Skip to main content

Main menu

  • Home
  • Content
    • Current
    • Ahead of print
    • Past Issues
    • JNM Supplement
    • SNMMI Annual Meeting Abstracts
    • Continuing Education
    • JNM Podcasts
  • Subscriptions
    • Subscribers
    • Institutional and Non-member
    • Rates
    • Journal Claims
    • Corporate & Special Sales
  • Authors
    • Submit to JNM
    • Information for Authors
    • Assignment of Copyright
    • AQARA requirements
  • Info
    • Reviewers
    • Permissions
    • Advertisers
  • About
    • About Us
    • Editorial Board
    • Contact Information
  • More
    • Alerts
    • Feedback
    • Help
    • SNMMI Journals
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI

User menu

  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Journal of Nuclear Medicine
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Journal of Nuclear Medicine

Advanced Search

  • Home
  • Content
    • Current
    • Ahead of print
    • Past Issues
    • JNM Supplement
    • SNMMI Annual Meeting Abstracts
    • Continuing Education
    • JNM Podcasts
  • Subscriptions
    • Subscribers
    • Institutional and Non-member
    • Rates
    • Journal Claims
    • Corporate & Special Sales
  • Authors
    • Submit to JNM
    • Information for Authors
    • Assignment of Copyright
    • AQARA requirements
  • Info
    • Reviewers
    • Permissions
    • Advertisers
  • About
    • About Us
    • Editorial Board
    • Contact Information
  • More
    • Alerts
    • Feedback
    • Help
    • SNMMI Journals
  • View or Listen to JNM Podcast
  • Visit JNM on Facebook
  • Join JNM on LinkedIn
  • Follow JNM on Twitter
  • Subscribe to our RSS feeds
OtherContinuing Education

Radiopharmaceutical Therapy for Palliation of Bone Pain from Osseous Metastases

Neeta Pandit-Taskar, Maria Batraki and Chaitanya R. Divgi
Journal of Nuclear Medicine August 2004, 45 (8) 1358-1365;
Neeta Pandit-Taskar
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Maria Batraki
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Chaitanya R. Divgi
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Bone metastasis occurs as a result of a complex pathophysiologic process between host and tumor cells leading to cellular invasion, migration adhesion, and stimulation of osteoclastic and osteoblastic activity. The process is mediated by parathyroid hormones, cytokines, and tumor-derived factors. Several sequelae occur as a result of osseous metastases and resulting bone pain can lead to significant debilitation. Pain associated with osseous metastasis is thought to be distinct from neuropathic or inflammatory pain. Several mechanisms—such as invasion of tumor cells, spinal cord astrogliosis, and sensitization of nervous system—have been postulated to cause pain. Pharmaceutical therapy of bone pain includes nonsteroidal analgesics and opiates. These drugs are associated with side effects, and tolerance to these agents necessitates treatment with other modalities. Bisphosphonates act by inhibiting osteoclast-mediated resorption and have been increasingly used in treatment of painful bone metastasis. While external beam radiation therapy remains the mainstay of pain palliation of solitary lesions, bone-seeking radiopharmaceuticals have entered the therapeutic armamentarium for the treatment of multiple painful osseous lesions. 32P has been used for >3 decades in the treatment of multiple osseous metastases. The myelosuppression caused by this agent has led to the development of other bone-seeking radiopharmaceuticals, including 89SrCl, 153Sm-ethylenediaminetetramethylene phosphonic acid (153Sm-EDTMP), 179mSnCl, and 166Ho-Labeled 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetramethylenephosphonate (166Ho-DOTMP). 89Sr is a bone-seeking radionuclide, whereas 153Sm-EDTMP is a bone-seeking tetraphosphonate; both have been approved by the Food and Drug Administration for the treatment of painful osseous metastases. While both agents have been shown to have efficacy in the treatment of painful osseous metastases from prostate cancer, they may also have utility in the treatment of painful osseous metastases from breast cancer and perhaps from non-small cell lung cancer. This article illustrates the salient features of these radiopharmaceuticals, including the approved dose, method of administration, and indications for use. We conclude with recommended guidelines for therapy and follow-up.

  • radiopharmaceutical therapy
  • bone-seeking radiopharmaceuticals
  • bone pain
  • osseous metastases

Bone metastasis is a major complication of several solid cancers; the prostate, breast, lung, kidney, and thyroid are cancers most associated with bone pain (most often, breast and prostate). Although bone metastases are often clinically silent, they can lead to serious sequelae, such as pain, fractures, and hypercalcemia. These complications often reduce performance status and decrease the quality of life. Bone pain due to metastasis is often considered a unique type of pain. It is initially a dull or aching pain, worse at night, that improves with physical activity (1). If pain increases with physical activity, the risk of imminent fracture is increased.

The pathophysiology of bone metastasis and related complications is complex. Normal bone undergoes continuous remodeling that is essential to maintain mechanical function. The process is performed by a multicellular unit formed by 2 different cell types, osteoclasts and osteoblasts. Osteoclasts resorb bone, whereas osteoblasts replace bone (2). Systemic factors—such as the parathyroid hormone, local osteoclast-activating cytokines, and growth factors—contribute to the process (3). During bone resorption, growth factors and mineral ions are released from the bone matrix. When bone metastases occurs, a cycle of signaling takes place that results in increased osteolytic activity. Tumor-derived factors stimulate osteoclast-mediated bone resorption, whereas the growth factors released from the bone matrix stimulate the tumor cells to grow and secrete additional cytokines. This leads to osteopenia and increased risk of developing fractures and, when lesions are in vertebrae, spinal cord compression. The calcium released from the bone matrix in the course of this process can lead to the hypercalcemia of malignancy (HCM), a serious metabolic condition.

Skeletal metastasis is a multifactorial process in which several biologic processes play a role leading to interaction between host and tumor cells. Cellular invasion and migration, cell matrix adhesion or cell-to-cell adhesions, interaction with endothelial cells, regulation of growth factor, and stimulation of osteoclasts and osteoblasts are thought to contribute to development of skeletal metastasis. Cadtherins, integrins, imuunoglobulins, selectins, and CD44 are some of the molecules implicated in loss of cellular adhesion that causes cell matrix detachment, invasion, and migration. Several isoforms of CD44, an adhesion molecule that binds hyaluronate, have been associated with lymphatic spread of the tumors and are expressed in breast and colon cancers. P selectins bind to a wide range of carcinoma cells, including breast, colon, and lung. Interaction with the endothelial cells leads to local arrest or further migration of the tumor cells. Platelet fibrin thrombi and inflammatory cytokines lead to adhesions and arrest of the tumor cells, whereas chemotactic factors lead to increased mobility of tumor cells. Invasiveness is mediated by several enzyme systems, including serine proteases, of which matrix metalloproteins and urokinase plasminogen activation system are thought to be most important. Cellular motility is critical for tumor cells to develop distant metastasis. Motility is mediated by several factors, including: growth factors, hyaluronians, components of matrix, and host- or tumor-secreted factors. Cytokinins such as autotoxin and hepatocyte growth factor are also implicated in inducing cell motility (4–7).

Pain from bone metastasis is of variable intensity and intermittent at onset but progresses to continuous low level pain with episodes of breakthrough pain, which later becomes chronic pain. Mechanical allodynia, in which normal nonpainful activity such as coughing and gentle limb movements can also be perceived as painful, can occur leading to significant limitation of activity. Bone pain is thought to be distinct from neuropathic or inflammatory pain, where there is upregulation of the glial fibrillary acidic protein in the spinal cord indicating astrogliosis. The exact mechanism of cancer pain is unknown and it is postulated that the pain may be due to the presence of tumor in the bone. Treatment with bisphosphonates supports the thought that osteolysis leads to bone pain, which is linked to the number and extent of osteoclastic activity. Bone cancer pain is also thought to occur due to sensitization of the nervous system. Sensory information from peripheral tissues is transmitted to the spinal cord and brain by primary afferent sensory neurons, nociceptors that detect stimuli that are perceived as harmful and convert into electrochemical signals that are transmitted to the central nervous system. Tumors secrete a variety of factors that sensitize or directly excite primary afferent neurons, causing the sensation of pain. Receptors for many of these factors are expressed by primary afferent neurons. Peripheral sensitization leads to increased release of substance P from nociceptors, even with minor stimulation. Sensitization also occurs centrally in the dorsal horn of the spinal cord, which leads to allodynia and hyperalgesia. The spinal cord may show marked astrocytosis, increased expression of c-Fos (a general marker of neuronal activity), and a peptide—dynorphin. Other factors that may contribute to the activation of sensory neurons, is the lower intracellular and extracellular pH of solid tumors, which can also cause pain in cancer patients. It is also thought that tumor growth may entrap and injure nerves, causing neuropathic pain (8–10).

Management of bone pain includes analgesia, radiation, radiofrequency (RF) ablation, hormones, chemotherapy, and surgery. Localized sites of involvement can be treated with surgery, RF ablation, or external beam radiation, whereas radiopharmaceuticals, hormones, and chemotherapy are used to treat more diffuse bone involvement.

Control of bone pain usually begins with analgesic medications used in a 3-step approach. Nonsteroidal antiinflammatory drugs (NSAIDs), including aspirin, ibuprofen, and naproxen, are used to relieve mild to moderate pain initially. With persistent or increased pain while on NSAIDs, treatment progresses to the second step, which utilizes weak opioids, such as codeine or hydrocodone, changing to higher doses or more potent opioids (step 3), such as morphine, hydromorphone, or fentanyl, if the pain persists or becomes more severe. The more potent narcotic agents have significant side effects, including constipation, limitations in mental and physical status, and addiction. Lethargy and constipation are particularly common symptoms. Increased doses also increase the cost significantly when used on a long-term basis. To limit the dose of these medications, radiation therapy and surgery are used for the treatment of localized bone metastases. Some patients with extensive disease in a portion of the body are candidates for hemibody irradiation. Hemibody radiation provides relief from pain associated with extensive bone involvement, but it is associated with a high incidence of side effects (11). Bone-seeking radionuclides are therefore preferred for widespread painful bone metastasis (12).

BISPHOSPHONATES

Bisphosphonates (Fig. 1) inhibit osteoclast-mediated bone resorption by binding to bone mineral, interfering with osteoclast activation. These agents also promote repair by stimulating osteoblast differentiation and bone formation. As a result, these agents are playing an increasing role in the treatment of painful bone metastases.

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Structure of bisphosphonate.

Bisphosphonates are used in the treatment of benign conditions, such as osteoporosis; in malignancies with osteoblastic and osteolytic bone metastases as well as HCM (13,14). The use of bisphosphonates has made a significant change in the therapeutic approach to management of bone metastasis and pain prevention in breast cancer and multiple myeloma (15). Bisphosphonates reduce skeletal complications (16): delay progression of existing bone metastases and reduce the development of new lesions in patients with breast cancer and multiple myeloma (17,18).

Pamidronate, zoledronate, and clodronate are some of the bisphosphonates that have been used clinically. Pamidronate is a second-generation bisphosphonate that is effective in patients with multiple myeloma or breast cancer, who primarily have osteolytic metastases (19,20), and is shown to reduce skeletal complication and morbidity in patients with breast cancer (20–22). Zoledronic acid is a newer nitrogen-containing bisphosphonate that has been evaluated in bone metastases from a variety of cancers. Zoledronic acid was shown to be as effective as pamidronate in reducing skeletal complications (23) but was superior in treating hypercalcemia (24). In breast cancer patients, bisphosphonate therapy is recommended for patients with radiographic evidence of bone destruction but is not recommended for treatment based an abnormal bone scan alone (25). This therapy also reduces skeletal-related events in hormone-refractory prostate cancer (26), non-small cell lung cancer, and urologic malignancies (27,28).

There has been concern regarding the efficacy of bone scanning in patients who were treated with bisphosphonates since methylene diphosphonate (MDP) is similar to the bisphosphonates. The literature is controversial, suggesting that the timing of the bone scan vis-à-vis the last dose of bisphosphonate is critical, though this has not been borne out by studies with etidronate and alendronate (29–31).

RADIATION THERAPY

External beam radiation therapy can often produce pain relief within 48 h of the start of therapy. External beam therapy is most effective when the disease is localized, allowing the highest dose to be delivered to the lesion. If multiple lesions at multiple sites require therapy, side effects—particularly bone marrow suppression—limit the dose that can be delivered. Hemibody radiation therapy has been used in patients with widespread lesions and pain but can lead to significant toxicity, including alopecia and radiation pneumonitis if the upper body is treated; or nausea, vomiting, and diarrhea if the lower body is treated. Myelosuppression can also occur especially in patients treated with whole-body radiation.

The mechanism of pain relief after radiation therapy is not known. Radiation causes a reduction in the number of viable tumor cells that may result in shrinkage of the tumor. Removal of the tumor from bone then enables osteoblastic repair, restoring integrity of the damaged bone. However, certain features—especially the lack of a dose-response relationship and the rapid pain relief, which may be seen especially after systemic radiation (within 24–48 h in up to 25% of patients)—make it unlikely that tumor shrinkage itself is responsible for the pain relief (32). It has also been suggested that radiation may have an analgesic effect through osteoclast inhibition, which provides an insight to bisphosphonate activity (33).

In about 80% of patients with local pain, a single dose of radiation will achieve pain relief within 4–6 wk. If a lesion recurs at that site, retreatment is effective if subradical doses were used (34–36). Wide-field external beam treatment aimed to cover areas roughly equivalent to the half-body above or below the diaphragm depending on the sites of pain may be given in those with widespread disease or where systemic treatment has failed or is inappropriate. Treatment is delivered as single doses of 6 Gy to the upper hemibody, which is limited by lung tolerance, and 8 Gy to the lower hemibody (37). Sequential hemibody radiotherapy, allowing 4–6 wk between each treatment, is given to achieve coverage of the whole skeleton with external beam radiotherapy. The interval between treatments is useful for bone marrow regeneration. After lower hemibody treatment, short-lived and self-limiting nausea and gastrointestinal upset are common; in upper hemibody treatment, pneumonitis is common if tolerance doses are exceeded. Symptomatic bone marrow depression may also occur and transfusion is often required to maintain the hemoglobin in the early weeks after treatment (38), though life-threatening complications such as neutropenia or thrombocytopenia are unusual.

RADIOPHARMACEUTICAL THERAPY

Since multiple sites of osseous metastases are common and some patients have multifocal bone pain, systemic targeted treatment of the skeletal metastatic sites offers the potential of pain relief with minimal side effects. Radiopharmaceuticals developed for the treatment of painful bone metastases use the following radionuclides: 32P, 89Sr, 186Re, 188Re, 153Sm, and 177Lu (Table 1). All agents have advantages and side effects. The agents differ in terms of efficacy; duration of pain palliation; tumoricidal effects; ability to repeat treatments; toxicity, and expense. Most studies with these agents have been conducted in prostate and breast cancer patients.

View this table:
  • View inline
  • View popup
TABLE 1

Radiopharmaceuticals

MECHANISM OF ACTION

The sources of radiation within bone differ with the radiopharmaceutical used: The metallic chelated radiotracers tend to chemically absorb to the trabecular surface, whereas 32P and 89Sr (as the chloride) distribute more widely throughout bone. Due to the heterogeneity of radiopharmaceutical uptake, spicule thickening, and tumor and marrow distribution, there is variation in dosimetry (39).

32P

32P is a reactor-produced, pure β-emitting radionuclide with a physical half-life of 14.3 d. The maximum and mean β-particle energies are 1.71 and 0.695 MeV, respectively, with the mean and maximum particle range in tissue of 3 and 8 mm, respectively. Oral and injectable 32P (usually the injectable sodium orthophosphate) was introduced for treatment of metastatic bone pain >50 y ago. 32P is incorporated into the hydroxyapatite molecule present in large amounts in most osseous metastases. The likely mechanism of action is DNA damage from decay to 32S, with subsequent cell death, as well as damage to cells producing pain modulators—for example, lymphocytes that produce lymphokines.

32P as sodium orthophosphate was one of the first radiopharmaceuticals used to reduce pain from bone metastases (40) and the most widely used radiotracer until the 1980s (41). Its uptake in malignant tissue was demonstrated by Lawrence et al. (42). Earlier studies used testosterone and androgen stimulation for 32P bone metastasis therapy (43–45). The response rates were between 59% and 93% for prostate cancer and between 52% and 94% for breast cancer, with overall response rates of 77% and 84%. No dose-response relationship was seen. Pain palliation occurred within 14 d, with a range of 2 d to 4 wk. A major problem with 32P therapy is the occurrence of toxicity, including myelosuppression and pancytopenia. Additional concerns include the very high β-energy and low lesion-to-normal bone ratio. Pancytopenia occurs around 4–5 wk, with recovery by 6–7 wk, and is dose dependent. However, this is not found to be clinically significant with either a single dose or even multiple doses up to 444 MBq (12 mCi). Acute leukemias have been reported with use of 32P in polycythemia vera; however, grade 4 leukopenia and thrombocytopenia are quite rare (41).

89SrCl

Strontium is an element that behaves biologically like calcium. It localizes in bone primarily in areas of osteoblastic activity. 89Sr has a physical half-life of 50.5 d and emits a β-particle with a maximum energy of 1.46 MeV and an average soft-tissue range of 2.4 mm. The usual therapeutic dose is 148 MBq (4 mCi).

After intravenous administration, 89Sr is concentrated in bone in proportion to osteoblastic activity. Of the 89Sr that is not concentrated in bone, about 80% is excreted through the kidneys and about 20% through the gastrointestinal system. Although the biologic half-life is 4–5 d, about 20% is retained in the body after 90 d (46).

89Sr therapy can relieve bone pain for up to 14 mo and is recommended for use in patients with moderate pain and a reasonable life expectancy. Response rates range from 60% to 84% (47). The onset of pain relief is generally within 7–21 d, with a mean duration of relief of about 6 mo. Transient increased bone pain (painful flare) may occur in the first 2–3 d after treatment. This is usually mild, self-limited, and controlled with analgesics. A flare usually heralds a good treatment response. Patients can be retreated after 90 d, though multiple therapies may lead to greater marrow toxicity. There is a dose-response relationship for complete relief of pain (though not for partial pain relief) (48). Several studies have demonstrated no significant differences between response to 89Sr and response to local or hemibody radiation (49,50). Concurrently administered chemotherapy is effective in enhancing pain relief and delaying the onset of new painful metastases (51). However, combining 89Sr with chemotherapy does not increase the overall pain relief response rates (complete plus partial response of 55%–80%). Although the agent is useful for osseous pain palliation, there is no survival benefit, even when a dose of 399.6 MBq (10.8 mCi) is used (49,52). However, in patients with androgen-independent prostate cancer and severe bone pain (53), combined treatment with 89Sr and doxorubicin resulted in a mean survival of 28 mo compared with 17 mo for patients treated with doxorubicin alone. The use of 89Sr can help reduce the lifetime health-care costs by decreasing both the need for radiotherapy and the need for narcotics and hospitalization (54,55).

153Sm

153Sm, a nuclide with a physical half-life of 1.9 d, decays by β-emission. The β-particle has a maximum energy of 0.81 MeV, a mean energy of 0.23 MeV, and an average soft-tissue range of 0.6 mm. The β-ray is accompanied by a 103-keV γ-ray, which is 28% abundant. 153Sm is complexed with ethylenediaminetetramethylene phosphonic acid to form 153Sm-EDTMP. This phosphonate complex concentrates in the skeleton, in proportion to osteoblastic activity. After intravenous injection, <1% remains in the blood at 5 h. About 65% of the dose remains in the skeleton. The urinary excretion is nearly complete by 6 h (56). The distribution of 153Sm-EDTMP is identical to that of bone-seeking radiopharmaceuticals such as 99mTc-MDP (99mTc-methylene diphosphonate) (57) (Fig. 2). 153Sm-EDTMP is usually administered at a dose of 37 MBq/kg (1 mCi/kg). An initial dose-escalation study demonstrated dose-limiting myelotoxicity, with a maximum tolerated dose of 37 MBq/kg (1 mCi/kg) (58). Platelet nadir occurred between days 16 and 45 (median, 28 d). Pain palliation occurs in 62%–74% of patients (56,59,60) with better overall responses at higher doses. Pretreatment platelet count, tumor type, prior hormonal therapy, and percentage of 153Sm-EDTMP uptake had more impact on myelotoxicity than the administered activity. Patients with extensive metastases were more likely to have myelotoxicity, probably due to a fragile marrow. Bone marrow suppression was generally mild, reversible, and not associated with grade 4 toxicity. Platelets and white blood cells reached nadir at 3 or 4 wk with both doses and recovered by 8 wk (61). A trend toward improved survival was also seen (59).

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

Targeting of bone metastases with 153Sm-EDTMP in patient with prostate cancer. ANT = anterior; POST = posterior. Arrow indicates uptake in pubic bone.

153Sm is the most widely used pain palliation radiopharmaceutical agent in the United States. Its ease of use, the ability to image its distribution, and its clinical results (including the ability to titrate the amount based on body weight) make it attractive, while issues with availability, radiation safety, and potential irreversible myelosuppression limit wider use.

Rhenium Isotopes

186Re-Labeled 1,1-hydroxyethylidene diphosphonate (186Re-HEDP/186Re-(Sn)HEDP or etidronate) and 188Re are investigational radiopharmaceuticals for bone pain palliation. 186Re has a physical half-life of 3.7 d and emits a β-particle with a maximum energy of 1.07 MeV, a mean energy of 0.349 MeV, and an average soft-tissue range of 1.1 mm. A study by Sciuto et al. with 186Re-HEDP showed overall pain palliation of 92% in breast cancer patients (47). Pain relief was established early and the tumor-to-bone marrow absorbed dose ratio was 14:1, suggesting its clinical relevance in patients with more compromised bone marrow reserve. Typical marrow recovery times range from 4 to 6 wk. Other reported side effects are mild pain flare phenomenon (6.5%), occurring in the first week after treatment and lasting 2–4 d (62). Several initial studies reported the safety and efficacy of use of 186Re-HEDP in bone pain palliation. The maximum tolerated administered activity of 186Re-HEDP in patients with metastatic breast cancer was found to be 2,405 MBq (65 mCi) with thrombocytopenia as dose-limiting toxicity (63), and a response rate of 77% has been reported (64).

Its role in androgen-independent prostate cancer and multiple other cancers has also been studied with reported response rates of about 50%–100% in various cancers (65,66). For a variety of reasons, however, the agent is not available for use in the United States.

188Re is an isotope of 186Re and is also been used for bone pain palliation. It is inexpensive, as it may be “milked” on demand from a 188W/188Re generator, and a kit is available for attaching it to HEDP. 188Re-(Sn)HEDP has similar biodistribution and radiation dosimetry characteristics as 186Re-(Sn)HEDP and appears to result in similar benefits and toxicities in patients with skeletal metastases. Dosimetric calculations asserted safe, same-day outpatient therapy to be feasible with 188Re-(Sn)HEDP (67). This has mainly been investigated for use in developing countries. It offers the possibility of repeated therapy without additional costs. The high energy of this β-emitter, with a maximal energy of 2.12 MeV, has the potential of killing tumor cells. There are limited data on the use of this agent. An initial study demonstrated pain palliation in 60%–75% of prostate cancer patients receiving a dose of ≥2.6 GBq (68). Recently, the effectiveness of repeated therapy using 188Re-HEDP has been demonstrated in 64 patients with progressive, hormone-resistant prostate cancer (69).

117mSn-Pentetate

The chelate 117mSn-diethylenetriaminepentaacetic acid (117mSn-DTPA) is an experimental radiopharmaceutical undergoing evaluation for treatment of painful bone metastases. 117mSn, a nuclide with a physical half-life of 13.6 d, decays by isomeric transition with emission of the dominant γ-ray at 156 keV. The γ-ray undergoes conversion and it is the conversion electrons that have the therapeutic potential. The energetic conversion electrons have a very short range in soft tissue (0.2−0.3 mm), which may explain the low incidence of myelosuppression seen with 117mSn-pentetate. 117mSn is injected as the pentetate (DTPA) chelate and has no affinity for hydroxyapatite. The mechanism of localization is postulated as precipitation of stannous oxide on bone surfaces or by a hydrolysis reaction with hydroxyapatite. In reported studies, the pain palliation rate is approximately 75%. There is no dose-response relationship; the onset of pain relief is also much earlier than that with the other agents described. At doses of >444 MBq (>12 mCi) (per 70 kg body weight), pain palliation has been noted as early as <1 wk after treatment (70).

PATIENT IDENTIFICATION AND REFERRAL

Patients referred for bone pain palliation therapy are most commonly breast and prostate cancer patients, referred for radiopharmaceutical treatment by the medical oncologists. Before treatment it is mandatory to check the complete blood count, total leukocyte count, and platelet count (Tables 2 and 3). Since the blood counts can change dramatically in these patients, especially if they are receiving other chemotherapy or radiotherapy, the blood counts should be performed within a week before therapy. Patients with myelosuppression should be evaluated carefully to determine if the benefits of treatment outweigh the risks of worsening the myelosuppression.

View this table:
  • View inline
  • View popup
TABLE 2

Common Indications and Contraindications

View this table:
  • View inline
  • View popup
TABLE 3

Checklist Before Administration of Radiopharmaceutical

A detailed history should be taken to evaluate other systemic illness that can affect treatment or increase side effects. An ideal patient is one who has multiple painful bone metastases positive on bone scan and who is clinically experiencing worsening of pain while on nonnarcotic analgesics. A bone scan is necessary to document metastatic disease at the site of pain. With 153Sm, images can be obtained after the dose is administered, making it possible to confirm uptake at the metastatic sites. The imaging is done the same day (2–4 h) or the next day (the latter has the advantage of better visualization of lumbosacral and pelvic disease, as there is clearance from the urinary system).

ADMINISTRATION OF RADIOPHARMACEUTICAL AGENT

Once eligibility is established, the patient is treated in an outpatient setting. A worksheet for assessing the eligibility may be used (Fig. 3). The patient is instructed about the radiation safety precautions. Informed consent is obtained. Patient identifiers are checked. An intravenous line is inserted and one must ensure good blood return and free flow of the saline flush to prevent extravasation of tracer. When 89Sr is used, a fixed dose of 148 MBq (4 mCi) is usually used. If 159Sm is used, the patient is weighed, and 37 MBq/kg (1 mCi/kg) is administered. The dose is administered by an attending physician. The tracer is injected as a slow intravenous injection over a period of 1–2 min and flushed with 10–20 mL of saline. A note is placed in the patient’s record describing the administration. The radiation safety officer is informed and present during the administration. If the patient is treated with samarium, images are usually recorded at 24 h to compare with the diagnostic bone scan.

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

Worksheet for radiopharmaceutical therapy for bone pain. LMP = last menstrual period; WBC = white blood cell count; RP = radiopharmaceutical.

FOLLOW-UP

Patients are usually seen by their medical oncologist in follow-up. Blood counts are obtained at least weekly for the first 6 wk and more frequently if there is evidence of toxicity. A drop in counts may be seen by 2–4 wk after treatment. Blood counts, notably thrombocyte and neutrophil counts, should be obtained until all counts are normal; return to pretreatment baseline may not always occur. Patients whose blood counts do return to pretreatment values may be retreated, generally not before 3 mo.

In summary, bone pain palliation therapy with radiopharmaceuticals is a cost-effective systemic therapy to relieve pain from skeletal metastases with a consequent decrease in morbidity with an improvement in quality of life; use of bone-seeking radiopharmaceuticals, by themselves or as part of a multimodality regimen, may also reduce disease progression. It should be actively considered especially in patients with widespread metastasis. The ability to use this treatment with other therapies with added benefit is definitely an asset. Although single therapy has been shown to be effective especially in metastatic prostate cancer, repeat therapies should be carefully administered as the effect of such therapy on marrow reserve has not yet been completely elucidated.

Footnotes

  • Received Apr. 15, 2004; accepted Apr. 15, 2004.

    For correspondence or reprints contact: Neeta Pandit-Taskar, MD, Nuclear Medicine Service, Memorial Sloan-Kettering Cancer Center, 1275 York Ave., New York, NY 10021.

    E-mail: pandit-n{at}mskcc.org

  • ↵* NOTE: FOR CE CREDIT, YOU CAN ACCESS THIS ACTIVITY THROUGH THE SNM WEB SITE (http://www.snm.org/ce_online) THROUGH AUGUST 2005.

REFERENCES

  1. ↵
    Casciato DA, Chansky HA. Bone and joint complications. In: Casiato DA, Lowitz BB, eds. Manual of Clinical Oncology. 4th ed. Philadelphia, PA: Lippincott Williams Wilkins; 2000:601–614.
  2. ↵
    Parfitt AM. Bone remodeling, normal and abnormal: a biological basis for the understanding of cancer-related bone disease and its treatment. Can J Oncol. 1995;5(suppl 1):1–10.
    OpenUrl
  3. ↵
    Raisz LG. Physiology and pathophysiology of bone remodeling. Clin Chem. 1999;45:1353–1358.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Choong PF. The molecular basis of skeletal metastases. Clin Orthop. 2003;415(suppl):S19–S31.
  5. Gunthert U, Hofmann M, Rudy W, et al. A new variant of glycoprotein CD44 confers metastatic potential to rat carcinoma cells. Cell. 1991;65:13–24.
    OpenUrlCrossRefPubMed
  6. Behrens J, Frixen U, Schipper J, Weidner M, Birchmeier W. Cell adhesion in invasion and metastasis. Semin Cell Biol. 1992;3:169–178.
    OpenUrlCrossRefPubMed
  7. ↵
    Li Y, Bhargava MM, Joseph A, Jin L, Rosen EM, Goldberg ID. Effect of hepatocyte growth factor/scatter factor and other growth factors on motility and morphology of non-tumorigenic and tumor cells. In Vitro Cell Dev Biol Anim. 1994;30A:105–110.
    OpenUrlPubMed
  8. ↵
    Honore P, Rogers SD, Schwei MJ, et al. Murine models of inflammatory, neuropathic and cancer pain each generates a unique set of neurochemical changes in the spinal cord and sensory neurons. Neuroscience. 2000;98:585–598.
    OpenUrlCrossRefPubMed
  9. Treede RD, Meyer RA, Raja SN, Campbell JN. Peripheral and central mechanisms of cutaneous hyperalgesia. Prog Neurobiol. 1992;38:397–421.
    OpenUrlCrossRefPubMed
  10. ↵
    Mantyh PW, Clohisy DR, Koltzenburg M, Hunt SP. Molecular mechanisms of cancer pain. Nat Rev Cancer. 2002;2:201–209.
    OpenUrlCrossRefPubMed
  11. ↵
    Kuban DA, Delbridge T, el-Mahdi AM, Schellhammer PF. Half-body irradiation for treatment of widely metastatic carcinoma of the prostate. J Urol. 1989;141:572–574.
    OpenUrlPubMed
  12. ↵
    Hamdy NA, Papapoulos SE. The palliative management of skeletal metastases in prostate cancer: use of bone-seeking radionuclides and bisphosphonates. Semin Nucl Med. 2001;31:62–68.
    OpenUrlCrossRefPubMed
  13. ↵
    Reid IR, Brown JP, Burckhardt P, et al. Intravenous zoledronic acid in postmenopausal women with low bone mineral density. N Engl J Med. 2002;346:653–661.
    OpenUrlCrossRefPubMed
  14. ↵
    Berenson JR, Rosen LS, Howell A, et al. Zoledronic acid reduces skeletal-related events in patients with osteolytic metastases: a double-blind, randomized dose-response study. Cancer. 2001;91:1191–1200.
    OpenUrlCrossRefPubMed
  15. ↵
    Body JJ, Bartl R, Burckhardt P, et al. Current use of bisphosphonates in oncology: International Bone and Cancer Study Group. J Clin Oncol. 1998;16:3890–3899.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    LoRusso P. Analysis of skeletal-related events in breast cancer and response to therapy. Semin Oncol. 2001;28(4 suppl 11):22–27.
    OpenUrl
  17. ↵
    McCloskey EV, MacLennan I, Drayson M, Chapman C, Dunn J, Kanis JA. Effect of clodronate on morbidity and mortality in myelomatosis. Br J Haematol. 1998;100:317–325.
    OpenUrlCrossRefPubMed
  18. ↵
    Powles TS, Paterson S, Kanis JA, et al. Randomized, placebo-controlled trial of clodronate in patients with primary operable breast cancer. J Clin Oncol. 2002;20:3219–3224.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Berenson JR, Lichtenstein A, Porter L, et al. Efficacy of pamidronate in reducing skeletal events in patients with advanced multiple myeloma: Myeloma Aredia Study Group. N Engl J Med. 1996;334:488–493.
    OpenUrlCrossRefPubMed
  20. ↵
    Hortobagyi GN, Theriault RL, Porter L, et al. Efficacy of pamidronate in reducing skeletal complications in patients with breast cancer and lytic bone metastases: Protocol 19—Aredia Breast Cancer Study Group. N Engl J Med. 1996;335:1785–1791.
    OpenUrlCrossRefPubMed
  21. Theriault RL, Lipton A, Hortobagyi GN, et al. Pamidronate reduces skeletal morbidity in women with advanced breast cancer and lytic bone lesions: a randomized, placebo controlled trial. J Clin Oncol. 1999;17:846–854.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Coleman RE. Should bisphosphonates be the treatment of choice for metastatic bone disease? Semin Oncol. 2001;28(4 suppl 11):35–41.
    OpenUrl
  23. ↵
    Rosen LS, Gordon D, Antonio BS, et al. Zoledronic acid versus pamidronate in the treatment of skeletal metastases in patients with breast cancer or osteolytic lesions of multiple myeloma: a phase III, double-blind, comparative trial. Cancer J. 2001;7:377–387.
    OpenUrlPubMed
  24. ↵
    Major P, Lortholary A, Hon J, et al. Zoledronic acid is superior to pamidronate in the treatment of hypercalcemia of malignancy: a pooled analysis of two randomized, controlled clinical trials. J Clin Oncol. 2001;19:558–567.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Hillner BE, Ingle JN, Chlebowski RT, et al. American Society of Clinical Oncology 2003 update on the role of bisphosphonates and bone health issues in women with breast cancer. J Clin Oncol. 2003;21:4042–4057.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Saad F, Gleason DM, Murray R, et al. A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma. J Natl Cancer Inst. 2002;94:1458–1468.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Rosen LS, Gordon D, Tchekmedyian S, 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.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Rosen L, Gordon D, Tchekmedyian S, et al. Zoledronic acid (Zol) significantly reduces skeletal-related events (SREs) in patients with bone metastases from solid tumors [abstract]. Proc Am Soc Clin Oncol. 2002;21:295a.
    OpenUrl
  29. ↵
    Sandler ED, Parisi MT, Hattner RS. Duration of etidronate effect demonstrated by serial bone scintigraphy. J Nucl Med. 1991;32:1782–1784.
    OpenUrlAbstract/FREE Full Text
  30. Pecherstorfer M, Schilling T, Janisch S, et al. Effect of clodronate treatment on bone scintigraphy in metastatic breast cancer. J Nucl Med. 1993;34:1039–1044.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Carrasquillo JA, Whatley M, Dyer V, Figg WD, Dahut W. Alendronate does not interfere with 99mTc-methylene diphosphonate bone scanning. J Nucl Med. 2001;42:1359–1363.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Hoskin PJ, Ford HT, Harmer CL. Hemibody irradiation (HBI) for metastatic bone pain in two histological distinct groups of patients. Clin Oncol. 1989;1:67–69.
  33. ↵
    Hoskin PJ, Stratford MRL, Folkes LK, Regan J, Yarnold JR. Effect of local radiotherapy for bone pain on urinary markers of osteoclast activity. Lancet. 2000;355:1428–1429.
    OpenUrlCrossRefPubMed
  34. ↵
    Hoskin PJ. Radiotherapy for bone pain. Pain. 1995;63:137–139.
    OpenUrlCrossRefPubMed
  35. Mithal NP, Needham PR, Hoskin PJ. Retreatment with radiotherapy for painful bone metastases. Int J Radiat Oncol Biol Phys. 1994;29:1011–1014.
    OpenUrlPubMed
  36. ↵
    Nielsen OS. Palliative radiotherapy of bone metastases: there is now evidence for the use of single fractions. Radiother Oncol. 1999;52:95–96.
    OpenUrlCrossRefPubMed
  37. ↵
    Hoskin PJ. Scientific and clinical aspects of radiotherapy in the relief of bone pain. Cancer Surv. 1988;7:69–86.
    OpenUrlPubMed
  38. ↵
    Dearnaley DP, Bayley RJ, A’Hern RP, et al. Palliation of bone metastases in prostate cancer: hemibody irradiation or strontium-89. Clin Oncol. 1992;4:101–107.
    OpenUrlCrossRef
  39. ↵
    Silberstein EB. Dosage and response in radiopharmaceutical therapy of painful osseous metastases. J Nucl Med. 1996;37:249–252.
    OpenUrlFREE Full Text
  40. ↵
    Friedell HL, Storaasli JP. The use of radioactive phosphorus in the treatment of carcinoma of the breast with widespread metastases to bone. AJR. 1950;64:559–575.
    OpenUrl
  41. ↵
    Silberstein EB. The treatment of painful osseous metastases with phosphorus-32-labeled phosphates. Semin Oncol. 1993;20(suppl 2):10–21.
    OpenUrl
  42. ↵
    Lawrence JH, Scott KG, Tuttle LW. Studies on leukemia with aid of radioactive phosphorus. Int Clin. 1939;3:33–58.
  43. ↵
    Maxfield JR, Maxfield JG, Maxfield WS. The use of radioactive phosphorus and testosterone in metastatic bone lesions from breast and prostate. South Med J. 1958;51:320–327.
    OpenUrlPubMed
  44. Miller AD. Radiophosphorus (P32) treatment in carcinoma of the breast and prostate: report of 39 cases. J Am Osteopath Assoc. 1974;74:217–222.
    OpenUrlPubMed
  45. ↵
    Cheung A, Driedger AA. Evaluation of radioactive phosphorus in the palliation of metastatic bone lesions from carcinoma of the breast and prostate. Radiology. 1980;134:209–212.
    OpenUrlPubMed
  46. ↵
    Blake GM, Zivanovic MA, McEwan AJ, et al. Sr-89 therapy: strontium kinetics in disseminated carcinoma of the prostate. Eur J Nucl Med. 1986;12:447–454.
    OpenUrlPubMed
  47. ↵
    Sciuto R, Festa A, Pasqualoni R, et al. Metastatic bone pain palliation with 89-Sr and 186-Re-HEDP in breast cancer patients. Br Cancer Res Treat. 2001;66:101–109.
    OpenUrlCrossRefPubMed
  48. ↵
    Mertens WC, Stitt L, Porter AT. Strontium-89 therapy and relief of pain in patients with prostate carcinoma metastatic to bone: a dose response relationship? Am J Clin Oncol. 1993;16:238–242.
    OpenUrlPubMed
  49. ↵
    Porter AT, McEwan AJB, Powe JE, et al. Results of a randomized phase III trial to evaluate the efficacy of strontium-89 adjuvant to local field external-beam irradiation in the management of endocrine-resistant metastatic prostate cancer. Int J Radiat Oncol Biol Phys. 1993;25:805–813.
    OpenUrlCrossRefPubMed
  50. ↵
    Quilty PM, Kirk D, Bolger JJ, et al. A comparison of the palliative effects of strontium-89 and external beam radiotherapy in metastatic prostate cancer. Radiother Oncol. 1994;31:33–40.
    OpenUrlCrossRefPubMed
  51. ↵
    Scioto R, Maini CL, Tofani A, et al. Radiosensitization with low-dose carboplatin enhances pain palliation in radioisotope therapy with strontium-89. Nucl Med Commun. 1996;17:799–804.
    OpenUrlCrossRefPubMed
  52. ↵
    Brundage MD, Crook JM, Lukka H. Use of strontium-89 in endocrine-refractory prostate cancer metastatic to bone: Provincial Genitourinary Cancer Disease Site Group. Cancer Prev Control. 1998;2:79–87.
    OpenUrlPubMed
  53. ↵
    Tu S, Millikan RE, Mengistu B, et al. Bone-targeted therapy for advanced androgen-independent carcinoma of the prostate: a randomised phase II trial. Lancet. 2001;357:336–341.
    OpenUrlCrossRefPubMed
  54. ↵
    McEwan AJ, Amyotte GA, McGowan DG, et al. A retrospective analysis of the cost effectiveness of treatment with Metastron in patients with prostate cancer metastatic to bone. Eur Urol. 1994;26(suppl 1):26–31.
  55. ↵
    Malmberg I, Persson U, Ask A, et al. Painful bone metastases in hormone-refractory prostate cancer: economic costs of strontium-89 and/or external radiotherapy. Urology. 1997;50:747–753.
    OpenUrlCrossRefPubMed
  56. ↵
    Eary JF, Collins C, Stabin M, et al. Samarium-153-EDTMP biodistribution and dosimetry estimation. J Nucl Med. 1993;34:1031–1036.
    OpenUrlAbstract/FREE Full Text
  57. ↵
    Singh A, Holmes RA, Farhanghi M, et al. Human pharmacokinetics of samarium-153 EDTMP in metastatic cancer. J Nucl Med. 1989;30:1814–1818.
    OpenUrlAbstract/FREE Full Text
  58. ↵
    Farhanghi M, Holmes RA, Volkert WA, Logan KW, Singh A. Samarium-153-EDTMP: pharmacokinetic, toxicity and pain response using an escalating dose schedule in treatment of metastatic bone cancer. J Nucl Med. 1992;33:1451–1458.
    OpenUrlAbstract/FREE Full Text
  59. ↵
    Collins C, Eary JF, Donaldson G, et al. Samarium-153-EDTMP in bone metastases of hormone refractory prostate carcinoma: a phase I/II trial. J Nucl Med. 1993;34:1839–1844.
    OpenUrlAbstract/FREE Full Text
  60. ↵
    Serafini AN, Houston SJ, Resche I, 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.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    Resche I, Chatal JF, Pecking A, 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.
  62. ↵
    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.
    OpenUrlCrossRefPubMed
  63. ↵
    de Klerk JM, van het Schip AD, Zonnenberg BA, et al. Phase 1 study of rhenium-186-HEDP in patients with bone metastases originating from breast cancer. J Nucl Med. 1996;37:244–249.
    OpenUrlAbstract/FREE Full Text
  64. ↵
    Maxon HR 3rd, Thomas SR, Hertzberg VS, et al. Rhenium-186 hydroxyethylidene diphosphonate for the treatment of painful osseous metastases. Semin Nucl Med. 1992;22:33–40.
    OpenUrlCrossRefPubMed
  65. ↵
    Graham MC, Scher HI, Liu GB, et al. Rhenium-186-labeled hydroxyethylidene diphosphonate dosimetry and dosing guidelines for the palliation of skeletal metastases from androgen-independent prostate cancer. Clin Cancer Res. 1999;5:1307–1318.
    OpenUrlAbstract/FREE Full Text
  66. ↵
    Li S, Liu J, Zhang H, Tian M, Wang J, Zheng X. Rhenium-188 HEDP to treat painful bone metastases. Clin Nucl Med. 2001;26:919–922.
    OpenUrlPubMed
  67. ↵
    Maxon HR 3rd, Schroder LE, Washburn LC, et al. Rhenium-188(Sn)HEDP for treatment of osseous metastases. J Nucl Med. 1998;39:659–663.
    OpenUrlAbstract/FREE Full Text
  68. ↵
    Palmedo H, Guhlke S, Bender H, et al. Dose escalation study with rhenium-188 hydroxyethylidene diphosphonate in prostate cancer patients with osseous metastases. Eur J Nucl Med. 2000;27:123–130.
    OpenUrlCrossRefPubMed
  69. ↵
    Palmedo H, Manka-Waluch A, Albers P, 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.
    OpenUrlAbstract/FREE Full Text
  70. ↵
    Srivastava SC, Atkins HL, Krishnamurthy GT, et al. Treatment of metastatic bone pain with tin-117m stannic diethylenetriaminepentaacetic acid: a phase I/II clinical study. Clin Cancer Res. 1998;4:61–68.
    OpenUrlAbstract
PreviousNext
Back to top

In this issue

Journal of Nuclear Medicine: 45 (8)
Journal of Nuclear Medicine
Vol. 45, Issue 8
August 1, 2004
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Journal of Nuclear Medicine.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Radiopharmaceutical Therapy for Palliation of Bone Pain from Osseous Metastases
(Your Name) has sent you a message from Journal of Nuclear Medicine
(Your Name) thought you would like to see the Journal of Nuclear Medicine web site.
Citation Tools
Radiopharmaceutical Therapy for Palliation of Bone Pain from Osseous Metastases
Neeta Pandit-Taskar, Maria Batraki, Chaitanya R. Divgi
Journal of Nuclear Medicine Aug 2004, 45 (8) 1358-1365;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Radiopharmaceutical Therapy for Palliation of Bone Pain from Osseous Metastases
Neeta Pandit-Taskar, Maria Batraki, Chaitanya R. Divgi
Journal of Nuclear Medicine Aug 2004, 45 (8) 1358-1365;
Twitter logo Facebook logo LinkedIn logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One
Bookmark this article

Jump to section

  • Article
    • Abstract
    • BISPHOSPHONATES
    • RADIATION THERAPY
    • RADIOPHARMACEUTICAL THERAPY
    • MECHANISM OF ACTION
    • PATIENT IDENTIFICATION AND REFERRAL
    • ADMINISTRATION OF RADIOPHARMACEUTICAL AGENT
    • FOLLOW-UP
    • Footnotes
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • THIS MONTH IN JNM
  • PubMed
  • Google Scholar

Cited By...

  • Prostate Cancer, Version 1.2014
  • Prostate Cancer
  • Bone metastases from differentiated thyroid carcinoma
  • High-Linear Energy Transfer Irradiation Targeted to Skeletal Metastases by the {alpha}-Emitter 223Ra: Adjuvant or Alternative to Conventional Modalities?
  • {beta}2-Microglobulin Is a Signaling and Growth-Promoting Factor for Human Prostate Cancer Bone Metastasis.
  • Google Scholar

More in this TOC Section

  • Approaches to Imaging Immune Activation Using PET
  • Large Language Models and Large Multimodal Models in Medical Imaging: A Primer for Physicians
  • Precision Oncology in Melanoma: Changing Practices
Show more Continuing Education

Similar Articles

SNMMI

© 2025 SNMMI

Powered by HighWire