<<

Biomedicine & Pharmacotherapy 93 (2017) 1277–1284

Available online at ScienceDirect www.sciencedirect.com

Review

Managing metastatic : New perspectives, different solutions

a,b, b,c d

Iwona Zaporowska-Stachowiak *, Jacek Łuczak , Karolina Hoffmann ,

e d,f b,c

Katarzyna Stachowiak , Wiesław Bryl , Maciej Sopata

a 

Chair and Department of Pharmacology, Poznan University of Medical Sciences, Poland

b 

Palliative Medicine In-Patient Unit, University Hospital of Lord’s Transfiguration, Poznan University of Medical Sciences, Poland

c 

Department of Palliative Medicine, Poznan University of Medical Sciences, Poland

d 

Arterial Hypertension and Metabolic Disorders In-patient Unit, University Hospital of Lord’s Transfiguration, Poznan University of Medical Sciences, Poland

e

Institute of Applied Linguistics, University of Warsaw, Poland

f

Department of Internal Diseases, Metabolic Disorders and Arterial Hypertension, Poznan University of Medical Sciences, Poland

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

Article history:

Received 6 April 2017 Bone metastases are the most frequent cause of -induced (CIBP). Although palliative

Received in revised form 28 June 2017 radiotherapy and pharmacotherapy conducted according to World Health Organization (WHO)

Accepted 5 July 2017

ladder are the treatment of choice for CIBP reduction, these methods are not always successful, especially

Keywords: with regard to alleviation of incidental pain. Antiresorptive drugs () are able to inhibit

Bone pain bone destruction (loss), proliferation of cancer cells and angiogenesis, but their prolonged use may lead

Bone metastases

to a spectrum of adverse effects. In this paper, types of bone metastases, their complications, as well as

Pathological fractures

diagnostic and therapeutic implications are presented. Moreover, the paper discusses presently used

Spinal cord compression

CIBP treatment methods and research directions for future methods, with special focus on bone

Antiresorptive drugs

metastases.

© 2017 Elsevier Masson SAS. All rights reserved.

Contents

1. Introduction ...... 1277

1.1. Types of bone metastases ...... 1277

1.1.1. Osteolytic metastases ...... 1278

1.1.2. Osteosclerotic (osteoblastic) metastases ...... 1278

2. Diagnostics of cancer induced bone pain (CIBP) ...... 1279

3. Therapeutic implications of CIBP ...... 1280

4. Summary ...... 1281

References ...... 1282

1. Introduction

1.1. Types of bone metastases

Bone metastases can be categorized on the basis of radiological

and histopathological image data and divided into osteolytic,



* Corresponding author at: Chair and Department of Pharmacology, Poznan osteosclerotic and mixed types.



University of Medical Sciences, Rokietnicka 5A, 60-806, Poznan, Poland. Palliative Osteolytic metastases are responsible for bone destruction.



Medicine In-patient Unit, University Hospital of Lord’s Transfiguration, Poznan

These types of lesions are related with breast, lung, thyroid, kidney



University of Medical Sciences, os. Rusa 55, 61-245, Poznan, Poland. Tel.: +48 609 26

and cancer.

70 73; fax: +48 61 873 83 06.

E-mail address: [email protected] (I. Zaporowska-Stachowiak).

http://dx.doi.org/10.1016/j.biopha.2017.07.023

0753-3322/© 2017 Elsevier Masson SAS. All rights reserved.

1278 I. Zaporowska-Stachowiak et al. / Biomedicine & Pharmacotherapy 93 (2017) 1277–1284

The second type of bone , referred to as the 1.1.1. Osteolytic metastases

osteosclerotic type is able to stimulate new bone tissue formation

and is associated with prostate and . are polynuclear cells adhered to the bone using

The third type, called mixed metastasis, occurs mainly with avb3 integrin. Due to the secretion of collagenases, proteases and

, breast and . This type of metastasis is hydrogen ions, they are able to demineralize the bone matrix.

related to local interactions of metastatic cells with osteoclasts and Osteoclasts’ main growth factors are called RANKL (Receptor

[1,2]. Activators of Nuclear Factor kb Ligand) and M-CSF (macrophage

Regardless of their type, metastases alter normal bone colony stimulating factor) [16,17]. RANKL activates osteoclasto-

architecture and increase the risk of complications referred to as genesis, prolongs the time of osteoclasts’ survival by binding on

skeletal-related events (SRE) [1–3]. The processes of bone these cells and their precursors’ surface [18–21]. In turn, M-CSF

resorption and formation are closely interrelated and, although increases RANKL expression on the surface of bone stromal cells,

they may be impaired due to ongoing cancerogenesis, they occur at has a chemotactic effect on osteoclasts and prolongs their life span

the site of the metastasis (independently of its type) in nearly all by inhibiting apoptosis [22].

types of cancer [4]. There are two exceptions from this rule. The IL-11 is another cellular factor that induces and escalates

first is (the 3rd most frequent type of cancer in osteoclastogenesis [23]. There are many other growth factors that

children and youth) which is related to pure osteosclerotic can directly or indirectly induce osteoclasts’ formation, stimulate

metastasis [5] and the second is multiple myeloma giving their activity or prolong their life span. One of them is Parathyroid

exclusively metastases of “purely” osteolytic character [6]. In all Hormone-Related Peptide (PTHRP1), produced by a majority of

other types of , the process of bone formation is secondary solid tumor cells (e.g. breast cancers). PTHrP binds to PTHRP1

to [7]. receptor (i.e. receptor of PTH) and induces RANKL expression on

Prostate and bladder cancers are responsible mainly for cells of stromal bone marrow (see Fig. 1) [24,25].

osteosclerotic (aka. osteoblastic) metastases [8], however intense TGFb, in turn, stimulates the production of PTHrP by breast

osteolysis is also observed in these types of cancers. Therefore, the cancer cells [26,27]. It has been found that PTHrP production was

application of antiresorptive drugs decreases pain and risk of increased in 92% of patients with metastases from breast to bone,

pathological fractures in patients with [9–11]. while the presence of this peptide was confirmed in only 50% of

Breast and lung cancers and lead to simultaneous patients from breast cancer without metastasis to bone

osteosclerotic and osteolytic metastases. These types of lesions are tissue [28].

therefore referred to as mixed- type metastases. Phenotypic similarity of breast cancer cells to osteoblasts is the

In patients with breast and lung cancer, osteolytic lesions reason why breast cancer tends to metastasize to the bone. Barns

prevail. In 25% of patients suffering from breast cancer, blastic et al. have shown ectopic expression of the transcription factor

metastasis is also present, while osteosclerotic metastases prevail referred to as the RUNX2 protein (Runt-Related Transcription

in 15–20% of such patients [1,3,12]. In the case of thyroid, kidney Factor 2) that stimulates bone sialoprotein (BSP) synthesis in

and multiple myeloma cancers, osteolytic metastasis is the most breast cancers [29]. BSP level correlates positively with the

frequent type of lesion [13]. frequency of metastasis to the bone [30]. BSP plays a role in

Biochemical markers of bone turnover (i.e. cell-secreted angiogenesis, micro-calcification and protection from comple-

peptides and proteins as well as cellular products of degradation) ment-induced cell lysis [31]. RUNX2 protein also stimulates

are assessed for quick evaluation of cellular activity in the bone osteoblasts’ differentiation. Breast cancer and multiple myeloma

[14]. The levels of the above-mentioned markers may be increased cells produce cytokines that inhibit osteoblasts’ activity, namely

in female breast cancer patients even in the absence of clinical activin A and DKK-1 (dikkopf-related protein 1) [32,33]. DKK-1 is a

manifestations of metastases [15].

Fig. 1. A simplified scheme of osteolytic bone metastases.

I. Zaporowska-Stachowiak et al. / Biomedicine & Pharmacotherapy 93 (2017) 1277–1284 1279

soluble inhibitor of the Wnt signal path which is important for the Osteolytic metastases may be a reason of such para-neoplastic

process of growth and differentiation of osteoblasts. symptoms, such as hypercalcemia (nausea, vomiting, constipation,

abdominal pain, debilitation, anorexia, body mass loss, polyuria,

nephrolithiasis, oscitance, impaired consciousness, coma and

1.1.2. Osteosclerotic (osteoblastic) metastases

death). These symptoms occur predominately in the course of

myeloma myelitis (in 33% of patients), breast cancer (19% of

The cells of some cancer types, such as adenocarcinoma, secrete

patients) and lung cancer [53,54].

or induce the secretion of various factors stimulating osteoblasts’

Neurological impairments (due to pressure on the nerve) and

proliferation [34,35], such as transforming growth factor b (TGFb)

pathological bone fractures causing pain and spinal cord compres-

and urokinase [36]. Other factors responsible for the formation and

sion are the most frequent results/complications of osteolytic and

activation of osteoblasts are Wnt (wingless int) proteins that

osteosclerotic metastases. These complications concern mainly

stimulate these cells’ differentiation and activation, prolong their

such long as femur or humerus, which are exposed to weight

life span and suppress osteoclastic activity [37]. Also Platelet-

bearing. Less frequent complications include: limited mobility

Derived Growth Factor (PDGF) increases osteoblasts’ activity and

(resulting in bedsores, clots, embolism, , muscle dystro-

stimulates angiogenesis [38].

phy) and spinal cord instability. Not only do the above-mentioned

Osteoblasts produce Insulin-Growth Factors (IGFs; and so do

complications significantly decrease the patients’ quality of life,

bone endothelial cells) and insulin-like growth factor-binding

but they are also life-threatening [55,56].

proteins (IGF-BPs). The IGF axis imbalance promotes tumor

invasion and tumor-induced angiogenesis. IGF balance depends

2. Diagnostics of cancer induced bone pain (CIBP)

on urokinase plasminogen activator (uPA), synthesized by tumor

and endothelial cells. IGF and uPA mutually boost their activity.

The majority (70–75%) of patients with metastases to the bone

High uPa levels are responsible for the degradation of IGF-BPs and

suffer from pain [57]. Thus, from the therapeutic point of view, pain

by the same token for the increase in IGFs. This in turn stimulates

diagnostics is of paramount importance.

the growth of metastatic cells and osteoblasts. IGF-1 also activates

Bone pain may be of localized nature and be present in one (in

metalloproteinases in cancer cells [39].

10% of patients) or a few areas of the bone system. It may also be

It was shown that Insulin-Growth Factors (IGF-1 and IGF-2)

generalized. The latter type of pain is frequent after bone marrow

stimulated the proliferation of osteoblasts and prolong their life

replacement and it is referred to as the bone marrow replacement

span [40]. Adrenomedulin (ADM) promotes angiogenesis. Prostate

syndrome [58–60]. The occurrence and intensity of CIBP depends

cancer cells were observed to produce osteogenic factors, including

among others on the location of the cancer and/or metastases and

Wnt proteins which are osteoinductive and oncogenic. DKK-1 is a

on the degree of bone tissue destruction [61]. However, some

Wnt antagonist and inhibits bone growth within osteoblastic

patients complain from CIBP weeks or months before bone

metastases [41]. Endothelin-1 (ET-1) stimulates ETAR and using N-

structure damage can be confirmed by radiology or scintigraphy

terminal portion of PTHrP down-regulates DKK-1 produced by

[62]. On the other hand, only 25% patients with metastases to the

tumor cells. It leads to osteoblasts’ proliferation, drop of

bone do not feel CIBP [63].

osteoclasts’ activity, to an increase in bone mineralization and

A very important issue that must be addressed when treating

to the activation of other pro-osteosclerotic growth factors [42,43].

cancer patients is the question of permanent cancer-induced bone

This is rooted in the fact that the endothelin system is crucial for

pain (localized, frequently described as “deep” and “dull”. In the

numerous physiological and pathological processes, including

early stage of the disease it may be of occasional nature, but the

cancerogenesis and osteoblastic metastases. Endothelins influence

patient’s motion and weight-bearing activities may intensify it.

the following signaling pathways: Ras, b-catenin/T-cell factor/

Also night pain and pain at rest are characteristic for patients

lymphoid enhancer factor, mitogen activated protein kinases,

with bone cancer.

nuclear factor-kB (NFkB), mammalian target of rapamycin (mTOR)

CIPB can be also felt during palpation and percussion

and the zinc-finger transcriptional factors (Snail) [44].

examination. Other typical syndromes are backbone deformation,

Also another particle, described as Vascular Endothelial Growth

oedema, increased muscle tonus in the lesion area, radiating pain

Factor (VEGF) is able to stimulate osteoblasts’ differentiation and is

and synalgia (e.g. presence of lesion in Th12-L1 vertebrae may be

involved in osteolysis [45]. Its expression is dependent of the

felt as lateral/bilateral pain of wing of ilium/sacro-iliac joint; see

mentioned IGF-1 levels in serum [39]. Another growth factor À

Table 1).

Fibroblast Growth Factor [2,6,25,46–48] stimulates not only

If metastases to the bone are suspected, their diagnostics should

osteoblasts’ differentiation but also their proliferation.

be performed as soon as possible by means of:

During bone cancerogenesis, the activated osteoblasts secrete

various growth factors that are later used by cancer cells to

 anamnesis and physical examination, imaging examination: x-

proliferate and to increase their odds for survival. These

ray, bone scintigraphy (displaying high sensitivity but low

proliferating cancer cells secrete increased numbers of pro-

specificity), computed tomography (CT), nuclear magnetic

osteoblastic factors that stimulate cancerogenesis. This path turns

resonance imaging (MRI, highly sensitive and specific, it is the

into a vicious circle [6]. Over a half a century ago, a theory was

diagnostic method of choice), positron emission tomography

proposed according to which all types of metastasis to the bone are

(PET),

preceded by osteolysis [49]. Although this theory has not been

confirmed to this day, it has been proven that by stopping

 laboratory tests: evaluation of peripheral morphology

osteolysis, cancer cells proliferation may be decreased (even in

(bone marrow cells suppression), assessment of plasma levels of:

cases of osteosclerotic metastases) thanks to the drop in the

ionized calcium, albumins (hipoalbuminemia À> increase of

secretion of cancer-inducing growth factors released from the

ionized calcium), urea, creatinine, alkaline phosphatase,

bone matrix [47].

 biopsy – histological and pathological confirmation of an

Apart from its impact on osteoclasts and osteoblasts, cancer

ongoing disease process.

cells in bone mobilize and modulate the function of bone marrow

cells (in particular platelets and white cells) as well as and

One should be aware of non-symptomatic vertebral fractures

cells responsible for angiogenesis [50–52].

that are discovered only in bone imaging [67]. In case of metastases

1280 I. Zaporowska-Stachowiak et al. / Biomedicine & Pharmacotherapy 93 (2017) 1277–1284

Table 1

Characteristics of bone [56,64–66]

Bone Cancer Pain Characteristics

Duration Permanent Pain (may be periodic at the beginning)

Incidental

1) Spontaneous at rest (pain without cause)

2) Breakthrough pain (BTP), dependent/independent of the patient (e.g. motion-induced/cough-induced)

So-called wandering pain (appears and disappears, sometimes completely, and shifts to another location; frequent in hematologic

cancers)

Nature Frequent descriptions:

1) Permanent – “dull”, “deep”

2) Incidental – “acute”, “throbbing”

Severity Severe (60% of patients), moderate (30%), weak (10%). Intensity of pain increases along with progression of disease

Site Backbone (in 13%, Th most frequently); generalized pain (metastases, multiple metastases in 10.2% of patients), pelvis bones (in 7.1%), thorax

(ribs in 6.8%), long bones (in 3.9%)

Radiation +

Synalgia +

(e.g. lesion in the pelvis is felt as a pain in the knee)

Other symptoms Paresthesia, mechanical and dynamical , hyper-/hipoalgesia

Pain triggers/aggravating Dependent and independent on the patient: motion, weight-bearing activities

factors

Alleviating factors See text below and Tables 2 and 3 (treatment)

to the bones, medical treatment is aimed to increase life quality The signi cance of bone resorption suppression in the

and to alleviate or to significantly reduce pain. Moreover, it should prevention of occurrence of new and of progression of diagnosed

prevent bedsore formation, immobilization and provide anti- metastases to the bone is still not fully elucidated. There are reports

fi –

clotting prophylaxis. indicating a bene cial impact of such suppression [71 74]. The

The treatment consists of: unique pathophysiology of metastases to the bone may be

confirmed by the fact that the application of antiresorptive drugs

 assessment of expected time of survival and physical fitness, does not correlate with tumor growth in soft tissues [75].

 assessment of fracture risk, Antiresorptive drugs used for the treatment of metastases are

 surgical consultation: assessment of usefulness of such inter- presented in Table 3.

ventions as vertebroplasty, use of orthopedic appliances, The currently used antiresorptive drugs (osteoclastogenesis

 radiotherapeutic consultation to assess the possibility of tele- inhibiting), i.e. bisphosphonates and (antibody bind-

radiotherapy and radioisotopic therapy. ing to the ligand of receptor activator of nuclear factor kappa-beta)

significantly decrease the number of bone-related complications in

Bone pain decreases the patient's mobility, causes anxiety, cases of metastases to the bones and display an additional

depression, increased risk of , throm- anticancer effect. However, in ca. 30 50% of patients with

boembolism and bedsore formation, thus decreasing the quality of metastases to the bone who use antiresorptive drugs, new

life [68]. metastases and complications are observed, as well as progression

of previously existing lesions [6,84,85]. The above clearly indicates

3. Therapeutic implications of CIBP a need for new therapeutic options. New drugs able to affect the

pathophysiology of metastasis to bone that are used currently or

The following procedures are applied in the prophylaxis and are at the stage of clinical trials are as follows:

treatment of metastases to the bone:

 Recombined, RANKL-blocking OPG drugs (OPG analogs) [86],



1 Systemic treatment that consists of hormone therapy, chemo- Soluble RANK-Fc suppressing RANKL [21,87] ,

 Â

therapy, radio-isotopic therapy (Sr89, Sm 153, Ren 186 decrease Oligonucleotides that block P2 7 receptor of NK-kB and

pain and have a cytoreductive effect), application of drugs suppress its activation [88],



preventing bone loss, such as bisphosphonates and denosumab. Humanized anti-PTHrP antibodies that stop RANKL-induced

Radium-223 dichloride (Ra-223) is also considered a bone- osteolysis [25],

 –

targeting agent whose intake results in pain decrease and, in Atrasentan inhibitor of endothelin ETA receptor able to stop

some patients, metastases stabilization or remission [70], osteosclerotic metastases in patients with prostate cancer [40],



2 Local treatment e.g. by means of palliative radiotherapy that Activin A (ACE-011) inhibitors [89,90] and inhibitors of metal-

leads to regression of metastases and to calcification of loproteinases of the bone matrix able to stop metastatic cells and

osteolytic lesions; another procedure is surgical treatment that the angiogenesis process [91]. In the case of metastases to the

is used to join bone shards, to introduce implants and bone, the bers of afferent CGRP and NF200 neurons undergo

endoprostheses, to perform vertebtoplasty and external fixation pathological reorganization referred to as sprouting. Among

of bones, sprouting CGRP and NF200 bers surrounded by bone marrow

3 Symptomatic treatment: treatment of chronic and acute pain cancer cells colony, increased numbers of TrkA receptors (high

(according to the WHO three-step ladder for cancer pain relief it af nity to nerve growth factor receptor/tropomiosin receptor

is the so-called “fourth-step” i.e. invasive, anesthetic methods kinase A) [92] and of GAP43 protein (growth associated protein

Table 2), hypercalcemia and treatment. 43) are observed. The TrkA receptor binds NGF (nerve growth

I. Zaporowska-Stachowiak et al. / Biomedicine & Pharmacotherapy 93 (2017) 1277–1284 1281

Table 2

Drugs used in Poland for the treatment of chronic and acute bone pain according to the WHO three-step pain relief ladder [69].

Group of drugs Drugs Comments

1 st choice drugs

Analgetics Nonsteroidal anti-inflammatory drugs KETOPROFEN, diclofenak, Efficacy in 75% of patients

(NSAIDs) ibuprofen, naprokxen

Opioids Morphine sulphate or Morphine and oxycodone are not recommended for patients

hydrochloride, phentanyl, with renal insufficiency

oxycodone, buprenorphine,

methadone

Glucocorticosteroids Dexamethasone, prednisone Recommended in case of efficacy or impaired efficacy of

NSAIDs; always recommended in case of possible or

performed compression of spinal cord – very high initial

dosage should be decreased with time to the lowest efficient

dose

2nd choice drugs

Antiresorptive drugs BISPHOSPHONATES Pamidronate, zolendronate Used depending on renal function and concentration of

ionized calcium level in blood

Antiepileptic drugs Gabapentin and other similar drugs gabapentin, pregabalin Recommended due to high neuropathic bone pain component

So-called 4th step of the Blocs of peripheral nerves and nerve Bupivacaine, lidocaine These local analgetics may be neurotoxic and cardiotoxic,

analgesic plexuses; other blocks: central, especially if used for prolonged periods of time!

ladderÀinvasive extradural, subarachnoid, paravertebral

anesthetic techniques

factor) which is essential for the survival of afferent and Bone Pain is responsible for patients’ limited mobility and negative

autonomic nerves, modulates neurotransmitter release and emotions, increased risk of respiratory system infection, embolism,

stimulates pathogenic proliferation of nerve fibers [93,94], bedsore formation and it significantly decreases the quality of life.

 Tanezumab is an antibody against NGF; it displays an analgetic Therefore it is of paramount importance to provide patients

effect and is a promising drug for the treatment of metastases to with an early integrated with oncological treatment,

the bone [95]. Also the GAP43 protein plays a role in the which will ensure an optimum pain alleviation strategy, con-

sprouting of nerve fibers and in the regeneration of central and sultations, treatments and physical therapy, offered simultaneous-

peripheral nerve fibers [96]. ly with comprehensive care taking into account the psycho-social

and spiritual components of suffering, as well as will offer support

for the persons caring for the patient.

4. Summary In the treatment of CIBP, palliative radiotherapy and analgetic

drugs given to the patient according to the WHO three-step ladder

Bone pain, caused mainly by metastases to the bone system (use of drugs with various targets having an impact on the receptor

(that are unfavorable prognostics of survival time), occurs most and neuropathic component of pain) are the treatment of choice

frequently in patients with malignant tumors. Cancer Induced

Table 3

Antiresorptive drugs used in the treatment of bone metastases.

Group of drugs/ Mechanism of action Comments

Drug

Bisphosphonates pamidronate They have a direct and indirect effect: by preventing bone loss and Recommendations for pamidronate: osteolytic metastases;

(BPs) cancer cell proliferation they stop the vicious circle of cancer cell shorter T1/2, lower risk of nephrotoxicity and bone necrosis than

and osteoclasts interaction in the microenvironment of the bone for zoledronate

zoledronate tissue. Bisphosphonates stimulate the differentiation of T-cell Zoledronate – recommendations: osteoblastic and osteolytic

subpopulation (gd cells) responsible for the destruction of cancer metastases, significant nephrotoxicity and risk of bone necrosis

cells – this effect is especially strong in zoledronate; they strongly [72]. Long-term effect; during application, temporary suppression

suppress osteoclastic activity, decrease cancer cells’ survival of bone formation may be observed leading to jaw necrosis (the

(complex mechanism); they stop angiogenesis and display effect is stronger in the case of zoledronate) Regular use of BPs as

immuno-modulative effect[76–79]. means of oncologic and palliative treatment leads to high

cumulative doses

Monoclonal denosumab It binds the ligand of receptor activator of nuclear factor kappa- Recommendations (apart from ):

Human beta (RANKL). It leads to the suppression of formation, activity and Skeletal-Related Events Prevention in adult patients with solid

Antibody survival of osteoclasts and to the drop in the resorption of tumor metastases to the bones (pathological fractures, need of

(IgG2) trabecular and cortical bone. It prevents the formation and treats bone radiation, pressure on spinal cord, need of surgical treatment

metastases to the bone; it prevents Skeletal Related Events (more of bones. The drug is administered in the dose of 120 mg s.c 1Â/4

effectively than BPs) and displays anticancer effect (confirmed in weeks. In case of treatment of adults or young people with a

patients with bone marrow cancer) [80–82] mature bone system and suffering from inoperable giant-cell

tumor of the bone, the dosage is 120 mg s.c. 1Â/4 weeks and on

day 8 and 15 of the treatment, additional doses of 120 mg are given

[83]. In comparison to zoledronate, the use of denosumab leads to

a longer delay in the occurrence of the first skeletal-related event

in patients with metastases of breast or prostate cancer to the

bone [73].

1282 I. Zaporowska-Stachowiak et al. / Biomedicine & Pharmacotherapy 93 (2017) 1277–1284

but are unfortunately often insufficient to effectively heal [21] J. Zhang, J. Dai, Z. Yao, Y. Lu, W. Dougall, E.T. Keller, Soluble receptor activator of

nuclear factor kappaB Fc diminishes prostate cancer progression in bone,

incidental pain.

Cancer Res. 63 (2003) 7883–7890.

Antiresorptive drugs used as co- (e.g. bisphospho-

[22] A.T. Mancino, V.S. Klimberg, M. Yamamoto, S.C. Manolagas, E. Abe, Breast

nates and denosumab) stop bone loss, cancer cell proliferation and cancer increases osteoclastogenesis by secreting M-CSF and upregulating

RANKL in stromal cells, J. Surg. Res. 100 (2001) 18–24, doi:http://dx.doi.org/

angiogenesis. However, their prolonged use may lead to adverse

10.1006/jsre.2001.6204.

effects. Therefore, there is an urgent need not only for further

[23] H. Morgan, A. Tumber, P.A. Hill, Breast cancer cells induce formation

research on these complicated and still not fully elucidated by stimulating host IL-11 production and downregulating granulocyte/

macrophage colony-stimulating factor, Int. J. Cancer 109 (2004) 653–660, doi: mechanisms of metastases formation but also for the search for

http://dx.doi.org/10.1002/ijc.20056.

new therapeutic measures in the treatment of Cancer-Induced

[24] T.A. Guise, W.M. Kozlow, A. Heras-Herzig, S.S. Padalecki, J.J. Yin, J.M. Chirgwin,

Bone Pain. Molecular mechanism of breast cancer metastases to bone, Clin. Breast Cancer

5 (2005) 46–53.

References [25] T.A. Guise, J.J. Yin, S.D. Taylor, Y. Kumagai, M. Dallas, B.F. Boyce, T. Yoneda, G.R.

Mundy, Evidence for a causal role of parathyroid hormone-related protein in

the pathogenesis of human breast cancer-mediated osteolysis, J. Clin. Invest.

[1] R.E. Coleman, Skeletal complications of malignancy, Cancer 80 (1997) 1588–

98 (1996) 1544–1549, doi:http://dx.doi.org/10.1172/JCI118947.

1594.

[26] J.J. Yin, K. Selander, J.M. Chirgwin, M. Dallas, B.G. Grubbs, R. Wieser, J.

[2] G.R. Mundy, Metastasis to bone: causes, consequences and therapeutic

Massagué, G.R. Mundy, T.A. Guise, TGF-b signaling blockade inhibits PTHrP

opportunities, Nat. Rev. Cancer 2 (2002) 584–593.

secretion by breast cancer cells and bone metastases development, J. Clin.

[3] D. Chappard, H. Libouban, E. Legrand, N. Ifrah, C. Masson, M.F. Baslé, M. Audran,

Invest. 103 (1999) 197–206, doi:http://dx.doi.org/10.1172/JCI3523.

Computed microtomography of bone specimens for rapid analysis of bone

[27] T.J. Martin, Manipulating the environment of cancer cells in bone: a novel

changes associated with malignancy, Anat. Rec. 293 (2010) 1125–1133, doi:

therapeutic approach, J. Clin. Invest. 110 (2002) 1399–1401, doi:http://dx.doi.

http://dx.doi.org/10.1002/ar.21150. org/10.1172/JCI17124.

[4] S. Fili, M. Karalaki, B. Schaller, Mechanism of : the role of

[28] G.J. Powell, J. Southby, J.A. Danks, R.G. Stillwell, J.A. Hayman, M.A. Henderson,

osteoprotegerin and of the host-tissue microenvironment-related survival

R.C. Bennett, T.J. Martin, Localization of parathyroid hormone-related protein

factors, Cancer Lett. 283 (2009) 10–19, doi:http://dx.doi.org/10.1016/j.

in breast cancer metastases: increased incidence in bone compared with other

canlet.2009.01.011.

sites, Cancer Res. 51 (1991) 3059–3061.

[5] M.P. Stokkel, M.F. Linthorst, J.J. Borm, A.H. Taminiau, E.K. Pauwels, A

[29] G.L. Barnes, A. Javed, S.M. Waller, M.H. Kamal, K.E. Hebert, M.Q. Hassan, A.

reassessment of bone scintigraphy and commonly tested pretreatment

Bellahcene, A.J. Van Wijnen, M.F. Young, J.B. Lian, G.S. Stein, L.C. Gerstenfeld,

biochemical parameters in newly diagnosed osteosarcoma, J. Cancer Res. Clin.

Osteoblast-related transcription factors Runx2 (Cbfa1/AML3) and MSX2

Oncol. 128 (2002) 393–399, doi:http://dx.doi.org/10.1007/s00432-002-0350-

mediate the expression of bone sialoprotein in human metastatic breast

5.

cancer cells, Cancer Res. 63 (2003) 2631–2637.

[6] G.D. Roodman, Mechanism of bone metastasis, N. Engl. J. Med. 350 (2004), doi:

[30] T.E. Kruger, A.H. Miller, A.K. Godwin, J. Wang, Bone sialoprotein and

http://dx.doi.org/10.1056/NEJMra030831 (1665-1664).

osteopontin in bone metastasis of osteotropic cancers, Crit. Rev. Oncol.

[7] G.A. Rodan, T.J. Martin, Therapeutic approaches to bone diseases, Science 289

Hematol. 89 (2014) 330–341, doi:http://dx.doi.org/10.1016/j.

(2000) 1508 1514, doi:http://dx.doi.org/10.1126/science.289.5484.1508. critrevonc.2013.08.013.

[8] M.P. Roudier, C. Morrissey, L.D. True, C.S. Higano, R.L. Vessella, S.M. Ott,

[31] N.S. Fedarko, B. Fohr, P.G. Robey, M.F. Young, L.W. Fisher, Factor H binding to

Histopathological assessment of prostate cancer bone osteoblastic metastases,

bone sialoprotein and osteopontin enables tumor cell evasion of complement-

J. Urol.180 (2008) 1154–1160, doi:http://dx.doi.org/10.1016/j.juro.2008.04.140.

mediated attack, J. Biol. Chem. 275 (2000) 16666–16672, doi:http://dx.doi.org/

[9] S.A. Charhon, M.C. Chapuy, E.E. Delvin, A. Valentin-Opran, C.M. Edouard, P.J. 10.1074/jbc.M001123200.

Meunier, Histomorphometric analysis of sclerotic bone metastases from

[32] G. Leto, L. Incorvaia, G. Badalamenti, F.M. Tumminello, N. Gebbia, C. Flandina,

prostatic carcinoma special reference to , Cancer 51 (1983) 918–

M. Crescimanno, G. Rini, Activin A circulating levels in patients with bone

924, doi:http://dx.doi.org/10.1002/1097-0142(19830301)51:5<918::AID-

metastasis from breast or prostate cancer, Clin. Exp. Metastasis 23 (2006) 117– CNCR2820510526>3.0.CO;2-J.

122, doi:http://dx.doi.org/10.1007/s10585-006-9010-5.

[10] G.M. Oades, J. Coxon, K.W. Colston, The potential role of bisphosphonates in

[33] E. Tian, F. Zhan, R. Walker, E. Rasmussen, Y. Ma, B. Barlogie, J.D. Shaughnessy Jr,

prostate cancer, Prostate Cancer Prostatic Dis. 5 (2002) 264–272, doi:http://dx.

The role of the wnt-signaling antagonist DKK1 in the development of

doi.org/10.1038/sj.pcan.4500607.

osteolytic lesions in multiple myeloma, N. Engl. J. Med. 349 (2003) 2483–2494,

[11] R.E. Coleman, Conclusion Bone markers in metastatic bone disease, Cancer doi:http://dx.doi.org/10.1056/NEJMoa030847.

Treat. Rev. 32 (2006) 27–28.

[34] T.A. Guise, G.R. Mundy, Cancer Bone Endocr. Rev. 19 (1998) 18–54, doi:http://

[12] R.E. Coleman, J.J. Seaman, The role of zolendronic acid in cancer: clinical dx.doi.org/10.1210/edrv.19.1.0323.

studies in the treatment and prevention of bone metastases, Semin. Oncol. 28

[35] R.L. Vessella, E. Corey, Targeting factors involved in bone remodeling as

(2001) 11–16.

treatment strategies in prostate cancer bone metastasis, Clin. Cancer Res. 12

[13] E.T. Keller, J. Zhang, C.R. Cooper, P.C. Smith, L.K. McCauley, K.J. Pienta, R.S.

(2006) 6285–6290, doi:http://dx.doi.org/10.1158/1078-0432.CCR-06-0813.

Taichman, Prostate carcinoma skeletal metastases: crosstalk between tumor

[36] Y. Matuo, N. Nishi, H. Takasuka, Y. Masuda, K. Nishikawa, J.T. Isaacs, P.S. Adams,

and bone, Cancer Metastasis Rev. 20 (2001) 333–349, doi:http://dx.doi.org/

W.L. McKeehan, G.H. Sato, Production and significance of TGF-beta in AT-3

10.1023/A:1015599831232.

metastatic cell line established from Dunning rat prostatic adenocarcinoma,

[14] L.J. Suva, C.H. Washam, R.W. Nicholas, R.J. Griffin, Bone metastasis: mechanism

Biochem. Biophys. Res. Commun. 166 (1990) 840–847.

of therapeutic opportunities, Nat. Rev. Endocrinol. 7 (2011) 208–218, doi:

[37] C.L. Hall, S. Kang, O.A. MacDougald, E.T. Keller, Role of Wnts in prostate cancer

http://dx.doi.org/10.1038/nrendo.2010.227.

bone metastases, J. Cell. Biochem. 97 (2006) 661–672, doi:http://dx.doi.org/

[15] L.M. Demers, L. Costal, V.M. Chinchilli, L. Gaydos, E. Curley, A. Lipton, 10.1002/jcb.20735.

Biochemical markers of bone turnover in patients with metastatic bone

[38] B. Yi, P.J. Williams, M. Niewolna, Y. Wang, T. Yoneda, Tumor-derived platalet-

disease, Clin. Chem. 41 (1995) 1489–1494.

derived growth factor-BB plays a critical role in osteosclerotic bone metastasis

[16] W.C. Dougall, M. Glaccum, K. Charrier, K. Rohrbach, K. Brasel, T. De Smedt, E.

in an animal model of human breast cancer, Cancer Res. 62 (2002) 917–923.

Daro, J. Smith, M.E. Tometsko, C.R. Maliszewski, A. Amstrong, V. Shen, S. Bain,

[39] A.A. Samani, S. Yakar, D. LeRoith, P. Brodt, The role of the IGF system in cancer

D. Cosman, D. Anderson, P.J. Morrisey, J.J. Peschon, J. Schuh, RANK is essential

growth and metastasis: overview and recent insights, Endocr. Rev. 28 (1)

for osteoclast and lymph node development, Genes Dev. 13 (1999) 2412–2424.

(2007) 20–47.

[17] M. Baud’huin, R. Renault, C. Charrier, A. Riet, A. Moreau, R. Brion, F. Gouin, L.

[40] K. Fizazi, J. Yang, S. Peleg, C.R. Sikes, E.L. Kreimann, D. Daliani, M. Olive, K.A.

Duplomb, D. Heymann, Interleukin-34 is expressed by giant cell tumours of

Raymond, T.J. Janus, C.J. Logothesis, G. Karsenty, N.M. Navone, Prostate cancer

bone and plays a key role in RANKL-induced osteoclastogenesis, J. Pathol. 221

cells- interaction shifts expression of growth/survival-related genes

(2010) 77–86, doi:http://dx.doi.org/10.1002/path.2684.

in prostate˛ cancer and reduces expression of osteoprotegerin in osteoblasts,

[18] H. Yasuda, N. Shima, N. Nakagawa, K. Yamaguchi, M. Kinosaki, S. Mochizuki, A.

Clin. Cancer Res. 9 (2003) 2587–2597.

Tomoyasu, K. Yano, M. Goto, A. Murakami, E. Tsuda, T. Morinaga, K. Higashio, N.

[41] K.T. Nanda, K.M. Chelsea, S. Murahari, S.S. Shu, L.G. Lanigan, J.L. Werbeck, E.T.

Udagawa, N. Takahashi, T. Suda, Osteoclast differentiation factor is a ligand for

Keller, L.K. McCauley, J.J. Pinzone, T.J. Rosol, DOCKKOPF-1 (DKK-1) stimulated

osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to

prostate cancer growth and metastasiss and inhibited bone formation in

RANCe/RANKL, Proc. Natl. Acad. Sci. U. S. A. 95 (1998) 3597–3602.

osteoblastic bone metastases, Prostate 71 (6) (2011) 615–625.

[19] D.L. Lacey, E. Timms, H.-L. Tan, M.J. Kelley, C.R. Dunstan, T. Burgess, R. Elliott, A.

[42] J.B. Nelson, Endothelin receptor antagonists, World J. Urol. 23 (2005) 19–27.

Colombero, G. Elliott, S. Scully, H. Hsu, J. Sullivan, N. Hawkins, E. Davy, C.

[43] P.H. Stern, A. Tatrai, D.E. Semler, S.K. Lee, P. Lakatos, P.J. Strieleman, G. Tarjan, J.L.

Capparelli, A. Eli, Y.-X. Qian, S. Kaufman, I. Sarosi, V. Shalhoub, G. Senaldi, J.

Sanders, Endothelin receptors, second messengers, and actions in bone, J. Nutr.

Guo, J. Delaney, W.J. Boyle, Osteoprotegerin ligand is a cytokine that regulates

125 (1995) 2028–2032.

osteoclast differentiation and activation, Cell 93 (1998) 165–176, doi:http://dx.

[44] R. Wang, R.H. Dashwood, Endothelins and their receptors in cancer:

doi.org/10.1016/S0092-8674(00)81569-X.

identification of therapeutic targets, Pharmacol. Res. 63 (6) (2011) 519–524.

[20] G.D. Roodman, W.C. Dougall, RANK ligand as a therapeutic target for bone

[45] Y. Kitagawa, J. Dai, J. Zhang, J.M. Keller, J. Nor, Z. Yao, E.T. Keller, Vascular

metastases and multiple myeloma, Cancer Treat. Rev. 34 (2008) 92–101, doi:

endothelial growth factor contributes to prostate cancer-mediated http://dx.doi.org/10.1016/j.ctrv.2007.09.002.

I. Zaporowska-Stachowiak et al. / Biomedicine & Pharmacotherapy 93 (2017) 1277–1284 1283

osteoblastic activity, Cancer Res. 65 (2005) 10921–10929, doi:http://dx.doi. prostatectomy, Oncotarget 8 (2017) 44131–44140, doi:http://dx.doi.org/

org/10.1158/0008-5472.CAN-05-1809. 10.18632/oncotarget.17311.

[46] M.P. Valta, T. Hentunen, Q. Qu, E.M. Valve, A. Harjula, J.A. Seppänen, H.K. [71] S. Vallet, M.R. Smith, N. Raje, Novel bone-targeted strategies in oncology, Clin.

Väänänen, P.L. Härkönen, Regulation of osteoblast differentiation: a novel Cancer Res. 16 (2010) 4084–4093.

function for fibroblast growth factor 8, Endocrinology 147 (2006) 2171–2182, [72] R.E. Coleman, A djuvant bisphosphonates in breast cancer: are we witnessing

doi:http://dx.doi.org/10.1210/en.2005-1502. the emergence of a new therapeutic strategy? Eur. J. Cancer 45 (2009) 1909–

[47] S.M. Virk, J.R. Lieberman, Tumor metastasis to bone, Arthritis Res. Ther. 9 1915.

(2007) 5, doi:http://dx.doi.org/10.1186/ar2169. [73] M. Gnant, B. Mlineritsch, W. Schippinger, G. Luschin-Ebengreuth, S.

[48] Y. Tang, X. Wu, W. Lei, L. Pang, C. Wan, Z. Shi, L. Zhao, T.R. Nagy, X. Peng, J. Hu, X. Pöstlberger, Ch. Menzel, J. Raimund, M. Seifert, M. Hubalek, V. Bjelic-

Feng, W. Van Hul, M. Wan, X. Cao, TGF-b1-induced migration of bone Radisic, H. Samonigg, Ch. Tausch, H. Eidtmann, G. Steger, W. Kwasny, P. Dubsky,

mesenchymal cells couples bone resorption with formation, Nat. Med. 15 M. Fridrik, F. Fitzal, M. Stierer, E. Rücklinger, R. Greil, Endocrine therapy plus

(2009) 757–765, doi:http://dx.doi.org/10.1038/nm.1979. zoledronic acid in premenopausal breast cancer, N. Engl. J. Med. 360 (2009)

[49] S.I. Roland, Calcium studies in ten cases of osteoblastic prostatic metastasis, J. 679–691.

Urol. 79 (1958) 339–342. [74] A.T. Stopeck, A. Lipton, J.J. Body, G.G. Steger, K. Tonkin, R.H. de Boer, M.

[50] K. Pantel, V. Müller, M. Auer, N. Nusser, N. Harbeck, S. Braun, Detection and Lichinitser, Y. Fujiwara, D.A. Yardley, M. Viniegra, M. Fan, Q. Jiang, R. Dansey, S.

clinical implications of early systemic tumor cell dissemination in breast Jun, A. Braun, Denosumab compared with zoledronic acid for the treatment of

cancer, Clin. Cancer Res. 9 (2003) 6326–6334. bone metastases in patients with advanced breast cancer: a randomized,

[51] R. Aft, M. Naughton, K. Trinkaus, M. Watson, L. Ylagan, M. Chavez-MacGregor, J. double-blind study, J. Clin. Oncol. 28 (2010) 5132–5139.

Zhai, S. Kuo, W. Shannon, K. Diemer, V. Herrmann, J. Dietz, A. Ali, M. Ellis, P. [75] T. Powles, S. Paterson, J.A. Kanis, E. McCloskey, S. Ashley, A. Tidy, K. Rosenqvist,

Weiss, T. Eberlein, C. Ma, P.M. Fracasso, I. Zoberi, M. Taylor, W. Gillanders, T. I. Smith, L. Ottestad, S. Legault, M. Pajunen, A. Nevantaus, E. Männistö, A.

Pluard, J. Mortimer, K. Weilbaecher, Effect of zolendronic acid on disseminated Suovuori, S. Atula, J. Nevalainen, L. Pylkkänen, Randomized, placebo-

tumour cells in women with locally advanced breast cancer: an open label controlled trial of clodronate in patients with primary operable breast cancer,

randomised, phase 2 trial, Lancet Oncol. 11 (2010) 421–428. J. Clin. Oncol. 20 (2002) 3219–3224.

[52] M. Chavez-Macgregor, A. Aviles-Salas, D. Green, A. Fuentes-Alburo, C. Gomez- [76] H.L. Neville-Webbe, I. Holen, R.E. Coleman, The anti-tumour activity of

Ruiz, A. Aguayo, Angiogenesis in the bone marrow of patients with breast bisphosphonates, Cancer Treat. Rev. 28 (2002) 305–319.

cancer, Clin. Cancer Res. 11 (2005) 5396–5400. [77] M.T. Valenti, L. Dalle Carbonare, F. Bertoldo, L. Donatelli, V. Lo Cascio, The

[53] R.E. Coleman, P. Smith, R.D. Rubens, Clinical course and prognostic factors effects on hTERT gene expression is an additional mechanism of amino-

following recurrence from breast cancer, Br. J. Cancer 77 (1998) 336–340. bisphosphonates in prostatic cancer cells, Eur. J. Pharmacol. 580 (2008) 36–42.

[54] J.A. Kanis, E.V. McCloskey, Bisphosphonates in the treatment of multiple [78] F. Dieli, D. Vermijlen, F. Fulfaro, N. Caccamo, S. Meraviglia, G. Cicero, A. Roberts,

myeloma, in: J.J. Body (Ed.), Tumor Bone Disease and Osteoporosis in Cancer S. Buccheri, M. D’Asaro, N. Gebbia, A. Salerno, M. Eberl, A.C. Hayday, Targeting

Patients, Marcel Dekker Inc., New York, 2000, pp. 457–482. human gd T cells with zoledronate and interleukin-2 for immunotherapy of

[55] A. So, J. Chin, N. Fleshner, F. Saad, Management of skeletal-related events in hormone-refractory prostate cancer, Cancer Res. 67 (2007) 7450–7457.

patients with advanced prostate cancer and bone metastases: incorporating [79] D.P. Dearnaley, M.D. Mason, M.K. Parmar, K. Sanders, M.R. Sydesc, Adjuvant

new agents into clinical practice, Can. Urol. Assoc. J. 6 (2012) 465–470. therapy with oral sodium clodronate in locally advanced and metastatic

 

[56] I. Filipczak-Bryniarska, M. Rucinska, K. Bryniarski, J. Woron, J. Wordliczek, Ból prostate cancer: long-term overall survival results from the MRC PR04 and

kostny, in: M. Malec-Milewska, M. Krajnik, J. Wordliczek (Eds.), Chory na PR05 randomised controlled trials, Lancet Oncol. 10 (2009) 872–876.

nowotwór. Kompendium leczenia bólu, Medical Education, Warszawa, 2013, [80] A. Lipton, G.G. Steger, J. Figueroa, C. Alvarado, P. Solal-Celigny, J.J. Body, R. de

pp. 399–412. Boer, R. Berardi, P. Gascon, K.S. Tonkin, R. Coleman, A.H. Paterson, M.C.

[57] T. Ibrahim, L. Mercatali, D. Amadori, Bone and cancer: the osteoncology, Clin. Peterson, M. Fan, A. Kinsey, S. Jun, Randomized active-controlled phase II study

Cases Miner. Bone Metab. 10 (2013) 121–123. of denosumab efficacy and safety in patients with breast cancer-related bone

[58] S. Hesselmann, O. Micke, U. Schaefer, N. Willich, Systemic mast cell disease metastases, J. Clin. Oncol. 25 (2007) 4431–4437.

(SMCD) and bone pain. A case treated with radiotherapy, Strahlenther. Oncol. [81] K. Fizazi, A. Lipton, X. Mariette, J.J. Body, Y. Rahim, J.R. Gralow, G. Gao, L. Wu, W.

178 (2002) 275–279. Sohn, S. Jun, Randomized phase II trial of denosumab in patients with bone

[59] O.G. Johnson, P. Sartain, J.M. Ducore, G.R. Buchanan, Bone pain as an initial metastases from prostate cancer, breast cancer, or other neoplasms after

symptom of childhood acute lymphoblastic : association with nearly intravenous bisphosphonates, J. Clin. Oncol. 27 (2009) 1564–1571.

normal hematologic indexes, J. Pediatr. 117 (1990) 233–237. [82] R. Vij, N. Horvath, A. Spencer, K. Taylor, S. Vadhan-Raj, R. Vescio, J. Smith, Y.

[60] J.T. Lin, E. Lachmann, W. Nagler, Low and in acute and Qian, H. Yeh, S. Jun, An open-label, phase 2 trial of denosumab in the treatment

relapsed mast cell leukemia: a case report, Arch. Phys. Med. Rehab. 83 (2002) of relapsed or plateau-phase multiple myeloma, Am. J. Hematol. 84 (2009)

860–863. 650–656.

[61] L. Doré-Savard, N. Beaudet, L. Tremblay, A micro-imaging study linking bone [83] C.L. Gaston, R.J. Grimer, M. Parry, Current status and unanswered questions on

cancer pain with tumor growth and bone resorption in a rat model, Clin. Exp. the use of Denosumab in giant cell tumor of bone, Clin. Sarcoma Res. 6 (2016),

Metastasis 30 (2013) 225–236. doi:http://dx.doi.org/10.1186/s13569-016-0056-0.

[62] N.M. Luger, D.B. Mach, M.A. Sevcik, P.W. Mantyh, Bone cancer pain: from [84] G.R. Mundy, Metastatic bone disease: clinical features, pathophysiology and

model to mechanism to therapy, J. Pain Symptom Manage. 29 (2005) 32–46. treatment strategies, Cancer Treat. Rev. 27 (2001) 165–176.

[63] G. Wagner, Frequency of pain in patients with cancer, Recent Results Cancer [85] J.J. Body, T. Facon, R.E. Coleman, A. Lipton, F. Geurs, M. Fan, D. Holloway, M.C.

Res. 89 (1984) 64–71. Peterson, P.J. Bekker, A study of the biological receptor activator of nuclear

[64] A. Caraceni, C. Martini, E. Zecca, R.K. Portenoy, M.A. Ashby, G. Hawson, K.A. factor-kappaB ligand inhibitor, denosumab, in patients with multiple

Jackson, N. Lickiss, N. Muirden, M. Pisasale, D. Moulin, V.N. Schulz, M.A. Rico myeloma or bone metastases from breast cancer, Clin. Cancer Res. 12 (2006)

Pazo, J.A. Serrano, H. Andersen, H.T. Henriksen, I. Mejholm, P. Sjogren, T. 1221–1228.

Heiskanen, E. Kalso, P. Pere, R. Poyhia, E. Vuorinen, I. Tigerstedt, P. Ruismaki, M. [86] J.J. Body, P. Greipp, R.E. Coleman, T. Facon, F. Geurs, J.P. Fermand, J.L.

Bertolino, F. Larue, J.Y. Ranchere, G. Hege-Scheuing, I. Bowdler, F. Helbing, E. Harousseau, A. Lipton, X. Mariette, C.D. Williams, A. Nakanishi, D. Holloway, S.

Kostner, L. Radbruch, K. Kastrinaki, S. Shah, S. Vijayaram, K.S. Sharma, P.S. Devi, W. Martin, C.R. Dunstan, P.J. Bekker, A phase I study of AMGN-0007 a

P.N. Jain, P.V. Ramamani, A. Beny, C. Brunelli, M. Maltoni, S. Mercadante, R. recombinant osteoprotegerin construct, in patients with multiple myeloma or

Plancarte, S. Schug, P. Engstrand, A.F. Ovalle, X. Wang, M.F. Alves, M.R. breast carcinoma related bone metastases, Cancer 1 (97) (2003) 887–892.

Abrunhosa, W.Z. Sun, L. Zhang, A. Gazizov, M. Vaisman, S. Rudoy, M. Gomez [87] B.T. Feeley, N.Q. Liu, A.H. Conduah, L. Krenek, K. Roth, W.C. Dougall, J. Huard, S.

Sancho, P. Vila, J. Trelis, P. Chaudakshetrin, M.L. Koh, R.T. Van Dongen, A. Dubinett, J.R. Lieberman, Mixed metastatic lung cancer lesions in bone are

Vielvoye-Kerkmeer, M.V. Boswell, T. Elliott, E. Hargus, L. Lutz, Breakthrough inhibited by noggin overexpression and RANK:Fc administration, J. Bone

pain characteristics and syndromes in patients with cancer pain. An Miner. Res. 21 (2006) 1571–1580.

international survey, Palliat. Med. 18 (2004) 177–183. [88] L. Penolazzi, E. Magri, E. Lambertini, G. Calò, M. Cozzani, G. Siciliani, R. Piva, R.

[65] P. Niscola, A. Tendas, L. Scaramucci, M. Giovannini, V. De Sanctis, Pain in blood Gambari, Local in vivo administration of a decoy oligonucleotide targeting NF-

cancers, Indian J. Palliat. Care 17 (2011) 175–183. kappaB induces apoptosis of osteoclasts after application of orthodontic forces

[66] A. Caraceni, R.K. Portenoy, An international survey of cancer pain to rat teeth, Int. J. Mol. Med. 18 (2006) 807–811.

characteristics and syndromes, IASP Task Force on Cancer Pain, [89] S. Vallet, S. Mukherjee, N. Vaghela, T. Hideshima, M. Fulciniti, S. Pozzi, L. Santo,

International Association for the Study of Pain, Pain 82 (1999) 263–274. D. Cirstea, K. Patel, A.R. Sohani, A. Guimaraes, W. Xie, D. Chauhan, J.A.

[67] T.H. Diamond, C.S. Higano, M.R. Smith, T.A. Guise, F.R. Singer, Osteoporosis in Schoonmaker, E. Attar, M. Churchill, E. Weller, N. Munshi, J.S. Seehra, R.

men with prostate carcinoma receiving androgen-deprivation therapy: Weissleder, K.C. Anderson, D.T. Scadden, N. Raje, Activin A promotes multiple

recommendations for diagnosis and therapies, Cancer 100 (2004) 892–899. myeloma-induced osteolysis and is a promising target for myeloma bone

[68] T. Rusteon, T. Moum, G. Padilla, S. Paul, C. Miaskowski, Predictors of quality of disease, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) 5124–5129.

life in oncology outpatients with pain from bone metastasis, J. Pain Symptom [90] S. Pozzi, H. Yan, S. Vallet, N. Vaghela, M. Fulciniti, D. Cirstea, L. Santo, S.

Manage. 20 (2005) 234–242. Mukherjee, T. Hideshima, L. Schirtzinger, S. Kuhstoss, N.C. Munshi, K.C.

[69] World Health Organization, Cancer Pain Relief, second ed., World Health Anderson, D. Scadden, N. Raje, Promoting osteoblastogenesis using a novel

Organization, Geneva, 1996 http://whqlibdoc.who.int/publications/ Dkk-1 neutralizing antibody in the treatment of multiple myeloma related

9241544821.pdf (Accessed 04 April 2017).. bone disease, Blood 112 (2008) 2739.

[70] V. Wenter, A. Herlemann, W.P. Fendler, H. Ilhan, N. Tirichter, P. Bartenstein, C.G. [91] P.N. Lara, W.M. Stadler, J. Longmate, D.I. Quinn, J. Wexler, M. Van Loan, P.

Stief, C. la Fougere, A. Rominger, G. Gratzke, Radium-223 for primary bone Twardowski, P.H. Gumerlock, N.J. Vogelzang, E.E. Vokes, H.J. Lenz, J.H.

metastases in patients with hormone-sensitive prostate cancer after radical Doroshow, D.R. Gandara, A randomized phase II trial of the matrix

1284 I. Zaporowska-Stachowiak et al. / Biomedicine & Pharmacotherapy 93 (2017) 1277–1284

metalloproteinase inhibitor BMS-275291 in hormone-refractory prostate adult in chronic prostate cancer-induced bone pain, J. Neurosci. 30

cancer patients with bone metastases, Clin. Cancer Res. 12 (2006) 1556–1563. (2010) 14649–14656.

[92] F.F. di Mola, H. Friess, Z.W. Zhu, A. Koliopanos, T. Bley, P. Di Sebastiano, P. [95] M. Sopata, N. Katz, W. Carey, M.D. Smith, D. Keller, K.M. Verburg, C.R. West, G.

Innocenti, A. Zimmermann, M.W. Büchler, Nerve growth factor and Trk high Wolfram, M.T. Brown, Efficacy and safety of tanezumab in the treatment of

affinity receptor (TrkA) gene expression in inflammatory bowel disease, Gut 46 pain from bone metastases, Pain 156 (2015) 1703–1713.

(2000) 670–679. [96] M. Caprini, A. Gomis, H. Cabedo, R. Planells-Cases, C. Belmonte, F. Viana, A.

[93] S. Pezet, S.B. McMahon, Neurotrophins: mediators and modulators of pain, Ferrer-Montiel, GAP43 stimulates inositol triphosphate-mediated calcium

Annu. Rev. Neurosci. 29 (2006) 507–538. release in response to hypotonicity, EMBO J. 22 (2003) 3004–3014.

[94] J.M. Jimenez-Andrade, A.P. Bloom, J.I. Stake, W.G. Mantyh, R.N. Taylor, K.T.

Freeman, J.R. Ghilardi, M.A. Kuskowski, P.W. Mantyh, Pathological sprouting of