Recent advances in bone-targeted therapy

Recent advances in bone-targeted therapy

Journal Pre-proof Recent advances in bone-targeted therapy Chen Shi, Tingting Wu, Yu He, Yu Zhang, Dehao Fu PII: S0163-7258(20)30001-2 DOI: https:...

6MB Sizes 1 Downloads 76 Views

Journal Pre-proof Recent advances in bone-targeted therapy

Chen Shi, Tingting Wu, Yu He, Yu Zhang, Dehao Fu PII:

S0163-7258(20)30001-2

DOI:

https://doi.org/10.1016/j.pharmthera.2020.107473

Reference:

JPT 107473

To appear in:

Pharmacology and Therapeutics

Please cite this article as: C. Shi, T. Wu, Y. He, et al., Recent advances in bone-targeted therapy, Pharmacology and Therapeutics(2020), https://doi.org/10.1016/ j.pharmthera.2020.107473

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

© 2020 Published by Elsevier.

Journal Pre-proof

P&T #23607 Recent advances in bone-targeted therapy Chen Shi a,# , Tingting Wu a,# , Yu He b , Yu Zhang a, Dehao Fu b,* a

Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong

University of Science & Technology (HUST), Wuhan, P.R. China Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong

f

b

Corresponding author:

pr

*

oo

University of Science & Technology (HUST), Wuhan, P.R. China

e-

Dehao Fu, Department of Orthopaedics, Union Hospital, Tongji Medical College,

Pr

Huazhong University of Science & Technology (HUST), 1277 Jiefang Avenue,

Wuhan 430022, P.R. China. Email: [email protected]

rn

al

These authors contributed equally to this work.

Jo u

#

1

Journal Pre-proof

Abstract

The coordination between bone resorption and bone formation plays an essential role in keeping the mass and microstructure integrity of the bone in a steady state. However, this balance can be disturbed in many pathological conditions of the bone. Nowadays, the classical modalities for treating bone-related disorders are being challenged by severe obstacles owing to low tissue

f

selectivity and considerable safety concerns. Moreover, as a highly mineralized tissue, the bone

oo

shows innate rigidity, low permeability, and reduced blood flow, features that further hinder the

pr

effective treatment of bone diseases. With the development of bone biology and precision

e-

medicine, one novel concept of bone-targeted therapy appears to be promising, with improved

Pr

therapeutic efficacy and minimized systematic toxicity. Here we focus on the recent advances in

bone-targeted treatment based on the unique biology of bone tissues. We summarize commonly

al

used bone targeting moieties, with an emphasis on bisphosphonates, tetracyclines, and biomimetic

rn

bone-targeting moieties. We also introduce potential bone-targeting strategies aimed at the bone

Jo u

matrix and major cell types in the bone. Based on these bone-targeting moieties and strategies, we discussed the potential applications of targeted therapy to treat bone diseases. We expect that this review will put together useful insights to help with the search for therapeutic efficacy in bone-related conditions.

Keywords: Bone targeting, osteoblasts, osteoclasts, bone remodeling, bone formation, bone resorption

2

Journal Pre-proof

Contents

1. Introduction 2. Bone structure and biology 3. Bone-targeting moieties 4. Bone-targeting strategies

f

5. Targeted therapy for bone-related diseases

oo

6. Conclusion and perspectives

pr

Conflicts of interest

e-

Acknowledgements

Jo u

rn

al

Pr

References

3

Journal Pre-proof

Abbreviations

Asp,

aspartic

acid;

ALN,

alendronate;

BMPs,

bone

morphogenetic

proteins;

BPs,

bisphosphonates; BSP, bone sialoprotein; BMD, bone mineral density; CAP, chondrocyte-homing peptide; CBDs, collagen binding domains; CDs, carbon dots; DC, doxycycline; DOX, doxorubicin; ET-1, endothelin-1; Glu, glutamic acid; HA, hydroxyapatite; MMPs, matrix

oo

f

metalloproteinases; MSCs, mesenchymal stem cells; OPG, osteoprotegerin; PDB, Paget's disease of bone; PTH, parathyroid hormone; PGE2, prostaglandin E2; PLGA, Poly(lactic-coglycolic

pr

acid); RANK, receptor activator of NF-κB; SAA, salvianic acid A; SAON, steroid-associated

e-

osteonecrosis; Sct, Salmon calcitonin; SERMs, selective estrogen receptor modulators; SELEX,

Pr

systematic evolution of ligands by exponential enrichment; TCs, tetracyclines; TKIs, tyrosine

Jo u

rn

al

kinase inhibitors; VEGF, vascular endothelial growth factor; ZA, zoledronic acid.

4

Journal Pre-proof

1. Introduction

The bone is a specified connective tissue with highly mineralized architecture and structure (Burr, 2019). As an internal support system, the bone provides a structural foundation for the body and sites for muscle attachment for locomotion. Additionally, the bone offers protection to the brain and splanchnic organs, as well as a house for hematopoietic tissues of the bone marrow. The

f

bone is also the primary source and depot of inorganic ions, especially phosphate and calcium,

oo

which can participate actively in mineral homeostasis and energy metabolism in the body

pr

(Florencio-Silva, et al., 2015). For the healthy development and maintenance of the skeleton, the

e-

bone undergoes a constant modeling and remodeling process through bone formation by

Pr

osteoblasts and bone resorption by osteoclasts (Low & Kopecek, 2012). Under healthy

physiological conditions, the balance between bone-resorption and rebuilding is well-coordinated,

al

which can keep the microstructure integrity and mass of the bone in a steady state. However, this

rn

equilibrium would be disturbed in many bone diseases (Rodan & Martin, 2000).

Jo u

As the most prevalent form of bone metabolic disorders, osteoporosis has a high incidence in women and men over 50, especially postmenopausal women. Osteosarcoma and bone metastasis, other types of imbalances in bone metabolism, could cause significant pain and increase mortality rates in patients. Other diseases, including osteoarthritis, osteomyelitis, and Paget's disease of bone (PDB), could amplify the economic burden and decrease the quality of life for millions of individuals worldwide. With an advanced understanding of bone biology, the treatment of bone diseases has been

improved but still faces significant obstacles in the clinic. Estrogen replacement therapy is usually utilized for osteoporosis in postmenopausal women. However, the treatment remains a source of 5

Journal Pre-proof

considerable concerns after prolonged administration, especially as a potential risk for breast cancer, uterus cancer, and endometritis (Astedt, 1982; Wallach & Gambrell, 1982). Parathyroid hormone (PTH), the most frequently used anabolic, is effective in stimulating bone formation. Unfortunately, the anabolic effect of PTH is only limited to two years, owing to a secondary osteoclastogenic effect that results in bone catabolism over anabolism (Cosman., et al., 2005;

f

Neer, et al., 2001). More significantly, patients receiving PTH may also carry an increased risk of

oo

developing osteosarcoma (Leder, 2017; Lindsay, et al., 1997; Neer, et al., 2001). An RNA

pr

interference (RNAi)-based therapy aimed at bone disease-associated pathogenic genes offers a

e-

new genetic medical approach; this therapy is potentially translational therapy for bone-related

Pr

diseases (Novina & Sharp, 2004). However, this modality requires a large therapeutic dose of

systematically administered siRNA, which would increase cost and safety concerns (Itaka, et al.,

al

2007). Moreover, the above chemicals or biotherapies are usually administered systematically

rn

when treating bone-related disorders. On the one hand, the therapeutic agents have a non-specific

Jo u

in vivo bio-distribution, causing undesired side effects to non-skeletal tissues. On the other hand, the low concentration of therapeutics in localized bone would maximize therapeutic efficiency (Carbone, et al., 2017). These limitations leave the field with a significant challenge to surmount in the current bone disease treatment strategies. Hence, the development of effective and innovative therapies against bone-related diseases is highly desirable. The most important clinical objective in treating clinical diseases is to realize precise therapeutic efficacy in defined pathological s ites. Specific treatment for bone-related disorders

also inspires abundant interests. Pierce et al (William M. Pierce, 1987) first proposed the concept of ‘bone-targeting’ in 1986, providing new insights into bone diseases. With years of concerted 6

Journal Pre-proof

efforts, targeted therapy has made considerable progress in bone disease treatment (He, et al., 2017; Hirabayashi & Fujisaki, 2003; Sousa & Clezardin, 2018; Zhang, et al., 2018). By endowing bone therapeutic agents with osteotropicity, their pharmacokinetic profile can be altered significantly to a favorable skeletal deposition. This therapeutic regiment in bone tissues would work sustainably and effectively to improve the local therapeutic index (Wang, et al., 2005).

f

Besides, the preferential accumulation of therapeutics in the bone can decrease the systematic

oo

distribution to other tissues, potentially minimizing systemic toxicity and long-term concerns

pr

remarkably (E. J. Carbone, et al., 2017). Also, the required dose of therapeutic agents in

e-

bone-targeted therapy for local application is smaller when compared to doses used in traditional

Pr

treatments (Cheng, et al., 2017). Given these unique advantages, bone-targeted treatment could

serve as a promising modality in the therapeutic field of bone diseases.

al

In this review, we will focus on the advances in recent findings on bone-targeted therapy

rn

based on the unique biology of bone tissues. We will strive to introduce the commonly used

Jo u

bone-targeting moieties, with emphasis on bisphosphonates (BPs), tetracyclines (TCs), and bone-targeting biologicals. We will also summarize the main bone-targeting strategies aimed at the bone matrix and major cell types, such as osteoblasts, bone lining cells, osteoclasts, and osteocytes in bone tissues. We will then discuss the potential and promising applications of targeted therapy to treat bone diseases, including osteoporosis, osteosarcoma and bone metastasis, osteoarthritis, bone fracture and others based on the bone-targeting moieties and strategies. We hope that this review will provide useful information towards overcoming the current dilemma faced by

bone-related therapies and offer new insights into finding ways to treat bone-related diseases efficiently. 7

Journal Pre-proof

2. Bone structure and biology 2.1. Composition of the bone matrix The composition of the bone varies with its diseases, age, anatomical location, and nutritional status. Generally, an adult bone is composed of an organic matrix (20-40%), inorganic mineral

f

(50-70%), water (5-10%), and lipids (1-5%) (Shea & Miller, 2005).

oo

Organic matrix

pr

The organic matrix of the bone consists primarily of collagen (approx. 90%), predominantly

e-

type I collagen, a triple-helical molecule composed of a single alpha-2 chain and two identical

Pr

alpha-1 chains. Collagen is deposited in layers of mature bones, known as lamellae, which endows

the bone tissue with a high density of collagenous filaments. Non-collagenous proteins, such as

al

osteonectin, osteopontin, and matrix GLA protein, within the bone matrix, may play a role in the

rn

matrix mineralization process, while others, like growth factors, bone morphogenetic proteins

Jo u

(BMPs), cytokines, and adhesion molecules have important signaling functions (Sommerfeldt & Rubin, 2001). Moreover, proteoglycans, including biglycan, osteoaderin, decorin, and lumican, are also located in the matrix (Florencio-Silva, et al., 2015). Inorganic mineral Bone mineral accounts for 50% and 75% of bone tissue by volume and weight, respectively. The majority of inorganic materials in the bone mineral are calcium and phosphate ions. Significant amounts of sodium, bicarbonate, citrate, potassium, magnesium, zinc, fluorite,

strontium, and barium are also present in the bone (Downey & Siegel, 2006). Notably, calcium and phosphate ions nucleate to form hydroxyapatite (HA) [Ca10 (PO4 )6 (OH)2 ], a plate-like crystal 8

Journal Pre-proof

that is 20-80 nm in length and 2-5 nm in thickness; this crystal can provide strength and rigidity to skeletal load-bearing features. HA also contains many impurities, such as carbonate and magnesium, that cause bone apatite to be crystallized poorly and carbonate substitutes apatite. This imperfection could render apatite more soluble and, thus, enable it to release ions for homeostasis (Shea & Miller, 2005).

f

2.2. Bone cell biology

oo

As a rigid structure, the bone carries a self-repairing ability that can remove and replace

pr

mineral stores rapidly during metabolic demand and reshape the structure after altering

e-

mechanical stimuli. Four distinctly different cell types participate in the formation, resorption, and

Pr

maintenance of the bone: osteoblasts, bone lining cells, osteoclasts, and osteocytes (Table 1). Osteoblasts

al

Osteoblasts are a group of cells with a cuboidal shape located along the surface of the bone.

rn

These cells account for 4-6% of total bone cells (Capulli, Paone, & Rucci, 2014). Osteoblasts are

Jo u

derived from a mesenchymal stem cell (MSC) lineage and can form a compact layer of cells on the bone surface together with their precursors. Structurally, these cuboidal-shaped cells contain plenty of rough endoplasmic reticula, well-developed Golgi apparatus, and diversified secretory vesicles. These morphological characteristics reflect the protein-synthesizing and secreting activity of these cells (Marks & Popoff, 1988). Functionally, osteoblasts are well known for their prominent bone-forming abilities through the deposition of the organic matrix and participation in subsequent bone mineralization. During this process, osteoblasts first produce collagen,

predominantly type I collagen, non-collagenous proteins (e.g., osteocalcin, bone sialoprotein (BSP) II, and osteopontin), and proteoglycan (e.g., biglycan and decorin) to form an organic 9

Journal Pre-proof

matrix (Florencio-Silva, et al., 2015). Mineralization then takes place in two phases following the organic matrix formation: vesicular phase and fibrillar phase (Burr, 2019; Yadav, et al., 2016). In the vesicular phase, matrix vesicles with variable sizes, ranging from 30 to 200 nm, are released by osteoblasts to the newly formed bone matrix. Osteoblasts then secrete enzymes and alkaline phosphatase (ALP) to release calcium ions from proteoglycan-binding compounds and phosphate

f

ions from phosphate-containing compounds, respectively, inside the vesicles, which can further

oo

form HA crystals (Arana-Chavez, Soares, & Katchburian, 1995). In the fibrillar phase, once the

pr

calcium and phosphate ions in matrix vesicles supersaturate, their structures are ruptured, and the

e-

HA crystals diffuse to surrounding matrixes (Boivin, et al., 2008; Silva & Bilezikian, 2015).

Pr

Bone lining cells

Bone lining cells are inactive osteoblasts at the bone surface. These quiescent cells differ

al

from active osteoblasts and do not undergo neither bone formation nor resorption.

rn

Morphologically, bone lining cells are flat-shaped cells with extremely flat nuclei and few

Jo u

cytoplasmic organelles that can extend well along the bone surface (Matic, et al., 2016). Depending on the physiological status of the bone, bone lining cells can reacquire secretory abilities, increase in size, and adopt a cuboidal shape (Miller, et al., 1989). When bone resorption does not initiate, these cells prevent direct contact between bone matrixes and osteoclasts and participate in osteoclast differentiation (Hienz, Paliwal, & Ivanovski, 2015). Bone lining cells are also a substantial portion of the temporary anatomical structure noted as the basic multicellular unit during the bone remodeling process (Everts, 2002).

Osteoclasts

10

Journal Pre-proof

Osteoclasts are polarized, multinucleated giant cells that originate from hematopoietic stem cell lineage. The most important characteristic of osteoclasts is their ability to resorb fully mineralized bones (Detsch & Boccaccini, 2015). Resorption begins with the activation of osteoclasts, which requires osteoclasts to attach to the bone surface (Teitelbaum, 2000). Bone resorption then occurs in a particular invaginated cell specialization zone known as the “ruffled

f

border,” the site of proteolytic enzyme and acid release for matrix degradation and mineral

oo

dissolution (Merolli, et al., 2018). Subsequently, the degraded matrix and mineral byproducts are

pr

transported to the extracellular environment at the opposite side of the cells from the ruffled

e-

border. After the degradation process, osteoclasts either become inactive or directly die via cell 3

Pr

apoptosis. Generally, an activated osteoclast can complete 200,000 µm worth of resorption per

day, which is nearly equal to the bone formation rate by 7 to 10 generations of osteoblasts with a

al

15 to 20 days lifespan (Sommerfeldt & Rubin, 2001). Reportedly, osteoclasts also possess other

Jo u

Osteocytes

rn

functions, such as the direct regulation of the hematopoietic stem cell niche (Julia F, 2014).

As the most abundant and long-living cells in bone cell populations, osteocytes comprise 90% to 95% of all bone cells and have an extremely long lifespan of up to 25 years (Franz-Odendaal, Hall, & Witten, 2006). Osteocytes are derived from MSCs through the differentiation of osteoblasts. Though the precise mechanisms of how osteoblasts become osteocytes remain elusive, it is believed that an embedding mechanism may explain the transmission process. Compared to neighboring cells, a subpopulation of osteoblasts on bone

surface slow down their matrix production and then become “buried alive” in the mineralized matrix produced by adjacent osteoblasts (Franz-Odendaal, Hall, & Witten, 2006). After 11

Journal Pre-proof

transmission, osteocytes display distinctly different structures and functionalities. Compared to osteoblasts, osteocytes have a smaller size, increased nucleus to cytoplasm ratio, and fewer cell organelles. Osteocytes also show a dendritic morphology with a higher number of filopodia (Sommerfeldt & Rubin, 2001). Though they cannot achieve bone formation directly like osteoblasts, they have several important roles. Typically, osteocytes can permit the diffusion of

f

mineral and fluids, helping to maintain bone mineral homeostasis (Shea & Miller, 2005).

oo

Additionally, osteocytes can detect mechanical pressures and serve as mechanosensors, promoting

pr

the adaptation of the bone to daily mechanical forces (Rochefort, Pallu, & Benhamou, 2010). This

Pr

Prideaux, Findlay, & Atkins, 2016).

e-

cell type can also regulate the activities of both osteoblasts and osteoclasts (Mori & Burr, 1993;

The interconnection between bone cells and matrix in bone microenvironment

al

It has been suggested that the crosstalk between bone cells should be a coordinate process for

rn

steady bone formation and resorption. Initially, the detailed mechanism of this crosstalk was not

Jo u

well understood for years. It was not until 1997 that a molecular bas is for the intercellular crosstalk was discovered in the form of osteoprotegerin (OPG) and its cognate ligand OPG-L. OPG is secreted by osteoblasts, while the OPG-L is expressed on osteoblasts. Both molecules can bind with the receptor activator of NF-κB (RANK) to activate the downstream signal, a typical transmembranous receptor expressed on osteoclast precursors. The binding of OPG-L (also known as RANK-L) on osteoblasts with RANK induces a signaling and gene expression cascade, promoting the formation of osteoclasts from precursors (Ahmed, 2015; Hofbauer, et al., 2000). In

contrast, OPG competes with RANK to bind with RANK-L, which can inhibit osteoclast formation and subsequent bone resorption effectively. 12

Journal Pre-proof

Recently, semaphorins (Sema) have been revealed as key molecules participating in the cell-cell communication between osteoblasts and osteoclasts (Hayashi, et al., 2012). Among the identified semaphorin families, two types are closely related to cell crosstalk. Semaphorin 3A (Sema3A) produced by osteoblasts inhibits osteoclasts, while Semaphorin 4D (Sema4D) secreted by osteoclasts inhibits osteoblasts (Negishi-Koga & Takayanagi, 2012). Other factors are also

f

involved in the bone remodeling process. EphrinB2 (Eph2), a membrane-bound molecule located

oo

on osteoclasts, can bind with ephrinB4 (Eph4) expressed on osteoblast membranes. Remarkably,

pr

their bindings transduce bidirectional signals. An Eph2/Eph4 binding facilitates the differentiation

e-

of osteoclasts, whereas a reversed Eph4/Eph2 signaling decreases osteoclastogenesis (Zhao, et al.,

Pr

2006).

Located within lacunae encompassed by highly mineralized bone matrices, osteocytes harbor

al

numerous cytoplasmic processes that function through small channels, expediting their

rn

interconnection with neighboring osteocytes, bone lining cells, and osteoblasts on the bone

Jo u

surface. Osteocytes, like osteoblasts, can secret RANK-L or OPG to promote or inhibit osteoclastogenesis, respectively. Also, upon mechanical stimulus, osteocytes secret secondary factors to modify bone physiology. Moreover, nitric oxide (NO), prostaglandin E2 (PGE2), and insulin-like growth factor-1 (IGF-1) can stimulate osteoblast activity, while sclerostin and the dickkopf WNT signaling pathway inhibitor (DKK-1) play a converse role. The extracellular matrix is closely associated with the bone cells and plays an essential role in bone homeostasis. The collagenous matrix generated by osteoblast-progenitors regulate growth

factors (e.g. BMPs), facilitating osteoblast differentiation and matrix maturation (Yang, et al., 2003). Osteoblasts/osteocytes produce matrix metalloproteinases (MMPs) which promote the 13

Journal Pre-proof

remodeling of matrix and development of the lacunocanalicular system. Osteocytes also secrete dentin matrix protein 1 (DMP1) and matrix extracellular phosphoglycoprotein (MEPE) to regulate calcium and phosphate metabolism in matrix (Alford, Kozloff, & Hankenson, 2015). Furthermore, various matrix proteins provide RGD binding sites to bind with integrins on osteoclast, enabling the formation of ruffled border and sealing off of resorptive spaces, where cathepsin K and

f

hydrogen ions are then surrounded to degrade type I collagen and demineralize inorganic

oo

components, respectively (Lakkakorpi, et al., 1991). Along with bone resorbing process,

pr

osteoclasts regulate extracellular calcium concentration, and in turn, the extracellular and

e-

intracellular calcium level influence the osteoclast activity (Li, Kong, & Qi, 2006). Apart from

Pr

bone cells, PTH act as a major regulator of the levels of calcium and phosphate in the bone matrix (Wein & Kronenberg, 2018).

al

As multipotent stromal cells, MSCs are capable of osteogenic differentiation (Grayson, et al.,

rn

2015). The bone marrow microenvironment created during bone remodeling directs the fate of

Jo u

MSCs. IGF-1 released from the bone matrix can stimulate the differentiation of MSCs to osteoblasts by activating mammalian target of rapamycin (Xian, et al., 2012). Transforming growth factor β (TGF-β), which is released and activated by osteoclasts, can recruit MSCs specifically to local site of repair for new bone formation. The expression of Sema4D on osteoclasts, on the other hand, inhibits MSC differentiation (Crane & Cao, 2014) (Figure 1).

14

pr

oo

f

Journal Pre-proof

e-

Figure 1. The interconnection between bone cells and matrix in bone microenvironment.

Pr

Osteoblasts express RANK-L or secret OPG to bind with RANK on osteoclast precursors, which

facilitate or inhibit osteoclast formation, respectively. Sema3A produced by osteoblasts inhibits

al

osteoclasts, while Sema4D secreted by osteoclasts inhibits osteoblasts. Eph2, a membrane-bound

rn

molecule present on osteoclasts, binds with Eph4 expressed on osteoblasts. Eph2/Eph4 binding

Jo u

facilitates the differentiation of osteoclasts, when a reversed Eph4/Eph2 signaling decreases osteoclastogenesis. Osteocytes secret RANK-L or OPG to promote or inhibit osteoclastogenesis, respectively. Osteocytes also produce NO, PGE2 and IGF-1 to stimulate osteoblast activity, while release sclerostin and

DKK-1

to

inhibit

osteoblast

activity.

For

matrix

regulation,

osteoblasts/osteocytes produce MMPs to promote the remodeling of matrix and development of the lacunocanalicular system. Cathepsin K and hydrogen ions in ruffled border degrade type I collagen and demineralize inorganic components, respectively. IGF-1 released from the bone

matrix stimulates MSCs differentiation to osteoblasts. TGF-β released by osteoclasts recruits

15

Journal Pre-proof

MSCs to local site of repair for bone formation while Sema4D inhibit MSC differentiation. More in-depth discussions please refer to the text.

3. Bone-targeting moieties Because of its unique structure and composition, the bone also provides opportunities for

f

varieties of agents called bone mineral seekers to selectively target skeletal tissues. Years of

oo

extensive investigations have demonstrated that different types of moieties, including many small

pr

molecules (< 1000 Da), as well as large macromolecular proteins, show high bone specificity.

e-

Typical bone-targeting moieties are classified in Table 2 and will be discussed in detail in the

Pr

following paragraphs.

3.1. Bisphosphonates

al

BPs are a class of analogs of naturally occurring pyrophosphates. Fleisch and colleagues first

rn

reported the biological function of BPs in the 1960s (Mühlbauer, 1968). Since then, BPs have

Jo u

been investigated extensively for biomedical applications. These analogs possess a similar ability to regulate bone mineralization as pyrophosphates but are much more chemically stable than pyrophosphates. Structurally, the two phosphonate groups (PO(O−)2) share a common oxygen molecule (P–O–P) in pyrophosphates, but the oxygen molecule in BPs is replaced by a carbon atom (P–C–P). The substitution of the P-C-P backbone in BPs does not affect its affinity for bone minerals but makes it more resistant to most chemical reagents and enzymatic degradation, resulting in an extended residence for BPs in skeletal tissues for up to years (Brown, et al., 2014).

The affinity of BPs for the bone is greatly affected by the molecular structure of BPs. Mechanistically, the P–C–P backbone presents two phosphonate groups to chelate with divalent 16

Journal Pre-proof calcium ions (Ca2+) in a bidentate structure on an HA surface (Yewle, 2012). The two remaining side chains (R1 and R2) on the P-C-P carbon atom can regulate BPs’ affinity for HA further. R1 side chains are responsible for interacting with the HA content of the bone, especially where a large amount of calcium is released from osteoporotic bone tissues. The existence of the hydroxyl or amine group at the R1 side chain can further increase bone affinity through additional 2+

f

interaction with Ca to generate a tridentate binding to HA (Lawson, et al., 2010).

oo

Based on the presence or absence of nitrogen side groups at the R2 position, BPs can be

pr

categorized into two groups: nitrogen-containing BPs (N-BPs) and non-nitrogen-containing BPs

e-

(non-N-BPs). It has been suggested that the N-BPs possess a higher binding affinity for the bone

Pr

than non-N-BPs (Nancollas, et al., 2006). This difference in affinity could be attributed to the

direct hydrogen bonding action between nitrogen functional groups and the hydroxyl on a HA

al

surface. Moreover, N-BPs can significantly inhibit the synthesis of farnesyl pyrophosphate that is

rn

necessary for osteoclast functioning (Drake, Clarke, & Khosla, 2008). The in vivo bone

Jo u

homeostasis will then be shifted toward bone formation due to the reduced osteoclast activity, which does not affect osteoblast bone formation. Non-N-BPs can be metabolized to toxic methylene-containing adenos ine triphosphate (ATP) analog in bone cells, including osteoclasts, inducing substantial cell apoptosis (Reyes, et al., 2016). Besides, the chemical groups positioned at the R1 and R2 chains can be conjugated with nonspecific bone therapeutic agents to obtain osteotropicity (Rotman, et al., 2018). The use of BPs as bone-targeting moieties still faces several potential complications. Because

BPs can inhibit bone resorption, atypical femur fractures (AFF) could be induced by long-term low bone turnover (Dell, et al., 2012). Compared with AFF, osteonecrosis of the jaw (ONJ) occurs 17

Journal Pre-proof

at a much lower level. However, the occurrence rate of ONJ increases when BPs are used under an oncological condition (Khan, et al., 2015). BPs can also cause extra-skeletal adverse effects, including gastrointestinal effects, cancer of the esophagus, and atrial fibrillation (Reyes, et al., 2016). The gastrointestinal side effects could be related to the poor gastrointestinal absorption of oral BP, which prolongs the exposure of gastric and intestinal mucosa to a large amount of BP

f

(Pazianas, et al., 2010). In the meantime, taking pills could induce persistent injury of the mucosa,

oo

which could lead, potentially, to dysplasia of the esophageal cells and increase the risk of cancer

pr

of the esophagus (Green, et al., 2010). Atrial fibrillation could result from the increased blood

e-

calcium level, together with the possible inflammation of arterial wall caused by BP (Abrahamsen,

Pr

Eiken, & Brixen, 2009).

The adverse effects of BPs are correlated with the given dosage. At high doses, BPs could

al

inhibit bone mineralization and induce osteomalacia. Patients who have never received BP therapy

rn

and are given a high dose of BP, may experience a typical acute-phase response with fevers up to

Jo u

39°C for 1 to 3 days and transient haematological changes. Besides, the fast intravenous injection of BPs at doses of more than 200 to 300 mg could induce severe renal failure (Adami & Zamberlan, 1996).

3.2. Tetracyclines TCs and their analogs are a family of compounds that display a basic chemical structure comprising a tetracyclic naphthacene carboxamide ring system surrounded by various functional groups and substituents. It is well-known that TCs can bind to bone apatite. This special attraction

to skeletal tissues is mainly attributed to the good metal-complexing abilities of TCs, which can chelate with Ca2+ in the HA of bone matrix (Albert & Rees, 1956). Van der Waals interactions and 18

Journal Pre-proof

hydrogen bonding between TCs and HA are also likely to induce surface complexation to bone apatite, contributing to an additional association between TCs and bones (Wang, Hu, & Zhang, 2010). TCs can equally be conjugated with estrone to develop a bone-targeted estrogen (XW-630). Compared to free estrone, TC-estrone seemed to be more efficient in improving trabecular bone connectivity (Lu, Qiu, & Zheng, 1998). Apart from targeting the skeleton, TCs can as well inhibit

f

collagenase with a potent anti-collagenolytic effect, that would reduce bone resorption.

oo

Furthermore, TCs can increase the expression of procollagen mRNA, elevating the number of

pr

activated osteoblasts (Reichert, et al., 2012). Interestingly, TCs have fluorescent properties and can

e-

label the surface of a growing bone formation area, which can be used for imaging and

Pr

quantifying new bone formation (Nakamura, et al., 2000).

Doxycycline (DC), a TC analogue, is also osteotropic with a high affinity for mineralized

al

bone. Moreover, DC shows potent benefits for connective tissue remodeling and healing. The

rn

microenvironment of chronic wounds is pro-inflammatory containing a high level of MMP. DC

et al., 2017).

Jo u

displays anti-inflammatory and MMP inhib itory effect , thus promoting normal tissue repair (Gomes,

There are several limitations to using TCs. The inherent biological activity involving heartburn, rash, dizziness, hypoglycemia, and nausea is a considerable concern in the practical use of TCs (Valentín, et al., 2009). Moreover, the chelation of TCs to skeletal tissues is permanent, which leads to undesired side effects, especially for teeth discoloration during dental development in children, and this permanent discoloration from yellow or gray to brown depends on the dose of

TCs (Sánchez, Rogers III, & Sheridan, 2004). Additionally, a high dose of TC may induce enamel hypoplasia during calcification (Bevelander, Rolle, & Cohlan, 1961). Despite high osteotropicity, 19

Journal Pre-proof

TCs show decreased affinity for pathologic skeletal sites with low bone turnover, making TCs suboptimal candidates for bone-related diseases, such as osteomyelitis (J. Hatzenbuehler, 2011). Besides, TCs are quite chemically unstable, which further hinders their clinical application. 3.3. Biomimetic bone-targeting moieties Bone-targeting peptides Recently, utilizing the repetitive acidic amino acids of natural bone proteins to interact with

oo

f

Ca2+ in mineralized tissues has emerged as a fundamental bone-targeting concept. Among these

pr

amino acids, acid oligopeptides, composed of repeating aspartic acid (Asp) or glutamic acid (Glu)

e-

sequences, natively occurring in osteopontin and osteocalcin, show a great affinity for HA

Pr

(Rotman, et al., 2018). Though the exact mechanism is still under debate, the structure of acid

oligopeptides has been demonstrated to correlate to the exclusive interaction between

al

oligopeptides and mineralized tissues. Acid oligopeptides with 4 to 10 Asp or Glu units (Asp4-10,

rn

Glu4-10) are reportedly considered as more biocompatible options for bone-targeting purposes

Jo u

(Junko, 2009). The optical isomeric form (L or D) of peptides affects in vivo biocompatibility to some extent. Compared with L-peptides, D-peptides are not readily recognized by the immune system and, thus, promote in vivo biocompatibility (Low & Kopecek, 2012). Oligopeptides’ binding affinity for the bone varies depending on the number of amino acids they contain, the binding rates of small peptides with HA are enhanced with increasing chain lengths of Asp and Glu. However, the binding affinity comes to a steady level when the chain is up to hexapeptides. As a result, acid oligopeptides with over six amino acids in length are the ideal candidates for

binding efficiency requirements. Meanwhile, there is evidence that the chirality of Asp and Glu (L or D) has no impact on mineral chelating properties (Sekido, et al., 2001). 20

Journal Pre-proof

Another oligopeptide, with six repeating sequences of Asp, serine, and serine ((AspSerSer)6), also shows a good skeletal affinity for the bone and has been widely used as a bone-targeting moiety. While Asp8 selectively binds to the bone resorption surface, (AspSerSer)6 displays selective binding to the bone formation surface, as it favorably chelates with osteoblast-mediated mineralizing nodules and amorphous calcium phosphate (Yarbrough, et al., 2010).

f

Apart from oligopeptides, other peptide sequences present osteotropicity. A peptide

oo

consisting of twelve amino acids, VTKHLNQISQSY (VT K), has been confirmed to have a high

pr

affinity for HA and skeleton-like materials. The phosphorylation of serine and the hydroxyl side

e-

group of tyrosine in the VTK peptide sequence are the major interacting domains involved in HA

Pr

binding (William N, 2010).

Bone-targeting proteins

al

The most abundant protein in the organic matrix is type I collagen, making it a highly

rn

potential targeting site in bone tissues. Collagen binding domains (CBDs), largely present in the

Jo u

collagenolytic proteases of microorganisms, can bind specifically with collagen (Watanabe, 2004). The cDNA of CBDs can be fused easily into the C-terminus or N- terminus of proteins of interest using standard molecular biology techniques. The fusion-protein, with an unimpaired protein structure and bioactivity, can then be obtained by expressing the recombinant protein. The expected proteins, such as PTH and stromal-derived factor-1-alpha (SDF-1α), can acquire the specific binding ability to collagen in the bone (Sun, et al., 2016; Tulasi, 2011). In addition to collagen, several other matrix proteins, including osteocalcin, sialoprotein, and

osteopontin, account for the minor components of the bone matrix. Interestingly, all of these proteins have been demonstrated to have a good affinity for Ca2+ and the mineral surface of the 21

Journal Pre-proof

bone (Ryuichi Fujisawa, 2012). Osteocalcin, a γ-carboxyglutamic acid-containing protein, is the most abundant non-collagenous protein in the bone. It can bind strongly to HA, instead of amorphous calcium phosphate, preventing crystallization. BSP is an acidic glycol-phosphoprotein in the bone that contains RGD (Arg-Gly-Asp) cell-attachment sequences near the C-terminal end; these sequences can be recognized by the presence of integrin αvβ3 and mediate selective

f

attachments to osteoblasts and fibroblasts (Oldberg, Franzen, & Heinegard, 1988). This protein

oo

also has polyglutamic acid sequences that are responsible for initiating the nucleation of the HA

pr

crystal (Hunter & Goldberg, 1993). Moreover, BSP shows a unique affinity for collagen, which

e-

further increases the potency for HA nucleation (Baht, Hunter, & Goldberg, 2008). Another

Pr

multifunctional protein, osteopontin, also contains RGD adhesive sequences, as well as Asp-rich

calcium-binding domains, which can bind to the surface of HA effectively. In contrast to BSP’s

rn

Bone-targeting cells

al

nucleation, osteopontin is likely to inhibit the formation and growth of the crystal (Boskey, 1995).

Jo u

With pluripotent nature, MSCs can be trafficked to injured tissues, also termed endogenous stem cell homing, and participate in subsequent tissue regeneration (Lin, et al., 2017). In bone tissue, MSCs can be recruited to fractured sites via (SDF)1/CXC chemokine receptor (CXCR) 4 signaling axis and improve healing by increasing the bone formation and bone content of the callus (Kitaori, et al., 2009). As described above, TGF-β released by osteoclasts can facilitate MSC recruitment in bone resorptive sites further. Besides, many integrins, including integrin α1, α2, α3, α4, α5, α6, α11 and β1 are expressed in MSCs (Guan, et al., 2012). Among them, the

overexpression of integrin α4 can promote MSC homing to bone (Mukherjee, et al., 2008).

22

Journal Pre-proof

Although using biomimetic moieties for bone-targeting presents fewer adverse effects in vivo, their limitations should also be noted. The poor stability of enzymatic cleaving and low oral bioavailability constitute the therapeutic concerns of these moieties in their practical utilization. The manufacturing process is similarly quite complicated, labor-intensive, and requires professional laboratory techniques, resulting in relatively high costs. Moreover, special conditions

f

for transport and storage are required (Aoki, et al., 2012; Sun, 2013).

oo

3.4. Other moieties

pr

In addition to the molecules mentioned above, a few other moieties can also be used to target

e-

the bone. The phosphonate groups in BPs are known to play an essential role in their interaction

properties.

Pr

with bone minerals. So, other phosphonate-containing compounds may likewise show similar

Tetraazacyclotetradecane-1,4,8,11-tetramethylene

phosphonic

acid

and

al

ethylenediamine tetra(methylene phosphonic acid), both containing four phosphonate groups, can

rn

chelate to Ca2+ on skeletal tissues (Lange, et al., 2016). Different from BPs, these compounds

Jo u

display a negligible physiological effect on bone metabolism (Rotman, et al., 2018). Recently, thanks to the systematic evolution of ligands by exponential enrichment (SELEX), aptamers consisting of short DNA/RNA oligonucleotides can be screened as targeting molecules with strong specificity and affinity. The SELEX method comprises three procedures: selection, partitioning, and amplification. Before selection, a library of oligonucleotides containing 1015 different unique sequences is first synthesized, then incubated with targeted-molecules for selection. After incubation, the unbound sequences are removed using different methods; then the

bound sequences are amplified by PCR and utilized for a new round of selection. Lastly, the specific cell-targeted aptamer candidates are obtained after several cycles (Zhuo, et al., 2017). In 23

Journal Pre-proof

this way, the CH6 aptamer with the inherent merits of high specificity and efficiency that can directly target osteoblasts is selected (Liang, et al., 2015). Radionuclides, such as Strontium and Lanthanides, due to their similar physical and chemical properties to those of calcium, can preferentially be deposited in bone tissues and participate in the metabolic process of bone minerals (Cawthray, et al., 2015).

oo

f

4. Bone-targeting strategies

Targeted therapy has been investigated extens ively for more than thirty years for use in the

pr

treatment of bone-related diseases. The advantages of the unique characteristics of targeted

e-

therapy show that it can improve drug utilization for increased therapeutic outcomes, as well as

Pr

reduce systemic toxicity-related side effects. Generally, bone-targeting strategies can be classified

rn

4.1. Bone matrix-targeting

al

into two types: bone matrix-targeting and bone cell-targeting strategies.

Thirty-five percent of the dry weight of the bone is organic collagen, and the rest is mainly

Jo u

2+

composed of inorganic HA, which contains 99% of the total Ca

in the body. Given their unique

characteristics, both collagen and HA can serve as potential targets of compounds or molecules deposited in bone tissues.

Fusing therapeutic proteins with CBDs is a strategy that is used frequently to target organic collagen in the bone. PTH, an essential protein in regulating bone remodeling, can activate osteoblasts directly. The protein’s functions are mainly mediated by the PTH receptor (PTH1R) that is expressed on osteocytes and osteoblasts (Silva & Bilezikian, 2015). Generally, CBDs consist of amino acids 861–981 of the ColH collagenase of Clostridium histolyticum (Matsushita, 24

Journal Pre-proof

et al., 1998). The fusion proteins of PTH can be synthesized by conjugating the carboxy-terminus of PTH directly with the amino-terminal of CBD (CBD-TH) or with CBD via an adjacent ColH domain known as a polycystic kidney disease domain (PKD, CBD-PKD-PTH). Both hybrid proteins can preferentially bind to type I collagen and increase cyclic Adenos ine monophosphate (cAMP) accumulation in PTH/PTHrP receptor transfected cells, resulting in an anabolic effect.

f

However, in practical use, PTH-CBD showed a 15% increase in bone mineral density (BMD),

oo

while PTH-PKD-CBD induced a modest increase (5%), with the PKD domain potentially

pr

reducing the availability of PTH to the PTH/PTHrP receptor. The results suggested that the direct

e-

link between PTH and CBD is more suitable for the treatment of bone diseases (Tulasi, 2011).

+

Pr

Besides PTH, CBD can also fuse with SDF-1α to selectively bind with collagen, with the resulting +

proteins potentially promoting CD34 and c-kit endogenous stem cells to home in the injured

al

region for bone regeneration (Shi, et al., 2016).

rn

Many compounds and molecules show a specific affinity for HA, demonstrating an excellent

Jo u

potential to target bone minerals. Thanks to the development of the chemical conjugating technique, therapeutic agents can be grafted easily with bone-targeting moieties by a linker element to acquire bone affinity. As analogs of natural pyrophosphates, BPs have a strong affinity for calcified tissues and can bind effectively to HA on the bone surface. PGE2 can stimulate bone formation after in vivo systematic administration (Chyun & Raisz, 1984); it can be coupled chemically to BPs via the C-1 carboxyl group or 15-hydroxyl group of PGE2. Besides, more than one equivalent of PGE2 utilizing a polysubstituted BP has been obtained by incorporating two moles of PGE2 per mole of a BP, with the tethered PEG2-BP found to have a good affinity for the bone and to liberate PGE2 at an acceptable rate to stimulate bone growth (Gil, et al., 1999). 25

Journal Pre-proof

However, the PGE2-BP conjugate has not develop a good medication for clinic, as it showed comparable bone formation effect to the parent PGE2 and limited accumulation in localized bone resorptive areas (Ossipov, 2015). Similarly, chemotherapeutic prodrugs have been prepared by conjugating drug molecules (camptothecin) to BPs through an ester-labile linkage to drugs, with a hydroxy group or carbonate-labile linkage binding of the amine functional group of drugs. The

f

prodrugs have shown a satisfactory binding ability to HA on the bone surface and can easily be

oo

activated hydrolytically in physiological conditions (Erez, et al., 2008).

pr

Apart from BPs, therapeutics can also be linked with other bone targeting moieties. TCs

e-

show a relatively complex chemical structure and poor stability during chemical conjugation. It is

Pr

possible to separate the HA binding domain from TC by structure-activity relationship study. The tricarbonylmethane group in ring A is sufficient for HA binding (Myers, Tochon-Danguy, &

al

Baud, 1983). Estradiol can be conjugated with this functional domain to enhance affinity for HA.

rn

This bone-targeting system improves the in vivo safety profile of estradiol after administration

Jo u

(Neale, et al., 2009). Estradiol can likewise be conjugated with aspartic acid oligopeptide. An estradiol prodrug has been synthesized by linking estradiol at position 3 to L-Asp-hexapeptide via succinate and was used to enhance the expression of the mRNAs of type I collagen α, osteopontin, and BSP, while only increasing BMD with negligible influence on the weights of the uterus and liver (Yokogawa, et al., 2001).

With high drug loading efficacy and small particle size, nanoscale devices are promising transporters of therapeutics to diseased portions of the bone and promote sustained drug release to targeted sites (Liu, et al., 2019). Using variable physicochemical properties, nanoparticles can be functionalized eas ily with bone-targeting ligands to improve the bone affinity of packaged 26

Journal Pre-proof

therapeutics. Poly(lactic-co-glycolic acid) (PLGA) is one of the most commonly used polymers with excellent in vivo biocompatibility and biodegradation and has been approved by the Food And Drug Administration (FDA) for biomedical applications; it is currently used for surgical sutures in the clinic (Lu, et al., 2009). Asp-linked PLGA nanoparticles can be fabricated by conjugating aspartic acid with PLGA nanoparticles directly. These osteotropic nanoparticles show

f

significant HA binding capability. A single Asp residue can serve as a targeting moiety for

oo

skeletal tissues (Erica J Carbone, et al., 2017). Another way to prepare Asp-functionalized PLGA

pr

nanoparticles is to synthesize Asp-PEG-PLGA copolymers first and then assemble the copolymers

e-

into nanoparticles. The use of these nanoparticles in previous in vivo bone affinity assay indicated

Pr

that Asp-PEG-PLGA nanoparticles had higher bone-targeting efficacy than nanoparticles that lack Asp (Fu, et al., 2014). By tagging fluorescein isothiocyanate (FITC) with an Asp peptide,

Besides, PLGA can be grafted to TC via an esterification reaction to form a TC-PLGA

rn

2014).

al

FITC-labeled Asp-PLGA nanoparticles can be prepared further for in vivo imaging (Jiang, et al.,

Jo u

copolymer, and the nanoparticles are then fabricated by a solvent emulsification method and display bone-targeting ability, improved curative effects, and reduced adverse effects to other organs (H. Wang, et al., 2015).

Recently, carbon dots (CDs) have attracted much attention for their convenient synthesis, colorful photoluminescence, and good biocompatibility, which have been used extensively for bioimaging, sensing, optoelectronics, catalysis, and energy conversion (J. Zhang & Yu, 2016). Fluorescent CDs for bone imaging can be fabricated using alendronate (ALN) without a nitrogen-doping precursor via a facile hydrothermal method. These ALN-based CDs have a strong

27

Journal Pre-proof

binding affinity for bone structures and extended skeletal fluorescence-retaining ability post-injection (Lee, et al., 2019).

4.2. Bone cell-targeting Because therapeutic agents are unable to differentiate between concrete cells in bone tissues, therapeutics will act on reachable cells with non-selectivity. Strategies that target specific cell

oo

f

types realize higher therapeutic efficiency and lower side effects, demonstrating great potential both in basic research and clinical application.

pr

Targeting osteoblasts

e-

Osteoblasts possess a prominent bone-forming ability and play an essential role in bone

Pr

homeostasis. Osteoblast-specific aptamers can be screened to bind osteoblasts specifically using

al

cell-SELEX. In a practical situation, a CH6 aptamer was selected after screening and post-inserted

rn

into the surface of osteogenic pleckstrin homology domain-containing family O member 1

Jo u

(Plekho1) siRNA-encapsulated lipid nanoparticles. The aptamer-functionalized nanoparticles promoted the selective uptake of Plekho1 s iRNA by osteoblasts and osteoblast-specific Plekho1 gene silencing, resulting in enhanced bone formation, improved bone microstructure, and ultimately stimulated bone recovery (Liang, et al., 2015).

Polypeptides have been used widely for targeted delivery owing to their simple spatial structure, small molecular weight, high specificity, strong penetrability, and low immunogenicity. As mentioned above, (AspSerSer)6 has a good affinity for the skeleton and can bind selectively to bone formation surfaces. (AspSerSer)6-attached cationic liposomes can be developed and used to deliver Plekho1 siRNAs to bone-formation surfaces with precision (Zhang, et al., 2012). Besides, 28

Journal Pre-proof

(AspSerSer)6-functionalized nanodevices can be used for selectively transporting Sema3A plasmids to osteoblasts (Yang, et al., 2018).

Phage display is a powerful selection technique for high-throughput screening of peptide or protein interactions. Using a phage or plasmid as the vector, exogenous peptide or protein gene could be fused with a bacteriophage coat protein and expressed on the surface of virion (Wu, et

f

al., 2016). Making use of this tool, Sun et al. ( Sun, et al., 2016) selected a peptide sequence

oo

(Ser-Asp-Ser-Ser-Asp, SDSSD) that showed a binding affinity for osteoblast-specific factor 2 and,

pr

thus, targeted osteoblasts in a ligand-receptor specific manner. The SDSSD peptide was then

Pr

osteoblast bone formation activity.

e-

modified on PU nanomicelles to deliver nucleic acids to osteoblasts accurately, promoting

Targeting osteoclasts

al

Osteoclasts have an excellent capability of resorbing the mineralized bones to maintain an in

rn

vivo stable state of the bone. Many factors participate in this process, making them attractive

Jo u

targets in osteoclasts. Integrin αvβ3, an adhesion receptor expressed on the surface of osteoclasts, can promote the adhesion of osteoclasts to the bone matrix for subsequent bone resorption activity. Recently, owing to the development of the Protein Chip technology, the inhibition of protein-protein interactions (PPIs) has been considered an emerging approach in disease treatment. IPS-02001, a small molecule inhibiting integrin αVβ3-osteopontin PPI, was screened a few years back using an in silico docking method-integrated ProteoChip technology; and it inhibited the maturation and resorptive activity of osteoclasts by blocking αVβ3 integrin signaling, which reduced ovariectomy-induced bone loss (Park, et al., 2016). Alongside integrin, the microRNAs

29

Journal Pre-proof

(miRNAs) in osteoclasts also play a critical role in regulating bone homeostasis. It has been demonstrated that elevated miR-214-3p in osteoclasts is closely associated with increased serum exosomal miR-214-3p and reduced bone formation in elderly fractured female and ovariectomized mice. Osteoclast-targeting platforms containing antagomiR-214-3p can be developed by conjugating a D-Asp8 peptide with a cationic liposome. This system inhibited the expression of

f

miR-214-3p, which in turn promoted bone formation in ovariectomized aging mice (Li, et al.,

oo

2016).

pr

Targeting osteocytes

e-

As the most abundant cells in the bone, osteocytes are attractive target sites for their

Pr

important function in bone metabolism. Osteocytes also play a significant role in the crosstalk of osteoclasts and cancer cells. This class of bone cells can manipulate the activity of early cancer

al

bone metastasis and subsequent osteoclastogenesis by expressing RANKL, OPG, and sclerostin,

rn

thereby providing a favorable bone milieu for the settlement of cancer cells (Cui, Evans, & Jiang,

Jo u

2016; Zhou, et al., 2015). Therefore, inhibiting osteocyte-mediated osteolysis would suppress the ingrowth and invasiveness of metastatic cancer.

Zoledronic acid (ZA), a third-generation BP, has a high affinity for bone tissues. More importantly, this BP can focus on sites of osteocytes and cancer cells during the early stage of bone metastasis. Additionally, ZA shows a synergistic effect in alleviating metastatic bone destruction alongside traditional Chinese medicine, plumbagin (PL) (Qiao, et al., 2016). A version of an upconversion nanoparticle that contains ZA and PL has also been fabricated, and it specifically targets osteocytes. This system inhibits the phosphorylation of osteocytic protein

30

Journal Pre-proof

kinase-a, cAMP-response element-binding protein, extracellular regulated protein kinase, and c-Jun N-terminal kinase, in that way decreasing the expression of osteoclastogenic RANKL and sclerostin (Qiao, et al., 2017).

Targeting stem cells

With unlimited self-renewal and excellent differentiating ability into multiple cell types, stem

oo

f

cells have been applied widely in regeneration engineering. Notably, in the aspect of bone injury, MSCs can differentiate into chondrocytes and osteoblasts, contributing to the repair process

pr

(Grayson, et al., 2015). The unique functions of stem cells make them potential targets in the

e-

bone. Particularly, various surface molecules on stem cells have been identified using large-scale

Pr

analyses, which has paved a way to develop specific affinity reagents for these molecules. Integrin α1, α2, α3, α4, α5, α6, α11, and β1 are expressed on the surfaces of MSCs. Among them, the

al

overexpression of integrin α4 can promote MSCs’ homing to the bone (Mukherjee, et al., 2008).

rn

Guan et al. (Guan, et al., 2012) synthesized a specific peptidomimetic ligand (LLP2A) against

Jo u

integrin α4β1 that had a high affinity for MSCs. By further combining LLP2A with ALN, the resulting compound can facilitate the in vitro migration and osteogenic differentiation of MSCs, as well as in vivo trabecular bone formation. Recently, a DNA aptamer, named Apt19S, was successfully developed using SELEX to target pluripotent stem cells specifically (Hou, et al., 2015). The aptamer was further immobilized on a bilayer scaffold for explic itly recognition and binding with MSCs. After implanting Apt19S into a defective osteochondral area, the aptamer-functionalized scaffold can facilitate the recruitment and binding with MSCs, promoting both the regeneration of defective cartilage and subchondral bone. Moreover, in vivo tests on rats

31

Journal Pre-proof

have indicated that this kind of scaffold displays excellent performance in the restoration of the osteochondral knee joint (Hu, et al., 2017).

5. Targeted therapy for bone-related diseases

Generally, the coordination between bone-absorbing osteoclasts and bone-forming osteoblasts keeps the bone in a steady state. This equilibrium is disturbed under many pathological

oo

f

conditions of the bone, including osteoporosis, osteosarcoma and bone metastasis, osteoarthritis, bone fracture, and other skeletal disorders. With the development of precision medicine, targeted

pr

therapy aimed at preselected sites has raised considerable interest in research and achieved much

Jo u

rn

al

Pr

e-

progress in bone-related diseases (Figure 2 and Table 3).

32

Journal Pre-proof

Figure 2. The schematic illustration of targeted therapy for bone-related diseases.

5.1. Osteoporosis

Osteoporosis is a common skeletal disease characterized by remarkably reduced bone mass and strength, as well as impaired skeletal microarchitecture that, in turn, increases the propensity for fragility fractures. As an emerging socioeconomic and medical threat, osteoporosis has a

oo

f

higher prevalence in the aging population, particularly postmenopausal women. A fracture

pr

caused-osteoporosis occurs mainly in the spine, hip, and wrist, which would result in the loss of

e-

mobility and autonomy. The risk of fracture in the lifetime of patients is as high as 40% (Rachner, Khosla, & Hofbauer, 2011). As a consequence, there is an urgent need for the effective prevention

Pr

and therapy of osteoporosis. Generally, the therapeutic strategies for osteoporosis can be classified

al

into two categories: the suppression of bone loss and enhancement of bone strength by inhibiting

rn

the antiresorptive activity of osteoclasts and the acceleration of bone formation and reverse of

Jo u

bone deterioration by stimulating osteoblasts.

Salmon calcitonin (sCT), an antiresorptive protein, can produce its antiresorptive effect by acting with calcitonin receptors (CTRs) expressed on osteoclasts (Chambers & Magnus, 1982).

However, sCT has low bone selectivity, given that CTRs are also expressed extensively in many non-skeletal tissues. To overcome this issue, sCT is, therefore, conjugated to thiolated-BP via an SMCC-chemistry to improve site-specificity to the bone. Consequently, the conjugate, with the restored bioactivity of sCT, shows enhanced specificity to mineralized tissues. Regrettably, a conclusive in vivo therapeutic efficacy is missing, meaning that further confirmation is required (Bhandari, et al., 2010). 33

Journal Pre-proof

With satisfactory bio-distribution and tissue-specific properties, nanosystems have been broadly

used

for

targeted

therapy.

Hengst

et

al

(Hengst,

et

al.,

2007)

used

cholesteryl-trioxyethylene-bisphosphonic acid as a targeting moiety and developed the bone-targeted liposomes to treat bone-related diseases, such as osteoporosis, with prolonged exposure to therapeutics and minimized systematic side effects. Ryu et al. (Ryu, et al., 2016)

f

designed a category of ALN conjugated nanodiamonds (ALN-NDs) for potential osteoporosis

oo

treatment. ALN-NDs can accumulate effectively in the bone after intravenous injection and be

pr

employed potentially for the efficient treatment of osteoporosis thanks to the presence of ALN.

e-

Estrogen plays a key role in regulating bone metabolism in both females and males (Khosla,

Pr

Amin, & Orwoll, 2008). Supposedly, estrogen therapy can prevent and treat osteoporosis effectively (Khos la & Hofbauer, 2017). However, low selectivity by the skeleton has restricted the

al

significant application of estrogen. One classical means to acquire accurate site-specificity to the

rn

bone is to couple estrogen with bone-targeting moieties. Conjugation with BP, enables estrogen to

Jo u

display a preferential profile in the bone, compared to a natural estrogen analog (17β-oestradiol). Therefore, targeted estrogens could potentially improve the compliance of osteoporosis patients by

reducing the medication frequency, as well as adverse effects (Tsushima, et al., 2000). Selective estrogen receptor modulators (SERMs) are derived from estrogen by medicinal chemistry approaches. Commercialized products, such as Raloxifene and Bazedoxifene, are emerging as relatively new classes of drugs for osteoporosis treatment. Interestingly, SERMs have an agonistic effect similar to that of estrogens in tissues, such as the bone, but an antagonistic or neutral outcome in other tissues, like the breast and uterus (Riggs & Hartmann, 2003). From this

34

Journal Pre-proof

perspective, SERMs themselves already have a targeting property attributed to the different cell responsiveness and their modulation of nuclear receptors instead of spatial targeting (Shang & Brown, 2002).

Unlike antiresorptive therapy, anabolic therapy, another typical modality for osteoporosis treatment, primarily enhances bone formation rather than reduce bone resorption. PTH is a

oo

f

representative anabolic drug that has been approved for treating postmenopausal osteoporosis. Despite its better short-term efficacy, compared to BPs, PTH remains only second-line therapy

pr

due to its considerable side effects (e.g., hypercalcemia) and inconvenient dosing (Neer, et al.,

e-

2001). There is an indication that hypercalcemia is avoidable, and reduced injection frequency is

Pr

possible if PTH targeting strategies are developed. Generally, this targeted system is constructed by fusing PTH with CBD. Ponnapakkam et al. (Ponnapakkam, et al., 2014) demonstrated that

al

PTH-CBD increased spinal BMD by 14.2% after 5 months without inducing hypercalcemia

rn

following monthly injections in ovariectomized rats. PTH-CBD also increased the level of ALP in

Jo u

serum, which is consistent with its anabolic effect. Even for a single subcutaneous administration, PTH-CBD showed a sustained bone anabolic effect for an extended period (9–12 months) with no

hypercalcemia (Ponnapakkam, et al., 2012).

PTH aside, the prostaglandin hormone, PGE2, shows an anabolic activity as well and can stimulate in vivo bone growth (Jee, Ke, & Li, 1991). Four receptor subtypes (EP1, 2, 3, and 4) interact with PGE2. Of the four receptor subtypes, EP4 plays a crucial role in the bone-forming

activity of PGE2. There is evidence that activating the EP4 receptor can suppress the apoptosis of osteoblasts and osteoid precursor cells in bone marrow (Machwate, et al., 2001). However, both 35

Journal Pre-proof

PGE2 and EP4 agonists have several side effects, including gastrointestinal disturbance and vasodilation. To avoid drawbacks and realize a sustained and concentrated impact in the bone, the EP4

receptor

agonist

is

conjugated

with

alendronic

acid

via

a

cleavable

dipeptide-para-aminobenzyl alcohol linker. This prodrug has synergistic bone-forming effects through the EP4a and inhibition of resorption by alendronic acid, possessing a significant potential

oo

f

to treat osteoporosis (Xie, Chen, & Young, 2017).

Stem cell therapy could potentially reduce the susceptibility of fracture and increase mineral

pr

density by differentiating into osteoblasts, which would be promising for the treatment of

e-

osteoporosis (Antebi, Pelled, & Gazit, 2014). Allogeneic MSCs isolated from the bone marrow

Pr

have been infused systematically into mice with dexamethasone-induced osteoporosis in the past, with the transfused MSCs homing and inhabiting in the recipient bone marrow for more than 4

(Sui, et al., 2016). Gene engineering is usually used for MSC modification to

rn

osteoblastogenesis

al

weeks. These donor cells committed to Osterix (Osx)+ osteoblast progenitors and induced

Jo u

enhance its functionality for tissue regeneration. Li and coworkers (Li, et al., 2016) constructed baculovirus-engineered MSCs expressing miR-140* or miR-214 sponges. These hybrid vectors

transfected MSCs continuously attenuated the cellular levels of miR-140*/miR-214, promoting osteogenesis and repressing osteoclast maturation in vitro. Compared with miR-140*, reducing miR-214 displayed more potent effects. In an osteoporotic rat model, the allotransplantation of miR-214 sponges-expressing MSCs promoted bone healing, remodeling and bone quality (BMD, trabecular thickness, trabecular numbers, and trabecular distance) after 4 weeks.

5.2. Osteosarcoma and bone metastasis 36

Journal Pre-proof

A malignant bone tumor is a highly aggressive tumor that induces destructive changes in the bone structure and causes severe bone pain and other skeletal events. Generally, aggressive bone tumors are classified into primary malignancies (e.g., osteosarcoma) and adaptive bone metastases from other malignant tumors. Osteosarcoma

f

Osteosarcoma, the most common type of primary sarcoma in the bone, is characterized by the

oo

presence of stromal cells that can generate bone-like tissues (Abarrategi, et al., 2016). Standard

pr

chemotherapy is used customarily as a first-line treatment and has improved the long-term overall

e-

survival of patients. However, owing to the rigidity, low blood flow, and reduced permeability of

Pr

skeletal tissues, a high dose of chemotherapeutic agents that would cause considerable toxicity is

required (Li, et al., 2016). A more efficient approach is to develop bone-targeted strategies.

al

Kopecek and co-workers (Low, Yang, & Kopecek, 2014) utilized D-ASP octapeptide and

rn

doxorubicin (DOX) to create bone-targeted micelles as potential osteosarcoma therapeutics. The

Jo u

micelles displayed rapid adsorption toward HA and potential cytotoxicity to osteosarcoma Saos-2 cells. Integrin avβ3 and avβ5 are expressed widely in osteosarcoma cells. RGD, a cell affinitive peptide, can interact with these integrins and, thus, be used for targeting osteosarcoma. On this basis, Fang et al. (Fang, et al., 2017) developed RGD-decorated biodegradable polymeric micelles loaded with DOX and used them to target MG63 osteosarcoma cells specifically, which resulted in higher cytotoxicity than that obtained with the use of their non-targeted counterparts. Folate receptors are also overexpressed on the surface of osteosarcoma cells. Liposome-packaging

antineoplastic agents (curcumin and C6 ceramide) were modified with folic acid in another

37

Journal Pre-proof

research to bind with folate receptors, and the targeting liposomes displayed enhanced cell cytotoxicity in osteosarcoma-ridden cells (Dhule, et al., 2014). Osteosarcoma often occurs in the distal femur, proximal humerus, and proximal tibia, all areas with abundant blood supply. This bone tumor depends on the development of new blood vessels, in a process known as angiogenesis, for growth and metastasis (Xie, Ji, & Guo, 2017).

f

Vascular endothelial growth factor (VEGF) is a key tumor-derived angiogenic factor that can

oo

stimulate angiogenesis while increasing vascular permeability, facilitating tumor progress (Quan

pr

& Choong, 2006). Besides, the VEGF level correlates with poor prognosis in osteosarcoma

e-

patients (DuBois & Demetri, 2007). Thanks to the advancement in biotechnology though, VEGF

Pr

receptor tyrosine kinase inhibitors (TKIs), the most typical anti-angiogenesis targeting therapy,

have been developed successfully and used to treat sarcoma in the clinic. Although T KIs have not

al

been applied in clinical osteosarcoma, clinical trials have shown promising outcomes. The Italian

rn

Sarcoma Group conducted a phase II trial using sorafenib to treat relapsed and unresectable

Jo u

osteosarcoma and showed that TKI-targeting therapy increased the 4-month progression-free survival from < 30-46% (Grignani, et al., 2012). There is evidence that several VEGF subtypes are involved in the VEGF family, including VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, and VEGF-F (Niu & Chen, 2010). Among them, the pathway mediated by VEGF-A and VEGF receptors plays a vital role in the pro-angiogenic function of tumors and is prioritized for developing anti-angiogenic therapy. Liang et al (Liang, et al., 2017) utilized CRISPR/Cas9, a powerful genome editing technology, to

generate CRISPR/Cas9 plasmids encoding VEGF-A gRNA. The researchers then screened a cell-specific aptamer (LC09) to improve tumor-targeted characteristics and subsequently 38

Journal Pre-proof

fabricated an LC09-functionalized PEG-PEI-Cholesterol lipopolymer encapsulating CRISPR/Cas9 plasmids for anti-angiogenesis therapy. This system decreased the expression and secretion of VEGF-A, as well as angiogenes is by effective VEGF-A genome editing in osteosarcoma, inhibiting both primary tumor and lung metastasis. miRNA-based therapy has emerged as an attractive strategy in osteosarcoma treatment

f

because of the critical role of miRNA in tumorigenesis and growth. Reportedly, upregulating

oo

miR-132 can inhibit the proliferation of osteosarcoma cells by arresting the cell cycle at the G1/S

pr

phase, which is mediated by reducing cyclin E1 expression (Wang, et al., 2014). Besides,

e-

miR-133a possesses an antitumor effect by decreasing the proliferation and promoting the

Pr

apoptosis of osteosarcoma cells, a role probably attributed to the suppressed expression of B-cell

lymphoma-extra large (Bcl-xL) and myeloid cell leukemia-1 (Mcl-1) (Ji, et al., 2013). In a recent

al

study, the loss of miR-34a, miR-93, and miR-200c was revealed to occur during tumor switch

rn

from avascular dormancy to the fast-growing angiogenic phenotype. Subsequently, dendritic

Jo u

polyglycerol nanopolyplexes were designed to package these miRNAs for targeting osteosarcoma. After reconstituting the miRNAs into osteosarcoma cells using nanopolyplexes, they were found to reduce angiogenetic factors and cell migration, including their target gene levels, MET proto-oncogene, hypoxia-inducible factor 1α, and moesin, thereby prolonging the dormancy period of progressive osteosarcomas (Tiram, et al., 2016). Given that osteosarcoma cells possess osteoblastic features, targeting osteoclast functions could be a potentially useful approach to treat osteosarcoma (Zhou, et al., 2014). ZOL, a member

of BPs, has shown a good affinity for bone tissue. Moreover, ZOL can reduce tumor-induced osteolysis by inhibiting osteoclasts directly and attenuating the expression of RANKL and 39

Journal Pre-proof

monocyte chemoattractant protein-1. These potential outcomes are in line with the significant suppression of in vivo tumor growth (Ohba, et al., 2014). With the advancement in gene engineering, it is convenient to engineer T cells genetically with tumor-associated antigens for immunotherapy. The human epidermal growth factor receptor (HER2) is a kind of oncogene expressed in around 60% of primary osteosarcoma. Ahmed and

f

colleagues’ (Ahmed, et al., 2009) adoptive transfusion of HER2-specific T cells induced

oo

regression of established osteosarcoma xenografts and increased the overall survival of

pr

tumor-bearing mice. In a follow-up study, the HER-2-specifc CAR T cells targeted drug resistant

e-

tumor-init iating cells (TICs) and remarkably reduce TICs in bulk tu mors (Rainusso, et al., 2012).

Pr

However, during the pathogenesis, progression and metastasis of osteosarcoma, there is a high

incidence of alterations in genetic information, causing tumor heterogeneity and therapeutic

al

obstacles (Wang, et al., 1998). Identifying and targeting mutated genes in osteosarcoma could

rn

boost the efficiency of cancer treatment. TP53 gene is a common mutant tumor suppressor gene in

Jo u

osteosarcoma and its mutation would lead to the loss of the tumor suppressor function of wild-type TP53 (Duffy, Synnott, & Crown, 2017). Tang et al. (Tang, et al., 2019) used a CRISPR-Cas9 system and a TP53 inhibitor (NSC59984) to knock-out mutant TP53 in osteosarcoma cells with specificity. Following the inhibition of mutant TP53, the migration, pro liferat ion, and tumor formation activity of osteosarcoma cells decreased efficiently. Moreover, the knockout of mutant TP53 reduced the level of oncogene (IGF-1R), anti-apoptotic proteins (Bcl-2), and Survivin in osteosarcoma cells.

Bone metastasis

Bone metastasis, which occurs in 70 % to 80 % of patients, is common in patients with aggressive cancer, especially prostate and breast cancers (Coleman, 2006). In the skeleton, 40

Journal Pre-proof

multi-step processes are involved in metastasis development. Tumor cells need to disseminate from the primary site, colonize and survive in the new microenvironment, escape from dormancy to proliferation state, and eventually grow uncontrollably (Croucher, McDonald, & Martin, 2016). Tumor metastasis in the bone would cause unbearable pain, pathologic fracture, spinal cord compression, and hypercalcemia, significantly reducing the quality of life of patients (Pentyala, et

f

al., 2000). With the advancement in precision medicine, new approaches and therapies have been

oo

developed to alter the course of bone metastasis. In a real-world setting with the use of

pr

bone-targeted denosumab among patients with metastatic tumors, greater compliance and longer

e-

persistence were observed, indicating that targeted therapy may improve treatment efficacy,

Pr

meaning that it holds significant potential for the treatment of bone metastasis (Qian, Bhowmik,

Kachru, & Hernandez, 2017).

al

Prostate cancer is one of the most common malignancies and ranks as the second leading

rn

cause of cancer-related deaths in men (Siegel, Miller, & Jemal, 2019). What is worse, almost 80%

Jo u

of patients develop aggressive bone metastasis, substantially reducing expected overall survival to less than 1 year (Cheville, et al., 2002). Unlike other cancer types, the initial metastasis of prostate cancer is limited to the skeleton that is often the only spread site in the late stage. Targeted therapy is promising in the way it controls bone metastasis owing to its unique characteristic of focusing on a given area. As the most abundant component in the bone, the bone matrix also contributes to initiation and support of cancer metastasis in the bone. Converting the bone matrix to a less favorable environment for metastatic tumor cells could be a potentially rewarding treatment

strategy.

41

Journal Pre-proof

The systemic administration of radiopharmaceuticals has a long history of alleviating bone metastasis. The FDA has approved Strontium-89 (Porter, et al., 1993) and samarium-153 (Sartor, et al., 2004) for commercialized use in treating metastatic prostate cancer, and these substances have shown preferential uptake in areas with high bone turnover, such as metastatic lesions, resulting in improved clinical outcomes in patients with bone metastasis. Besides, rhenium-186, Sr, and rhenium-188 all show a satisfactory effect on palliating bone metastasis in clinical trials.

f

89

oo

Another radiopharmaceutical radium-223 exhibits delayed symptomatic skeletal events, improved

pr

pain control, favorable toxicity profile, and a remarkable overall survival benefit in men with

e-

metastatic prostate cancer (Blacksburg, Witten, & Haas, 2015). In a phase II study of patients with

Pr

hormone-refractory prostate cancer and bone pain, radium-223 treatment prolonged the median

time of prostate-specific-antigen progression to 26 weeks compared to 8 weeks for placebo.

al

Moreover, the median overall survival was 65.3 weeks for radium-223-treated patients while that

rn

for the placebo group was shorten to 46.4 weeks (Nilsson, et al., 2007).

Jo u

The metastasis of prostate cancer to the bone is more likely to be osteoblastic instead of lytic, indicating that inhibiting the increased bone deposition by osteoblasts could be an appealing therapeutic option. Endothelin-1 (ET-1), a potent vasoconstrictor, can stimulate osteoblasts into phenotypes of osteoblastic bone lesions in metastatic prostate cancer (Nelson, et al., 1995). Based on these findings, antagonizing ET-1 is considered a promising therapy. In 2018, a phase III clinical trial (SWOG S0421) introduced a strategy combining chemotherapeutic docetaxel with an ET-1 antagonist (Atrasentan) to treat metastatic castration-resistant prostate cancer. Though no

significant difference was found, the therapy with Atrasentan showed 44.0% pain palliation and

42

Journal Pre-proof

28.7% functional status compared to 41.7% and 24.2% in the control group, respectively (Unger, et al., 2017). According to the microscopic analysis of skeletal metastatic foci, bone resorption mediated by osteoclasts could prime the bone for osteoblastic metastasis. Hence, targeting osteoclasts could serve as an alternative approach to control metastasis development. BPs, with a high affinity for

f

the skeleton and an excellent ability to inhibit osteoclast activity, have been used in metastatic

oo

cancer treatment. Notably, zoledronate has been approved for treating bone metastases of prostate

pr

cancer in the clinic. Because the RANK/RANKL/OPG signaling pathway is essential for

e-

osteoclast-mediated bone resorption, using OPG-Fc containing an OPG binding domain could

Pr

block the RANK pathway from activating osteoclasts, suppressing the growth of metastatic

tumors (Yonou, et al., 2003).

al

Breast cancer, which is the most common cancer type in women, invades distant organs

rn

preferentially, especially the bone. Killing tumor cells in the bone directly is an effective way of

Jo u

treating metastatic cancer. Cytotoxic chemotherapeutics can cause cytotoxicity to tumor cells and attenuate metastatic tumor progress. However, low selectivity and high dose-related toxicity have

restricted the therapeutic efficacy of these drugs significantly. Reportedly, nanotechnology and bone targeting moieties could help resolve this dilemma to some extent. Zhou et al. (Zhao, et al., 2019) used Glu6 -RGD as a targeting ligand to fabricate liposomes loaded with paclitaxel, which showed extraordinary targeting activity on metastatic cancer cells, inducing higher cytotoxicity. Similarly, Liu’s group (Zhu, et al., 2018) developed a class of ALN-functionalized micelles encapsulating bortezomib-catechol conjugates, and this prodrug showed an acid-responsive drug

43

Journal Pre-proof

release performance and minimized systematic toxicity. Moreover, this targeted platform remarkably suppressed the growth of cancer cells at metastatic bone lesions and decreased the destruction of the metastatic bone structure.

As described above, osteoclasts play an essential role in bone metastasis from primary tumors. A therapeutic modality simultaneously working on bone resorption and tumor growth

oo

f

could be a meaningful strategy to treat breast cancer-related bone metastasis. ALN-modified polymer nanoparticles have been fabricated to package cisplatin prodrugs to treat bone metastasis

pr

originating from breast cancer. This platform not only inhibited tumor growth in the bone

e-

effectively but also decreased osteoclastic bone destruction efficiently (He, et al., 2017).

Pr

ALN-functionalized nanoparticles have similarly been developed to load DOX and have shown suppressed tumor growth and reduced bone resorption with negligible systematic toxicity (Zhao,

Jo u

5.3. Osteoarthritis

rn

al

et al., 2017).

Osteoarthritis is a degenerative and heterogeneous joint disease that can cause pain, stiffness, and even disability to patients. Specifically, this “joint disorder” is characterized by articular

cartilage degradation, subchondral bone sclerosis, osteophyte formation, and synovial inflammation (Herrero-Beaumont & Roman-Blas, 2013). Several targeted approaches have been explored in osteoarthritis treatment based on the development and features of osteoarthritis.

Increasing evidence shows that subchondral bone remodeling hightens during the progression of osteoarthritis (Bellido, et al., 2010; Goldring & Goldring, 2010), and this subchondral bone 44

Journal Pre-proof

resorption usually occurs in the early phase of osteoarthritis development. Hence, inhibiting osteoclastic bone resorption could be a potentially useful therapeutic option. With a critical role in bone turnover, BPs can serve as both targeting moieties and efficient therapeutic agents, providing benefits

for

osteoarthritis

treatment.

Moreau

and

colleagues

found

that

tiludronate

(TLN) treatment resulted in less joint effusion, reduced level of PGE2 in synovial fluids, and

f

larger subchondral bone surfaces when compared with placebo controls. TLN also decreased

oo

inflammatory factors, including matrix MMP-13 and ADAMTS5 in cartilage and cathepsin K in

pr

the subchondral bone, further alleviating the symptoms of osteoarthritis (Mor eau, et al., 2011).

e-

Alongside preclinical investigations, BPs display promising outcomes in clinical application.

Pr

According to annually collected patient data from the NIH Osteoarthritis Initiative cohort over 4

years, BP users had lesser joint space narrowing over time (Laslett, et al., 2014).

al

Repetitive mechanical injury is a vital signal for the occurrence and progression of

rn

osteoarthritis. Chondrocytes are generally accepted as the target of these abnormal biomechanical

Jo u

factors, making them ideal targets for osteoarthritis treatment (Goldring, 2000). Pi et al. (Pi, et al., 2011) identified a chondrocyte-homing peptide (CAP, DWRVIIPPRPSA) with a two-step

biopanning using synovium and cartilage explants from healthy rabbits and conjugated this CAP covalently with polyethylenimine (PEI) to construct a non-viral vector. The PEI-CAP copolymers were then used to condense siRNA in nanoparticles to silence hypoxia-inducible factor-2α, a key factor in chondrocyte catabolic state. The cartilage-targeting nanoparticles downregulated catabolic

factors in vitro, maintained cartilage integrity, and alleviated synovium inflammation in vivo (Pi, et al., 2015).

45

Journal Pre-proof

NF-κB signaling pathways are involved extensively in inflammatory diseases and play an essential role in subchondral bone resorption. Notably, synoviocytes and chondrocytes secrete pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6), which trigger osteoblasts to release RANKL to bind with RANK. NF-κB transcription factors are then activated for subsequent bone resorptive activity (Rigoglou, Papavassiliou, & biology, 2013). Therefore, targeted therapies

f

inhibiting NF-κB pathways could be acceptable approaches to treat osteoarthritis. In a

oo

staphylococcus aureus-induced osteoarthritis model, combinational therapies, composed of

pr

antibiotics with RANK-Fc or OPG-Fc, reduced the expression of bone resorption makers,

e-

including osteocalcin, the C-terminal telopeptide of type I collagen, and TRACP5b, mitigating

Pr

cartilage/bone destruction (Verdrengh et al., 2010). To block the NF-kB signaling pathway further

and accurately, Doschak et al. (Doschak et al., 2009) conjugated OPG with BP chemically, and

al

after systemic administration, OPG-BP conjugates accumulated highly in the bone, suggesting that

rn

they could, potentially, be applicable in the treatment of osteoarthritis with active bone

Jo u

remodeling.

5.4. Bone fracture

Bone fractures pose major challenges to orthopedic practice. Around 5-10% of fractures result in delayed healing o r non-union (Novicoff, et al., 2008). Despite the fact that autologous bone graft is regarded as a gold-standard treatment in fracture non-union, their application can lead to donor site morbidity (Kneser, et al., 2006). Alternatively, recombinant human bone morphogenetic protein (rhBMP)-2 and rhBMP-7 are used for bone harvesting. However,

rhBMP treatments have a very narrow indication and require megadoses of the protein (Virk 46

Journal Pre-proof

& Lieberman, 2012). With the latest signs of progress made in cell and molecular biology, more

effective modalities are developed to restore the structural integrity of bone.

As previously mentioned, MSCs can traffic to injured tissues and differentiate into the osteoblasts and chondrocytes for bone healing. Therefore, MSC-based cell therapy could serve as a potential strategy in fracture repair. Reportedly, the concentration and counts of injected MSCs

oo

f

are in proportion to the healing rate. A practical investigation showed that the fracture of patients who received less than 1000 MSCs per mL and total 30,000 MSCs did not heal, whereas those of

e-

pr

patients given 1500 MSCs per mL and total 54,000 MSCs healed (Gomez-Barrena, et al., 2015).

Although MSCs have extraordinary performance in tissue engineering, their differentiation

Pr

and homing ability remain limited for efficient bone regeneration. To improve this condition, Xu

al

et al. (Xu, et al., 2015) developed Sry-related high-mobility group box 11 (Sox11)-modified

rn

MSCs by transducing lentivirus carrying Sox11 into MSCs for fracture healing. The results

Jo u

demonstrated that the overexpression of Sox11 enhanced the BMP/Smad signaling pathway and activated the promoter activity of Run x2 and CXCR4. Co mpared with control MSCs, more So x11-overexpressed MSCs migrated to the fracture area, initiated callus ossification, and promoted fracture healing in an open femur fracture model. Salvianic acid A (SAA) is a water-soluble active

compound extracted from salvia miltiorrhizae (Danshen), which has potent anabolic ability (Cui, et al., 2009). However, the clinical translation of SAA is greatly impeded by its short half-life in serum and its lack of osteotropicity. To deal with these conditions, Liu et al. (Liu, et al., 2018)

develop a bone-targeting liposome formulation to deliver SAA specifically to the bone. The author synthesized pyrophosphorylated cholesterol by conjugating pyrophosphate to cholesterol via a 47

Journal Pre-proof

triethylene glycol linker and used it as the targeting ligand. The SAA-loaded liposomes were then

prepared using a reverse-phase evaporation method with pyrophosphorylated cholesterol and lecithin as the backbone. The targeted-liposome formulation showed a high affinity for HA in vitro and robust ability of bone-targeting and long retention in vivo. In a delayed femur fracture union mouse model, SAA-loaded targeting liposome was found to shorten healing time

f

remarkably (from > 64 days to 42 days), improve histologic and histomorphometric parameters

oo

and mechanical property outcomes.

e-

pr

5.5. Other bone-related diseases

Apart from the skeletal maladies mentioned above, several other bone-related conditions also

Pr

benefit from targeted therapy. PDB is a common skeletal disorder with increased and inordinate

al

bone remodeling, leading to osteolytic and osteosclerotic lesions (Vallet & Ralston, 2016). BP is

rn

one of the first-line medications for treating PDB because of its extraordinary inhibitory effects on

Jo u

bone turnover and innate bone affinity. Reportedly, the RANKL/OPG ratio in serum lessened after BP treatment, decreasing subsequent bone resorption and attaining highly efficient PDB therapy (Martini, et al., 2007).

Corticosteroids are routinely used to treat inflammatory and autoimmune diseases but their use is limited by steroid-associated osteonecrosis (SAON), a severe complication that often induces subchondral bone destruction (Chan, et al., 2006). Chen et al. (Chen, et al., 2018) developed a class of Asp8-functionalized liposomes loaded with a small phytomolecule (icaritin) for SAON prevention and found that liposomes alleviated steroids-treated rats of SAON

48

Journal Pre-proof

efficiently, with remarkably decreased osteocyte apoptosis, as well as increased osteoclastogenesis and osteogenesis.

Osteomyelitis is an inflammatory disease accompanied by severe bone destruction (Lew & Waldvogel, 2004). Current therapy for osteomyelitis focuses mainly on antimicrobial issues. However, even therapeutic efficiency in these instances is usually restricted by the limited

oo

f

concentrations of antibiotics that reach the bone. Promisingly, Sedghizadeh and coworkers have established an osteoadsorptive BP-ciprofloxac in conjugate that demonstrates a significantly

pr

improved therapeutic index in osteomyelitis treatment compared to ciprofloxacin (Xie, et al.,

6. Conclusion and perspectives

Pr

e-

2017).

al

In this review, we summarized the recent progress in bone-targeted therapy. Targeted therapy

rn

for bone-related diseases has several advantages over standard treatment procedures. Therapeutic

Jo u

agents are concentrated in the targeted skeleton to a maximum extent, decreasing the required dosage significantly, and promoting the therapeutic efficacy of the agents. Additionally, the retention time of the drugs is prolonged in the targeted area with improved bioavailability.

Furthermore, the systematic toxicities of these agents are substantially reduced in the controlled non-specific and unintended distribution to other tissues. We also presented the composition of skeletal tissues, including the extracellular matrix and cells, in detail. With the understanding of bone biology, we next summarized the commonly used bone-targeting moieties, with emphasis on BPs, TCs, and biomimetic proteins and peptides, as well as targeting strategies in the bone. Lastly,

49

Journal Pre-proof

we discussed the potential application of targeted therapy in the treatment of bone-related diseases. It appears that a targeting strategy is a promising approach to skeletal disease treatment.

Though the existing targeted therapy has so far been a hugely promising success, there are still considerable obstacles that hinder its practical application. Unlike other soft tissues, the bone is characterized by its rigidity, low permeability, and reduced blood flow, features that limit the

oo

f

reach of most of the current targeting moieties, such as receptors and biomolecules, to the bone. However, bits of existing targeting moieties that do reach the bone are slowly or even hardly

pr

cleared from the body, which would cause long-term safety concerns. Moreover, most of the

e-

targeted therapies are still at laboratory phases of investigations, and researchers are finding it

Pr

challenging to advance them to the clinical stage. Therefore, more researches need to be made to resolve the dilemma faced in the use of bone-targeted therapy. With a comprehensive

al

understanding of bone biology and the rapid development of biotechnology, it is possible to

rn

explore more effective moieties to target the skeleton in the future. Potential options are now in

Jo u

the numbers, meaning that the suboptimal targeting moieties could be substituted by safer ones. Furthermore, clinical translations could be accelerated using advanced development in genomics,

genetics, and cell biology. Exploiting effective models to minimize the physiological differences between animals and humans may also shorten the period between the basic research and clinical application. Nevertheless, targeted therapy has various merits, exhibits satisfactory benefits, and could be an appealing strategy in bone-related disease treatments.

Conflicts of interest The authors declare that there is no conflict of interest. 50

Journal Pre-proof

Acknowledgements This work was supported financially by the National Natural Science Foundation of China [Grant

Jo u

rn

al

Pr

e-

pr

oo

f

number: #81672216, #81874026]

51

Journal Pre-proof

Table 1. Summary of bone cells

Cell type

Osteoblasts

Resources

MSC lineage

Ref

Cuboidal shaped cells with plenty of rough endoplasmic reticula, well-developed Golgi apparatus, and diversified secretory vesicles

Bone-forming abilities through the deposition of the organic matrix and participation in subsequent bone mineralization

f o

(Marks & Popoff, 1988; Florencio-Silva, et al., 2015; Burr, 2019; Yadav, et al., 2016)

Reacquire secretory abilities; Prevent direct contact between bone matrixes and osteoclasts and participate in osteoclast differentiation

(Matic, et al., 2016; Miller, et al., 1989; Hienz, Paliwal, & Ivanovski, 2015)

Resorb fully mineralized bones

(Detsch & Boccaccini, 2015; Teitelbaum, 2000; Merolli, et al., 2018)

Permit the diffusion of mineral and fluids; Detect mechanical pressures; Regulate osteoblasts and osteoclasts

(Sommerfeldt & Rubin, 2001; Shea & Miller, 2005; Rochefort, Pallu, & Benhamou, 2010; Mori & Burr, 1993; Prideaux, Findlay, & Atkins, 2016)

l a n

Osteoblasts

o r p

e

r P

r u o

Hematopoietic stem cell lineage

J Osteocytes

Functions

Flat-shaped cell with extremely flat nuclei and few cytoplasmic organelles

Bone lining cells Inactive osteoblasts

Osteoclasts

Structure

Polarized and multinucleated giant cells

A dendritic morphology with a higher number of filopodia

52

Journal Pre-proof

Table 2. Summary of bone-targeting moieties

Classifications

BPs

Targeting moieties

Targeting site

BPs

TCs

Biomimetic bone-targeting moieties

Acid oligopeptides composed of repeating Asp or Glu

J

HA

References

Induce atypical femur fractures; Osteonecrosis of the jaw; Target HA surface; Shift bone Gastrointestinal effects; Cancer of homeostasis toward bone esophagus and atrial fibrillation; formation; Induce osteoclast Inhibit bone mineralization and apoptosis induce osteomalacia, typical acute-phase response and renal failure at high dose

(Yewle, 2012; Lawson, et al., 2010; Drake, Clarke, & Khosla, 2008; Dell, et al., 2012; Khan, et al., 2015; Adami & Zamberlan, 1996)

Target skeletal tissues; Have broad spectrum antibiotic activity; Reduce bone resorption; Label the surface of growing bone formation area

Cause inherent biological activity including heartburn, rash, dizziness, hypoglycemia and nausea; Stain teeth in dental developing children; Being chemical unstable; Induce enamel hypoplasia during calcification at a high dose

(Albert & Rees, 1956; Reichert, et al., 2012; Nakamura, et al., 2000; Valentín, et al., 2009; Bevelander, Rolle, & Cohlan, 1961)

Have good affinity toward HA

Poorly stable to enzymatic cleavage; Low oral bioavailability; Complicated production process;

(Yarbrough, et al., 2010; William N, 2010; Tulasi, 2011,

al

o r p

e

r P

rn

u o

HA

limitations

f o

HA

TCs

Bioactivities

53

Journal Pre-proof

(AspSerSer)6

VTK

HA

CBD

Collagen

Produce targeting

Osteocalcin

HA

Bind to HA; Prevent the growth of crystal

BSP

Osteoblasts, fibroblasts, collagen

Target osteoblasts, fibroblasts and collagen; Initiate the nucleation of HA crystal

Osteopontin

HA

MSCs

Injured bone

l a n

r u o

J

Other moieties

Chelate with osteoblast-mediated mineralizing nodules and amorphous calcium phosphate Target HA and skeleton-like materials

Bone formation surface

fusion-protein

Harsh conditions for transport and storage

f o

for

Ryuichi Fujisawa, 2012; Hunter & Goldberg, 1993; Boskey, 1995; Aoki, et al., 2012; Kitaori, et al., 2009)

o r p

e

r P

Bind to the surface of HA; Inhibit the formation and growth of crystal Home to fractured sites in bone 2+

HA

Chelate with Ca on skeletal tissues; No effect on bone metabolism

Long-term safety concerns

(Lange, et al., 2016)

CH6 aptamer

Osteoblasts

Target osteoblasts

High cost; Complicated production process

(Liang, et al., 2015)

Radionuclides

HA

Deposit in bone and partic ipate in metabolic process of bone Long-term safety concerns minerals

Phosphonate-containing compounds

54

(Cawthray, et al., 2015)

Journal Pre-proof

Table 3. A brief summary of targeted therapy for bone-related diseases

Bone diseases

Osteoporosis

Osteosarcoma

Bone metastasis

Therapeutics

Targets

Effect

Reference (Ryu, et al., 2016) (Riggs & Hartmann, 2003) (Ponnapakkam, et al., 2012) (Xie, Chen, & Young, 2017)

ALN-NDs

Osteoclasts

Accumulate in bone and possess potential to treat osteoporosis

SERMs

Osteoclasts

Targeted therapy for osteoporosis

PTH-CBD

Osteoblasts

Has high bone affinity and sustained

Alendronic acid-conjugated EP4 receptor agonist

Osteoblasts

Bone-targeting and synergistic bone forming effects

RGD-decorated micelles

Osteosarcoma cells

Induce higher cytotoxicity to tumor cells

Injured bone

Promoted bone healing, remodeling and bone quality (BMD, trabecular thickness, trabecular numbers, and trabecular distance)

(Li, et al., 2016)

Decrease VEGF expression and inhibiting tumor growth

(Liang, et al., 2017)

Reduce factors for angiogenesis and cell migration

(Tiram, et al., 2016)

Baculovirus-engineered MSCs LC09 aptamer-functionalized lipopolymers MicroRNAs-packaged nanopolyplexes Mutant TP53 knockout CRISPR-Cas9 system ZOL

l a n

J

r u o VEGF

ro

miRNA in osteosarcoma Mutant TP53

Osteoclasts

Radium-223

Bone matrix

OPG-Fc

Osteoclasts

f o

p e

anabolic effect

r P

Inhibit the migration, proliferation, and tumor formation activity of osteosarcoma cells Reduce tumor-induced osteolysis Improve the symptomatic skeletal events and pain control as well as reducing toxicity Block RANK pathway and inhibit metastatic tumor 55

(Fang, et al., 2017)

(Tang, et al., 2019)

(Ohba, et al., 2014) (Blacksburg, Witten, & Haas, 2015) (Yonou, et al., 2003)

Journal Pre-proof

Glu6-RGD-decorated liposomes ALN-modified nanoparticles

f o

Tiludronate Osteoarthritis

Metastatic cancer Induce higher cytotoxicity cells Inhibit tumor growth and decrease osteoclastic bone Osteoclasts destruction Subchondral Decrease inflammatory factors and alleviate the symptom of bone osteoarthritis Silence HiF-2α; Downregulate catabolic factors, maintain Chondrocytes cartilage integrity and alleviate synovium inflammation Block RANK/RANKL pathway and induce active bone NF-κB remodeling for osteoarthritis treatment Migrate to the fracture area, initiate callus ossification and Fracture area promote fracture healing Show high affinity for HA; Shorten healing time; Improve Bone matrix therapeutic outcomes

CAP-PEI based nanoparticles

BP-conjugated OPG Sox11-modified MSCs

Bone fracture SAA-loaded liposomes

l a n

o r p

e

r P

PDB

BP

Osteoclasts

SAON

Asp8-functionalized liposomes

Osteomyelitis

BP conjugated ciprofloxacin

Osteocytes and Decrease osteocytes apoptosis and increase osteoclatsogenesis osteoclasts and osteogenesis High turnover Improve the therapeutic index for osteomyelitis site

o J

ur

Reduce RANKL/OPG ratio

56

(Zhao, et al., 2019) (He, et al., 2017) (Moreau, et al., 2011) (Pi, et al., 2015) (Doschak, et al., 2009)

(Xu, et al., 2015)

(Liu, et al., 2018) (Martini, et al., 2007) (Chen, et al., 2018) (Xie, et al., 2017)

Journal Pre-proof

References

Abarrategi, A., Tornin, J., Martinez-Cruzado, L., Hamilton, A., Martinez-Campos, E., Rodrigo, J. P., Gonzalez, M. V., Baldini, N., Garcia-Castro, J., & Rodriguez, R. (2016). Osteosarcoma: Cells-of-origin, cancer stem cells, and targeted therapies. Stem Cells Int, 2016, 3631764. Abrahamsen, B., Eiken, P., & Brixen, K. (2009). Atrial fib rillation in fracture patients treated with oral

oo

f

bisphosphonates. J Intern Med, 265, 581-592. Adami, S., & Zamberlan, N. (1996). Adverse effects of bisphosphonates. A comparative review. Drug

pr

Saf, 14, 158-170.

e-

Ahmed, N., Salsman, V. S., Yvon, E., Louis, C. U., Perlaky, L., Wels, W. S., Dishop, M. K., Kleinerman,

Pr

E. E., Pule, M., Rooney, C. M ., Heslop, H. E., & Gottschalk, S. (2009). Immunotherapy for osteosarcoma: genetic modificat ion of T cells overcomes low levels of tu mor antigen

al

expression. Mol Ther, 17, 1779-1787.

Jo u

280-303.

rn

Ahmed, R. (2015). Regulatory mechanisms of bone development and function. Int J Adv Res, 3,

Albert, A., & Rees, C. W. (1956). Avidity of the tetracyclines for the cations of metals. Nature, 177, 433-434.

Alford, A. I., Ko zloff, K. M., & Hankenson, K. D. (2015). Ext racellular matrix networks in bone remodeling. Int J Biochem Cell Biol, 65, 20-31. Antebi, B., Pelled, G., & Gazit, D. (2014). Stem cell therapy for osteoporosis. Curr Osteoporos Rep, 12, 41-47. Aoki, K., Alles, N., Soysa, N., & Ohya, K. (2012). Peptide-based delivery to bone. Adv Drug Deliv Rev, 64, 1220-1238. 57

Journal Pre-proof

Arana-Chavez, V. E., Soares, A. M. V., & Katchburian, E. (1995). Junctions between early developing osteoblasts of rat calvaria as revealed by freeze-fracture and ultrathin section electron microscopy. %J Archives of histology and cytology. Arch Histol Cytol, 3. Åstedt, B. (1982). On the role of estrogens in endometrial carcinogenesis. Acta Obstet Gynecol Scand, 61, 33-35.

oo

hydroxyapatite nucleation. Matrix Biol, 27, 600-608.

f

Baht, G., Hunter, G., & Goldberg, H. J. M. B. (2008). Bone sialoprotein-collagen interaction promotes

pr

Bellido, M., Lugo, L., Roman-Blas, J. A., Castaneda, S., Caeiro, J. R., Dapia, S., Calvo, E., Largo,

e-

R., & Herrero-Beaumont, G. (2010). Subchondral bone microstructural damage by

Arthritis Res Ther, 12, R152.

Pr

increased remodelling aggravates experimental osteoarthritis preceded by osteoporosis.

al

Bevelander, G., Rolle, G. K., & Cohlan, S. Q. J. J. o. D. R. (1961). The effect of the administration of

rn

tetracycline on the development of teeth. J Dent Res, 40, 1020-1024.

Jo u

Bhandari, K. H., Newa, M., Uludag, H., & Doschak, M. R. J. I. j. o. p. (2010). Synthesis, characterization and in vitro evaluation of a bone targeting delivery system for salmon calcitonin. Int J Pharm, 394, 26-34. Blacksburg, S. R., Witten, M. R., & Haas, J. A. (2015). Integrating bone targeting radiopharmaceuticals into the management of patients with castrate-resistant prostate cancer with symptomatic bone metastases. Current Treatment Options in Oncology, 16, 11. Boivin, G., Bala, Y., Doublier, A., Farlay, D., Ste-Marie, L. G., Meunier, P. J., & Delmas, P. D. (2008).

The role of mineralization and organic matrix in the microhardness of bone tissue from controls and osteoporotic patients. Bone, 43, 532-538. 58

Journal Pre-proof

Boskey, A. L. (1995). Osteopontin and related phosphorylated sialoproteins: effects on mineralizationa. Ann N Y Acad Sci, 760, 249-256. Brown, J. P., Morin, S., Leslie, W., Papaioannou, A., Cheung, A. M., Davison, K. S., Goltzman, D., Hanley, D. A., Hodsman, A., Josse, R., Jovaisas, A., Juby, A., Kaiser, S., Karaplis, A., Kendler, D., Khan, A., Ngui, D., Olszynski, W., Ste-Marie, L. G., & Adachi, J. (2014). Bisphosphonates

f

for treatment of osteoporosis: expected benefits, potential harms, and drug holidays. Can Fam

oo

Physician, 60, 324-333.

pr

Burr, D. B. (2019). Bone morphology and organization. In D. B. Burr & M. R. Allen (Eds.), Basic and

e-

Applied Bone Biology (pp. 3-26): Academic Press.

Biochem Biophys, 561, 3-12.

Pr

Capulli, M., Paone, R., & Rucci, N. (2014). Osteoblast and osteocyte: games without frontiers. Arch

al

Carbone, E. J., Rajpura, K., Allen, B. N., Cheng, E., Ulery, B. D., & Lo, K. W. (2017). Osteotropic

rn

nanoscale drug delivery systems based on small molecule bone-targeting moieties.

Jo u

Nanomedicine, 13, 37-47.

Carbone, E. J., Rajpura, K., Jiang, T., Kan, H.-M., Yu, X., Lo, K. W.-H. J. J. o. N., & Nanotechnology. (2017). Osteotropic nanoscale drug delivery system via a single aspartic acid as the bone-targeting moiety. J Nanosci Nanotechnol, 17, 1747-1752. Cawthray, J. F., Creagh, A. L., Haynes, C. A., & Orvig, C. (2015). Ion exchange in hydroxyapatite with lanthanides. Inorg Chem, 54, 1440-1445. Chambers, T., & Magnus, C. J. T. J. o. p. (1982). Calcitonin alters behaviour of isolated osteoclasts. J

Pathol, 136, 27-39.

59

Journal Pre-proof

Chan, M. H., Chan, P. K., Griffith, J. F., Chan, I. H., Lit, L. C., Wong, C., Antonio, G. E., Liu, E. Y., Hui, D. S., & Suen, M. W. J. P. (2006). Steroid-induced osteonecrosis in severe acute respiratory syndrome: a retrospective analysis of biochemical markers of bone metabolism and corticosteroid therapy. Pathology, 38, 229-235. Chen, S., Zheng, L., Zhang, J., Wu, H., Wang, N., Tong, W., Xu, J., Huang, L., Zhang, Y., Yang, Z.,

f

Lin, G., Wang, X., & Qin, L. (2018). A novel bone targeting delivery system carrying

oo

phytomolecule icaritin for prevention of steroid-associated osteonecrosis in rats. Bone, 106,

pr

52-60.

e-

Cheng, H., Chawla, A., Yang, Y., Li, Y., Zhang, J., Jang, H. L., & Khademhosseini, A. (2017).

Pr

Development of nanomaterials for bone-targeted drug delivery. Drug Discov Today, 22,

1336-1350.

al

Cheville, J. C., Tindall, D., Boelter, C., Jenkins, R., Lohse, C. M., Pankratz, V. S., Sebo, T. J., Davis, B.,

rn

& Blute, M. L. J. C. (2002). Metastatic prostate carcinoma to bone: clinical and pathologic

Jo u

features associated with cancer-specific survival. Cancer, 95, 1028-1036. Chyun, Y. S., & Raisz, L. G. J. P. (1984). Stimulation of bone formation by prostaglandin E2. Prostaglandins, 27, 97-103. Coleman, R. E. (2006). Clinical features of metastatic bone disease and risk of skeletal morbidity. Clin Cancer Res, 12, 6243s-6249s. Cosman., F., Nieves., J., Zion., M., Woelfert., L., Luckey., M., & Lindsay., R. (2005). Daily and cyclic parathyroid hormone in women receiving alendronate. N Engl J Med, 353, 566–575. Crane, J. L., & Cao, X. J. T. J. o. c. i. (2014). Bone marrow mesenchymal stem cells and TGF-β signaling in bone remodeling. J Clin Invest, 124, 466-472. 60

Journal Pre-proof

Croucher, P. I., McDonald, M. M., & Martin, T. J. (2016). Bone metastasis: the importance of the neighbourhood. Nat Rev Cancer, 16, 373-386. Cui, L., Liu, Y.-y., Wu, T., A i, C.-m., & Chen, H.-q. J. A. P. S. (2009). Osteogenic effects of D (+) β-3, 4-dihydro xyphenyl lactic acid (salv ianic acid A, SAA) on osteoblasts and bone marrow stromal cells of intact and prednisone-treated rats. Acta Pharmacol Sin, 30, 321.

f

Cui, Y.-X., Evans, B. A., & Jiang, W. G. J. A. r. (2016). New roles of osteocytes in proliferation,

oo

migration and invasion of breast and prostate cancer cells. Anticancer Res, 36, 1193-1201.

pr

Dell, R. M., Adams, A. L., Greene, D. F., Funahashi, T. T., Silverman, S. L., Eisemon, E. O., Zhou, H.,

e-

Burchette, R. J., & Ott, S. M. (2012). Incidence of atypical nontraumatic diaphyseal fractures

Pr

of the femur. J Bone Miner Res, 27, 2544-2550.

Detsch, R., & Boccaccini, A. R. (2015). The role of osteoclasts in bone tissue engineering. J Tissue Eng

al

Regener Med, 9, 1133-1149.

rn

Dhule, S. S., Penfornis, P., He, J., Harris, M. R., Terry, T., John, V., & Pochampally, R. (2014). The

Jo u

combined effect of encapsulating curcumin and C6 ceramide in liposomal nanoparticles against osteosarcoma. Mol Pharm, 11, 417-427. Doschak, M. R., Kucharski, C. M., Wright, J. E., Zernicke, R. F., & Uludag, H. (2009). Improved bone delivery of osteoprotegerin by bisphosphonate conjugation in a rat model of osteoarthritis. Mol Pharm, 6, 634-640. Downey, P. A., & Siegel, M. I. (2006). Bone biology and the clinical implications for osteoporosis. Phys Ther, 86, 77-91.

Drake, M. T., Clarke, B. L., & Khosla, S. (2008). Bisphosphonates: Mechanism of action and role in clinical practice. Mayo Clin Proc, 83, 1032-1045. 61

Journal Pre-proof

DuBois, S., & Demetri, G. (2007). Markers of angiogenesis and clinical features in patients with sarcoma. Cancer, 109, 813-819. Duffy, M. J., Synnott, N. C., & Crown, J. (2017). Mutant p53 as a target for cancer treat ment. Eur J Cancer, 83, 258-265. Erez, R., Ebner, S., Attali, B., Shabat, D. J. B., & letters, m. c. (2008). Chemotherapeutic bone-targeted

f

bisphosphonate prodrugs with hydrolytic mode of activation. Bioorg Med Chem Lett, 18,

oo

816-820.

pr

Everts, V. (2002). The bone lining cell: its role in cleaning Howship's lacunae and initiating bone

e-

formation. J Bone Miner Res, 1, 77-90.

Pr

Fang, Z. H., Sun, Y. P., Xiao, H., Li, P., Liu, M., Ding, F., Kan, W. S., & Miao, R. S. (2017). Targeted

osteosarcoma chemotherapy using RGD peptide-installed doxorubicin-loaded biodegradable

al

polymeric micelle. Biomed Pharmacother, 85, 160-168.

rn

Florencio-Silva, R., Sasso, G. R. d. S., Sasso-Cerri, E., Simões, M. J., & Cerri, P. S. (2015). Biology of

17.

Jo u

bone tissue: structure, function, and factors that influence bone cells. BioMed Res Int, 2015,

Franz-Odendaal, T. A., Hall, B. K., & Witten, P. E. (2006). Buried alive: How osteoblasts become osteocytes. Dev Dyn, 235, 176-190. Fu, Y.-C., Fu, T.-F., Wang, H.-J., Lin, C.-W., Lee, G.-H., Wu, S.-C., & Wang, C.-K. J. A. b. (2014). Aspartic acid-based modified PLGA–PEG nanoparticles for bone targeting: In vitro and in vivo evaluation. Acta Biomater, 10, 4583-4596.

62

Journal Pre-proof

Gil, L., Han, Y., Opas, E. E., Rodan, G. A., Ruel, R., Seedor, J. G., Tyler, P. C., Young, R. N. J. B., & chemistry, m. (1999). Prostaglandin E2-bisphosphonate conjugates: potential agents for treatment of osteoporosis. Bioorg Med Chem, 7, 901-919. Goldring, M. B. (2000). The role of the chondrocyte in osteoarthritis. Arthritis Rheum, 43, 1916-1926. Goldring, S. R., & Goldring, M. B. (2010). Bone and cartilage in osteoarthritis: is what's best for one

f

good or bad for the other? In: BioMed Central.

oo

Go mes., K. d. N., A lves., A. P. N. N., Dutra., P. G. P., & Viana., G. S. d. B. (2017). Do xycycline

pr

induces bone repair and changes in Wnt signalling. Int J Oral Sci, 9, 158.

e-

Go mez-Barrena, E., Rosset, P., Lo zano, D., Stanovici, J., Ermthaller, C., & Gerbhard, F. (2015). Bone

Pr

fracture healing: Cell therapy in delayed unions and nonunions. Bone, 70, 93-101.

Grayson, W. L., Bunnell, B. A., Martin, E., Frazier, T., Hung, B. P., & Gimble, J. M. J. N. R. E. (2015).

al

Stromal cells and stem cells in clinical bone regeneration. Nat Rev Endocrinol, 11, 140.

rn

Green, J., Czanner, G., Reeves, G., Watson, J., Wise, L., & Beral, V. (2010). Oral bisphosphonates and

Jo u

risk o f cancer of oesophagus, stomach, and colorectu m: case-control analysis within a UK primary care cohort. BMJ-Br Med J, 341, c4444. Grignani, G., Palmerini, E., Dileo, P., Asaftei, S. D., D'Ambrosio, L., Pignochino, Y., Mercuri, M., Picci, P., Fagioli, F., Casali, P. G., Ferrari, S., & Aglietta, M. (2012). A phase II trial of sorafenib in relapsed and unresectable high-grade osteosarcoma after failure of standard multimodal therapy: an Italian Sarcoma Group study. Ann Oncol, 23, 508-516. Guan, M., Yao, W., Liu, R., Lam, K. S., Nolta, J., Jia, J., Panganiban, B., Meng, L., Zhou, P., &

Shahnazari, M. J. N. m. (2012). Directing mesenchymal stem cells to bone to augment bone formation and increase bone mass. Nat Med, 18, 456. 63

Journal Pre-proof

Hayashi, M., Nakashima, T., Taniguchi, M., Kodama, T., Kumanogoh, A., & Takayanagi, H. J. N. (2012). Osteoprotection by semaphorin 3A. Nature, 485, 69-74. He, Y., Huang, Y., Huang, Z., Jiang, Y., Sun, X., Shen, Y., Chu, W., & Zhao, C. (2017). Bisphosphonate-functionalized coordination polymer nanoparticles for the treatment of bone metastatic breast cancer. J Control Release, 264, 76-88.

f

Hengst, V., Oussoren, C., Kissel, T., & Storm, G. J. I. j. o. p. (2007). Bone targeting potential of

oo

bisphosphonate-targeted liposomes: Preparation, characterization and hydroxyapatite binding

pr

in vitro. Int J Pharm, 331, 224-227.

e-

Herrero-Beaumont, G., & Roman-Blas, J. A. (2013). Osteoarthritis: Osteoporotic OA: a reasonable

Pr

target for bone-acting agents. Nat Rev Rheumatol, 9, 448-450.

Hienz, S. A., Paliwal, S., & Ivanovski, S. (2015). Mechanisms of bone resorption in periodontitis. J

al

Immunol Res, 2015, 615486.

rn

Hirabayashi, H., & Fujisaki, J. (2003). Bone-specific drug delivery systems: approaches via chemical

Jo u

modification of bone-seeking agents. Clin Pharmacokinet, 42, 1319-1330. Hofbauer, L., Khosla, S., Dunstan, C., Lacey, D., Boyle, W., & Riggs, B. J. J. B. M. R. (2000). The roles of osteoprotegerin and osteoprotegerin ligand in the paracrine regulation of bone resorption. J Bone Miner Res, 15, 2-12. Hou, Z., Meyer, S., Propson, N. E., Nie, J., Jiang, P., Stewart, R., & Thomson, J. A. J. C. r. (2015). Characterization and target identification of a DNA aptamer that labels pluripotent stem cells. Cell Res, 25, 390.

64

Journal Pre-proof

Hu, X., Wang, Y., Tan, Y., Wang, J., Liu, H., Wang, Y., Yang, S., Shi, M., Zhao, S., & Zhang, Y. J. A. M. (2017). A difunctional regeneration scaffold for knee repair based on aptamer‐directed cell recruitment. Adv Mater, 29, 1605235. Hunter, G. K., & Goldberg, H. A. (1993). Nucleation of hydroxyapatite by bone sialoprotein. Proc Natl Acad Sci, 90, 8562-8565.

f

Itaka, K., Ohba, S., Miyata, K., Kawaguchi, H., Nakamura, K., Takato, T., Chung, U. I., & Kataoka, K.

oo

(2007). Bone regeneration by regulated in vivo gene transfer using biocompatible polyplex

pr

nanomicelles. Mol Ther, 15, 1655-1662.

e-

J. Hatzenbuehler, T. J. P. (2011). Diagnosis and management of osteomyelitis. Am Fam Physician, 84,

Pr

1027-1033.

Jee, W. S., Ke, H. Z., & Li, X. J. (1991). Long-term anabolic effects of prostaglandin-E2 on tibial

al

diaphyseal bone in male rats. Bone Miner, 15, 33-55.

rn

Ji, F., Zhang, H., Wang, Y., Li, M., Xu, W., Kang, Y., Wang, Z., Wang, Z., Cheng, P., Tong, D., Li, C., &

Jo u

Tang, H. (2013). MicroRNA-133a, downregulated in osteosarcoma, suppresses proliferation and promotes apoptosis by targeting Bcl-xL and Mcl-1. Bone, 56, 220-226. Jiang, T., Yu, X., Carbone, E. J., Nelson, C., Kan, H. M., & Lo, K. W.-H. J. I. j. o. p. (2014). Poly aspartic acid peptide-linked PLGA based nanoscale particles: potential for bone-targeting drug delivery applications. Int J Pharm, 475, 547-557. Julia F, C. (2014). Osteoclasts: more than 'bone eaters'. Trends Mol Med, 8. Junko, I. (2009). Selective drug delivery to bone using acidic oligopeptides. %J Journal of bone and

mineral metabolism. J Bone Miner Metab, 1.

65

Journal Pre-proof

Khan, A. A., Morrison, A., Hanley, D. A., Felsenberg, D., McCauley, L. K., O'Ryan, F., Reid, I. R., Ruggiero, S. L., Taguchi, A., Tetradis, S., Watts, N. B., Brandi, M. L., Peters, E., Guise, T., Eastell, R., Cheung, A. M., Morin, S. N., Masri, B., Cooper, C., Morgan, S. L., Obermayer-Pietsch, B., Langdahl, B. L., Al Dabagh, R., Davison, K. S., Kendler, D. L., Sándor, G. K., Josse, R. G., Bhandari, M., El Rabbany, M., Pierroz, D. D., Sulimani, R.,

f

Saunders, D. P., Brown, J. P., Compston, J., & Jaw, o. b. o. t. I. T. F. o. O. o. t. (2015).

oo

Diagnosis and Management of Osteonecrosis of the Jaw: A systematic review and

pr

international consensus. J Bone Miner Res, 30, 3-23.

e-

Khosla, S., Amin, S., & Orwoll, E. (2008). Osteoporosis in men. Endocr Rev, 29, 441-464.

Pr

Khosla, S., & Hofbauer, L. C. (2017). Osteoporosis treatment: recent developments and ongoing

challenges. Lancet Diabetes Endocrinol, 5, 898-907.

al

Kitaori, T., Ito, H., Schwarz, E. M ., Tsutsumi, R., Yoshitomi, H., Oishi, S., Nakano, M., Fujii, N.,

rn

Nagasawa, T., Nakamura, T. J. A., & Rheu matology, R. O. J. o. t. A. C. o. (2009). Stro mal

Jo u

cell-derived factor 1/ CXCR4 signaling is crit ical for the recru it ment of mesenchyma l stem cells to the fracture site during skeletal repair in a mouse model. Arth Rheum, 60, 813-823. Kneser, U., Schaefer, D. J., Po lykandriotis, E., & Horch, R. E. (2006). Tissue engineering of bone: the reconstructive surgeon's point of view. J Cell Mol Med, 10, 7-19. Nanomedicine, 14, 2271-2282. Lakkakorpi, P., Horton, M ., Helfrich, M., Karhukorp i, E., & Väänänen, H. J. T. J. o. c. b . (1991). Vit ronectin receptor has a ro le in bone resorption but does not mediate t ight sealing zone attachment of osteoclasts to the bone surface. J Cell Biol, 115, 1179-1186.

66

Journal Pre-proof

Lange, R., ter Heine, R., Knapp, R., de Klerk, J. M. H., Bloemendal, H. J., & Hendrikse, N. H. (2016). Pharmaceutical and clinical development of phosphonate-based radiopharmaceuticals for the targeted treatment of bone metastases. Bone, 91, 159-179. Laslett, L. L., Kingsbury, S. R., Hensor, E. M. A., Bowes, M. A., & Conaghan, P. G. (2014). Effect of bisphosphonate use in patients with symptomatic and radiographic knee osteoarthritis: data

f

from the Osteoarthritis Initiative. Annals of the Rheumatic Diseases, 73, 824-830.

oo

Lawson, M. A., Xia, Z., Barnett, B. L., Triffitt, J. T., Phipps, R. J., Dunford, J. E., Locklin, R. M.,

pr

Ebetino, F. H., & Russell, R. G. G. (2010). Differences between bisphosphonates in binding

e-

affinities for hydroxyapatite. J Biomed Mater Res B Appl Biomater, 92B, 149-155.

Pr

Leder, B. Z. (2017). Parathyroid hormone and parathyroid hormone-related protein analogs in

osteoporosis therapy. Curr Osteoporos Rep, 15, 110-119.

al

Lee, K. K., Lee, J.-G., Park, C. S., Lee, S. H., Raja, N., Yun, H.-s., Lee, J.-S., & Lee, C.-S. J. R. a.

rn

(2019). Bone-targeting carbon dots: effect of nitrogen-doping on binding affinity. RSC

Jo u

Advances, 9, 2708-2717.

Lew, D. P., & Waldvogel, F. A. (2004). Osteomyelitis. Lancet, 364, 369-379. Li, C. j., Liu, X. z., Zhang, L., Chen, L. b., Shi, X., Wu, S. j., & Zhao, J. n. J. O. s. (2016). Advances in bone-targeted drug delivery systems for neoadjuvant chemotherapy for osteosarcoma. Orthop Surg, 8, 105-110. Li, D., Liu, J., Guo, B., Liang, C., Dang, L., Lu, C., He, X., Cheung, H. Y.-S., Xu, L., & Lu, C. J. N. c. (2016). Osteoclast-derived exosomal miR-214-3p inhibits osteoblastic bone formation. Nat

Commun, 7, 10872. Li, Z., Kong, K., & Qi, W. (2006). Osteoclast and its roles in calciu m metabolis m and bone 67

Journal Pre-proof

development and remodeling. Biochem Biophys Res Commun, 343, 345-350. Li., K.-C., Chang., Y.-H., Yeh., C.-L., & Hu., Y.-C. (2016). Healing of osteoporotic bone defects by baculovirus-engineered bone marro w-derived MSCs expressing MicroRNA sponges. Biomaterials, 74, 155-166. Liang, C., Guo, B., Wu, H., Shao, N., Li, D., Liu, J., Dang, L., Wang, C., Li, H., Li, S., Lau, W. K.,

f

Cao, Y., Yang, Z., Lu, C., He, X., Au, D. W., Pan, X., Zhang, B. T., Lu, C., Zhang, H., Yue, K.,

oo

Qian, A., Shang, P., Xu, J., Xiao, L., Bian, Z., Tan, W., Liang, Z., He, F., Zhang, L., Lu, A., &

pr

Zhang, G. (2015). Aptamer-functionalized lipid nanoparticles targeting osteoblasts as a novel

e-

RNA interference-based bone anabolic strategy. Nat Med, 21, 288-294.

Pr

Liang, C., Li, F., Wang, L., Zhang, Z. K., Wang, C., He, B., Li, J., Chen, Z., Shaikh, A. B., Liu, J., Wu,

X., Peng, S., Dang, L., Guo, B., He, X., Au, D. W. T., Lu, C., Zhu, H., Zhang, B. T., Lu, A., &

al

Zhang, G. (2017). Tumor cell-targeted delivery of CRISPR/Cas9 by aptamer-functionalized

rn

lipopolymer for therapeutic genome editing of VEGFA in osteosarcoma. Biomaterials, 147,

Jo u

68-85.

Lin, W., Xu, L., Zwingenberger, S., Gibon, E., Good man, S. B., & Li, G. (2017). Me senchymal stem cells homing to improve bone healing. J Orthop Translat, 9, 19-27. Lindsay, R., Nieves, J., Formica, C., Henneman, E., Woelfert, L., Shen, V., Dempster, D., & Cosman, F. (1997). Randomised controlled study of effect of parathyroid hormone on vertebral-bone mass and fracture incidence among postmenopausal women on oestrogen with osteoporosis. Lancet, 350, 550-555.

Liu, X., Wang, D., Zhang, P., & Li, Y. (2019). Recent advances in nanosized drug delivery systems for overcoming the barriers to anti-PD immunotherapy of cancer. Nano Today. 68

Journal Pre-proof

Liu, Y., Jia, Z., Akhter, M. P., Gao, X., Wang, X., Wang, X., Zhao, G., Wei, X., Zhou, Y., Wang, X., Hartman, C. W., Fehringer, E. V., Cui, L., & Wang, D. (2018). Bone-targeting liposome formulation of Salvianic acid A accelerates the healing of delayed fracture Union in Mice. Low, S. A., & Kopecek, J. (2012). Targeting polymer therapeutics to bone. Adv Drug Deliv Rev, 64, 1189-1204.

f

Low, S. A., Yang, J. Y., & Kopecek, J. (2014). Bone-targeted acid-sensitive doxorubicin conjugate

oo

micelles as potential osteosarcoma therapeutics. Bioconjugate Chem, 25, 2012-2020.

pr

Lu, B., Qiu, M., & Zheng, H. (1998). Co mparat ive study of tetracycline-estrone and estrone effects on

e-

bone histomorphometric parameters in ovariecto mized rats. ZhongHua Fu Chan Ke Za Zhi, 33,

Pr

31-34.

Lu, J. M., Wang, X. W., Marin-Muller, C., Wang, H., Lin, P. H., Yao, Q. Z., & Chen, C. Y. (2009).

al

Current advances in research and clinical applications of PLGA-based nanotechnology. Expert

rn

Rev Mol Diagn, 9, 325-341.

Jo u

Machwate, M., Harada, S., Leu, C., Seedor, G., Labelle, M., Gallant, M., Hutchins, S., Lachance, N., Sawyer, N., & Slipetz, D. J. M. p. (2001). Prostaglandin receptor EP4 mediates the bone anabolic effects of PGE2. Mol Pharmacol, 60, 36-41. Marks, S. C., Jr., & Popoff, S. N. (1988). Bone cell biology: the regulation of development, structure, and function in the skeleton. Am J Anat, 183, 1-44. Martini, G., Gennari, L., Merlotti, D., Salvadori, S., Franci, M. B., Campagna, S., Avanzati, A., De Paola, V., Valleggi, F., & Nuti, R. (2007). Serum OPG and RANKL levels before and after

intravenous bisphosphonate treatment in Paget's disease of bone. Bone, 40, 457-463.

69

Journal Pre-proof

Matic, I., Matthews, B. G., Wang, X., Dyment, N. A., Worthley, D. L., Rowe, D. W., Grcevic, D., & Kalajzic, I. (2016). Quiescent bone lining cells are a major source of osteoblasts during adulthood. Stem Cells, 34, 2930-2942. Matsushita, O., Jung, C.-M., Minami, J., Katayama, S., Nishi, N., & Okabe, A. J. J. o. B. C. (1998). A study of the collagen-binding domain of a 116-kDa clostridium histolyticum collagenase. J

f

Biol Chem, 273, 3643-3648.

oo

Merolli, A., Fung, S., Murthy, N. S., Pashuck, E. T., Mao, Y., Wu, X., Steele, J. A. M., Martin, D.,

pr

Moghe, P. V., Bromage, T., & Kohn, J. J. J. o. M. S. M. i. M. (2018). “Ruffled border”

e-

formation on a CaP-free substrate: A first step towards osteoclast-recruiting bone-grafts

Pr

materials able to re-establish bone turn-over. J Mater Sci Mater Med, 29, 38.

Miller, S. C., de Saint-Georges, L., Bowman, B. M., & Jee, W. S. (1989). Bone lining cells: structure

al

and function. Scanning Microsc, 3, 953-960; discussion 960-951.

rn

Moreau, M., Rialland, P., Pelletier, J. P., Martel-Pelletier, J., Lajeunesse, D., Boileau, C., Caron, J.,

Jo u

Frank, D., Lussier, B., del Castillo, J. R., Beauchamp, G., Gauvin, D., Bertaim, T., Thibaud, D., & Troncy, E. (2011). Tiludronate treatment improves structural changes and symptoms of osteoarthritis in the canine anterior cruciate ligament model. Arthritis Res Ther, 13, R98. Mori, S., & Burr, D. B. (1993). Increased intracortical remodeling following fatigue damage. Bone, 14, 103-109. Mühlbauer, H. F. G. G. R. B. A. C. C. (1968). The influence of pyrophosphate analogues (diphosphonates) on the precipitation and dissolution of calcium phosphate in vitro and in

vivo. Calcif Tissue Res, 2, 10.

70

Journal Pre-proof

Mukherjee, S., Raje, N., Schoonmaker, J. A., Liu, J. C., Hideshima, T., Wein, M. N., Jones, D. C., Vallet, S., Bouxsein, M. L., & Pozzi, S. J. T. J. o. c. i. (2008). Pharmacologic targeting of a stem/progenitor population in vivo is associated with enhanced bone regeneration in mice. J Clin Invest, 118, 491-504. Myers, H., Tochon-Danguy, H., & Baud, C. J. C. t. i. (1983). IR absorption spectrophotometric analysis

f

of the complex formed by tetracycline and synthetic hydroxyapatite. Calcif Tissue Int, 35,

oo

745-749.

pr

Nakamura, Y., Simpo, S., Lee, M., Oikawa, T., Yoshii, T., Noda, K., Kuwahara, Y., & Kawasaki, K.

Pr

in the rat. Biotech Histochem, 75, 1-6.

e-

(2000). Histology and tetracycline labeling of a single section of alveolar bone of first molars

Nancollas, G. H., Tang, R., Phipps, R. J., Henneman, Z., Gulde, S., Wu, W., Mangood, A., Russell, R.

al

G. G., & Ebetino, F. H. (2006). Novel insights into actions of bisphosphonates on bone:

rn

Differences in interactions with hydroxyapatite. Bone, 38, 617-627.

Jo u

Neale, J. R., Richter, N. B., Merten, K. E., Taylor, K. G., Singh, S., Waite, L. C., Emery, N. K., Smith, N. B., Cai, J., Pierce Jr, W. M. J. B., & letters, m. c. (2009). Bone selective effect of an estradiol conjugate with a novel tetracycline-derived bone-targeting agent. Bioorg Med Chem Lett, 19, 680-683. Neer, R. M., Arnaud, C. D., Zanchetta, J. R., Prince, R., Gaich, G. A., Reginster, J.-Y., Hodsman, A. B., Eriksen, E. F., Ish-Shalom, S., Genant, H. K., Wang, O., Mellström, D., Oefjord, E. S., Marcinowska-Suchowierska, E., Salmi, J., Mulder, H., Halse, J., Sawicki, A. Z., & Mitlak, B.

H. (2001). Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med, 344, 1434-1441. 71

Journal Pre-proof

Negishi-Koga, T., & Takayanagi, H. (2012). Bone cell communication factors and Semaphorins. BoneKEy reports, 1, 183-183. Nelson, J. B., Hedican, S. P., George, D. J., Reddi, A. H., Piantadosi, S., Eisenberger, M. A., & Simons, J. W. J. N. m. (1995). Identification of endothelin–1 in the pathophysiology of metastatic adenocarcinoma of the prostate. Nat Med, 1, 944.

f

Nilsson, S., Franzen, L., Parker, C., Tyrrell, C., Blom, R., Tennvall, J., Lennernas, B., Petersson, U.,

oo

Johannessen, D. C., Sokal, M., Pigott, K., Yachnin, J., Garkavij, M., Strang, P., Harmenberg,

pr

J., Bolstad, B., & Bruland, O. S. (2007). Bone-targeted radium-223 in symptomatic,

e-

hormone-refractory prostate cancer: a randomised, multicentre, placebo-controlled phase II

Pr

study. Lancet Oncol, 8, 587-594.

Niu, G., & Chen, X. Y. (2010). Vascular Endothelial Growth Factor as an Anti-Angiogenic Target for

al

Cancer Therapy. Curr Drug Targets, 11, 1000-1017.

rn

Novicoff, W. M., Manaswi, A., Hogan, M. V., Brubaker, S. M., M ihalko, W. M., & Saleh, K. J. (2008).

Jo u

Critical analysis of the evidence for current technologies in bone-healing and repair. J Bone Joint Surg Am, 90 Suppl 1, 85-91. Novina, C. D., & Sharp, P. A. (2004). The RNAi revolution. Nature, 430, 161-164. Ohba, T., Cole, H. A., Cates, J. M., Slosky, D. A., Haro, H., Ando, T., Schwartz, H. S., Schoenecker, J. G. J. J. o. B., & Research, M. (2014). Bisphosphonates inhibit osteosarcoma‐mediated osteolysis via attenuation of tumor expression of MCP‐1 and RANKL. J Bone Miner Res, 29, 1431-1445.

Oldberg, A., Franzen, A., & Heinegard, D. (1988). The primary structure of a cell-binding bone sialoprotein. J Biol Chem, 263, 19430-19432. 72

Journal Pre-proof

Ossipov, D. A. (2015). Bisphosphonate-modified b io materials for drug delivery and bone tissue engineering. Expert Opin Drug Deliv, 12, 1443-1458.

Park, D., Park, C.-W., Choi, Y., Lin, J., Seo, D.-H., Kim, H.-S., Lee, S. Y., & Kang, I.-C. J. B. (2016). A novel small-molecule PPI inhibitor targeting integrin αvβ3-osteopontin interface blocks bone resorption in vitro and prevents bone loss in mice. Biomaterials, 98, 131-142.

f

Pazianas, M., Cooper, C., Ebetino, F. H., & Russell, R. G. G. (2010). Long -term treat ment with

oo

bisphosphonates and their safety in postmenopausal osteoporosis. Ther Clin Risk Manage, 6,

pr

325-343.

e-

Pentyala, S. N., Lee, J., Hsieh, K., Waltzer, W. C., Trocchia, A., Musacchia, L., Rebecchi, M. J., &

Pr

Khan, S. A. (2000). Prostate cancer: a comprehensive review. Med Oncol, 17, 85-105. Pi, Y., Zhang, X., Shao, Z., Zhao, F., Hu, X., & Ao, Y. J. G. t. (2015). Intra -articular delivery of

al

anti-Hif-2α siRNA by chondrocyte-homing nanoparticles to prevent cartilage degeneration in

rn

arthritic mice. Gene Ther, 22, 439.

Jo u

Pi, Y. B., Zhang, X., Shi, J. J., Zhu, J. X., Chen, W. Q., Zhang, C. G., Gao, W. W., Zhou, C. Y., & Ao, Y. F. (2011). Targeted delivery of non-viral vectors to cartilage in vivo using a chondrocyte-homing peptide identified by phage display. Biomaterials, 32, 6324-6332. Ponnapakkam, T., Katikaneni, R., Sakon, J., Stratford, R., & Gensure, R. C. (2014). Treating osteoporosis by targeting parathyroid hormone to bone. Drug Discov Today, 19, 204-208. Ponnapakkam, T., Katikaneni, R., Suda, H., Miyata, S., Matsushita, O., Sakon, J., & Gensure, R. C. J. C. t. i. (2012). A single injection of the anabolic bone agent, parathyroid hormone–collagen

binding domain (PTH–CBD), results in sustained increases in bone mineral density for up to 12 months in normal female mice. Calcif Tissue Int, 91, 196-203. 73

Journal Pre-proof

Porter, A. T., McEwan, A. J., Powe, J. E., Reid, R., McGowan, D. G., Lukka, H., Sathyanarayana, J. R., Yakemchuk, V. N., Thomas, G. M., Erlich, L. E., & et al. (1993). 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, 25, 805-813.

f

Prideaux, M., Findlay, D. M., & Atkins, G. J. (2016). Osteocytes: The master cells in bone remodelling.

oo

Curr Opin Pharmacol, 28, 24-30.

pr

Qian, Y., Bhowmik, D., Kachru, N., & Hernandez, R. K. (2017). Longitudinal patterns of bone-targeted

Pr

Care Cancer, 25, 1845-1851.

e-

agent use among patients with solid tumors and bone metastases in the United States. Support

Qiao, H., Cui, Z., Yang, S., Ji, D., Wang, Y., Yang, Y., Han, X., Fan, Q., Qin, A., & Wang, T. J. A. n.

al

(2017). Targeting osteocytes to attenuate early breast cancer bone metastasis by theranostic

rn

upconversion nanoparticles with responsive plumbagin release. ACS Nano, 11, 7259-7273.

Jo u

Qiao, H., Wang, T., Yu, Z., Han, X., Liu, X., Wang, Y., Fan, Q., Qin, A., Tang, T. J. C. d., & disease. (2016). Structural simulation of adenosine phosphate via plumbagin and zoledronic acid competitively targets JNK/Erk to synergistically attenuate osteoclastogenesis in a breast cancer model. Cell Death Dis, 7, e2094. Quan, G. M., & Choong, P. F. (2006). Anti-angiogenic therapy for osteosarcoma. Cancer Metastasis Rev, 25, 707-713. Rachner, T. D., Khosla, S., & Hofbauer, L. C. J. T. L. (2011). Osteoporosis: now and the future. Lancet,

377, 1276-1287. Rainusso, N., Brawley, V. S., Ghazi, A., Hicks, M. J., Gottschalk, S., Rosen, J. M., & Ah med, N. (2012). 74

Journal Pre-proof

Immunotherapy targeting HER2 with genetically mod ified T cells eliminates tumor-initiat ing cells in osteosarcoma. Cancer Gene Ther, 19, 212-217.

Reichert, J., Cipitria, A., Epari, D., Saifzadeh, S., Krishnakanth, P., Berner, A., Woodruff, M., Schell, H., Mehta, M., Schuetz, M., Duda, G., & Hutmacher, D. J. S. T. M. (2012). A tissue engineering solution for segmental defect regeneration in load-bearing long bones. Sci Transl

f

Med, 4, 141ra193.

oo

Reyes, C., Hitz, M., Prieto-Alhambra, D., & Abrahamsen, B. (2016). Risks and benefits of

pr

bisphosphonate therapies. J Cell Biochem, 117, 20-28.

e-

Riggs, B. L., & Hartmann, L. C. J. N. E. J. o. M. (2003). Selective estrogen-receptor

Pr

modulators—mechanisms of action and application to clinical practice. N Engl J Med, 348,

618-629.

al

Rigoglou, S., Papavassiliou, A. G. J. T. i. j. o. b., & biology, c. (2013). The NF-κB signalling pathway

rn

in osteoarthritis. Int J Biochem Cell Biol, 45, 2580-2584.

Jo u

Rochefort, G., Pallu, S., & Benhamou, C. J. O. I. (2010). Osteocyte: the unrecognized side of bone tissue. Osteoporosis Int, 21, 1457-1469. Rodan, G. A., & Martin, T. J. (2000). Therapeutic Approaches to Bone Diseases. Science, 289, 1508-1514. Rotman, S. G., Grijpma, D. W., Richards, R. G., Moriarty, T. F., Eglin, D., & Guillaume, O. (2018). Drug delivery systems functionalized with bone mineral seeking agents for bone targeted therapeutics. J Control Release, 269, 88-99.

75

Journal Pre-proof

Ryu, T. K., Kang, R. H., Jeong, K. Y., Jun, D. R., Koh, J. M., Kim, D., Bae, S. K., & Choi, S. W. (2016). Bone-targeted delivery of nanodiamond-based drug carriers conjugated with alendronate for potential osteoporosis treatment. J Control Release, 232, 152-160. Ryuichi Fujisawa, M. T. (2012). Acidic bone matrix proteins and their roles in calcification. Front Biosci, 17, 1891-1903.

f

Sánchez, A. R., Rogers III, R. S., & Sheridan, P. J. J. I. j. o. d. (2004). Tetracycline and other

oo

tetracycline‐derivative staining of the teeth and oral cavity. Int J Dermatol, 43, 709-715.

pr

Sartor, O., Reid, R. H., Hoskin, P. J., Quick, D. P., Ell, P. J., Coleman, R. E., Kotler, J. A., Freeman, L.

e-

M., Olivier, P., & Quadramet 424Sm10/11 Study, G. (2004). Samarium-153-Lexidronam

Pr

complex for treatment of painful bone metastases in hormone-refractory prostate cancer.

Urology, 63, 940-945.

al

Sekido, T., Sakura, N., Higashi, Y., Miya, K., Nitta, Y., Nomura, M., Sawanishi, H., Morito, K.,

rn

Masamune, Y., Kasugai, S., Yokogawa, K., & Miyamoto, K.-I. (2001). Novel drug delivery

Jo u

system to bone using acidic oligopeptide: pharmacokinetic characteristics and pharmacological potential. J Drug Targeting, 9, 111-121. Shang, Y., & Brown, M. (2002). Molecular determinants for the tissue specificity of SERMs. Science, 295, 2465-2468. Shea, J. E., & Miller, S. C. (2005). Skeletal function and structure: implications for tissue-targeted therapeutics. Adv Drug Deliv Rev, 57, 945-957. Shi, J., Sun, J., Zhang, W., Liang, H., Shi, Q., Li, X., Chen, Y., Zhuang, Y., Dai, J. J. A. a. m., &

interfaces. (2016). Demineralized bone matrix scaffolds modified by CBD-SDF-1α promote

76

Journal Pre-proof

bone regeneration via recruiting endogenous stem cells. ACS Appl Mater Interfaces, 8, 27511-27522. Siegel, R. L., Miller, K. D., & Jemal, A. (2019). Cancer statistics, 2019. CA Cancer J Clin, 69, 7-34. Silva, B. C., & Bilezikian, J. P. J. C. o. i. p. (2015). Parathyroid hormone: anabolic and catabolic actions on the skeleton. Curr Opin Pharmacol, 22, 41-50.

oo

function of the skeleton. Eur Spine J, 10 Suppl 2, S86-95.

f

Sommerfeldt, D. W., & Rubin, C. T. (2001). Biology of bone and how it orchestrates the form and

pr

Sousa, S., & Clezardin, P. (2018). Bone-targeted therapies in cancer-induced bone disease. Calcif

e-

Tissue Int, 102, 227-250.

Pr

Sui, B., Hu, C., Zhang, X., Zhao, P., He, T., Zhou, C., Qiu, X., Chen, N., Zhao, X., & Jin, Y. J. S. c. t. m. (2016). Allogeneic mesenchymal stem cell therapy promotes osteoblastogenesis and prevents

al

glucocorticoid‐induced osteoporosis. Stem Cells Transl Med, 5, 1238-1246.

rn

Sun, J., Zhao, Y., Li, Q., Chen, B., Hou, X., Xiao, Z., & Dai, J. (2016). Controlled release of

Jo u

collagen-binding SDF-1α improves cardiac function after myocardial infarction by recruiting endogenous stem cells. Sci Rep, 6, 26683. Sun, L. (2013). Modern chemistry and applications peptide-based drug development advantages and disadvantages. Mod Chem Appl, 1, 1-2. Sun, Y., Ye, X., Cai, M., Liu, X., Xiao, J., Zhang, C., Wang, Y., Yang, L., Liu, J., & Li, S. J. A. n. (2016). Osteoblast-targeting-peptide modified nanoparticle for siRNA/microRNA delivery. ACS Nano, 10, 5759-5768. Tang, F., Min, L., Seebacher, N. A., Li, X. Y., Zhou, Y. B., Hornicek, F. J., Wei, Y. Q., Tu, C. Q., & Duan, Z. F. (2019). Targeting mutant TP53 as a potential therapeutic strategy for the treat ment 77

Journal Pre-proof

of osteosarcoma. J Orthop Res, 37, 789-798.

Teitelbaum, S. L. (2000). Bone Resorption by Osteoclasts. Science, 289, 1504-1508. Tiram, G., Segal, E., Krivitsky, A., Shreberk-Hassidim, R., Ferber, S., Ofek, P., Udagawa, T., Edry, L., Shomron, N., Roniger, M., Kerem, B., Shaked, Y., Aviel-Ronen, S., Barshack, I., Calderon, M., Haag, R., & Satchi-Fainaro, R. (2016). Identification of dormancy-associated microRNAs

f

for the design of osteosarcoma-targeted dendritic polyglycerol nanopolyplexes. ACS Nano, 10,

oo

2028-2045.

pr

Tsushima, N., Yabuki, M., Harada, H., Katsumata, T., Kanamaru, H., Nakatsuka, I., Yamamoto, M., &

e-

Nakatsuka, M. (2000). Tissue distribution and pharmacological potential of sm‐16896, a

Pr

novel oestrogen‐bisphosphonate hybrid compound. J Pharm Pharmacol, 52, 27-37.

Tulasi, P. (2011). Monthly administration of a novel PTH-collagen binding domain fusion protein is

al

anabolic in mice. %J Calcified tissue international. Calcif Tissue Int, 6.

rn

Unger, J. M., Griffin, K., Donaldson, G. W., Baranowski, K. M., Good, M. J., Reburiano, E., Hussain,

Jo u

M., Monk, P. J., Van Veldhuizen, P. J., Carducci, M. A., Higano, C. S., Lara, P. N., Tangen, C. M., Quinn, D. I., Wade, J. L., III, Vogelzang, N. J., Thompson, I. M., Jr., & Moinpour, C. M. (2017). Patient-reported outcomes for patients with metastatic castration-resistant prostate cancer receiving docetaxel and Atrasentan versus docetaxel and placebo in a randomized phase III clinical trial (SWOG S0421). J Patient Rep Outcomes, 2, 27. Valentín, S., Morales, A., Sánchez, J. L., & Rivera, A. (2009). Safety and efficacy of doxycycline in the treatment of rosacea. Clin, Cosmet Invest Dermatol, 2, 129-140.

Vallet, M., & Ralston, S. H. (2016). Biology and treatment of Paget's disease of bone. J Cell Biochem, 117, 289-299. 78

Journal Pre-proof

Verdrengh, M., Bokarewa, M., Ohlsson, C., Stolina, M., & Tarkowski, A. (2010). RANKL-targeted therapy inhibits bone resorption in experimental Staphylococcus aureus-induced arthritis. Bone, 46, 752-758. Virk, M. S., & Lieberman, J. R. (2012). Biologic adjuvants for fracture healing. Arthritis Res Ther, 14, 225.

oo

replacement therapy. Fertil Steril, 37, 457-474.

f

Wallach, E. E., & Gambrell, R. D. (1982). The menopause: benefits and risks of estrogen-progestogen

pr

Wang, D., Miller, S. C., Kopeckova, P., & Kopecek, J. (2005). Bone-targeting macromolecular

e-

therapeutics. Adv Drug Deliv Rev, 57, 1049-1076.

Pr

Wang, D. G., Fan, J. B., Siao, C. J., Berno, A., Young, P., Sapolsky, R., Ghandour, G., Perkins, N., Winchester, E., Spencer, J., Kruglyak, L., Stein, L., Hsie, L., Topaloglou, T., Hubbell, E.,

al

Robinson, E., M ittmann, M., Morris, M. S., Shen, N., Kilburn, D., Riou x, J., Nusbaum, C.,

rn

Rozen, S., Hudson, T. J., Lipshutz, R., Chee, M., & Lander, E. S. (1998). Large -scale

Jo u

identification, mapping, and genotyping of single-nucleotide polymorphisms in the human genome. Science, 280, 1077-1082. Wang, H., Liu, J., Tao, S., Chai, G., Wang, J., Hu, F.-Q., & Yuan, H. J. I. j. o. n. (2015). Tetracycline-grafted PLGA nanoparticles as bone-targeting drug delivery system. Int J Nanomed, 10, 5671. Wang, J., Hu, J., & Zhang, S. (2010). Studies on the sorption of tetracycline onto clays and marine sediment from seawater. J Colloid Interface Sci, 349, 578-582.

Wang, J., Xu, G., Shen, F., & Kang, Y. (2014). miR-132 targeting cyclin E1 suppresses cell proliferation in osteosarcoma cells. Tumour Biol, 35, 4859-4865. 79

Journal Pre-proof

Watanabe, K. (2004). Collagenolytic proteases from bacteria. Appl Microbiol Biotechnol, 63, 520-526. Wein, M. N., & Kronenberg, H. M. J. C. S. H. p. i. m. (2018). Regulation of bone remodeling by parathyroid hormone. Cold Spring Harbor Perspect Med, 8, a031237. William M. Pierce, J. W. M. P., Jr. (1987). Bone-targeted carbonic anhydrase inhibitors: effect of a proinhibitor on bone resorption in vitro. Exp Biol Med, 186, 96-102.

f

William N, A. (2010). Phosphorylation-dependent mineral-type specificity for apatite-binding peptide

oo

sequences. Biomaterials, 36.

pr

Wu, C.-H., Liu, I.-J., Lu, R.-M., & Wu, H.-C. J. J. o. b. s. (2016). Advancement and applications of

e-

peptide phage display technology in biomedical science. J Biomed Sci, 23, 8.

Pr

Xian, L., Wu, X., Pang, L., Lou, M., Rosen, C. J., Qiu, T., Crane, J., Frassica, F., Zhang, L., & Rodriguez, J. P. J. N. m. (2012). Matrix IGF-1 maintains bone mass by activation of mTOR in

al

mesenchymal stem cells. Nat Med, 18, 1095.

rn

Xie, H., Chen, G., & Young, R. N. (2017). Design, synthesis, and pharmacokinetics of a bone-targeting

Jo u

dual-action prodrug for the treatment of osteoporosis. J Med Chem, 60, 7012-7028. Xie, L., Ji, T., & Guo, W. (2017). Anti-angiogenesis target therapy for advanced osteosarcoma (Review). Oncol Rep, 38, 625-636. Xu, L., Huang, S., Hou, Y., Liu, Y., Ni, M., Meng, F., Wang, K., Ru i, Y., Jiang, X., & Li, G. (2015). So x11-modified mesenchymal stem cells (MSCs) accelerate bone fracture healing: So x11 regulates differentiation and migration of MSCs. FASEB J, 29, 1143-1152. Yadav, M. C., Bottini, M., Cory, E., Bhattacharya, K., Kuss, P., Narisawa, S., Sah, R. L., Beck, L.,

Fadeel, B., Farquharson, C., & Millan, J. L. (2016). Skeletal mineralization deficits and

80

Journal Pre-proof

impaired biogenesis and function of chondrocyte-derived matrix vesicles in phospho1(-/-) and phospho1/pi t1 double-knockout mice. J Bone Miner Res, 31, 1275-1286. Yang, K., Miron, R., Bian, Z., & Zhang, Y. J. B. (2018). A bone-targeting drug-delivery system based on Semaphorin 3A gene therapy ameliorates bone loss in osteoporotic ovariectomized mice. Bone, 114, 40-49.

f

Yang, S., Wei, D., Wang, D., Ph imphilai, M., Krebsbach, P. H., Franceschi, R. T. J. J. o. B., & Research,

oo

M. (2003). In vitro and in vivo synergistic interactions between the Run x2/Cbfa1 transcription

pr

factor and bone morphogenetic protein-2 in stimulat ing osteoblast differentiation. J Bone

e-

Miner Res, 18, 705-715.

Pr

Yarbrough, D. K., Hagerman, E., Eckert, R., He, J., Choi, H., Cao, N., Le, K., Hedger, J., Qi, F.,

Anderson, M., Rutherford, B., Wu, B., Tetradis, S., & Shi, W. (2010). Specific binding and

al

mineralization of calcified surfaces by small peptides. Calcif Tissue Int, 86, 58-66. Chemistry:

rn

Yewle, J. N. (2012). Bifunctional bisphosphonates for delivering biomolecules to bone. In

Jo u

KENTUCKY UNIV LEXINGTON, 2012. Yokogawa, K., Miya, K., Sekido, T., Higashi, Y., Nomura, M., Fujisawa, R., Morito, K., Masamune, Y., Waki, Y., & Kasugai, S. J. E. (2001). Selective delivery of estradiol to bone by aspartic acid oligopeptide and its effects on ovariectomized mice. Endocrinology, 142, 1228-1233. Yonou, H., Kanomata, N., Goya, M., Kamijo, T., Yokose, T., Hasebe, T., Nagai, K., Hatano, T., Ogawa, Y., & Ochiai, A. (2003). Osteoprotegerin/osteoclastogenesis inhibitory factor decreases human prostate cancer burden in human adult bone implanted into nonobese diabetic/severe

combined immunodeficient mice. Cancer Res, 63, 2096-2102.

81

Journal Pre-proof

Zhang, G., Guo, B., Wu, H., Tang, T., Zhang, B.-T., Zheng, L., He, Y., Yang, Z., Pan, X., & Chow, H. J. N. m. (2012). A delivery system targeting bone formation surfaces to facilitate RNAi-based anabolic therapy. Nat Med, 18, 307. Zhang, J., & Yu, S.-H. J. M. T. (2016). Carbon dots: large-scale synthesis, sensing and bioimaging. Mater Today, 19, 382-393.

f

Zhang, L., Gray, K. P., Shaw, G., Evan, C., Francini, E., & Sweeney, C. (2018). Bone targeted therapy

oo

and skeletal related events in the era of modern therapies for castration resistant prostate

pr

cancer with bone metastases. J Clin Oncol, 36, 5049-5049.

e-

Zhao, C., Irie, N., Takada, Y., Shimoda, K., Miyamoto, T., Nishiwaki, T., Suda, T., & Matsuo, K.

Pr

(2006). Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis. Cell Metab, 4,

111-121.

al

Zhao, Y. P., Ye, W. L., Liu, D. Z., Cui, H., Cheng, Y., Liu, M., Zhang, B. L., Mei, Q. B., & Zhou, S. Y.

rn

(2017). Redox and pH dual sensitive bone targeting nanoparticles to treat breast cancer bone

Jo u

metastases and inhibit bone resorption. Nanoscale, 9, 6264-6277. Zhao, Z., Zhao, Y., Xie, C., Chen, C., Lin, D., Wang, S., Lin, D., Cui, X., Guo, Z., & Zhou, J. (2019). Dual-active targeting liposomes drug delivery system for bone metastatic breast cancer: Synthesis and biological evaluation. Chem Phys Lipids, 223, 104785. Zhou, J. Z., Riquelme, M. A., Gao, X., Ellies, L. G., Sun, L.-Z., & Jiang, J. X. J. O. (2015). Differential impact of adenosine nucleotides released by osteocytes on breast cancer growth and bone metastasis. Oncogene, 34, 1831.

Zhou, W., Hao, M., Du, X., Chen, K., Wang, G., & Yang, J. (2014). Advances in targeted therapy for osteosarcoma. Discov Med, 17, 301-307. 82

Journal Pre-proof

Zhu, J., Huo, Q., Xu, M., Yang, F., Li, Y., Shi, H., Niu, Y., & Liu, Y. (2018). Bortezomib-catechol conjugated prodrug micelles: combining bone targeting and aryl boronate-based pH-responsive drug release for cancer bone-metastasis therapy. Nanoscale, 10, 18387-18397. Zhuo, Z., Yu, Y., Wang, M., Li, J., Zhang, Z., Liu, J., Wu, X., Lu, A., Zhang, G., & Zhang, B. (2017). Recent advances in selex technology and aptamer applications in biomedicine. Int. J. Mol.

Jo u

rn

al

Pr

e-

pr

oo

f

Sci., 18, 2142.

83

Figure 1

Figure 2