effective alternative [94,95,96,97,98] (Figure 4, Table 5). Strontium-89 chloride, Samarium-153-ethylenediamine tetramethylene phosphonic acid (EDTMP), Rhenium-186-hydroxyethylidine diphosphonic acid (HEDP), and Radium-233 dichloride have been approved for the treatment of bone pain due to osteoblastic or mixed bone metastases, mainly from prostate and breast cancers (most common indications) and other tumours presenting with painful osteoblastic lesions, confirmed by whole-body bone scintigraphy performed within at least 8 weeks before therapy [98,99,100,101]. These agents mainly accumulate in osteosclerotic and osteoblastic bone metastases, whereas they are not suitable for treating osteolytic and osteoclastic bone metastases. Furthermore, in metastatic bones vulnerable to fracture, local therapy such as surgery or radiation therapy should be performed prior to radionuclide therapy [102].
The mechanism of pain palliation resulting from beta particles or alpha particles emitted by radionuclides is not clear yet. Beta or alpha particles kill tumour cells; therefore, pain relief occurs because of mechanical pressure reduction (Figure 5). Pain control, however, frequently occurs before the tumour mass shrinks. Lymphocytes, which are radiation sensitive cells, secrete a variety of cytokines causing pain. The death of lymphocytes cells at the tumour site may also contribute to pain relief [94].

4.1. Several Radionuclides Are Used in Clinical Practice, and Many of Them Are under Investigation

4.1.1. Strontium-89 Chloride

There is solid data on the efficacy of Strontium-89 Chloride for bone pain relief in patients with prostate and breast cancers, with a pain relief rate of 63–88% [94,103,104,105,106,107,108,109,110]. Symptomatic improvement usually occurred within 6 weeks after administration, with a mean duration of the pain-free period of about 6 months [111], with no dose-response relationship [112]. Retreatment for responders is possible at time intervals of not less than 12 weeks [112,113].

4.1.2. Samarium-153-EDTMP

Reduction of bone pain occurs in 62–78% of patients with bone metastases within 1 week of Samarium-153-EDTMP administration, with a definite dose-response relationship [109,114,115,116,117,118,119,120,121], with a mean duration of approximately 3–8 months. In several phase II/III clinical trials this radionuclide has shown significant efficacy for bone pain alteration in patients with various types of cancer, including lung, prostate, and breast cancer as well as osteosarcoma [122,123,124]. The minimum interval for retreatment should be 8 weeks [112,113].

4.1.3. Radium-223-Dichloride

A phase III randomized double-blind placebo-controlled trial, ALSYMPCA, investigated the effectiveness of Radium-223-dichloride in 921 patients with metastatic castration-resistant prostate cancer with symptomatic bone metastases, previous use of analgesics or radiotherapy to bones, and no visceral metastasis. There was a significant improvement in median overall survival in the Radium-223-dichloride group vs. the placebo group (14.9 vs. 11.3 months respectively) and median time to the first symptomatic skeletal event (15.6 vs. 9.8 months, respectively) [125,126,127]. Median time to initial opioid use was significantly longer in the Ra-223 group, with risk reduction of 38%, compared to placebo. Less Ra-223 patients (36%) than placebo patients (50%) required opioids for pain relief. The QOL pain score showed reduced pain for Ra-223 patients relative to placebo patients at week 16 [128].

4.1.4. Rhenium-186-HEDP

Clinical studies assessing Rhenium-186-HEDP were mainly performed on patients with prostate cancer and breast cancer [129,130,131,132,133]. General response rates for pain palliation ranged between 38% and 82% [95,99,134,135,136,137,138,139]. A pain response occurred 1–3 weeks after administration, with a durability of 5–12 months. No definite relationship between dose and pain response was observed [138]. Retreatment for responders is possible at time intervals of not less 6–8 weeks [140,141].

4.1.5. Rhenium-188-HEDP

In a study by Palmedo et al. [142], with Rhenium-188-HEDP pain relief occurred in 64% of prostate cancer patients with bone metastases. Mean duration of the effect was 7.5 weeks [142]. In a study by Liepe et al. [143], pain relief was achieved in 76% of patients with hormone-refractory prostate carcinoma treated with the radionuclide. Therefore, in patients with progressive hormone-resistant prostate carcinoma and bone pain, repeated Rhenium-188-HEDP administration revealed the pain response rate of 92% vs. 60% and durability of response of 5.66 months, compared to 2.55 months for the single treatment group, respectively [144].

4.2. Side Effects of Radionuclide Therapy

The above-mentioned agents may produce some side effects such as gastrointestinal ulceration, enhanced bleeding, neutropenia, and disturbed renal function [94]. In about 10% of cases, regardless of the agent used, there is a possibility of a flare (a painful response with an increase of pain insensitivity). Usually, within 72 h of administration, these symptoms (typically temporary, mild, and self-limiting) should be gone. When osseous metastases involve the cervical spine, a low risk of post-therapy spinal cord compression exists and prophylactic corticosteroids should be given [97,98,99].

5. Radiation Therapy for Painful Primary/Regional/Metastatic Solid Tumours Other Than Bone Tumours

Progressing soft tissue tumours may also produce mild to severe pain. Primary or metastatic brain tumours, particularly those presenting with a larger mass effect or surrounding oedema, are frequently associated with pain. Surgery and radiotherapy are under consideration in the group of patients, depending on different factors (performance status of the patient, presence of uncontrolled extracranial disease, expected survival, etc.). Brain radiation therapy, among others—whole brain radiation therapy—may help reduce tumour mass and volume of oedema, thus leading to pain relief. Lung cancer patients may suffer from severe pain resulting from the invasion of brachial plexus by the direct apical tumour. Meta-analysis of 14 randomized clinical trials evaluating palliative radiation therapy for lung cancer patients revealed satisfactory symptomatic relief, among others—pain reduction [150]. Various fractionation schedules were used in the trials as follows: 10 Gy in 1 fraction, 17 Gy in 2 fractions, and 20 Gy in 5 fractions, as well as long courses, such as 30–45 Gy in 10–15 fractions [150]. Locally advanced gastrointestinal cancer patients may also suffer from the pain of different intensity. Administration of IMRT for a gastric tumour in patients with satisfactory performance status leads to a decrease of symptoms, among others—pain—in more than 70% of patients [151]. Pain may be experienced by advanced rectal cancer patients. Palliative radiation therapy to the pelvis, both using conventional fractionation (45 Gy in 25 fractions) and hypofractionated radiotherapy (30 Gy in 6 fractions) produces satisfactory pain relief in approximately 70% of patients [152,153]. As many as 40% of pancreatic cancer patients are diagnosed with locoregionally advanced disease or progress during the course of the disease [154]. The infiltration of nerves in the area surrounding the pancreas is observed in 43–72% of patients, which causes severe pain [155]. Delivering median 28 Gy (25–33 Gy) in 5 fractions using SBRT resulted in abdominal pain relief in 78% of advanced pancreatic cancer patients [156].

6. Pain Assessment after Radiation Therapy

There is no uniform pain assessment in cancer patients receiving radiation therapy. Some authors report pain control as subjective physician/patient reports [75,76,102,157]. Validated Brief Pain Inventory (BPI) was also used [158]. The International Bone Metastases Consensus Working Party proposed pain response categories in palliative radiation therapy [159]. They defined four response categories:
  • Complete response: A pain score of 0 at the treated site and no concomitant increase in analgesic intake, which means stable or reduced analgesics in daily oral morphine equivalent (OMED).
  • Partial response: Pain decrease of 2 or more at the treated site on a scale of 0 to 10 without analgesic increase, or analgesic dose decrease of 25% or more from the baseline without an increase in pain intensity.
  • Pain progression: Increase in pain score of 2 or more above the baseline at the treated site with stable OMED, or an increase of 25% or more OMED in comparison to the baseline with the pain score stable or 1 point above the baseline.
  • Intermediate response: Any response that is not captured by those defined above [159].

7. Future Directions

New and high-quality prospective data is awaited that will answer several questions and allow for definite statements regarding different combinations of radiation therapy (IMRT, SBRT, brachytherapy, and radionuclide therapy) with surgery (kyphoplasty or vertebroplasty for spine metastases or intramedullary fixation or endoprosthetic reconstruction for long bone metastases) and/or novel molecularly targeted agents/immunotherapy. Prospective phase III randomized studies will define the optimal use (in terms of efficacy and toxicity) of SBRT for treatment of newly diagnosed or recurrent painful spinal metastases. The STEREO-OS trial, which assesses the effect of standard systemic treatment in oligometastatic (3–5 sites) prostate, breast, and/or lung cancer patients in combination with SBRT for painful bone metastases [160], may serve as an example. Biological image-guided SBRT for painful bone metastases, with non-homogenous dose escalation based on FDG-PET(18F-Fluorodeoxyglucose—positron emission tomography) results, is another interesting concept [161]. Delivering high biological dose in the tumour region using IMRT with integrated boost for painful spinal bone metastases is the subject of the IRON-2 trial (Intensity-modulated Radiotherapy With Integrated-boost in Patients With Spinal Bone Metastases) [162]. Furthermore, the VERTICAL study is ongoing, assessing the analgesic effect of SBRT comparing to standard low dose EBRT [163]. Prospective cohorts of patients with painful bone metastases (the PRESENT study—Prospective Evaluation of Interventional Studies on Bone Metastases) or exclusively with long bone metastases (the OPTIMAL study) are formed to better guide personalized treatment in terms of improved quality of life and analgesic effect of radiation therapy, surgery, or combined treatment modalities [164,165].
A novel concept is a radiosurgical hypophysectomy for intractable bone metastases pain. Australian researchers will assess whether delivery of a single high dose (150 Gy) of radiation therapy to a small area of the pituitary gland and pituitary stalk in a highly precise manner may be helpful in reducing intractable pain from bone metastases [166].
The Palliative Radiotherapy and Inflammation Study (PRAIS) aims to find predictive factors associated with inflammation for palliative RT for cancer-induced pain response [167]. Furthermore, the latest findings over genetic biomarkers of different aspects of palliative RT for painful bone metastases are very interesting. Namely, Furfari et al. [168,169,170] identified genes’ profiles for changes in quality of life and pain relief, pain flare, and dexamethasone response following RT.
Development of higher quality data will further help find the best combinations of EBRT with bisphosphonates, radiopharmaceuticals, and novel biological agents reducing formation/activity of osteoclasts and bone resorption, like monoclonal antibody directed against RANKL—Receptor Activator for Nuclear Factor κ B Ligand (e.g., denosumab or novel agents, like JMT103) [171,172] or reducing cancer-induced bone osteolysis c-src inhibitors (dasatinib, bosutinib) [173].

8. Conclusions

Radiation therapy plays an important role in pain relief in cancer patients. As the radiation oncology field evolved, a number of challenges appeared. Important issues exist which have to be resolved, such as the following: Inconsistent endpoints of trials, difficulty in measuring the response, reluctance to practice evidence-based medicine (e.g., the choice of optimal regimen, re-treatment fractionation), differences in physicians’ and patients’ perspectives, as well as incorporating systemic treatment in combination with radiation therapy (bisphosphonates, nanotechnology, etc.).
Evidence-based treatment guidelines should be established and followed. Collaboration in multidisciplinary tumour boards provides the best, personalized, holistic care, which leads to, among others, pain reduction and an improved quality of patients’ life.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Agarawal, J.P.; Swangsilpa, T.; van der Linden, Y.; Rades, D.; Jeremic, B.; Hoskin, P.J. The Role of External Beam Radiotherapy in the Management of Bone Metastases. Clin. Oncol. 2006, 18, 747–760. [Google Scholar] [CrossRef]
  2. Chow, E.; Zeng, L.; Salvo, N.; Dennis, K.; Tsao, M.; Lutz, S. Update on the Systematic Review of Palliative Radiotherapy Trials for Bone Metastases. Clin. Oncol. 2012, 24, 112–124. [Google Scholar] [CrossRef]
  3. Konopka-Filippow, M.; Zabrocka, E.; Wõjtowicz, A.; Skalij, P.; Wojtukiewicz, M.Z.; Sierko, E. Pain management during radiotherapy and radiochemotherapy in oropharyngeal cancer patients: Single-institution experience. Int. Dent. J. 2015, 65, 242–248. [Google Scholar] [CrossRef]
  4. Lutz, S.; Berk, L.; Chang, E.; Chow, E.; Hahn, C.; Hoskin, P.; Howell, D.; Konski, A.; Kachnic, L.; Lo, S.; et al. Palliative radiotherapy for bone metastases: An ASTRO evidence-based guideline. Int. J. Radiat. Oncol. Biol. Phys. 2011, 79, 965–976. [Google Scholar] [CrossRef]
  5. Kougioumtzopoulou, A.; Zygogianni, A.; Liakouli, Z.; Kypraiou, E.; Kouloulias, V. The role of radiotherapy in bone metastases: A critical review of current literature. Eur. J. Cancer Care (Engl). 2017, 26, e12724. [Google Scholar] [CrossRef]
  6. Wei, R.L.; Mattes, M.D.; Yu, J.; Thrasher, A.; Shu, H.-K.; Paganetti, H.; De Los Santos, J.; Koontz, B.; Abraham, C.; Balboni, T. Attitudes of radiation oncologists toward palliative and supportive care in the United States: Report on national membership survey by the American Society for Radiation Oncology (ASTRO). Pract. Radiat. Oncol. 2017, 7, 113–119. [Google Scholar] [CrossRef]
  7. Arcangeli, G.; Pinnarò, P.; Rambone, R.; Giannarelli, D.; Benassi, M. A phase III randomized study on the sequencing of radiotherapy and chemotherapy in the conservative management of early-stage breast cancer. Int. J. Radiat. Oncol. Biol. Phys. 2006, 64, 61–167. [Google Scholar] [CrossRef]
  8. Sharma, K.; Bahadur, A.; Mohanta, P.; Singh, K.; Rathi, A. Palliative treatment of painful bone metastases: Does fractionation matter? Indian J. Palliat. Care 2008, 14, 7. [Google Scholar] [CrossRef]
  9. Harstell, W.F.; Santosh, Y. Palliation of bone metastases. In Principles and Practice of Radiotion Oncology, Volume 1.; Halperin, E., Perez, C., Brady, L., Eds.; Lippincott Williams and Wilkin: Philadelphia, PA, USA, 2013; pp. 1778–1791. [Google Scholar]
  10. Johnstone, C.; Lutz, S.T. External beam radiotherapy and bone metastases. Ann. Palliat. Med. 2014, 3, 114–122. [Google Scholar]
  11. Lutz, S. The Role of radiation therapy in controlling painful bone metastases. Curr. Pain Headache Rep. 2012, 16, 300–306. [Google Scholar] [CrossRef]
  12. Goblirsch, M.J.; Zwolak, P.P.; Clohisy, D.R. Biology of Bone Cancer Pain. Clin. Cancer Res. 2006, 12, 6231s–6235s. [Google Scholar] [CrossRef]
  13. Needham, P.R.; Mithal, N.P.; Hoskin, P.J. Radiotherapy for bone pain. J. R. Soc. Med. 1994, 87, 503–505. [Google Scholar]
  14. Goblirsch, M.; Mathews, W.; Lynch, C.; Alaei, P.; Gerbi, B.J.; Mantyh, P.W.; Clohisy, D.R. Radiation treatment decreases bone cancer pain, osteolysis and tumor size. Radiat. Res. 2004, 161, 228–234. [Google Scholar] [CrossRef]
  15. Wu, J.S.Y.; Wong, R.K.S.; Lloyd, N.S.; Johnston, M.; Bezjak, A.; Whelan, T. Radiotherapy fractionation for the palliation of uncomplicated painful bone metastases—An evidence-based practice guideline. BMC Cancer 2004, 4, 71. [Google Scholar] [CrossRef]
  16. Sprave, T.; Verma, V.; Förster, R.; Schlampp, I.; Hees, K.; Bruckner, T.; Bostel, T.; El Shafie, R.A.; Welzel, T.; Nicolay, N.H.; et al. Bone density and pain response following intensity-modulated radiotherapy versus three-dimensional conformal radiotherapy for vertebral metastases—Secondary results of a randomized trial. Radiat. Oncol. 2018, 13, 212. [Google Scholar] [CrossRef]
  17. Koswig, S.; Budach, V. Remineralization and pain relief in bone metastases after after different radiotherapy fractions (10 times 3 Gy vs. 1 time 8 Gy). A prospective study. Strahlenther. Onkol. 1999, 175, 500–508. [Google Scholar] [CrossRef]
  18. Hoskin, P.J.; Stratford, M.R.L.; Folkes, L.K.; Regan, J.; Yarnold, J.R. Effect of local radiotherapy for bone pain on urinary markers of osteoclast activity. Lancet 2000, 355, 1428–1429. [Google Scholar] [CrossRef]
  19. Chow, E.; DeAngelis, C.; Chen, B.E.; Azad, A.; Meyer, R.M.; Wilson, C.; Kerba, M.; Bezjak, A.; Wilson, P.; Nabid, A.; et al. Effect of re-irradiation for painful bone metastases on urinary markers of osteoclast activity (NCIC CTG SC.20U). Radiother. Oncol. 2015, 115, 141–148. [Google Scholar] [CrossRef]
  20. Sapkaroski, D.; Osborne, C.; Knight, K.A. A review of stereotactic body radiotherapy—Is volumetric modulated arc therapy the answer? J. Med. Radiat. Sci. 2015, 62, 142–151. [Google Scholar] [CrossRef]
  21. Falkmer, U.; Järhult, J.; Wersäll, P.; Cavallin-Ståhl, E. A systematic overview of radiation therapy effects in skeletal metastases. Acta Oncol. (Madr). 2003, 42, 620–633. [Google Scholar] [CrossRef]
  22. Yarnold, J.R. 8 Gy single fraction radiotherapy for the treatment of metastatic skeletal pain: Randomised comparison with a multifraction schedule over 12 months of patient follow-up. Radiother. Oncol. 1999, 52, 111–121. [Google Scholar] [CrossRef]
  23. Foro Arnalot, P.; Fontanals, A.V.; Galcerán, J.C.; Lynd, F.; Latiesas, X.S.; de Dios, N.R.; Castillejo, A.R.; Bassols, M.L.; Galán, J.L.; Conejo, I.M.; et al. Randomized clinical trial with two palliative radiotherapy regimens in painful bone metastases: 30 Gy in 10 fractions compared with 8 Gy in single fraction. Radiother. Oncol. 2008, 89, 150–155. [Google Scholar] [CrossRef]
  24. Steenland, E.; Leer, J.; Van Houwelingen, H.; Post, W.J.; Van den Hout, W.B.; Kievit, J.; De Haes, H.; Martijn, H.; Oei, B.; Vonk, E.; et al. The effect of a single fraction compared to multiple fractions on painful bone metastases: A global analysis of the Dutch Bone Metastasis Study. Radiother. Oncol. 1999, 52, 101–109. [Google Scholar] [CrossRef]
  25. Nielsen, O.S.; Bentzen, S.M.; Sandberg, E.; Gadeberg, C.C.; Timothy, A.R. Randomized trial of single dose versus fractionated palliative radiotherapy of bone metastases. Radiother. Oncol. 1998, 47, 233–240. [Google Scholar] [CrossRef]
  26. Gaze, M.N.; Kelly, C.G.; Kerr, G.R.; Cull, A.; Cowie, V.J.; Gregor, A.; Howard, G.C.; Rodger, A. Pain relief and quality of life following radiotherapy for bone metastases: a randomised trial of two fractionation schedules. Radiother. Oncol. 1997, 45, 109–116. [Google Scholar] [CrossRef]
  27. Nongkynrih, A.; Dhull, A.K.; Kaushal, V.; Atri, R.; Dhankhar, R.; Kamboj, K. Comparison of Single Versus Multifraction Radiotherapy in Palliation of Painful Bone Metastases. World J. Oncol. 2018, 9, 91–95. [Google Scholar] [CrossRef]
  28. Loblaw, D.A.; Wu, J.S.; Kirkbride, P.; Panzarella, T.; Smith, K.; Aslanidis, J.; Warde, P. Pain flare in patients with bone metastases after palliative radiotherapy—A nested randomized control trial. Support. Care Cancer 2007, 15, 451–455. [Google Scholar] [CrossRef]
  29. Price, P.; Hoskin, P.J.; Easton, D.; Austin, D.; Palmer, S.G.; Yarnold, J.R. Prospective randomised trial of single and multifraction radiotherapy schedules in the treatment of painful bony metastases. Radiother. Oncol. 1986, 6, 247–255. [Google Scholar] [CrossRef]
  30. Cole, D.J. A randomized trial of a single treatment versus conventional fractionation in the palliative radiotherapy of painful bone metastases. Clin. Oncol. 1989, 1, 59–62. [Google Scholar] [CrossRef]
  31. Roos, D.E.; Turner, S.L.; O’Brien, P.C.; Smith, J.G.; Spry, N.A.; Burmeister, B.H.; Hoskin, P.J.; Ball, D.L. Randomized trial of 8 Gy in 1 versus 20 Gy in 5 fractions of radiotherapy for neuropathic pain due to bone metastases (Trans-Tasman Radiation Oncology Group, TROG 96.05). Radiother. Oncol. 2005, 75, 54–63. [Google Scholar] [CrossRef]
  32. Harstell, W.F.; Scott, C.B.; Bruner, D.W.; Scarantino, C.W.; Ivker, R.A.; Roach, M.; Suh, J.H.; Demas, W.F.; Movsas, B.; Petersen, I.A.; et al. Randomized trial of short- versus long-course radiotherapy for palliation of painful bone metastases. J. Natl. Cancer Inst. 2005, 97, 798–804. [Google Scholar]
  33. Fairchild, A.; Barnes, E.; Ghosh, S.; Ben-Josef, E.; Roos, D.; Hartsell, W.; Holt, T.; Wu, J.; Janjan, N.; Chow, E. International Patterns of Practice in Palliative Radiotherapy for Painful Bone Metastases: Evidence-Based Practice? Int. J. Radiat. Oncol. Biol. Phys. 2009, 75, 1501–1510. [Google Scholar] [CrossRef]
  34. Fischer-Valuck, B.W.; Baumann, B.C.; Apicelli, A.; Rao, Y.J.; Roach, M.; Daly, M.; Dans, M.C.; White, P.; Contreras, J.; Henke, L.; et al. Palliative radiation therapy (RT) for prostate cancer patients with bone metastases at diagnosis: A hospital-based analysis of patterns of care, RT fractionation scheme, and overall survival. Cancer Med. 2018, 7, 4240–4250. [Google Scholar] [CrossRef]
  35. Kachnic, L.; Berk, L. Palliative Single-Fraction Radiation Therapy: How Much More Evidence Is Needed? JNCI 2005, 97, 786–788. [Google Scholar] [CrossRef]
  36. Chow, E.; van der Linden, Y.M.; Roos, D.; Hartsell, W.F.; Hoskin, P.; Wu, J.S.Y.; Brundage, M.D.; Nabid, A.; Tissing-Tan, C.J.A.; Oei, B.; et al. Single versus multiple fractions of repeat radiation for painful bone metastases: A randomised, controlled, non-inferiority trial. Lancet Oncol. 2014, 15, 164–171. [Google Scholar] [CrossRef]
  37. Van den Hout, W.B.; van der Linden, Y.M.; Steenland, E.; Wiggenraad, R.G.J.; Kievit, J.; de Haes, H.; Leer, J.W.H. Single- versus multiple-fraction radiotherapy in patients with painful bone metastases: Cost-utility analysis based on a randomized trial. J. Natl. Cancer Inst. 2003, 95, 222–229. [Google Scholar] [CrossRef]
  38. Konski, A.; James, J.; Hartsell, W.; Leibenhaut, M.H.; Janjan, N.; Curran, W.; Roach, M.; Watkins-Bruner, D. Economic analysis of Radiation Therapy Oncology Group 97-14: Multiple versus single fraction radiation treatment of patients with bone metastases. Am. J. Clin. Oncol. Cancer Clin. Trials 2009, 32, 423–428. [Google Scholar] [CrossRef]
  39. Collinson, L.; Kvizhinadze, G.; Nair, N.; McLeod, M.; Blakely, T. Economic evaluation of single-fraction versus multiple-fraction palliative radiotherapy for painful bone metastases in breast, lung and prostate cancer. J. Med. Imaging Radiat. Oncol. 2016, 60, 650–660. [Google Scholar] [CrossRef] [Green Version]
  40. Bekelman, J.E.; Epstein, A.J.; Emanuel, E.J. Single- vs Multiple-Fraction Radiotherapy for Bone Metastases From Prostate Cancer. JAMA 2013, 310, 1501. [Google Scholar] [CrossRef]
  41. Schreiber, D.; Safdieh, J.; Becker, D.J.; Schwartz, D. Patterns of care and survival outcomes of palliative radiation for prostate cancer with bone metastases: Comparison of ≤5 fractions to ≥10 fractions. Ann. Palliat. Med. 2017, 6, 55–65. [Google Scholar] [CrossRef]
  42. Olson, R.A.; Tiwana, M.S.; Barnes, M.; Kiraly, A.; Beecham, K.; Miller, S.; Hoegler, D.; Olivotto, I. Use of single-versus multiple-fraction palliative radiation therapy for bone metastases: Population-based analysis of 16,898 courses in a Canadian Province. Int. J. Radiat. Oncol. Biol. Phys. 2014, 89, 1092–1099. [Google Scholar] [CrossRef]
  43. Olson, R.A.; Tiwana, M.; Barnes, M.; Cai, E.; McGahan, C.; Roden, K.; Yurkowski, E.; Gentles, Q.; French, J.; Halperin, R.; et al. Impact of using audit data to improve the evidence-based use of single-fraction radiation therapy for bone metastases in British Columbia presented at the Canadian Association of Radiation Oncology 2014 Annual Meeting, Aug 25–28, 2014, St. John’s, Newfoun. Int. J. Radiat. Oncol. Biol. Phys. 2016, 94, 40–47. [Google Scholar] [CrossRef]
  44. Morgan, K.J.; Anghelescu, D.L. A review of adult and pediatric neuropathic pain assessment tools. Clin. J. Pain 2017, 33, 844–852. [Google Scholar] [CrossRef]
  45. Roberto, A.; Deandrea, S.; Greco, M.T.; Corli, O.; Negri, E.; Pizzuto, M.; Ruggeri, F. Prevalence of Neuropathic Pain in Cancer Patients: Pooled Estimates from a Systematic Review of Published Literature and Results from a Survey Conducted in 50 Italian Palliative Care Centers. J. Pain Symptom Manag. 2016, 51, 1091–1102e4. [Google Scholar] [CrossRef]
  46. Vadalouca, A.; Raptis, E.; Moka, E.; Zis, P.; Sykioti, P.; Siafaka, I. Pharmacological Treatment of Neuropathic Cancer Pain: A Comprehensive Review of the Current Literature. Pain Pract. 2012, 12, 219–251. [Google Scholar] [CrossRef]
  47. Fallon, M.T.; Colvin, L. Neuropathic pain in cancer. Br. J. Anaesth. 2013, 111, 105–111. [Google Scholar] [CrossRef] [Green Version]
  48. Saito, T.; Tomitaka, E.; Toya, R.; Matsuyama, T.; Ninomura, S.; Watakabe, T.; Oya, N. A neuropathic pain component as a predictor of improvement in pain interference after radiotherapy for painful tumors: A secondary analysis of a prospective observational study. Clin. Transl. Radiat. Oncol. 2018, 12, 34–39. [Google Scholar] [CrossRef]
  49. Lu, C.-W.; Shao, J.; Wu, Y.-G.; Wang, C.; Wu, J.-H.; Lv, R.-X.; Ding, M.-C.; Shi, Z.-C.; Mao, N.-F. Which Combination Treatment Is Better for Spinal Metastasis. Am. J. Ther. 2019, 26, e38–e44. [Google Scholar] [CrossRef]
  50. Patchell, R.A.; Tibbs, P.A.; Regine, W.F.; Payne, R.; Saris, S.; Kryscio, R.J.; Mohiuddin, M.; Young, B. Direct decompressive surgical resection in the treatment of spinal cord compression caused by metastatic cancer: A randomised trial. Lancet 2005, 366, 643–648. [Google Scholar] [CrossRef]
  51. Maranzano, E.; Bellavita, R.; Rossi, R.; De Angelis, V.; Frattegiani, A.; Bagnoli, R.; Mignogna, M.; Beneventi, S.; Lupattelli, M.; Ponticelli, P.; et al. Short-course versus split-course radiotherapy in metastatic spinal cord compression: Results of a phase III, randomized, multicenter trial. J. Clin. Oncol. 2005, 23, 3358–3365. [Google Scholar] [CrossRef]
  52. Arcangeli, G.; Giovinazzo, G.; Saracino, B.; D’Angelo, L.; Giannarelli, D.; Micheli, A. Radiation therapy in the management of symptomatic bone metastases: The effect of total dose and histology on pain relief and response duration. Int. J. Radiat. Oncol. Biol. Phys. 1998, 42, 1119–1126. [Google Scholar] [CrossRef]
  53. Lutz, S.; Balboni, T.; Jones, J.; Lo, S.; Petit, J.; Rich, S.E.; Wong, R.; Hahn, C. Palliative radiation therapy for bone metastases: Update of an ASTRO Evidence-Based Guideline. Pract. Radiat. Oncol. 2017, 7, 4–12. [Google Scholar] [CrossRef]
  54. Pal, S.; Dutta, S.; Adhikary, S.; Bhattacharya, B.; Ghosh, B.; Patra, N. Hemi body irradiation: An economical way of palliation of pain in bone metastasis in advanced cancer. South Asian J. Cancer 2014, 3, 28. [Google Scholar] [CrossRef]
  55. Delinikolas, P.; Patatoukas, G.; Kouloulias, V.; Dilvoi, M.; Plousi, A.; Efstathopoulos, E.; Platoni, K. A novel Hemi-Body Irradiation technique using electron beams (HBIe−). Phys. Medica 2018, 46, 16–24. [Google Scholar] [CrossRef]
  56. Palma, D.A.; Salama, J.K.; Lo, S.S.; Senan, S.; Treasure, T.; Govindan, R.; Weichselbaum, R. The oligometastatic state-separating truth from wishful thinking. Nat. Rev. Clin. Oncol. 2014, 11, 549–557. [Google Scholar] [CrossRef]
  57. Osborn, V.W.; Lee, A.; Yamada, Y. Stereotactic Body Radiation Therapy for Spinal Malignancies. Technol. Cancer Res. Treat. 2018, 17. [Google Scholar] [CrossRef]
  58. Chang, U.K.; Cho, W.I.; Kim, M.S.; Cho, C.K.; Lee, D.H.; Rhee, C.H. Local tumor control after retreatment of spinal metastasis using stereotactic body radiotherapy; comparison with initial treatment group. Acta Oncol. (Madr). 2012, 51, 589–595. [Google Scholar] [CrossRef] [Green Version]
  59. Ryu, S.; Jin, R.; Jin, J.Y.; Chen, Q.; Rock, J.; Anderson, J.; Movsas, B. Pain Control by Image-Guided Radiosurgery for Solitary Spinal Metastasis. J. Pain Symptom Manag. 2008, 35, 292–298. [Google Scholar] [CrossRef]
  60. Sprave, T.; Verma, V.; Förster, R.; Schlampp, I.; Bruckner, T.; Bostel, T.; Welte, S.E.; Tonndorf-Martini, E.; Nicolay, N.H.; Debus, J.; et al. Randomized phase II trial evaluating pain response in patients with spinal metastases following stereotactic body radiotherapy versus three-dimensional conformal radiotherapy. Radiother. Oncol. 2018, 128, 274–282. [Google Scholar] [CrossRef]
  61. Nguyen, Q.N.; Shiu, A.S.; Rhines, L.D.; Wang, H.; Allen, P.K.; Wang, X.S.; Chang, E.L. Management of Spinal Metastases From Renal Cell Carcinoma Using Stereotactic Body Radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 2010, 76, 1185–1192. [Google Scholar] [CrossRef]
  62. Gibbs, I.C.; Kamnerdsupaphon, P.; Ryu, M.R.; Dodd, R.; Kiernan, M.; Chang, S.D.; Adler, J.R. Image-guided robotic radiosurgery for spinal metastases. Radiother. Oncol. 2007, 82, 185–190. [Google Scholar] [CrossRef]
  63. Pan, H.; Simpson, D.R.; Mell, L.K.; Mundt, A.J.; Lawson, J.D. A survey of stereotactic body radiotherapy use in the United States. Cancer 2011, 117, 4566–4572. [Google Scholar] [CrossRef] [PubMed]
  64. Sahgal, A.; Ma, L.; Weinberg, V.; Gibbs, I.C.; Chao, S.; Chang, U.K.; Werner-Wasik, M.; Angelov, L.; Chang, E.L.; Sohn, M.J.; et al. Reirradiation human spinal cord tolerance for stereotactic body radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, 107–116. [Google Scholar] [CrossRef] [PubMed]
  65. Kim, M.S.; Keum, K.C.; Cha, J.H.; Kim, J.H.; Seong, J.S.; Lee, C.G.; Nam, K.C.; Koom, W.S. Stereotactic Body Radiotherapy with Helical Tomotherapy for Pain Palliation in Spine Metastasis. Technol. Cancer Res. Treat. 2013, 12, 363–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Ryu, S.; Pugh, S.L.; Gerszten, P.C.; Yin, F.F.; Timmerman, R.D.; Hitchcock, Y.J.; Movsas, B.; Kanner, A.A.; Berk, L.B.; Followill, D.S.; et al. RTOG 0631 phase 2/3 study of image guided stereotactic radiosurgery for localized (1-3) spine metastases: Phase 2 results. Pract. Radiat. Oncol. 2014, 4, 76–81. [Google Scholar] [CrossRef] [PubMed]
  67. McGee, H.M.; Carpenter, T.; Ozbek, U.; Kirkwood, K.; Tzu-Chi, T.; Blacksburg, S.; Germano, I.M.; Green, S.; Buckstein, M. Analysis of Local Control and Pain Control Following Spine Stereotactic Radiosurgery Reveals Inferior Outcomes for Hepatocellular Carcinoma compared to other Radioresistant Histologies. Pract. Radiat. Oncol. 2018. [Google Scholar]
  68. Ryu, S.; Jin, J.-Y.; Jin, R.; Rock, J.; Ajlouni, M.; Movsas, B.; Rosenblum, M.; Kim, J.H. Partial volume tolerance of the spinal cord and complications of single-dose radiosurgery. Cancer 2007, 109, 628–636. [Google Scholar] [CrossRef] [Green Version]
  69. Guckenberger, M.; Mantel, F.; Gerszten, P.C.; Flickinger, J.C.; Sahgal, A.; Létourneau, D.; Grills, I.S.; Jawad, M.; Fahim, D.K.; Shin, J.H.; et al. Safety and efficacy of stereotactic body radiotherapy as primary treatment for vertebral metastases: A multi-institutional analysis. Radiat. Oncol. 2014, 9, 226. [Google Scholar] [CrossRef]
  70. Nieder, C.; Grosu, A.L.; Andratschke, N.H.; Molls, M. Update of human spinal cord reirradiation tolerance based on additional data from 38 patients. Int. J. Radiat. Oncol. Biol. Phys. 2006, 66, 1446–1449. [Google Scholar] [CrossRef] [PubMed]
  71. Mantel, F.; Flentje, M.; Guckenberger, M. Stereotactic body radiation therapy in the re-irradiation situation—A review. Radiat. Oncol. 2013, 8, 7. [Google Scholar] [CrossRef]
  72. Myrehaug, S.; Sahgal, A.; Hayashi, M.; Levivier, M.; Ma, L.; Martinez, R.; Paddick, I.; Régis, J.; Ryu, S.; Slotman, B.; et al. Reirradiation spine stereotactic body radiation therapy for spinal metastases: systematic review. J. Neurosurg. Spine 2017, 27, 428–435. [Google Scholar] [CrossRef]
  73. Choi, C.Y.H.; Adler, J.R.; Gibbs, I.C.; Chang, S.D.; Jackson, P.S.; Minn, A.Y.; Lieberson, R.E.; Soltys, S.G. Stereotactic radiosurgery for treatment of spinal metastases recurring in close proximity to previously irradiated spinal cord. Int. J. Radiat. Oncol. Biol. Phys. 2010, 78, 499–506. [Google Scholar] [CrossRef] [PubMed]
  74. Garg, A.K.; Wang, X.-S.; Shiu, A.S.; Allen, P.; Yang, J.; McAleer, M.F.; Azeem, S.; Rhines, L.D.; Chang, E.L. Prospective evaluation of spinal reirradiation by using stereotactic body radiation therapy. Cancer 2011, 117, 3509–3516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Damast, S.; Wright, J.; Bilsky, M.; Hsu, M.; Zhang, Z.; Lovelock, M.; Cox, B.; Zatcky, J.; Yamada, Y. Impact of dose on local failure rates after image-guided reirradiation of recurrent paraspinal metastases. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, 819–826. [Google Scholar] [CrossRef] [PubMed]
  76. Mahadevan, A.; Floyd, S.; Wong, E.; Jeyapalan, S.; Groff, M.; Kasper, E. Stereotactic body radiotherapy reirradiation for recurrent epidural spinal metastases. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, 1500–1505. [Google Scholar] [CrossRef]
  77. Hashmi, A.; Guckenberger, M.; Kersh, R.; Gerszten, P.C.; Mantel, F.; Grills, I.S.; Flickinger, J.C.; Shin, J.H.; Fahim, D.K.; Winey, B.; et al. Re-irradiation stereotactic body radiotherapy for spinal metastases: a multi-institutional outcome analysis. J. Neurosurg. Spine 2016, 25, 646–653. [Google Scholar] [CrossRef] [PubMed]
  78. Kirkpatrick, J.P.; van der Kogel, A.J.; Schultheiss, T.E. Radiation Dose-Volume Effects in the Spinal Cord. Int. J. Radiat. Oncol. Biol. Phys. 2010, 76, S42–S49. [Google Scholar] [CrossRef] [PubMed]
  79. Rades, D.; Stalpers, L.J.A.; Veninga, T.; Hoskin, P.J. Spinal reirradiation after short-course RT for metastatic spinal cord compression. Int. J. Radiat. Oncol. Biol. Phys. 2005, 63, 872–875. [Google Scholar] [CrossRef] [PubMed]
  80. Nieder, C.; Grosu, A.L.; Andratschke, N.H.; Molls, M. Proposal of human spinal cord reirradiation dose based on collection of data from 40 patients. Int. J. Radiat. Oncol. Biol. Phys. 2005, 61, 851–855. [Google Scholar] [CrossRef] [PubMed]
  81. Laufer, I.; Rubin, D.G.; Lis, E.; Cox, B.W.; Stubblefield, M.D.; Yamada, Y.; Bilsky, M.H. The NOMS Framework: Approach to the Treatment of Spinal Metastatic Tumors. Oncologist 2013, 18, 744–751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Zuckerman, S.L.; Lim, J.; Yamada, Y.; Bilsky, M.H.; Laufer, I. Brachytherapy in Spinal Tumors: A Systematic Review. World Neurosurg. 2018, 118, e235–e244. [Google Scholar] [CrossRef]
  83. Cardoso, E.R.; Ashamalla, H.; Weng, L.; Mokhtar, B.; Ali, S.; Macedon, M.; Guirguis, A. Percutaneous tumor curettage and interstitial delivery of samarium-153 coupled with kyphoplasty for treatment of vertebral metastases. J. Neurosurg. Spine 2009, 10, 336–342. [Google Scholar] [CrossRef] [PubMed]
  84. Yang, Z.; Tan, J.; Zhao, R.; Wang, J.; Sun, H.; Wang, X.; Xu, L.; Jiang, H.; Zhang, J. Clinical Investigations on the Spinal Osteoblastic Metastasis Treated by Combination of Percutaneous Vertebroplasty and 125I Seeds Implantation Versus Radiotherapy. Cancer Biother. Radiopharm. 2013, 28, 58–64. [Google Scholar] [CrossRef] [Green Version]
  85. Cao, Q.; Wang, H.; Meng, N.; Jiang, Y.; Jiang, P.; Gao, Y.; Tian, S.; Liu, C.; Yang, R.; Wang, J.; et al. CT-guidance interstitial 125Iodine seed brachytherapy as a salvage therapy for recurrent spinal primary tumors. Radiat. Oncol. 2014, 9, 301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Li, T.; Li, J.; Wang, Z.; Liu, B.; Han, D.; Wang, P. A preliminary comparative clinical study of vertebroplasty with multineedle or single-needle interstitial implantation of 125I seeds in the treatment of osteolytic metastatic vertebral tumors. J. Neurosurg. Spine 2014, 20, 430–435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Wang, S.; Shi, G.; Meng, X. Clinical curative effect of percutaneous vertebroplasty combined with 125I-seed implantation in treating spinal metastatic tumor. Pak. J. Pharm. Sci. 2015, 28, 1039–1042. [Google Scholar] [PubMed]
  88. Qian, J.; Bao, Z.; Zou, J.; Yang, H. Effect of pedicle fixation combined with 125I seed implantation for metastatic thoracolumbar tumors. J. Pain Res. 2016, 9, 271–278. [Google Scholar] [PubMed] [Green Version]
  89. Huang, H.; Xu, S.; Du, Z.; Li, F.; Wang, L. Treatment of metastatic thoracolumbar tumors by percutaneous vertebroplasty combined with interstitial implantation of 125I seeds. Zhonghua Zhong Liu Za Zhi 2014, 36, 228–231. [Google Scholar]
  90. Chow, E.; Ling, A.; Davis, L.; Panzarella, T.; Danjoux, C. Pain flare following external beam radiotherapy and meaningful change in pain scores in the treatment of bone metastases. Radiother. Oncol. 2005, 75, 64–69. [Google Scholar] [CrossRef] [PubMed]
  91. Hird, A.; Chow, E.; Zhang, L.; Wong, R.; Wu, J.; Sinclair, E.; Danjoux, C.; Tsao, M.; Barnes, E.; Loblaw, A. Determining the Incidence of Pain Flare Following Palliative Radiotherapy for Symptomatic Bone Metastases: Results From Three Canadian Cancer Centers. Int. J. Radiat. Oncol. Biol. Phys. 2009, 75, 193–197. [Google Scholar] [CrossRef]
  92. McDonald, R.; Chow, E.; Rowbottom, L.; De Angelis., C.; Soliman, H. Incidence of pain flare in radiation treatment of bone metastases: A literature review. J. Bone Oncol. 2014, 3, 84–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Gomez-Iturriaga, A.; Cacicedo, J.; Navarro, A.; Morillo, V.; Willisch, P.; Carvajal, C.; Hortelano, E.; Lopez-Guerra, J.L.; Illescas, A.; Casquero, F.; et al. Incidence of pain flare following palliative radiotherapy for symptomatic bone metastases: Multicenter prospective observational study. BMC Palliat. Care 2015, 14, 48. [Google Scholar] [CrossRef] [PubMed]
  94. Ma, Y.-B.; Yan, W.-L.; Dai, J.-C.; Xu, F.; Yuan, Q.; Shi, H.-H. Strontium-89: A desirable therapeutic for bone metastases of prostate cancer. Zhonghua Nan Ke Xue 2008, 14, 819–822. (In Chinese) [Google Scholar] [PubMed]
  95. Ogawa, K.; Washiyama, K. Bone Target Radiotracers for Palliative Therapy of Bone Metastases. Curr. Med. Chem. 2012, 19, 3290–3300. [Google Scholar] [CrossRef] [PubMed]
  96. Serafini, A.N. Therapy of metastatic bone pain. J. Nucl. Med. 2001, 42, 895–906. [Google Scholar] [PubMed]
  97. Finlay, I.G.; Mason, M.D.; Shelley, M. Radioisotopes for the palliation of metastatic bone cancer: A systematic review. Lancet Oncol. 2005, 6, 392–400. [Google Scholar] [CrossRef]
  98. Pandit-Taskar, N.; Batraki, M.; Divgi, C.R. Radiopharmaceutical therapy for palliation of bone pain from osseous metastases. J. Nucl. Med. 2004, 45, 1358–1365. [Google Scholar] [PubMed]
  99. Paes, F.M.; Serafini, A.N. Systemic Metabolic Radiopharmaceutical Therapy in the Treatment of Metastatic Bone Pain. Semin. Nucl. Med. 2010, 40, 89–104. [Google Scholar] [CrossRef] [PubMed]
  100. Silberstein, E.B.; Taylor, A.T. EANM procedure guidelines for treatment of refractory metastatic bone pain. Eur. J. Nucl. Med. Mol. Imaging 2003, 30, BP7–BP11. [Google Scholar]
  101. Bodei, L.; Lam, M.; Chiesa, C.; Flux, G.; Brans, B.; Chiti, A.; Giammarile, F. EANM procedure guideline for treatment of refractory metastatic bone pain. Eur. J. Nucl. Med. Mol. Imaging 2008, 35, 1934–1940. [Google Scholar] [CrossRef]
  102. Choi, J.Y. Treatment of Bone Metastasis with Bone-Targeting Radiopharmaceuticals. Nucl. Med. Mol. Imaging 2018, 52, 200–207. [Google Scholar] [CrossRef] [PubMed]
  103. Fettich, J.; Padhy, A.; Nair, N.; Morales, R.; Tanumihardja, M.; Riccabonna, G.; Nair, G. Comparative clinical efficacy and safety of Phosphorus-32 and Strontium-89 in the palliative treatment of metastatic bone pain: Results of an IAEA Coordinated Research Project. World J. Nucl. Med. 2003, 34, 226–231. [Google Scholar]
  104. Sciuto, R.; Festa, A.; Pasqualoni, R.; Semprebene, A.; Rea, S.; Bergomi, S.; Maini, C.L. Metastatic bone pain palliation with 89-Sr and 186-Re-HEDP in breast cancer patients. Breast Cancer Res. Treat. 2001, 66, 101–109. [Google Scholar] [CrossRef] [PubMed]
  105. Turner, S.L.; Gruenewald, S.; Spry, N.; Gebski, V. Less pain does equal better quality of life following Strontium-89 therapy for metastatic prostate cancer. Br. J. Cancer 2001, 84, 297–302. [Google Scholar] [CrossRef] [PubMed]
  106. Dafermou, A.; Colamussi, P.; Giganti, M.; Cittanti, C.; Bestagno, M.; Piffanelli, A. A multicentre observational study of radionuclide therapy in patients with painful bone metastases of prostate cancer. Eur. J. Nucl. Med. 2001, 28, 788–798. [Google Scholar] [CrossRef] [PubMed]
  107. Ashayeri, E.; Omogbehin, A.; Sridhar, R.; Shankar, R.A. Strontium 89 in the treatment of pain due to diffuse osseous metastases: a university hospital experience. J. Natl. Med. Assoc. 2002, 94, 706–711. [Google Scholar]
  108. Baczyk, M.; Milecki, P.; Baczyk, E.; Sowiński, J. The effectivness of strontium 89 in palliative therapy of painful prostate cancer bone metastases. Ortop. Traumatol. Rehabil. 2003, 5, 364–368. [Google Scholar] [PubMed]
  109. Liepe, K.; Kotzerke, J. A comparative study of Re-HEDP, Re-HEDP, Sm-EDTMP and Sr in the treatment of painful skeletal metastases. Nucl. Med. Commun. 2007, 28, 623–630. [Google Scholar] [CrossRef]
  110. Zenda, S.; Nakagami, Y.; Toshima, M.; Arahira, S.; Kawashima, M.; Matsumoto, Y.; Kinoshita, H.; Satake, M.; Akimoto, T. Strontium-89 (Sr-89) chloride in the treatment of various cancer patients with multiple bone metastases. Int. J. Clin. Oncol. 2014, 19, 739–743. [Google Scholar] [CrossRef]
  111. Laing, A.H.; Ackery, D.M.; Bayly, R.J.; Buchanan, R.B.; Lewington, V.J.; McEwan, A.J.B.; Macleod, P.M.; Zivanovic, M.A. Strontium-89 chloride for pain palliation in prostatic skeletal malignancy. Br. J. Radiol. 1991, 64, 817–822. [Google Scholar] [CrossRef] [PubMed]
  112. Silberstein, E.B.; Williams, C. Strontium-89 therapy for the pain of osseous metastases. J. Nucl. Med. 1985, 26, 345–348. [Google Scholar] [PubMed]
  113. Mertens, W.C.; Stitt, L.; Porter, A.T. Strontium 89 therapy and relief of pain in patients with prostatic carcinoma metastatic to bone: A dose response relationship? Am. J. Clin. Oncol. Cancer Clin. Trials 1993, 16, 238–242. [Google Scholar] [CrossRef]
  114. Dolezal, J. Systemic radionuclide therapy with Samarium-153-EDTMP for painful bone metastases. Nucl. Med. Rev. Cent. East. Eur. 2000, 3, 161–163. [Google Scholar] [PubMed]
  115. Wang, R.F.; Zhang, C.L.; Zhu, S.L.; Zhu, M. A comparative study of samarium-153-ethylenediaminetetramethylene phosphonic acid with pamidronate disodium in the treatment of patients with painful metastatic bone cancer. Med. Princ. Pract. 2003, 12, 97–101. [Google Scholar] [CrossRef] [PubMed]
  116. Sapienza, M.T.; Ono, C.R.; Guimarães, M.I.C.; Watanabe, T.; Costa, P.A.; Buchpiguel, C.A. Retrospective evaluation of bone pain palliation after samarium-153-EDTMP therapy. Rev. Hosp. Clin. Fac. Med. Sao Paulo 2004, 59, 321–328. [Google Scholar] [CrossRef] [PubMed]
  117. Sá de Camargo Etchebehere, E.C.; Cunha Pereira Neto, C.A.; Lopes de Lima, M.C.; de Oliveira Santos, A.; Ramos, C.D.; Silva, C.M.; Camargo, E.E. Treatment of bone pain secondary to metastases using samarium-153-EDTMP. Sao Paulo Med. J. 2004, 122, 208–212. [Google Scholar] [CrossRef]
  118. Sartor, O.; Reid, R.H.; Hoskin, P.J.; Quick, D.P.; Ell, P.J.; Coleman, R.E.; Kotler, J.A.; Freeman, L.M.; Olivier, P. Samarium-153-lexidronam complex for treatment of painful bone metastases in hormone-refractory prostate cancer. Urology 2004, 63, 940–945. [Google Scholar] [CrossRef]
  119. Tripathi, M.; Singhal, T.; Chandrasekhar, N.; Kumar, P.; Bal, C.; Jhulka, P.K.; Bandopadhyaya, G.; Malhotra, A. Samarium-153 ethylenediamine tetramethylene phosphonate therapy for bone pain palliation in skeletal metastases. Indian J. Cancer 2006, 43, 86–92. [Google Scholar] [CrossRef]
  120. Ripamonti, C.; Fagnoni, E.; Campa, T.; Seregni, E.; Maccauro, M.; Bombardieri, E. Incident pain and analgesic consumption decrease after samarium infusion: A pilot study. Support. Care Cancer 2007, 15, 339–342. [Google Scholar] [CrossRef]
  121. Dolezal, J.; Vizda, J.; Odrazka, K. Prospective evaluation of samarium-153-EDTMP radionuclide treatment for bone metastases in patients with hormone-refractory prostate cancer. Urol. Int. 2007, 78, 50–57. [Google Scholar] [CrossRef]
  122. Resche, I.; Chatal, J.F.; Pecking, A.; Ell, P.; Duchesne, G.; Rubens, R.; Fogelman, I.; Houston, S.; Fauser, A.; Fischer, M.; et al. A dose-controlled study of 153Sm-ethylenediaminetetramethylenephosphonate (EDTMP) in the treatment of patients with painful bone metastases. Eur. J. Cancer 1997, 33, 1583–1591. [Google Scholar] [CrossRef]
  123. Serafini, A.N.; Houston, S.J.; Resche, I.; Quick, D.P.; Grund, F.M.; Ell, P.J.; Bertrand, A.; Ahmann, F.R.; Orihuela, E.; Reid, R.H.; et al. Palliation of pain associated with metastatic bone cancer using samarium-153 lexidronam: A double-blind placebo-controlled clinical trial. J. Clin. Oncol. 1998, 16, 1574–1581. [Google Scholar] [CrossRef]
  124. Tian, J.H.; Zhang, J.M.; Hou, Q.T.; Oyang, Q.H.; Wang, J.M.; Luan, Z.S.; Chuan, L.; He, Y.J. Multicentre trial on the efficacy and toxicity of single-dose samarium-153-ethylene diamine tetramethylene phosphonate as a palliative treatment for painful skeletal metastases in China. Eur. J. Nucl. Med. 1999, 26, 2–7. [Google Scholar] [CrossRef]
  125. Parker, C.; Nilsson, S.; Heinrich, D.; Helle, S.I.; O’Sullivan, J.M.; Fosså, S.D.; Chodacki, A.; Wiechno, P.; Logue, J.; Seke, M.; et al. Alpha Emitter Radium-223 and Survival in Metastatic Prostate Cancer. N. Engl. J. Med. 2013, 369, 213–223. [Google Scholar] [CrossRef]
  126. Parker, C.; Zhan, L.; Cislo, P.; Reuning-Scherer, J.; Vogelzang, N.J.; Nilsson, S.; Sartor, O.; O’Sullivan, J.M.; Coleman, R.E. Effect of radium-223 dichloride (Ra-223) on hospitalisation: An analysis from the phase 3 randomised Alpharadin in Symptomatic Prostate Cancer Patients (ALSYMPCA) trial. Eur. J. Cancer 2017, 71, 1–6. [Google Scholar] [CrossRef] [Green Version]
  127. Parker, C.; Heinrich, D.; O’Sullivan, J.M.; Fossa, S.; Chodacki, A.; Demkow, T.; Cross, A.; Bolstad, B.; Garcia-Vargas, J.; Sartor, O. Sartor Overall survival benefit of radium-223 chloride (Alpharadin) in the treatment of patients with symptomatic bone metastases in Castration-resistant Prostate Cancer (CRPC): A phase III randomized trial (ALSYMPCA). Eur. J. Cancer 2011, 47, 3. [Google Scholar] [CrossRef]
  128. Nilsson, S.; Sartor, A.O.; Bruland, O.S.; Fang, F.; Aksnes, A.-K.; Parker, C. Pain analyses from the phase III randomized ALSYMPCA study with radium-223 dichloride (Ra-223) in castration-resistant prostate cancer (CRPC) patients with bone metastases. J. Clin. Oncol. 2013, 31, 5038. [Google Scholar] [CrossRef]
  129. Quirijnen, J.M.; Han, S.H.; Zonnenberg, B.A.; Klerk, J.M.; het Schip, A.D.; Dijk, A.; Kroode, H.F.; Blijham, G.H.; Rijk, P.P. Efficacy of rhenium-186-etidronate in prostate cancer patients with metastatic bone pain. J. Nucl. Med. 1996, 37, 1511–1515. [Google Scholar]
  130. Maxon, H.R.; Schroder, L.E.; Thomas, S.R.; Hertzberg, V.S.; Deutsch, E.A.; Scher, H.I.; Samaratunga, R.C.; Libson, K.F.; Williams, C.C.; Moulton, J.S. Re-186(Sn) HEDP for treatment of painful osseous metastases: initial clinical experience in 20 patients with hormone-resistant prostate cancer. Radiology 1990, 176, 155–159. [Google Scholar] [CrossRef]
  131. Giannakenas, C.; Kalofonos, H.P.; Apostolopoulos, D.J.; Zarakovitis, J.; Kosmas, C.; Vassilakos, P.J. Preliminary results of the use of Re-186-HEDP for palliation of pain in patients with metastatic bone disease. Am. J. Clin. Oncol. 2000, 23, 83–88. [Google Scholar] [CrossRef]
  132. Kolesnikov-Gauthier, H.; Carpentier, P.; Depreux, P.; Vennin, P.; Caty, A.; Sulman, C. Evaluation of toxicity and efficacy of 186Re-hydroxyethylidene diphosphonate in patients with painful bone metastases of prostate or breast cancer. J. Nucl. Med. 2000, 41, 1689–1694. [Google Scholar]
  133. Sciuto, R.; Tofani, A.; Festa, A.; Giannarelli, D.; Pasqualoni, R.; Maini, C.L. Short- and long-term effects of 186Re-1,1-hydroxyethylidene diphosphonate in the treatment of painful bone metastases. J. Nucl. Med. 2000, 41, 647–654. [Google Scholar] [PubMed]
  134. Paes, F.M.; Ernani, V.; Hosein, P.; Serafini, A.N. Radiopharmaceuticals: When and how to use them to treat metastatic bone pain. J. Support. Oncol. 2011, 9, 197–205. [Google Scholar] [CrossRef] [PubMed]
  135. Liepe, K.; Kotzerke, J. Internal radiotherapy of painful bone metastases. Methods 2011, 55, 258–270. [Google Scholar] [CrossRef] [PubMed]
  136. Maxon III, H.R.; Schroder, L.E.; Hertzberg, V.S.; Thomas, S.R.; Englaro, E.E.; Samaratunga, R.; Smith, H.; Moulton, J.S.; Williams, C.C.; Ehrhardt, G.J.; et al. Rhenium-186(Sn)HEDP for treatment of painful osseous metastases: Results of a double-blind crossover comparison with placebo. J. Nucl. Med. 1991, 32, 1877–1881. [Google Scholar]
  137. Maxon, H.R.; Thomas, S.R.; Hertzberg, V.S.; Schroder, L.E.; Englaro, E.E.; Samaratunga, R.; Scher, H.I.; Moulton, J.S.; Deutsch, E.A.; Deutsch, K.F.; et al. Rhenium-186 hydroxyethylidene diphosphonate for the treatment of painful osseous metastases. Semin. Nucl. Med. 1992, 22, 33–40. [Google Scholar] [CrossRef]
  138. Han, S.H.; Zonneberg, B.A.; de Klerk, J.M.; Quirijnen, J.M.; van het Schip, A.D.; van Dijk, A.; Blijham, G.H.; van Rijk, P.P. 186Re-etidronate in breast cancer patients with metastatic bone pain. J. Nucl. Med. 1999, 40, 639–642. [Google Scholar]
  139. Han, S.H.; de Klerk, J.M.H.; Tan, S.; van het Schip, A.D.; Derksen, B.H.; van Dijk, A.; Kruitwagen, C.L.J.J.; Blijham, G.H.; van Rijk, P.P.; Zonnenberg, B.A. The PLACORHEN study: A double-blind, placebo-controlled, randomized radionuclide study with (186)Re-etidronate in hormone-resistant prostate cancer patients with painful bone metastases. Placebo Controlled Rhenium Study. J. Nucl. Med. 2002, 43, 1150–1156. [Google Scholar]
  140. Fuster, D.; Herranz, R.; Alcover, J.; Mateos, J.J.; Martín, F.; Vidal-Sicart, S.; Pons, F. Treatment of metastatic bone pain with repeated doses of strontium-89 in patients with prostate neoplasm. Rev. Esp. Med. Nucl. 2000, 19, 270–274. [Google Scholar] [CrossRef]
  141. Sartor, O.; Reid, R.H.; Bushnell, D.L.; Quick, D.P.; Ell, P.J. Safety and efficacy of repeat administration of samarium Sm-153 lexidronam to patients with metastatic bone pain. Cancer 2007, 109, 637–643. [Google Scholar] [CrossRef] [Green Version]
  142. Palmedo, H.; Guhlke, S.; Bender, H.; Sartor, J.; Schoeneich, G.; Risse, J.; Grünwald, F.; Knapp, F.F.; Biersack, H.J. Dose escalation study with rhenium-188 hydroxyethylidene diphosphonate in prostate cancer patients with osseous metastases. Eur. J. Nucl. Med. 2000, 27, 123–130. [Google Scholar] [CrossRef]
  143. Liepe, K.; Hliscs, R.; Kropp, J.; Gruning, T.; Runge, R.; Koch, R.; Knapp, F.F.J.; Franke, W.G. Rhenium-188-HEDP in the palliative treatment of bone metastases. Cancer Biother. Radiopharm. 2000, 15, 261–265. [Google Scholar] [CrossRef]
  144. Palmedo, H.; Manka-Waluch, A.; Albers, P.; Schmidt-Wolf, I.G.H.; Reinhardt, M.; Ezziddin, S.; Joe, A.; Roedel, R.; Fimmers, R.; Knapp, F.F.; et al. Repeated bone-targeted therapy for hormone-refractory prostate carcinoma: Randomized phase II trial with the new, high-energy radiopharmaceutical rhenium-188 hydroxyethylidenediphosphonate. J. Clin. Oncol. 2003, 21, 2869–2875. [Google Scholar] [CrossRef]
  145. Nilsson, S.; Larsen, R.H.; Fosså, S.D.; Balteskard, L.; Borch, K.W.; Westlin, J.E.; Salberg, G.; Bruland, Ø.S. First clinical experience with α-emitting radium-223 in the treatment of skeletal metastases. Clin. Cancer Res. 2005, 11, 4451–4459. [Google Scholar] [CrossRef]
  146. Bruland, Ø.S.; Nilsson, S.; Fisher, D.R.; Larsen, R.H. High-linear energy transfer irradiation targeted to skeletal metastases by the α-emitter 223Ra: Adjuvant or alternative to conventional modalities? Clin. Cancer Res. 2006, 12, 6250–6258. [Google Scholar] [CrossRef]
  147. Nilsson, S.; Franzén, L.; Parker, C.; Tyrrell, C.; Blom, R.; Tennvall, J.; Lennernäs, B.; Petersson, U.; Johannessen, D.C.; Sokal, M.; et al. Bone-targeted radium-223 in symptomatic, hormone-refractory prostate cancer: A randomised, multicentre, placebo-controlled phase II study. Lancet Oncol. 2007, 8, 587–594. [Google Scholar] [CrossRef]
  148. Coleman, R.; Aksnes, A.K.; Naume, B.; Garcia, C.; Jerusalem, G.; Piccart, M.; Vobecky, N.; Thuresson, M.; Flamen, P. A phase IIa, nonrandomized study of radium-223 dichloride in advanced breast cancer patients with bone-dominant disease. Breast Cancer Res. Treat. 2014, 145, 411–418. [Google Scholar] [CrossRef] [Green Version]
  149. Prelaj, A.; Rebuzzi, S.E.; Buzzacchino, F.; Pozzi, C.; Ferrara, C.; Frantellizzi, V.; Follacchio, G.A.; Civitelli, L.; De Vincentis, G.; Tomao, S.; et al. Radium-223 in patients with metastatic castration-resistant prostate cancer: Efficacy and safety in clinical practice. Oncol. Lett. 2019, 17, 1467–1476. [Google Scholar] [CrossRef]
  150. Fairchild, A.; Harris, K.; Barnes, E.; Wong, R.; Lutz, S.; Bezjak, A.; Cheung, P.; Chow, E. Palliative thoracic radiotherapy for lung cancer: A systematic review. J. Clin. Oncol. 2008, 26, 4001–4011. [Google Scholar] [CrossRef]
  151. Tey, J.; Choo, B.A.; Leong, C.N.; Loy, E.Y.; Wong, L.C.; Lim, K.; Lu, J.J.; Koh, W.Y. Clinical outcome of palliative radiotherapy for locally advanced symptomatic gastric cancer in the modern era. Medicine (Baltimore) 2014, 93, e118. [Google Scholar] [CrossRef]
  152. Rominger, C.J.; Gelber, R.D.; Gunderson, L.L.; Conner, N. Radiation therapy alone or in combination with chemotherapy in the treatment of residual or inoperable carcinoma of the rectum and rectosigmoid or pelvic recurrence following colorectal surgery. Radiation therapy oncology group study (76-16). Am. J. Clin. Oncol. Cancer Clin. Trials 1985, 8, 118–127. [Google Scholar] [CrossRef]
  153. Bae, S.H.; Park, W.; Choi, D.H.; Nam, H.; Kang, W.K.; Park, Y.S.; Park, J.O.; Chun, H.K.; Lee, W.Y.; Yun, S.H.; et al. Palliative radiotherapy in patients with a symptomatic pelvic mass of metastatic colorectal cancer. Radiat. Oncol. 2011, 6, 52. [Google Scholar] [CrossRef]
  154. Rosati, L.M.; Herman, J.M. Role of Stereotactic Body Radiotherapy in the Treatment of Elderly and Poor Performance Status Patients With Pancreatic Cancer. J. Oncol. Pract. 2017, 13, 157–166. [Google Scholar] [CrossRef] [PubMed]
  155. Ng, S.P.; Koay, E.J. Current and emerging radiotherapy strategies for pancreatic adenocarcinoma: stereotactic, intensity modulated and particle radiotherapy. Ann. Pancreat. Cancer 2018, 1, 22. [Google Scholar] [CrossRef]
  156. Ryan, J.F.; Rosati, L.M.; Groot, V.P.; Le, D.T.; Zheng, L.; Laheru, D.A.; Shin, E.J.; Jackson, J.; Moore, J.; Narang, A.K.; et al. Stereotactic body radiation therapy for palliative management of pancreatic adenocarcinoma in elderly and medically inoperable patients. Oncotarget 2018, 9, 16427–16436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Lam, T.C.; Tseng, Y. Defining the radiation oncologist’s role in palliative care and radiotherapy. Ann. Palliat. Med. 2018, 7, 1002. [Google Scholar] [CrossRef]
  158. Wang, X.S.; Rhines, L.D.; Shiu, A.S.; Yang, J.N.; Selek, U.; Gning, I.; Liu, P.; Allen, P.K.; Azeem, S.S.; Brown, P.D.; et al. Stereotactic body radiation therapy for management of spinal metastases in patients without spinal cord compression: A phase 1-2 trial. Lancet Oncol. 2012, 13, 395–402. [Google Scholar] [CrossRef]
  159. Chow, E.; Hoskin, P.; Mitera, G.; Zeng, L.; Lutz, S.; Roos, D.; Hahn, C.; Van Der Linden, Y.; Hartsell, W.; Kumar, E. Update of the international consensus on palliative radiotherapy endpoints for future clinical trials in bone metastases. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, 1730–1737. [Google Scholar] [CrossRef] [PubMed]
  160. NCT03143322 Standard Treatment +/− SBRT in Solid Tumors Patients With Between 1 and 3 Bone-only Metastases. Available online: https://www.cochranelibrary.com/central/doi/10.1002/central/CN-01580971/full (accessed on 3 March 2019).
  161. Biological Image Guided Antalgic Stereotactic Body Radiotherapy of Bone Metastases. Available online: https://clinicaltrials.gov/ct2/show/NCT01429493 (accessed on 3 March 2019).
  162. Sprave, T.; Welte, S.E.; Bruckner, T.; Förster, R.; Bostel, T.; Schlampp, I.; Nicolay, N.H.; Debus, J.; Rief, H. Intensity-modulated radiotherapy with integrated-boost in patients with bone metastasis of the spine: Study protocol for a randomized controlled trial. Trials 2018, 19, 59. [Google Scholar] [CrossRef] [PubMed]
  163. Randomized Trial Comparing Conventional Radiotherapy With Stereotactic Radiotherapy in Patients With Bone Metastases—VERTICAL Study (VERTICAL). Available online: https://clinicaltrials.gov/ct2/show/NCT02364115 (accessed on 3 March 2019).
  164. Prospective Evaluation of Interventional Studies on Bone Metastases—The PRESENT Cohort. Available online: https://clinicaltrials.gov/ct2/show/NCT02356497 (accessed on 3 March 2019).
  165. Leiden University Medical, C. The OPTIMAL Study—A Prospective Cohort of Patients With Bone Metastases of the Long Bones. Available online: https://clinicaltrials.gov/show/NCT02705157 (accessed on 3 March 2019).
  166. Radiosurgical Hypophysectomy for Bone Metasteses Pain. Available online: https://clinicaltrials.gov/ct2/show/NCT03377517 (accessed on 3 March 2019).
  167. Habberstad, R.; Frøseth, T.C.S.; Aass, N.; Abramova, T.; Baas, T.; Mørkeset, S.T.; Caraceni, A.; Laird, B.; Boland, J.W.; Rossi, R.; et al. The Palliative Radiotherapy and Inflammation Study (PRAIS)—protocol for a longitudinal observational multicenter study on patients with cancer induced bone pain. BMC Palliat. Care 2018, 17, 110. [Google Scholar] [CrossRef] [PubMed]
  168. Furfari, A.; Wan, B.A.; Ding, K.; Wong, A.; Zhu, L.; Bezjak, A.; Wong, R.; Wilson, C.F.; DeAngelis, C.; Azad, A.; et al. Genetic biomarkers associated with response to palliative radiotherapy in patients with painful bone metastases. Ann. Palliat. Med. 2017, 6, S233–S239. [Google Scholar] [CrossRef] [PubMed]
  169. Furfari, A.; Wan, B.A.; Ding, K.; Wong, A.; Zhu, L.; Bezjak, A.; Wong, R.; Wilson, C.F.; DeAngelis, C.; Azad, A.; et al. Genetic biomarkers associated with changes in quality of life and pain following palliative radiotherapy in patients with bone metastases. Ann. Palliat. Med. 2017, 6, S248–S256. [Google Scholar] [CrossRef]
  170. Furfari, A.; Wan, B.A.; Ding, K.; Wong, A.; Zhu, L.; Bezjak, A.; Wong, R.; Wilson, C.F.; DeAngelis, C.; Azad, A.; et al. Genetic biomarkers associated with pain flare and dexamethasone response following palliative radiotherapy in patients with painful bone metastases. Ann. Palliat. Med. 2017, 6, S240–S247. [Google Scholar] [CrossRef] [PubMed]
  171. Rosen, L.S.; Gordon, D.; Tchekmedyian, S.; Yanagihara, R.; Hirsh, V.; Krzakowski, M.; Pawlicki, M.; de Souza, P.; Zheng, M.; Urbanowitz, G.; et al. Zoledronic acid versus placebo in the treatment of skeletal metastases in patients with lung cancer and other solid tumors: A phase III, double-blind, randomized trial—The Zoledronic Acid Lung Cancer and Other Solid Tumors Study Group. J. Clin. Oncol. 2003, 21, 3150–3157. [Google Scholar] [CrossRef] [PubMed]
  172. Safety, Tolerability and PK/PD of JMT103 in Patients With Bone Metastases From Tumors. Available online: https://www.clinicaltrials.gov/ct2/show/NCT03550508?cond=Bone+Metastases&draw=2&rank=1 (accessed on 3 March 2019).
  173. Sousa, S.; Clézardin, P. Bone-Targeted Therapies in Cancer-Induced Bone Disease. Calcif. Tissue Int. 2018, 102, 227–250. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Simulation fields (pink rectangle) for irradiation of painful bone metastases with megavoltage photon external beams: (A) humeral bone, (B) spine, (C) foot bones.
Figure 1. Simulation fields (pink rectangle) for irradiation of painful bone metastases with megavoltage photon external beams: (A) humeral bone, (B) spine, (C) foot bones.
Cancers 11 00390 g001
Figure 2. Isodose distribution in three-dimensional conformal radiation therapy (3D-CRT) for painful metastasis in dorsal vertebra: (A) one beam, (B) two oblique beams, (C) volumetrically modulated arch therapy (VMAT).
Figure 2. Isodose distribution in three-dimensional conformal radiation therapy (3D-CRT) for painful metastasis in dorsal vertebra: (A) one beam, (B) two oblique beams, (C) volumetrically modulated arch therapy (VMAT).
Cancers 11 00390 g002
Figure 3. Simulation fields (pink rectangle) for irradiation of painful bone metastases with megavoltage photon external beams. (A,B)—metastases to the femur after surgical stabilization, (C)—metastasis to the vertebral body after percutaneous vertebroplasty.
Figure 3. Simulation fields (pink rectangle) for irradiation of painful bone metastases with megavoltage photon external beams. (A,B)—metastases to the femur after surgical stabilization, (C)—metastasis to the vertebral body after percutaneous vertebroplasty.
Cancers 11 00390 g003
Figure 4. Usage of radioactive isotopes for multiple bone metastases. Abbreviations: 223Ra—Radium-223, 89Sr—Strontium-89, 153Sm—Samarium-153, α—alpha particle, β—beta-minus particle.
Figure 4. Usage of radioactive isotopes for multiple bone metastases. Abbreviations: 223Ra—Radium-223, 89Sr—Strontium-89, 153Sm—Samarium-153, α—alpha particle, β—beta-minus particle.
Cancers 11 00390 g004
Figure 5. Local bone analgesic activity of alpha radiation delivered with Radium-223-dichloride.
Figure 5. Local bone analgesic activity of alpha radiation delivered with Radium-223-dichloride.
Cancers 11 00390 g005
Table 1. Factors influencing the radiation technique and fractionation schemes in the treatment of painful bone metastases in clinical practice.
Table 1. Factors influencing the radiation technique and fractionation schemes in the treatment of painful bone metastases in clinical practice.
Patient-Related FactorsTumour-Related FactorsLogistic Issues
  • Performance status
  • Patient’s mobility
  • Clinical circumstances
    • painful uncomplicated bone metastases
    • pathologic fractures
    • spinal cord compression
    • re-irradiation
  • Compliance to treatment
  • Life expectancy
  • Socioeconomic status
  • Patient’s preferences
  • Pain intensity
  • Histologic type of primary tumour
  • Time elapsed from primary diagnosis to bone metastases
  • Multiplicity of metastases
  • Time of developing pain or neurologic deficits before RT 1
  • Treatment duration
  • Significance of family members’ assistance
  • Hospital location
  • Distance from patient’s home to radiation therapy department
  • Cost of therapy
  • Reimbursement issues
  • Organization of radiation departments
  • Availability of multidisciplinary tumour board
1 Radiation therapy.
Table 2. Clinical studies demonstrating the analgesic effect of radiation therapy on painful bone metastases depending on dose and fractionation regimen.
Table 2. Clinical studies demonstrating the analgesic effect of radiation therapy on painful bone metastases depending on dose and fractionation regimen.
TrialNumber of PatientsFractionationComplete or Partial Pain ResponseComplete Pain Response
Price et al., 1986 [29]2881 × 8 Gy
10 × 3 Gy
73%
64%
45%
28%
Cole et al., 1989 [30]291 × 8 Gy
6 × 4 Gy
88%
85%
NR 2
NR 2
Gaze et al., 1997 [26]2801 × 10 Gy
5 × 4.5 Gy
84%
89%
39%
48%
Nielsen et al., 1998 [25]2411 × 8 Gy
5 × 4 Gy
44%
46%
15%
17%
Steenland et al., 1999 [24]11711 × 8 Gy
6 × 4 Gy
72%
69%
37%
33%
Koswig et al., 1999 [17]1071 × 8 Gy
10 × 3 Gy
79%
82%
31%
33%
BPTWP 1 1999 [22]2721 × 8 Gy
5 × 4 Gy
72%
68%
52%
51%
Roos 2005 [31]2751 × 8 Gy
10 × 3 Gy
61%
53%
15%
18%
Hartsell et al., 2005 [32]9981 × 8 Gy
10 × 3 Gy
65%
66%
15%
18%
Foro Arnalot et al., 2008 [23]1601 × 8 Gy
10 × 3 Gy
75%
86%
15%
13%
Nongkynrih et al., 2018 [27]601 × 8 Gy
5 × 4 Gy
10 × 3 Gy
80%
75%
85%
20%
20%
20%
1 BPTWP—Bone Pain Trial Working Party, 2 NR—not reported.
Table 3. Pain response after re-irradiation of painful spine metastases [58,73,74,75,76,77].
Table 3. Pain response after re-irradiation of painful spine metastases [58,73,74,75,76,77].
StudyInitial RT Dose (Median)Re-Irradiation ModalityRe-Treatment DosePain Response
Choi et al., 2010 [73]40 Gy (24.2–50.4)CyberKnifeMedian marginal dose 20 Gy/2fr (range 18/1–25/5)
30 Gy/5fr
65% improvement in pain
Garg et al., 2011 [74]30 Gy (30–45)IG-IMRT27 Gy/3fr
20 Gy/5fr
Improvement in pain at 6 months
Damast et al., 2011 [75]30 Gy (8–66)IG-IMRT20 Gy/5fr
30 Gy/5fr
77% improvement in pain
Mahadevan et al., 2011 [76]30 Gy (8–46)CyberKnife25–30 Gy/5fr
24 Gy/3fr
79% improvement in pain
Chang et al., 2012 [58]39Gy Gy2 1CyberKnife20.6 Gy/1fr (18.2–23.7)80.8% pain control rate at 1 year
Hashmi et al., 2016 [77]30 Gy/10frIG-IMRT16.6 Gy/fr
24 Gy/3fr
74.3% improvement in pain
1 Gy is a way to normalize radiation doses that may have been given in a different dose/fractionation schedule. It converts all doses to the equivalent dose in 2 Gy per fraction. Abbreviations: fr—fraction, RT—radiation therapy, IG-IMRT—image-guided intensity modulated radiation therapy.
Table 4. Influence of brachytherapy on pain control in painful spine tumours [83,84,85,86,87,88,89]. 125I-iodine, 153Sm-samarium, SM—spinal metastases, VAS-visual-analogue scale of pain assessment.
Table 4. Influence of brachytherapy on pain control in painful spine tumours [83,84,85,86,87,88,89]. 125I-iodine, 153Sm-samarium, SM—spinal metastases, VAS-visual-analogue scale of pain assessment.
StudyBrachytherapy InterventionPain Control
VAS (Mean +/− Standard Deviation)
PretreatmentPosttreatment
Cardoso et al., 2009 [83]Percutaneous curettage of SM cement augmentation, and bone cement injection with 153Sm8.5 +/− 22.6 +/− 3.1
Yang et al., 2013 [84]Cement augmentation and percutaneous 125I seed implantation8.73 +/− 0.311.32 +/− 0.37
Cao et al., 2014 [85]Percutaneous 125I seed implantation4.48 +/− 2.031.18 +/− 1.38
Huang et al., 2014 [89]Cement augmentation and percutaneous 125I seed implantation7.12 +/− 1.482.26 +/− 1.07
Li et al., 2014 [86]Cement augmentation and percutaneous 125I seed implantation7.7 +/− 1.3 (SN)
8.0 +/− 1.2 (MN)
2.6 +/− 1.0
2.4 +/− 1.1
Wang et al., 2015 [87]Cement augmentation and percutaneous 125I seed implantation6.37 +/− 1.671.32 +/− 0.75
Qian et al., 2016 [88]Pedicle fixation of affected vertebra and implantation of 125I seeds via needles7.43 +/− 0.984.29 +/− 0.98
Abbreviations: 125I—iodine, 153Sm—samarium, SM—spinal metastases, VAS-visual-analogue scale of pain assessment.
Table 5. Radioactive isotopes used for the treatment of painful bone metastases.
Table 5. Radioactive isotopes used for the treatment of painful bone metastases.
Radioactive IsotopeTrialStudy TypeNumber of PatientsCancerAnalgesic EffectDuration of Analgesic Effect
Radium-223-ChlorideNilsson et al., 2005 [145]phase I25 ptsbreast, prostate50% pts1 WP 52%,
4 WP 60%,
8 WP 56%
Bruland et al., 2006 [146]phase I6 ptsprostaterepeated administration was well toleratedNS
Nilsson et al., 2007 [147]phase II randomized64 ptsprostate10 versus 16 (placebo) reported bone pain after injectionNS
Coleman et al., 2014 [148]phase IIa23 ptsbreastBPI pain severity index at week 17 was 0.6NS
Parker et al., 2011 ALSYMPCA [127]phase III randomized921 ptscastration-resistant prostateNSNS
Prelaj at al., 2019 [149]retrospective32 ptsprostate71%NS
Rhenium-186-HEDPMaxon et al., 1991 [136]double-blind20 ptsprostate80% ptsNS
Maxon et al., 1992 [137]prospective43 ptsbreast, prostate77% pts initial injection
50% second in injection
7 weeks
Han et al., 1999 [138]prospective30 ptsbreast58% ptsNS
Han et al., 2002
PLACORHEN [139]
double-blind randomized111 ptsprostate0–96% (mean 27%)NS
Rhenium-188-HEDPPalmedo et al., 2000 [142]prospective22 ptsprostate64%7.5 weeks
Liepe et al., 2000 [143]prospective15 ptsprostate76%NS
Samarium-153-EDTMPDolezal et al., 2000 [114]prospective33 ptsprostate, breast, other70%NS
Wang et al., 2003 [115]Comparative randomized9 ptsprostate, breast, other78%3.5 +/− 2.3 months
Sapienza et al., 2004 [116]retrospective73 ptsprostate, breast76%NS
Samarium-153-EDTMPEtchebehere et al., 2004 [117]retrospective58 ptsprostate, breast, other78%5.75–6 months
Sartor et al., 2004 [118]phase III randomized152 ptsprostate64%NS
Tripathi et al., 2006 [119]prospective86 ptsprostate, breast, other73%2–8 months
Ripamonti et al., 2007 [120]prospective13 ptsprostate, breast61,5%NS
Liepe et al., 2007 [109]prospective15 ptsprostate, breast73%10 +/− 1 weeks
Dolezal et al., 2007 [121]prospective32 ptsprostate72%3 months
Strontium-89 DichlorideSciuto. et al., 2001 [104]randomized51 ptsbreast 84%2–14 months
Turner et al., 2001 [105]prospective93 ptsprostate63%NS
Dafermou et al., 2001 [106]multicentre observational527 ptsprostate59.8%5.0 +/− 3.5 months
Ashayeri et al., 2002 [107]prospective27 ptsprostate, breast81%up to 1 year
Baczyk et al., 2003 [108]prospective70 ptsprostate88%3–12 months
Fettich et al., 2003 [103]prospective93 ptsbone mts75%NS
Liepe et al., 2007 [109]prospective15 ptsprostate, breast72%9 +/− 2 weeks
Ma et al., 2008 [94]prospective116 ptsprostate83.6%3–12 months
Zenda et al., 2014 [110]prospective54 pts26 pts prostate/breast
28 pts other malignancies (lung, head and neck, colorectal, other)
69.2%
73.1%
2–6 months
Abbreviations: PTS—patients, NS—not stated, WP—week point, BPI—Brief Pain Inventory, MTS—metastases.

Share and Cite

MDPI and ACS Style

Sierko, E.; Hempel, D.; Zuzda, K.; Wojtukiewicz, M.Z. Personalized Radiation Therapy in Cancer Pain Management. Cancers 2019, 11, 390. https://doi.org/10.3390/cancers11030390

AMA Style

Sierko E, Hempel D, Zuzda K, Wojtukiewicz MZ. Personalized Radiation Therapy in Cancer Pain Management. Cancers. 2019; 11(3):390. https://doi.org/10.3390/cancers11030390

Chicago/Turabian Style

Sierko, Ewa, Dominika Hempel, Konrad Zuzda, and Marek Z. Wojtukiewicz. 2019. "Personalized Radiation Therapy in Cancer Pain Management" Cancers 11, no. 3: 390. https://doi.org/10.3390/cancers11030390

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop