HHS Public Access
Author manuscript
Author Manuscript
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Published in final edited form as:
Br J Pharmacol. 2021 May ; 178(9): 2008–2025. doi:10.1111/bph.15251.
Bisphosphonates for delivering drugs to bone
Shuting Sun1,*, Jianguo Tao2,*, Parish P. Sedghizadeh3,!, Philip Cherian1, Adam F. Junka4,
Esmat Sodagar3, Lianping Xing2, Robert K. Boeckman Jr.5, Venkatesan Srinivasan5,
Zhenqiang Yao2, Brendan F. Boyce2, Brea Lipe6, Jeffrey D. Neighbors1,7, R. Graham G.
Russell8,9, Charles E. McKenna10, Frank H. Ebetino1,5,!
1.BioVinc,
Pasadena, California, CA, USA
Author Manuscript
2.Department
of Pathology and Laboratory Medicine, University of Rochester Medical Center,
Rochester, NY, USA
3.Center
for Biofilms, Herman Ostrow School of Dentistry, University of Southern California, Los
Angeles, CA, USA
4.Department
of Pharmaceutical Microbiology and Parasitology, Medical University of Wroclaw;
Wroclaw Research Centre EIT, Wroclaw, Poland
5.Department
of Chemistry, University of Rochester, Rochester, NY, USA
6.Department
of Medicine, University of Rochester Medical Center, Rochester, NY, USA
7.Department
Author Manuscript
of Pharmacology and Medicine, Pennsylvania State University College of Medicine,
Hershey, PA, USA
8.The
Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology &
Musculoskeletal Sciences, University of Oxford
9.The
Mellanby Centre for Bone Research, Department of Oncology and Metabolism, University
of Sheffield, Sheffield, United Kingdom
10.Department
of Chemistry, University of Southern California, Los Angeles, California, USA
Abstract
Author Manuscript
Advances in the design of potential bone-selective drugs for the treatment of various bone-related
diseases are creating exciting new directions for multiple unmet medical needs. For bone-related
cancers, off-target/non-bone toxicities with current drugs represent a significant barrier to the
quality of life of affected patients. For bone infections and osteomyelitis, bacterial biofilms on
!
Correspondence: Frank H. Ebetino, BioVinc, 2265 E. Pasadena California 91107 513 532 4084,
[email protected]; Parish
P. Sedghizadeh, University of Southern California; 925 West 34th Street # 4110, Los Angeles, CA 90089; Ph: (213) 740-2704
[email protected].
*Co-first authors: Shuting Sun and Jianguo Tao are co-first authors.
Nomenclature of Targets and Ligands
Key protein targets and ligands in this article are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the
common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Harding et al., 2018), and are permanently archived in
the Concise Guide to PHARMACOLOGY 2019/20 (Alexander et al., 2019).
DISCLOSURE
S.S., P.C., and F.H.E are employees of BioVinc LLC; S.S., F.H.E., and C.E.M. are founders and shareholders of BioVinc LLC; P.P. and
J.D.N. are consultants to BioVinc LLC; RKB is a consultant for Novartis. The other authors do not have disclosures.
Sun et al.
Page 2
Author Manuscript
infected bones limit the efficacy of antibiotics because it is hard to access the bacteria with current
approaches. Promising new experimental approaches to therapy, based on bone-targeting of drugs,
have been used in animal models of these conditions and demonstrate improved efficacy and
safety. The success of these drug-design strategies bodes well for the development of therapies
with improved efficacy for the treatment of diseases affecting the skeleton.
Keywords
Bone targeting; multiple myeloma; osteomyelitis; biofilm; bisphosphonate; antibiotic; conjugate;
bone resorption
INTRODUCTION
Author Manuscript
Author Manuscript
The ultimate goal of drug design is to develop therapies that work directly on the tissues,
cells, and biochemical targets relevant to any specific disease, and do not affect the
biochemistry at any other non-diseased compartment of the body. Calcified tissues of
the skeleton are targeted with great specificity by the bisphosphonate drug class to treat
bone-related diseases with minimal side effects on soft tissues. This has been evident for
many years with the potent nitrogen-containing bisphosphonates for the treatment of bone
diseases, including osteoporosis and cancers that have metastasized to bone (Pazianas,
Cooper, Ebetino & Russell, 2010). Bisphosphonates are a group of compounds with the
general structure: (HO)2P(O)CR1R2P(O)(OH)2 (Figure 1A). Their strong bone-binding
affinity is predominantly determined by the two phosphonate groups (P-C-P), which form
strong bi- and tri-dentate interactions with calcium, to which the R1 and R2 groups on
the bridging carbon also contribute to a certain extent (Ebetino et al., 2011; Russell,
Watts, Ebetino & Rogers, 2008). Delivery of other drug classes specifically to bone by
linking them to bisphosphonates is an intriguing therapeutic approach that has not yet
found clinical utility (Cole, Vargo-Gogola & Roeder, 2016; Farrell, Karpeisky, Thamm &
Zinnen, 2018; Kempfle et al., 2018; Young & Grynpas, 2018). Although this topic has
been covered recently in general reviews (Cole, Vargo-Gogola & Roeder, 2016; Farrell,
Karpeisky, Thamm & Zinnen, 2018; McKenna, Haratipour, Duro & Ebetino, 2020; Xing
et al., 2020), in this paper we assess the success of this approach to two areas of bone
disease that continue to be unmet medical needs. Specifically, we review work in which
bisphosphonates are chemically linked to drugs used clinically to treat multiple myeloma
(Wang, Xiao et al., 2018; Wang et al., 2020) or bone infections (osteomyelitis) (Sedghizadeh
et al., 2017b) that have been used successfully as drug-releasing conjugates in animal
models of these diseases.
Author Manuscript
The ability of bisphosphonate-based pharmaceuticals to detect bone cancers using skeletal
scintigraphy has been recognized for many years. Technetium-labeled bisphosphonate
complexes have been used in clinical bone scanning for decades (Cole, Vargo-Gogola
& Roeder, 2016). Recent developments in the targeting of a nucleoside monophosphate
with etidronate have led to a clinical candidate for bone cancers (Zinnen, Karpeisky, Von
Hoff, Plekhova & Alexandrov, 2019). Our team at the University of Rochester has also
demonstrated enhanced efficacy and evidence of better off-target safety by combining
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 3
Author Manuscript
Author Manuscript
bortezomib with a pharmacologically inactive bisphosphonate for the treatment of multiple
myeloma in a mouse model (Wang, Cai, et al., 2018). Thus, bisphosphonate-based bone
targeting to create potent and selective therapies to treat bone neoplasms and other bone
diseases is receiving increasing attention. The challenges inherent in combining two drugs
into one molecule are also being identified and addressed. For example, it is clear that the
use of relatively inert bisphosphonates for this purpose reduces the chance of confounding
results in vivo, particularly on bone resorption. Additionally, an ideal linkage between the
bisphosphonate and the drug “warhead” being delivered to the bone surface should have a
slow rate of cleavage in plasma to allow delivery to the bone compartment, but must then
be labile once adsorbed to the bone surface to allow sufficiently rapid release of the drug to
achieve a therapeutically effective local concentration (Figure 1A). Certain properties of the
microenvironment in bone and osteoclast resorption lacunae, e.g., the generally acidic pH
environment, and some specific enzymes secreted by osteoclasts, the cells that degrade bone,
make selective cleavable linkage design possible.
1. Changes in Normal Bone Remodeling
Author Manuscript
The bone remodeling process is regulated by two major cells: osteoclasts mediate bone
resorption, while osteoblasts form bone. In a normal skeleton, it has been shown by
Fogelman and others with bisphosphonate-based technetium scanning that there is a
greater % of bone remodeling in the trabecular regions of bone and therefore greater
uptake of bisphosphonate because of the higher percentage of exposed mineral surface
and mineralizing osteoid matrix to which bisphosphonates bind (Fogelman, 1982). The
bisphosphonates coordinate and chemisorb exquisitely well with the calcium ions of
hydroxyapatite surfaces and either bone resorption or formation events lead to exposed
bone mineral. Higher uptake of bisphosphonates on the skeleton is therefore associated with
bone turnover. Abnormal bone remodeling is characterized by either an imbalance of the
resorption and formation cycle and/or by a more rampant rate of either or both processes.
In many diseases, such as Paget’s disease, or in the presence of osteomyelitis or bone
metastases, this excessive rate of remodeling can be observed focally at the site of lesions.
Thus, more remodeling leads to greater exposure of bone mineral (hydroxyapatite surface
area) and therefore greater uptake of bisphosphonates focally at diseased skeletal sites.
Multiple Myeloma
Author Manuscript
Multiple myeloma (MM) is a hematologic malignancy caused by the expansion of malignant
plasma cells within the bone marrow (BM), thereby crowding out the normal marrow
cells and leading to anemia and other cytopenias, and to high levels of monoclonal
proteins in serum and urine (Al-Farsi, 2013; El Arfani, De Veirman, Maes, De Bruyne
& Menu, 2018; Shain, Dalton & Tao, 2015). Myeloma cells are highly dependent on the
BM microenvironment for growth, survival and activation through interactions particularly
with BM stromal cells (Xu, De Veirman, De Becker, Vanderkerken & Van Riet, 2018),
osteoblasts (Kawano et al., 2015) and osteoclasts (Lawson et al., 2015). However, myeloma
cells also damage the BM microenvironment (Ghobrial, Detappe, Anderson & Steensma,
2018) involving multiple cell types (Al-Farsi, 2013; Mahindra, Hideshima & Anderson,
2010). In particular, MM cells inhibit osteoblastogenesis by secreting soluble factors,
such as Dkk1, sFRP2 and sclerostin, and suppressing Runx-Related Transcription Factor 2
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 4
Author Manuscript
(Runx2) activity through direct cell-to-cell contact with the involvement of VLA-4/VCAM-1
interaction. Concurrently, MM cells promote osteoclast function by releasing numerous
cytokines, such as RANKL (Giuliani & Rizzoli, 2007; Terpos, Ntanasis-Stathopoulos,
Gavriatopoulou & Dimopoulos, 2018). Thus, myeloma cells increase bone resorption
and decrease bone formation, inducing a severe imbalance in bone remodeling and
contributing to the development of myeloma-associated bone diseases, including focal
lytic lesions, pathological fractures, and hypercalcemia (Paton-Hough, Chantry & Lawson,
2015). Although recent advances in treatment have greatly improved patient outcomes, MM
remains largely incurable (Goldschmidt, Ashcroft, Szabo & Garderet, 2019).
Author Manuscript
Author Manuscript
Author Manuscript
1.1 Bortezomib—Bortezomib (Btz, Velcade®) is a drug used to treat patients with
MM and mantle cell lymphoma (Jin et al., 2018; Manasanch & Orlowski, 2017; Robak
et al., 2015), and its ability to treat other types of cancer is being tested in clinical
trials (clinicaltrials.gov, 2019). Btz is a very specific 26S proteasome inhibitor, which
reversibly but strongly binds the threonine proteases of the 20S subunit, enabling inhibition
of the hydrolyzing catalytic activities of the 26S proteasome (Painuly & Kumar, 2013).
Mechanistically, Btz directly inhibits proteasomal degradation of the large amounts of
immunoglobulin produced by myeloma cells. This allows the accumulation of these
useless/ineffective proteins, leading to endoplasmic reticulum stress, activation of the
unfolded protein response, and eventually to myeloma cell apoptosis (Brewer & Diehl,
2000; Manasanch & Orlowski, 2017; Obeng, Carlson, Gutman, Harrington, Lee & Boise,
2006; Zinszner et al., 1998). Btz may also inhibit NF-κB signaling by protecting IκBα
from degradation, thus preventing its pro-survival activity and promoting myeloma cell
apoptosis (Roy, Sarkar & Basak, 2018; Vrabel, Pour & Sevcikova, 2019). However, other
studies have reported that instead of inhibiting IκBα degradation, Btz can decrease the
expression of IκBα, resulting in NF-κB activation in various tumor cell lines, including
MM (Hideshima et al., 2009; Li et al., 2010). Btz also inhibits osteoclast formation by
modulating p38, activator protein-1, and NF-κB signaling pathways (von Metzler et al.,
2007; Zavrski et al., 2005), and promotes osteoblast differentiation by stabilizing the key
osteoblast transcription factor, Runx2. Therefore, Btz also suppresses bone resorption and
stimulates bone formation, which can ameliorate the abnormal bone homeostasis caused
by MM (Giuliani et al., 2007; Pennisi, Li, Ling, Khan, Zangari & Yaccoby, 2009). These
complementary functions of Btz on myeloma cells and bone cells place it as an ideal
drug for treating patients with MM. Btz is administrated intravenously or subcutaneously.
Similar to other anti-cancer drugs given by systemic administration, the off-target effects
of Btz on non-skeletal tissues, mainly peripheral neuropathy (Cavaletti et al., 2007) and
thrombocytopenia (Lonial et al., 2005; Murai et al., 2014; Shi et al., 2014), have restricted
the amounts of the drug that can be administered. More importantly, patients develop
inherent or acquired Btz resistance (BR), which limits its clinical efficacy (Dispenzieri,
Jacobus, Vesole, Callandar, Fonseca & Greipp, 2010; Kuhn et al., 2012; Murray, Auger &
Bowles, 2014; Richardson et al., 2003; Richardson et al., 2005; Ruschak, Slassi, Kay &
Schimmer, 2011). The development of BR involves multiple mechanisms, such as activation
of chemo-resistance pathways, immunoproteasomes, and mutations in proteasome subunits
(Niewerth, Jansen, Assaraf, Zweegman, Kaspers & Cloos, 2015). Agents targeting these
molecular pathways have been tested in preclinical studies using BR myeloma cell lines and
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 5
Author Manuscript
in a set of CD138+ cells obtained from BM of patients with relapsed/refractory myeloma
(Chauhan et al., 2010; Richardson et al., 2014; Zangari & Suva, 2016).
Author Manuscript
1.2 Autophagy and its inhibitors, Chloroquine and Hydroxychloroquine, in
treatment of multiple myeloma—Recent studies report that autophagy may play a role
in drug resistance in cancer patients (Sui et al., 2013) including MM (Abdel Malek et al.,
2015; Hamouda et al., 2014; Zhang et al., 2015). Autophagy is a lysosome-dependent
protein degradation process. Autophagy degrades unnecessary or dysfunctional cellular
components and releases nucleotides, amino acids or fatty acids from them (Yun et al.,
2017) as a source of energy to ensure cell survival under critical conditions, such as
hypoxia and starvation (Verbaanderd et al., 2017). However, extensive autophagy may also
lead to cell death (Tsujimoto & Shimizu, 2005). In myeloma cells, massive amounts of
immunoglobulin that are not degraded because of Btz-mediated proteasome inhibition are
proposed to serve as an energy source via autophagy, thereby exerting a pro-survival effect
under drug treatment (Attar-Schneider, Drucker, Zismanov, Tartakover-Matalon, Rashid
& Lishner, 2012; Auner & Cenci, 2015; Dykstra, Allen, Born, Tong & Holstein, 2015;
Hoang, Benavides, Shi, Frost & Lichtenstein, 2009; Zeng, Chen, Zhao & Cui, 2012). Thus,
inhibition of autophagy may kill drug-resistant myeloma cells by limiting energy supply.
Author Manuscript
Author Manuscript
Chloroquine (CQ) and Hydroxychloroquine (HCQ) are FDA-approved autophagy inhibitors
and have been used to treat patients with malarial infections (CQ), systemic lupus
erythematosus and rheumatoid arthritis (HCQ) for decades. Recent reports indicate that
both CQ and HCQ reduce bone resorption by inhibiting osteoclast formation (Both et al.,
2018; Xiu et al., 2014). They have been extensively studied both in vitro and in vivo
in various cancer types. Results show that CQ or HCQ effectively increase the efficacy
and limit drug resistance of standard anti-cancer therapies (Verbaanderd et al., 2017).
Recently, 4 clinical studies (ClinicalTrials.gov Identifier: NCT01438177; NCT00568880;
NCT01396200; NCT01689987 (Verbaanderd et al., 2017)) have investigated the use
of CQ or HCQ in combination with other anti-MM drugs in patients with relapsed/
refractory myeloma. A phase I clinical trial (NCT00568880) led by Dr. Dan Vogl (Vogl
et al., 2014) and a phase II trial (NCT01438177) led by Dr. Amitabha Mazumder
(Montanari, Lu, Marcus, Saran, Malankar & Mazumder, 2014) examined the effects of a
combination of HCQ and Btz in 25 cases with refractory or relapsed MM, or CQ, Btz,
and Cyclophosphamide in 11 similar patients. Both trials report promising results and
demonstrated that CQ or HCQ was able to partially restore Btz sensitivity (Montanari,
Lu, Marcus, Saran, Malankar & Mazumder, 2014; Vogl et al., 2014). However, some
serious adverse events, including hypoglycemia (Goyal & Bordia, 1995; Prabha, 1996), BM
suppression (Nagaratnam, Chetiyawardana & Rajiyah, 1978), cardiomyopathy (CostedoatChalumeau et al., 2007) and irreversible retinal toxicity (Browning, 2014) have been
associated with long-term treatment with CQ or HCQ. Some subjects suffered from the
toxic side effects of the combined regimen (Montanari, Lu, Marcus, Saran, Malankar &
Mazumder, 2014) that led some of them to stop therapy (Vogl et al., 2014). Low doses of
CQ or HCQ in combination with Btz and/or other drugs limited by the systemic adverse
effects have been proposed to be responsible for hindering achievement of more robust
clinical responses (Scott et al., 2017; Vogl et al., 2014).
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 6
Author Manuscript
Author Manuscript
Although some advances were achieved by using the above bone-targeted agents to treat
MM or metastatic bone tumors, the possibly reduced systemic adverse effects of these drugs,
especially the Btz derived compositions, have not been formally investigated. Furthermore,
most studies have used an antiresorptive BP, such as etidronate (Reinholz et al., 2010;
Zinnen, Rowinsky, Alexandrov, Plekhova, Roudas & Karpeisky, 2017) or alendronate
(Agyin, Santhamma & Roy, 2013; Swami et al., 2014; Zhu et al., 2018). With this approach,
it can be difficult to distinguish the pharmacological effect of the BP component from that
of released Btz. Recently, we linked Btz to a bisphosphonate (BP) (Figure 1B) that binds
avidly to bone, but is not antiresorptive, using a novel covalently bonded linker to generate a
BP-linked Btz (named BP-Btz) conjugate and demonstrated that BP-Btz, but not Btz, bound
to bone slices and inhibited the growth of myeloma cells in vitro. BP-Btz more effectively
reduced tumor burden and bone loss than Btz in the 5TGM1 mouse model of MM (Wang,
Author Manuscript
1.3 Preclinical studies using bone-targeted strategies to deliver Bortezomib
or other drugs to bone marrow in mouse models of MM—Given that MM
primarily originates in the BM and destroys bone, selective delivery of effective anticancer
drugs such as Btz specifically to bone has been a focus of myeloma research. Bone targeting
should reduce the toxic side effects arising from systemic distribution of Btz and make
dose escalation possible in order to improve therapeutic outcomes. In 2010, Reinholz et
al. linked the chemotherapeutic drug arabinocytidine-5′-phosphate to a bisphosphonate
(BP) drug, etidronate, and generated a series of bone-targeted anti-cancer compounds,
of which the leading candidate was named MBC-11. MBC-11 was designed to target
both osteoclasts (etidronate) and tumor cells (arabinocytidine-5′-phosphate), and it reduced
bone metastases and bone tumor burden in mice-bearing 4T1/luc mouse breast cancer
cells following mammary gland inoculation. It inhibited bone destruction and increased
overall survival in mice bearing KAS-6/1-MIP1α human myeloma cells following tail vein
injection (Reinholz et al., 2010). Subsequently, MBC-11 advanced to a phase I oncology
clinical trial (NCT02673060) in 2014 for patients with cancer-induced bone disease and
achieved partial efficacy (Zinnen, Rowinsky, Alexandrov, Plekhova, Roudas & Karpeisky,
2017; Zinnen, Karpeisky, Von Hoff, Plekhova & Alexandrov, 2019). In 2013, Agyin et
al. synthesized a series of BP (alendronate)-proteasome inhibitor conjugates and showed
that they had similar effects as their non-bone-targeted counterparts to kill MM cells in
vitro (Agyin, Santhamma & Roy, 2013). In 2014, Swami et al. engineered bone-homing
polymeric nanoparticles (NPs), BP (alendronate)-conjugated polymer PLGA-b-PEG-Ald, to
load Btz and demonstrated that the bone-targeted NPs could bind to bone fragments in vitro
and retain and accumulate in bone in vivo. Pretreatment of Btz loaded into bone-targeted
NPs enhanced survival and decreased tumor burden in mice bearing Luc+/GFP+ MM1S
human myeloma cells (Swami et al., 2014). In 2018, 2 studies reported the synthesis of
bone-targeted nanoparticles (NPs), loaded Btz with a pH-sensitive aryl boric acid ester
linkage that offers a pH-responsive drug release mechanism (Wang, Cai, et al., 2018; Zhu
et al., 2018). In both studies, in vitro release assays showed that only 20-30% of Btz was
released from the conjugated drugs in neutral buffer (pH 7.4), while under acidic conditions
of around pH 5.0, the release rate of Btz was faster, delivering as much as 60-70% of the
drug load. In mice with intra-tibial administration of MDA-MB-231 cells, bone-targeted Btz
significantly reduced tumor burden and bone destruction relative to free Btz-treated mice.
Author Manuscript
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 7
Author Manuscript
Xiao et al., 2018). In particular, we also demonstrated that BP-Btz generated significantly
less systemic adverse effects, such as thrombocytopenia (Wang, Xiao, et al., 2018) and
peripheral neuropathy (Wang et al., 2020), compared with Btz alone.
Author Manuscript
Author Manuscript
BPs bind preferentially, but not solely, to skeletal sites with high bone turnover (Russell,
Watts, Ebetino & Rogers, 2008). Thus, some BP-conjugates which might be delivered to
other skeletal sites could cause bone side effects. For example, BP-conjugates that utilize
BPs with significant antiresorptive function (Agyin, Santhamma & Roy, 2013; Reinholz
et al., 2010; Zinnen, Karpeisky, Von Hoff, Plekhova & Alexandrov, 2019) could in theory
exhibit adverse effects that have been associated with other bisphosphonates, including
osteonecrosis of the jaw, severe suppression of bone turnover, and atypical subtrochanteric
femoral fractures (Abrahamsen, 2010; Keller, 2014). Therefore, in our conjugate approach,
we designed BP moieties, which could bind avidly to the bone, but lack significant
antiresorptive activity, to limit any chance of side effects and to clarify the source of
pharmacological effect in our studies. Furthermore, even the effects of delivery to lower
bone turnover skeletal sites would likely be relatively minimal, since less turnover involving
osteoclast activity would likely lead to a slower release of the active warhead at those sites.
Regarding the free drug, Btz promotes osteoblast differentiation, and reduces osteoclast
formation (Giuliani et al., 2007; Uy et al., 2007), while chloroquine (Xiu et al., 2014) or
hydroxychloroquine (Both et al., 2018) reduces osteoclastogenesis, which are features that
may be beneficial to MM therapy, since a drastic loss of bone is observed in patients
with this cancer. Although 99m-technetium bone scans indicate a high preference for
bisphosphonates to localize at sites of higher turnover where their primary effects are likely
to occur, we do not currently have data to indicate how much BP or free drug will be
delivered to bone sites with lower bone turnover and what side effects this might lead to.
Isotope labeling and related pharmacokinetic and pharmacodynamic studies will help to
evaluate these important questions.
Author Manuscript
1.4 Effects of BP-Btz and BP-CQ or BP-HCQ on Bortezomib-resistant MM—
As mentioned above, previous studies suggest that Btz resistance might be attributed to
autophagy (Abdel Malek et al., 2015; Hamouda et al., 2014; Zhang et al., 2015), and
the autophagy inhibitors, CQ or HCQ, were able to improve sensitivity to Btz or other
anti-MM drugs to relapsed/refractory MM in clinical studies (Montanari, Lu, Marcus, Saran,
Malankar & Mazumder, 2014; Vogl et al., 2014). However, the efficacy was limited due to
several systemic adverse effects, hindering the use of higher doses of CQ or HCQ or Btz
(Vogl et al., 2014). In view of the findings that targeting Btz to bone successfully improved
its efficacy and ameliorated off-target adverse effects (Wang, Xiao et al., 2018), we also
generated a bone-targeted CQ (BP-CQ) and a bone-targeted HCQ (BP-HCQ) by conjugating
them to a bisphosphonate using linker chemistry similar to that for BP-Btz (Figure 1B)
(Boeckman, Boyce, Xiao, YAO & Ebetino, 2017; Xing et al., 2020; Yao et al., 2016). Our
rationale is that delivering Bortezomib or chloroquine conjugates to bone using a BP with
high bone affinity, but no significant antiresorptive effects, could result in high and persistent
local drug concentrations at sites of bone diseases, thereby killing more myeloma cells and
preventing bone loss, with fewer adverse effects.
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 8
Author Manuscript
Author Manuscript
Author Manuscript
In our initial study, we examined if BP-Btz plus BP-CQ or BP-HCQ could more effectively
kill BR myeloma cells than individual drug alone in vitro. We used a BR U266 human
myeloma cell line (Mitra et al., 2016) and found that they were resistant to both Btz and
BP-Btz to a similar extent (Figure 1C&D). Treatment of the BR U266 human myeloma
cell line with 6 nM Btz or 30 μM CQ for 72 hrs killed 40% of the cells, while Btz+CQ
killed 70%, and BP-Btz and BP-CQ had similar effects in vitro (Figure 1E&F). We also
demonstrated that the equimolar concentration of the BP, including the linker structure that
is likely to be generated on release of Btz, had no greater effect on myeloma cells than
on vehicle-treated cells (Figure 1F). We then tested the effects of BP-Btz plus BP-CQ on
primary myeloma cells that were isolated from BM of 3 patients with relapsed/refractory
MM (Figure 1G). We used the same doses of Btz (6 nM) and CQ (30 μM) for cells from
patient #1 and #2 and found that BP-Btz or BP-CQ alone killed 20-30% of myeloma cells,
while a combination of these killed 50%. To determine if increasing the dose of Btz or
CQ could achieve a better effect, we treated cells from patient #3 with high doses of Btz
(12 nM) and CQ (40 μM) and found that they had a similar killing effect as the lower
doses. We also examined the effects of BP-Btz and BP-CQ on CD138+ myeloma cells from
2 newly diagnosed MM patients. Cells from one patient responded well to Btz, with 6
nM killing 65%, while 30 μM BP-CQ killed 70%, and the combination killed 90% of the
patient’s myeloma cells (Figure 1H). However, cells from another patient responded poorly
to BP-Btz, with 6 nM BP-Btz killing 15%, while 20 μM BP-CQ or BP-HCQ killed 25%,
and the combination killed 50% of the patient’s myeloma cells (Figure 1H), suggesting that
myeloma cells from the second case may already be Btz-resistant. These data that have been
presented in several international conferences (Boyce, 2019; Tao et al., 2018) and discussed
in our recent review paper (Xing et al., 2020) suggest that combined BP-Btz and BP-CQ
or BP-HCQ therapy may overcome BR myeloma and represent a new treatment for relapsed/
refractory MM patients or newly diagnosed MM patients who are inherently BR.
Author Manuscript
Studies in the literature have estimated the local pH under the tight sealing zone of the
active osteoclast on the bone surface. Mechanistically, it is accepted that the pH in the
resorption lacunae is acidic to account for rapid dissolution of bone mineral mediated by the
cell. Silver et al (Silver, Murrills & Etherington, 1988) reported that the local pH under the
active osteoclast sealing zone is as low as 4.5. A more recent paper reported a pH range of
4-6 in the resorption pit (Kowada et al., 2011). Thus, we designed the linkage in BP-Btz
to be acid sensitive. The in vitro experiments were performed at physiological pH (around
7.2), and we did not observe a significant difference between the effect of free drug and
bone-targeted drug on inhibition of MM cell growth. Since free drug appears to be released
primarily at an acidic pH, we speculate that the free drug is released under the osteoclast
or within lysosomes or other cell organelles where the pH is low after the entire conjugate
enters the cell. These release mechanisms could be explored further using fluorescent- or
isotope-labeled BPs and BP-Btz.
1.5 Conclusion—Despite current clinical treatments for MM, including Btz, showing
remarkable efficacy, drug resistance and off-target effects have restricted the use of Btz
clinically. Several clinical studies have also shown some promising results of the autophagy
inhibitors, CQ and HCQ, to overcome Btz resistance. However, the side effects of CQ
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 9
Author Manuscript
or HCQ limit the use of more effective doses, hindering achievement of more robust
clinical responses. Development of novel bone-targeted therapeutic agents in recent years
has significantly improved efficacy in preclinical studies in the treatment of MM. Although
some critical information, such as drug release kinetics, possible synergies of BP-drug
payload interactions, mechanisms of action, tissue distribution, and toxicity, still need to be
gathered, the BP conjugates represent a promising therapeutic strategy.
2. Osteomyelitis
Author Manuscript
Osteomyelitis is a limb- and life-threatening infection of bone that is difficult to treat
clinically (Lew & Waldvogel, 2004). These infections are associated with fractures
(1.8-27%), with open fractures of the leg produced by high energy impact injury being most
common (incidence 27%) (Chen & Vallier, 2016; Pollak, Jones, Castillo, Bosse, MacKenzie
& Group, 2010). Other significant sources of these infections are joint replacement surgery
(arthroplasty) and secondary infection of the bone after cutaneous infections, which occur
for example in diabetic patients. As an example of the burden of this disease, the incidence
of osteomyelitis joint infection following total knee or hip replacement is 1.0-3.0% and
0.3-2.4% respectively (Kamath et al., 2015). Among patients who required knee joint
replacement or joint prosthesis revision, the primary reason was infection (Bozic et al.,
2015). Osteomyelitis is also a major reason for hip joint revision (Kamath et al., 2015).
When infection was the reason for joint revision surgery, the associated mortality was
reported to be 18% (Choi & Bedair, 2014; Choi, Beecher & Bedair, 2013).
Author Manuscript
Author Manuscript
2.1 Current antimicrobial treatment for osteomyelitis—A number of Grampositive and Gram-negative bacteria, as well as fungi and mycobacteria, can cause
osteomyelitis, but by far the most common organism implicated in bone and joint infections
is Staphylococcus aureus (SA), both methicillin-susceptible (MSSA) and methicillin–
resistant (MRSA) (Dubost et al., 2014; Ferrand et al., 2016). For jawbone infections,
Aggregatibacter actinomycetemcomitans (Aa) is a key organism (Stoodley et al., 2011).
The standard of care for bone and joint infections usually requires systemic administration
of antibiotics, typically vancomycin for MRSA and multidrug-resistant MRSA strains, and
fluoroquinolones (ciprofloxacin, moxifloxacin etc.) for gram-negative pathogens (Fraimow,
2009). For acute infections, intravenous antibiotics are generally prescribed for 2 – 6
weeks. Prolonged courses of oral antibiotics may follow for chronic infections, or infections
associated with retained implanted hardware. Both for acute and chronic infections, these
extended courses of therapy can lead to drug-related adverse events in a significant
percentage of patients – 15% in one estimate for a cohort treated for infections with MSSA.
This is particularly an issue with vancomycin where nephrotoxicity can occur in as many
as 43% of patients (Carreno, Kenney & Lomaestro, 2014). In general, these antibiotic
treatments have poor bone pharmacokinetics and limited osseous absorption in vivo (Fong,
Ledbetter, Vandenbroucke, Simbul & Rahm, 1986; Kim, Kim & Oh, 2014). Systemic
toxicity or adverse effects, and resistance, are clinical issues associated with prolonged
dosing regimens.
The inadequate efficacy of current antimicrobial treatments for osteomyelitis has been
ascribed to the limited access of systemically administered antibiotics to sites where
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 10
Author Manuscript
causative bacteria can reside as biofilms on bone surfaces, even surfaces deep within the
osteocytic canalicular network (de Mesy Bently et al., 2017). Sedghizadeh et al. recently
demonstrated that osteomyelitis pathogens can invade bone, establish chronic biofilms on
bone surfaces, and directly destroy and resorb bone without participation of host immunity
or osteoclastogenesis (Junka et al., 2017). These observations add to the difficulty in treating
osteomyelitis biofilms, and to the urgency of resolving an unmet medical need in this
indication. Therefore, any improvement in bone bioavailability of therapeutic antibiotics
would be a significant advance in treating osteomyelitis.
Author Manuscript
Author Manuscript
Local antibiotic delivery to treat bone and joint infections can be attempted at the time of
surgical debridement using antibiotic-impregnated beads or antibiotic-impregnated cement
(Uskokovic, 2015). However, currently marketed beads are not bio-absorbable; therefore,
a second procedure is required to remove them. They also tend to release antibiotics in
an initial burst pattern that quickly depletes the bulk of the drug from the carrier beads,
followed by a slow release of lower concentrations that may not be adequate to control
infection and may foster development of resistance. While there have been recent advances
in the use of bio-stimuli responsive release strategies (Lavrador, Gaspar & Mano, 2018;
Lin, Caldwell, Bhaduri, Goel & Agarwal, 2017), these concerns limit the usefulness of
this approach in the majority of bone and joint infections (Bozhkova, Novokshonova &
Konev, 2015; Uskokovic, 2015). In the realm of prosthetic joint infection (PJI), antibioticimpregnated cement is used commonly at the time of first debridement of an infected
implant to improve control of the infection. A recent study showed that this can be
efficacious in the control of the infection in two-stage hip revision surgeries (Staats et
al., 2017). Different cement substrates are available and several antibiotics are used in
these treatments, with vancomycin being the most common. Variable anti-bacterial loading
and cement characteristics can lead to changes in mechanical properties (Slane, Gietman
& Squire, 2018) of the cement implant as well as varying anti-microbial elution rates
(Boelch et al., 2018). Concerns about prolonged sub-therapeutic antibiotic concentrations
and selection of resistant organisms also apply to cement (Corona, Espinal, RodriguezPardo, Pigrau, Larrosa & Flores, 2014; Walker, Baker, Holleyman & Deehan, 2017).
Author Manuscript
2.2 Bone-targeted antimicrobial therapy for osteomyelitis—BP drugs have a
high affinity for bone and preferentially accumulate at sites of active bone disease or biofilm
infection, resorption, and remodeling (Cheong et al., 2014; Ebetino et al., 2011). BPs also
penetrate the canalicular network to osteocytes and osteocytic lacunae where no blood flow
exists and where S. aureus biofilm organisms are known to embed (de Mesy Bentley et al.,
2017). In addition, the BP class of drugs has a long track-record of clinical use and a good
safety profile relative to most drug classes, and have thus been utilized for pharmacotherapy
in several bone diseases, such as osteoporosis, multiple myeloma, metastatic cancer to bone,
and Paget’s disease of bone. To exploit this BP affinity for bone, we are investigating a
“target and release” chemistry approach for treating bone infections which involves delivery
of antibiotics to the hydroxyapatite (HA) mineral of the skeleton via conjugation to BPs,
utilizing serum-stable drug-BP linkers that metabolize and release the antibiotic at bone
surfaces and within the canalicular network. Here, we have also utilized pharmacologically
“inert” BPs for this purpose to avoid confounding antiresorptive effects from the carrier BP.
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 11
Author Manuscript
We recently reported impressive in vivo efficacy in animal studies with a novel
bisphosphonate-carbamate-ciprofloxacin (BCC) conjugate, BV600022 (Sedghizadeh et al.,
2017b) (Figure 2A). Quantitative determination of the colony forming units (CFU) of
bacteria from resected peri-prosthetic osteomyelitis tissue showed that a single dose of 10
mg/kg of conjugate gave ~2 log reduction of CFU or 99% bacterial killing efficacy and
nearly an order of magnitude greater activity than ciprofloxacin alone given in multiple
doses (10 mg/kg X 3) (Figure 2B). Furthermore, the effects of a single dose of 10 mg/kg
of conjugate in vivo were significantly greater (p=0.0005, unpaired t-test) than the untreated
control arm. Also, we synthesized and tested a non-cleavable (non-antibiotic releasing)
amide-linked BP conjugate (BAC) BV600026 (Figure 2A), which was found to be nearly
inactive against osteomyelitis biofilms grown on HA surfaces in vitro (Sedghizadeh et al.,
2017b).
Author Manuscript
Author Manuscript
Author Manuscript
Additional in vitro assays of S. aureus and Aa (jawbone) osteomyelitis pathogens in both
planktonic and biofilm assays have been performed with BCC treatment as well as with
a few other novel BP-antibiotic conjugates (Figure 2A). A new fluoroquinolone carbamate
conjugate, bisphosphonate-carbamate-moxifloxacin (BCX, BV600052), based on the later
generation and more potent moxifloxacin fluoroquinolone, was compared to BCC in these
assays (Sedghizadeh et al., 2017a; Sedghizadeh et al., 2019; Sedghizadeh et al., 2017b;
Sedghizadeh et al., 2020). In addition, some of the most important oxazolidinones are
currently used against gram-positive pathogens, including superbugs such as MRSA. These
antibiotics are considered clinically as choices of last resort when other antibiotics have
failed. We therefore also designed, synthesized, and tested two new BP-oxazolidinone
conjugates, including a novel BP-carbamate-tedizolid conjugate (BCT, BV600037), and
a related more rapidly cleavable BP-ester-tedizolid conjugate (BET, BV600039), for activity
against S. aureus. Tedizolid is a recent FDA-approved oxazolidinone antibiotic indicated for
the treatment of skin infections, particularly those caused by both methicillin-susceptible S.
aureus and methicillin-resistant S. aureus pathogens, which cause the majority of clinical
cases of skin infection (Ebetino, Sun, McKenna, Sadrerafi & Cherian, 2020). Since most
osteomyelitis cases are also caused by similar Staphylococcal species, the application of
tedizolid to bone infections could prove efficacious. An ester-linked conjugate is expected to
have more rapid linker release kinetics than a carbamate-linked conjugate, and we projected
it could be a good comparison for the identification of the optimal linkage. We also tested
various unconjugated antibiotics, including vancomycin (V) and minocycline (M), which
are used clinically in the setting of infectious bone disease, in addition to the parent drugs
of these conjugates (ciprofloxacin (C), moxifloxacin (X) and tedizolid (T), Figure 2A) for
direct comparison with our conjugates in preventing or eradicating planktonic and biofilm
osteomyelitis pathogens in vitro and ex vivo.
2.2.1 Antimicrobials tested.: The following antibiotics (Figure 2A) were tested:
minocycline (M), ciprofloxacin (C), moxifloxacin (X), tedizolid (T), and vancomycin (V);
the following experimental conjugates were tested: BCC, BCX, BCT and BET. These
conjugates were prepared as described in our published work (Ebetino, Sun, McKenna,
Sadrerafi & Cherian, 2020; Sedghizadeh et al., 2017b; Sedghizadeh et al., 2020). All
antimicrobials were tested initially in standard susceptibility studies with the protocol, as
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 12
Author Manuscript
detailed in the reference (Sedghizadeh et al., 2017b). Based on the results and efficacy data
from these initial studies, various compounds were chosen for further testing in biofilm
preventative and eradication experiments using HA.
Author Manuscript
2.2.2 Antimicrobial susceptibility testing.: All tested microbial strains (S. aureus and
Aa) were screened for their sensitivity to the antibiotics and conjugates listed above
per standard microdilution methods described in EUCAST guidelines and detailed in
reference (Sedghizadeh et al., 2017b). Minimum inhibitory concentrations (MIC90) were
calculated and refer to a specific concentration of antimicrobial where 90% of bacterial
growth was inhibited. For biofilm experiments, the MBEC90 (minimum biofilm eradication
concentration), defined as the minimum concentration necessary to eradicate 90% of already
formed biofilm, was calculated instead of the MIC90. The MIC90 and MBEC90 results for
the conjugates (BCC, BCX, BCT, BET) were calculated based on the amount of their
parent antibiotic to allow molar comparison with the parent drug. MIC90 and MBEC90 of
tested antibiotics and conjugates against S. aureus and Aa strains is presented in Table 1.
Author Manuscript
2.2.3 Affinity of tested compounds to HA (Ebetino, Sun, McKenna, Sadrerafi &
Cherian, 2020; Sedghizadeh et al., 2020).: Having established reference values for MIC90
and MBEC90, we evaluated the relative binding affinity of tested antibiotics and conjugates
for HA powder. 1 μg/mL of each compound was added to an aqueous solution containing
10 μg/mL of HA powder (pH 6.7 - 6.9) and incubated for 4h/37°C under magnetic stirring.
Next, HA powder was allowed to sediment for 1h/4°C, and the unbound antibiotic in
the supernatant was measured using HPLC (Sedghizadeh et al., 2017b). 1 μg/mL of each
compound incubated at the same condition, but without HA, served as a control sample.
Affinity of compounds for HA powder was calculated as follows: 100% - peak area of tested
compound detected/peak area of control sample *100%. The results obtained were expressed
as [%] bound and were as follows: BET=92.7 ≈ BCT=92.1 ≈ BCX=91.8 ≈ BCC=91.2 >
M=49.1 > X=40.3 ≈ C=36.2 > V=28.2 > T=23.6. The highest affinity for HA was seen
with these BP-antibiotic conjugates as compared to the non-conjugated or parent antibiotics
alone. Of the individual non-conjugated antibiotics, minocycline demonstrated the highest
affinity for HA. Tedizolid demonstrated the lowest affinity for HA, but when conjugated
with BP also demonstrated very high affinity.
Author Manuscript
2.2.4 Infection prevention experiment with HA.: To assess the ability of the conjugates
and antibiotics to prevent bone infection in an in vitro model, we saturated HA spherules
with concentrations of compounds equal to the MIC90 for each pathogen, removed the
non-adherent compounds by three cycles of centrifugation-rinsing-decanting, and then
introduced a solution containing pathogens to this experimental setting (Sedghizadeh et
al., 2017b). Results are expressed as Planktonic Survival Rate [%] for the MIC90 values
of compounds tested against S. aureus and Aa, as shown in Figure 3 (A-B). In this HA
experimental setting against S. aureus, all of the BP conjugates (BCC, BET, BCX), but
none of the non-conjugated/parent antibiotics, demonstrated statistically significant (K-W
test, p<0.05) efficacy in preventing bacterial growth when compared to controls. Against
Aa, only the BCX prevented bacterial growth at a statistically significant level (K-W test,
p<0.05) compared to controls.
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 13
Author Manuscript
Author Manuscript
2.2.5 Biofilm eradication experiment with HA.: In this experimental setting, we looked
for differences in the ability of the conjugates and non-BP-conjugated antibiotics to
penetrate through established biofilms and the underlying matrix to inhibit microbial cells.
5ml containing 105 CFU/mL of each tested pathogen was introduced to HA pellets and the
samples were subjected to incubation for 24h/37°C under magnetic stirring. Next, specific
concentrations of tested compounds were added to samples (n=10 for each pathogen)
in a manner that their final concentration was equivalent to the MBEC90 for biofilm
forms of pathogens, as determined above. The subsequent procedures of incubation (5h/
37°C), centrifugation, and spectrometric analysis utilizing a TTC assay were performed, as
described elsewhere (Dydak et al., 2018). Results are expressed as Biofilm Survival Rate
[%] for the MBEC90 values of compounds tested against S. aureus and Aa as shown in
Figure 3 (C-D). In this HA experimental setting, several antibiotics and conjugates (BCC,
BET, BCX) demonstrated statistically significant (K-W test, p<0.05) efficacy in eradicating
established S. aureus biofilms compared to the controls, with the greatest relative efficacy
seen with the conjugates compared to the non-conjugated antibiotics. Against Aa, only
the BCX demonstrated statistically significant (K-W test, p<0.05) efficacy in eradicating
biofilms when compared to controls.
Author Manuscript
Author Manuscript
2.2.6 Ex vivo bone biofilm experiment.: Prevention and eradication experiments utilizing
bones (rat femurs and jaws) were performed in a manner analogous to those where HA
was used, as described in the sections above for S. aureus and Aa, and again the survival
rate of pathogens subjected to antimicrobials was calculated. The only differences were that
no magnetic stirring was applied and experiments were carried out in 6-well plates. The
key considerations of this experiment are: 1) The number of untreated cells (not exposed to
antimicrobials) was considered 100%; survival rate was calculated as follows: the number
of treated cells/number of untreated cells*100%; 2) Since the process of bone cleansing
could introduce bacterial/fungal contamination, sterilization was required before adding the
microbial inoculum to bone. To confirm efficacy of sterilization, testing of bone sterility
was performed (as described in Figure 4 caption); 3) After cleansing and sterilization,
bones were kept at −80°C in sterile containers to prevent their decomposition and microbial
contamination. Bones were taken out of −80°C immediately before all procedures related to
biofilm culturing; 4) Bones were used as a surface for biofilm growth (culture S. aureus on
femoral bones and Aa on jawbones). Since use of bone samples with high variance in weight
could lead to non-comparable results with regard to the number of biofilm-forming colonies,
we decided to use only those bone samples that did not differ significantly and established
the weight of bone as a basic criterion for similarity. The average weight of cleansed rat
femora and jaws was 0.1901g and 0.3213g, respectively. For further experiments, only these
bones for which the weight differed by less than 10% were used. However, it should be
noted that due to differences in bone shapes and weights in individual rat anthropometrics,
results obtained in this section only serve as a proof of principle of higher affinity to
bone and antimicrobial activity of the conjugates in the bone environment in comparison to
commonly used antibiotics. These findings are consistent with our in vitro results presented
herein. S. aureus was chosen for experiments performed on long bones as this is the most
common long bone osteomyelitis pathogen, and Aa was chosen for experiments performed
on rat jaw since this is a common jawbone osteomyelitis pathogen. Results of preventative
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 14
Author Manuscript
and eradication experiments on ex vivo bones are shown in Figure 4. In S. aureus long
bone biofilm prevention and eradication settings ex vivo, the greatest efficacy was seen with
the BET in both settings and tedizolid was also highly effective at eradicating established
biofilms. In Aa jawbone biofilm prevention and eradication settings ex vivo, significant
efficacy was only seen with the BCC in both settings (K-W test, p<0.05).
Author Manuscript
Author Manuscript
2.2.7 Discussion.: In standard antimicrobial susceptibility testing (planktonic cultures and
biofilm cultures grown on polystyrene), we found that each tested antibiotic or parent
drug was more efficacious than the conjugates BCC, BCX, BCT and BET against long
bone (S. aureus) and jawbone (Aa) osteomyelitis pathogens, as supported by MIC90 and
MBEC90 data (Table 1). However, when HA was used instead of polystyrene to grow
the same planktonic pathogens or biofilms, the tested conjugates were more efficacious
in both infection prevention and treatment experiments (Figure 3). Similar results were
also observed from the ex vivo experiments using rat femoral and jaw bones (Figure 4).
This is most likely due to the stronger bone affinity of the conjugates, and thus higher
local concentrations of antibiotic in the infected bone compartment compared to non-BPconjugated antibiotics, supported by the HA affinity assays (section 2.2.3). Since BPs
are known to target high turnover sites of the skeleton, these concentrations are further
magnified under biofilms where bone resorption is known to occur. This bone binding and
retention with sustained release of antibiotic over time confers advantages to the conjugates
in this setting as compared to antibiotics without BP conjugation. Of the non-BP-conjugated
antibiotics, minocycline demonstrated the greatest affinity to HA, which is consistent with
the known relatively high bone affinity of the tetracycline class in general, as compared
to other antibiotic classes. These findings lend support to the common clinical practice in
dentistry of using minocycline, often topically, for treatment of jawbone infections, such as
periodontitis, osteomyelitis, and osteonecrosis (Karasneh, Al-Eryani, Clark & Sedghizadeh,
2016).
Author Manuscript
The mechanism of action of individual antibiotics, such as those tested here, is well-known
(Aldred, Kerns & Osheroff, 2014; Kisgen, Mansour, Unger & Childs, 2014). However, the
exact mechanisms of activity for the novel conjugates remain to be elucidated. Conjugation
is a chemical modification that can alter the biochemical interactions of the antibiotic prior
to release from the BP moiety. As a result, properties of the parent drug, including its
pharmacodynamics, can be altered by such modification until released. It has been shown
in previous studies that conjugates in this class can retain the antibiotic activity of the
parent drug (Herczegh et al., 2002), albeit at lower levels, which we also confirmed here.
However, our non-cleavable BAC analog did not show any significant activity against
osteomyelitis biofilms grown on HA surfaces in vitro (Sedghizadeh et al., 2017b) (Table
1). Also, as discussed above, a single dose of BCC was more efficacious than multiple
doses of ciprofloxacin in vivo (Figure 2B). Together with the additional in vitro and ex
vivo preventative/eradication experiments discussed above (Figure 3, 4), it is likely that
release of antibiotic at the infected sites occurs, and these conjugates have a likely depot
effect providing a longer term release of antibiotic at infected sites. Bacteria at those sites
should therefore be subjected to a relatively higher and sustained concentration of the
antibiotic. We are making progress at magnifying this effect, as we have demonstrated
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 15
Author Manuscript
enhanced efficacy with conjugates, based on more potent fluoroquinolones. Indeed, the in
vitro HA biofilm prevention and eradication results presented herein indicated that the most
efficacious conjugate (BCX) against both S. aureus and Aa pathogens is also derived from
the more potent fluoroquinolones tested (Table 1, Figure 3). In addition, the release rate
is also a critical feature, as faster release could reduce the depot benefit, but slow release
could reduce the critical local concentration at any given time point. We have observed in the
MIC90 assays against S. aureus on polystyrene (Table 1) that an ester-linked BP-antibiotic
conjugate (BET) leads to greater in vitro antimicrobial activity than a carbamate-linked
conjugate (BCT), presumably due to faster release kinetics associated with ester hydrolysis
as compared to carbamate linkages. Whether this will translate to greater potency in vivo
remains to be studied.
Author Manuscript
These recent data show that this class of drug-releasing conjugates, incorporating osteoadsorptive BPs with high bone affinity and fluoroquinolone or oxazolidinone antibiotics
for bone-targeted delivery to treat osteomyelitis biofilm pathogens, constitutes an effective
and promising approach to providing higher antimicrobial potency than unconjugated
antimicrobial agents at high turnover bone sites, while minimizing systemic exposure
and toxicity. In addition, bone affinity, antibiotic potency, and conjugation schemes are
important for antimicrobial efficacy against osteomyelitis pathogens because they impact
bone binding, antibiotic activity and release. These findings will aid the chemistry
optimization for antimicrobial effects in future iterations of conjugates in this class and
offer bisphosphonates and useful linkages for use in the design of new selective therapies for
other bone-related diseases.
SUMMARY
Author Manuscript
An ideal therapeutic drug for bone diseases should act only on bone tissue with no
pharmacological activity at other anatomical sites, as a further refinement of the “magic
bullet” concept formulated by Ehrlich in 1900 (Tan & Grimes, 2010). Even more ideal is
a drug that targets the most diseased sites of the skeleton, which is an attribute that BPs
provide with their propensity to favor higher bone turnover sites. Research into bone-seeking
medicinal agents, such as the work presented here is progressively laying the foundation for
next-generation BP “target and release” type 'magic bullets' that minimize systemic exposure
or toxicity and maximize drug efficacy at the targeted site.
ACKNOWLEDGEMENTS
Author Manuscript
This work was supported by grants (R41DE025789, R42DE025789, R43AI125060 and R43AR073727,
R21AR069789, R21AR070984, R01AR063650) from the National Institutes of Health (NIDCR, NIAID, NIAMS).
ABBREVIATIONS
(Aa)
Aggregatibacter actinomycetemcomitans
(BP)
Bisphosphonate
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 16
Author Manuscript
(BM)
Bone marrow
(Btz)
Bortezomib
(BR)
Bortezomib resistance
(BHI)
Brain-Heart Infusion
(CCK8)
Cell counting kit-8
Author Manuscript
(CQ)
Chloroquine
(C)
Ciprofloxacin
(EUCAST)
European Committee on Antimicrobial Susceptibility Testing
(HA)
Hydroxyapatite
Author Manuscript
(HCQ)
Hydroxychloroquine
(MSSA)
Methicillin-susceptible Staphylococcus aureus
(MRSA)
Methicillin-resistant Staphylococcus aureus
(MIC90)
Minimum inhibitory concentrations where 90% of bacterial growth was inhibited
Author Manuscript
(MBEC90)
Minimum biofilm eradication concentration necessary to eradicate 90% of already formed
biofilm
(M)
Minocycline
(X)
Moxifloxacin
(MM)
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 17
Multiple myeloma
Author Manuscript
(MLC1,2)
Myosin light chain 1,2
(NPs)
Nano particles
(PJI)
Prosthetic joint infection
(Runx2)
Runt Related Transcription Factor 2
Author Manuscript
(T)
Tedizolid
(V)
Vancomycin
REFERENCES
Author Manuscript
Author Manuscript
Abdel Malek MA, Jagannathan S, Malek E, Sayed DM, Elgammal SA, Abd El-Azeem HG, et al.
(2015). Molecular chaperone GRP78 enhances aggresome delivery to autophagosomes to promote
drug resistance in multiple myeloma. Oncotarget 6: 3098–3110. doi: 10.18632/oncotarget.3075
[PubMed: 25605012]
Abrahamsen B (2010). Adverse effects of bisphosphonates. Calcif Tissue Int 86: 421–435. doi:
10.1007/s00223-010-9364-1 [PubMed: 20407762]
Agyin JK, Santhamma B, & Roy SS (2013). Design, synthesis, and biological evaluation of bonetargeted proteasome inhibitors for multiple myeloma. Bioorg Med Chem Lett 23: 6455–6458. doi:
10.1016/j.bmcl.2013.09.043 [PubMed: 24119559]
Al-Farsi K (2013). Multiple Myeloma: An Update. Oman Medical Journal 28. doi: 10.5001/
omj.2013.02
Aldred KJ, Kerns RJ, & Osheroff N (2014). Mechanism of quinolone action and resistance.
Biochemistry 53: 1565–1574. doi: 10.1021/bi5000564 [PubMed: 24576155]
Alexander SPH, Fabbro D, Kelly E, Mathie A, Peters JA, Veale EL, et al. (2019). THE CONCISE
GUIDE TO PHARMACOLOGY 2019/20: Enzymes. Br J Pharmacol 176 Suppl 1: S297–S396.
[PubMed: 31710714]
Attar-Schneider O, Drucker L, Zismanov V, Tartakover-Matalon S, Rashid G, & Lishner M (2012).
Bevacizumab attenuates major signaling cascades and eIF4E translation initiation factor in multiple
myeloma cells. Lab Invest 92: 178–190. doi: 10.1038/labinvest.2011.162 [PubMed: 22083671]
Auner HW, & Cenci S (2015). Recent advances and future directions in targeting the secretory
apparatus in multiple myeloma. Br J Haematol 168: 14–25. doi: 10.1111/bjh.13172 [PubMed:
25296649]
Boeckman RK Jr., Boyce BF, Xiao L, YAO Z, & Ebetino FH (2017). Phosphonate-chloroquine
conjugates and methods using same.
Boelch SP, Rueckl K, Fuchs C, Jordan M, Knauer M, Steinert A, et al. (2018). Comparison of
Elution Characteristics and Compressive Strength of Biantibiotic-Loaded PMMA Bone Cement
for Spacers: Copal(R) Spacem with Gentamicin and Vancomycin versus Palacos(R) R+G with
Vancomycin. Biomed Res Int 2018: 4323518. doi: 10.1155/2018/4323518 [PubMed: 30410931]
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 18
Author Manuscript
Author Manuscript
Author Manuscript
Author Manuscript
Both T, Zillikens MC, Schreuders-Koedam M, Vis M, Lam WK, Weel A, et al. (2018).
Hydroxychloroquine affects bone resorption both in vitro and in vivo. J Cell Physiol 233: 1424–
1433. doi: 10.1002/jcp.26028 [PubMed: 28556961]
Boyce BF (2019). Delivery of chloroquine and anti-cancer agents to bone. JBMR plus 3: 14. [PubMed:
30680359]
Bozhkova SA, Novokshonova AA, & Konev VA (2015). Current trends in local antibacterial
therapy of periprosthetic infection and osteomyelitis. Travmatol Ortop Ross: 92–107. doi:
10.21823/2311-2905-2015-0-3-92-107
Bozic KJ, Kamath AF, Ong K, Lau E, Kurtz S, Chan V, et al. (2015). Comparative Epidemiology
of Revision Arthroplasty: Failed THA Poses Greater Clinical and Economic Burdens Than
Failed TKA. Clin Orthop Relat Res 473: 2131–2138. doi: 10.1007/s11999-014-4078-8 [PubMed:
25467789]
Brewer JW, & Diehl JA (2000). PERK mediates cell-cycle exit during the mammalian unfolded protein
response. Proc Natl Acad Sci U S A 97: 12625–12630. doi: 10.1073/pnas.220247197 [PubMed:
11035797]
Browning DJ (2014). Pharmacology of Chloroquine and Hydroxychloroquine. In Hydroxychloroquine
and Chloroquine Retinopathy. ed Browning DJ Springer New York: New York, NY, pp 35–63,
DOI: 10.1007/978-1-4939-0597-3_2.
Carreno JJ, Kenney RM, & Lomaestro B (2014). Vancomycin-associated renal dysfunction: where are
we now? Pharmacotherapy 34: 1259–1268. doi: 10.1002/phar.1488 [PubMed: 25220436]
Cavaletti G, Gilardini A, Canta A, Rigamonti L, Rodriguez-Menendez V, Ceresa C, et al. (2007).
Bortezomib-induced peripheral neurotoxicity: a neurophysiological and pathological study in the
rat. Exp Neurol 204: 317–325. doi: 10.1016/j.expneurol.2006.11.010 [PubMed: 17214983]
Chauhan D, Singh AV, Aujay M, Kirk CJ, Bandi M, Ciccarelli B, et al. (2010). A novel orally active
proteasome inhibitor ONX 0912 triggers in vitro and in vivo cytotoxicity in multiple myeloma.
Blood 116: 4906–4915. doi: 10.1182/blood-2010-04-276626 [PubMed: 20805366]
Chen AT, & Vallier HA (2016). Noncontiguous and open fractures of the lower extremity:
Epidemiology, complications, and unplanned procedures. Injury 47: 742–747. doi: 10.1016/
j.injury.2015.12.013 [PubMed: 26776462]
Cheong S, Sun S, Kang B, Bezouglaia O, Elashoff D, McKenna CE, et al. (2014). Bisphosphonate
uptake in areas of tooth extraction or periapical disease. J Oral Maxillofac Surg 72: 2461–2468.
doi: 10.1016/j.joms.2014.07.004 [PubMed: 25262401]
Choi HR, & Bedair H (2014). Mortality following revision total knee arthroplasty: a matched
cohort study of septic versus aseptic revisions. J Arthroplasty 29: 1216–1218. doi: 10.1016/
j.arth.2013.11.026 [PubMed: 24405619]
Choi HR, Beecher B, & Bedair H (2013). Mortality after septic versus aseptic revision total hip
arthroplasty: a matched-cohort study. J Arthroplasty 28: 56–58. doi: 10.1016/j.arth.2013.02.041
Bortezomib clinicaltrials. [Online] Available from https://clinicaltrials.gov/ct2/results/browse?
term=bortezomib&brwse=cond_alpha_all. [Accessed].
Cole LE, Vargo-Gogola T, & Roeder RK (2016). Targeted delivery to bone and mineral deposits
using bisphosphonate ligands. Adv Drug Deliv Rev 99: 12–27. doi: 10.1016/j.addr.2015.10.005
[PubMed: 26482186]
Corona PS, Espinal L, Rodriguez-Pardo D, Pigrau C, Larrosa N, & Flores X (2014). Antibiotic
susceptibility in gram-positive chronic joint arthroplasty infections: increased aminoglycoside
resistance rate in patients with prior aminoglycoside-impregnated cement spacer use. J
Arthroplasty 29: 1617–1621. doi: 10.1016/j.arth.2014.03.029 [PubMed: 24798194]
Costedoat-Chalumeau N, Hulot JS, Amoura Z, Delcourt A, Maisonobe T, Dorent R, et al. (2007).
Cardiomyopathy related to antimalarial therapy with illustrative case report. Cardiology 107: 73–
80. doi: 10.1159/000094079 [PubMed: 16804295]
de Mesy Bentley KL, Trombetta R, Nishitani K, Bello-Irizarry SN, Ninomiya M, Zhang L, et
al. (2017). Evidence of Staphylococcus Aureus Deformation, Proliferation, and Migration in
Canaliculi of Live Cortical Bone in Murine Models of Osteomyelitis. J Bone Miner Res 32: 985–
990. doi: 10.1002/jbmr.3055 [PubMed: 27933662]
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 19
Author Manuscript
Author Manuscript
Author Manuscript
Author Manuscript
Dispenzieri A, Jacobus S, Vesole DH, Callandar N, Fonseca R, & Greipp PR (2010). Primary therapy
with single agent bortezomib as induction, maintenance and re-induction in patients with high-risk
myeloma: results of the ECOG E2A02 trial. Leukemia 24: 1406–1411. doi: 10.1038/leu.2010.129
[PubMed: 20535147]
Dubost JJ, Couderc M, Tatar Z, Tournadre A, Lopez J, Mathieu S, et al. (2014). Three-decade trends
in the distribution of organisms causing septic arthritis in native joints: single-center study of 374
cases. Joint Bone Spine 81: 438–440. doi: 10.1016/j.jbspin.2014.05.001 [PubMed: 24956987]
Dydak K, Junka A, Szymczyk P, Chodaczek G, Toporkiewicz M, Fijalkowski K, et al.
(2018). Development and biological evaluation of Ti6Al7Nb scaffold implants coated with
gentamycin-saturated bacterial cellulose biomaterial. PLoS One 13: e0205205. doi: 10.1371/
journal.pone.0205205 [PubMed: 30356274]
Dykstra KM, Allen C, Born EJ, Tong H, & Holstein SA (2015). Mechanisms for autophagy
modulation by isoprenoid biosynthetic pathway inhibitors in multiple myeloma cells. Oncotarget
6: 41535–41549. doi: 10.18632/oncotarget.6365 [PubMed: 26595805]
Ebetino FH, Hogan AM, Sun S, Tsoumpra MK, Duan X, Triffitt JT, et al. (2011). The relationship
between the chemistry and biological activity of the bisphosphonates. Bone 49: 20–33. doi:
10.1016/j.bone.2011.03.774 [PubMed: 21497677]
Ebetino FH, Sun S, McKenna CE, Sadrerafi K, & Cherian P (2020). Bone targeted
antimicrobial oxazolidinone related compounds, formulations thereof, and uses thereof BioVinc
LLC, p 112pp. WO2020005964A1, https://patents.google.com/patent/WO2020005964A1/en?
oq=WO2020005964A1
El Arfani C, De Veirman K, Maes K, De Bruyne E, & Menu E (2018). Metabolic Features of Multiple
Myeloma. Int J Mol Sci 19. doi: 10.3390/ijms19041200
Farrell KB, Karpeisky A, Thamm DH, & Zinnen S (2018). Bisphosphonate conjugation for bone
specific drug targeting. Bone Rep 9: 47–60. doi: 10.1016/j.bonr.2018.06.007 [PubMed: 29992180]
Ferrand J, El Samad Y, Brunschweiler B, Grados F, Dehamchia-Rehailia N, Sejourne A, et al. (2016).
Morbimortality in adult patients with septic arthritis: a three-year hospital-based study. BMC
Infect Dis 16: 239. doi: 10.1186/s12879-016-1540-0 [PubMed: 27246346]
Fogelman I (1982). Diphosphonate bone scanning agents--current concepts. Eur J Nucl Med 7: 506–
509. doi: 10.1007/bf00257217 [PubMed: 6217074]
Fong IW, Ledbetter WH, Vandenbroucke AC, Simbul M, & Rahm V (1986). Ciprofloxacin
concentrations in bone and muscle after oral dosing. Antimicrob Agents Chemother 29: 405–408.
doi: 10.1128/AAC.29.3.405. [PubMed: 2940971]
Fraimow HS (2009). Systemic antimicrobial therapy in osteomyelitis. Seminars in plastic surgery 23:
90–99. doi: 10.1055/s-0029-1214161 [PubMed: 20567731]
Ghobrial IM, Detappe A, Anderson KC, & Steensma DP (2018). The bone-marrow niche in MDS
and MGUS: implications for AML and MM. Nat Rev Clin Oncol 15: 219–233. doi: 10.1038/
nrclinonc.2017.197 [PubMed: 29311715]
Giuliani N, Morandi F, Tagliaferri S, Lazzaretti M, Bonomini S, Crugnola M, et al. (2007).
The proteasome inhibitor bortezomib affects osteoblast differentiation in vitro and in vivo in
multiple myeloma patients. Blood 110: 334–338. doi: 10.1182/blood-2006-11-059188 [PubMed:
17371942]
Giuliani N, & Rizzoli V (2007). Myeloma cells and bone marrow osteoblast interactions: Role in the
development of osteolytic lesions in multiple myeloma. Leukemia & Lymphoma, 48, 2323–2329.
doi: 10.1080/10428190701648281 [PubMed: 18067006]
Goldschmidt H, Ashcroft J, Szabo Z, & Garderet L (2019). Navigating the treatment landscape in
multiple myeloma: which combinations to use and when? Ann Hematol 98: 1–18. doi: 10.1007/
s00277-018-3546-8
Goyal V, & Bordia A (1995). The hypoglycemic effect of chloroquine. J Assoc Physicians India 43:
17–18. [PubMed: 9282631]
Hamouda MA, Belhacene N, Puissant A, Colosetti P, Robert G, Jacquel A, et al. (2014). The
small heat shock protein B8 (HSPB8) confers resistance to bortezomib by promoting autophagic
removal of misfolded proteins in multiple myeloma cells. Oncotarget 5: 6252–6266. doi:
10.18632/oncotarget.2193 [PubMed: 25051369]
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 20
Author Manuscript
Author Manuscript
Author Manuscript
Author Manuscript
Harding SD, Sharman JL, Faccenda E, Southan C, Pawson AJ, Ireland S, Gray AJG, Bruce L,
Alexander SPH, Anderton S, Bryant C, Davenport AP, Doerig C, Fabbro D, Levi-Schaffer F,
Spedding M, Davies JA, NC-IUPHAR (2018). The IUPHAR/BPS guide to pharmacology in 2018:
Updates and expansion to encompass the new guide to immunopharmacology. Nucleic Acids
Research 46, D1091–D1106. doi:10.1093/nar/gkx1121. [PubMed: 29149325]
Herczegh P, Buxton TB, McPherson JC 3rd, Kovacs-Kulyassa A, Brewer PD, Sztaricskai F, et al.
(2002). Osteoadsorptive bisphosphonate derivatives of fluoroquinolone antibacterials. J Med Chem
45: 2338–2341. doi: 10.1021/jm0105326 [PubMed: 12014972]
Hideshima T, Ikeda H, Chauhan D, Okawa Y, Raje N, Podar K, et al. (2009). Bortezomib induces
canonical nuclear factor-kappaB activation in multiple myeloma cells. Blood 114: 1046–1052. doi:
10.1182/blood-2009-01-199604 [PubMed: 19436050]
Hoang B, Benavides A, Shi Y, Frost P, & Lichtenstein A (2009). Effect of autophagy on multiple
myeloma cell viability. Mol Cancer Ther 8: 1974–1984. doi: 10.1158/1535-7163.MCT-08-1177
[PubMed: 19509276]
Jin J, Okamoto R, Yoon SS, Shih LY, Zhu J, Liu T, et al. (2018). Bortezomib-based therapy for
transplant-ineligible East Asian patients with newly diagnosed mantle-cell lymphoma. Onco
Targets Ther 11: 3869–3882. doi: 10.2147/OTT.S150339 [PubMed: 30013367]
Junka A, Szymczyk P, Ziolkowski G, Karuga-Kuzniewska E, Smutnicka D, Bil-Lula I, et al. (2017).
Bad to the Bone: On In Vitro and Ex Vivo Microbial Biofilm Ability to Directly Destroy
Colonized Bone Surfaces without Participation of Host Immunity or Osteoclastogenesis. PLoS
One 12: e0169565. doi: 10.1371/journal.pone.0169565 [PubMed: 28076372]
Kamath AF, Ong KL, Lau E, Chan V, Vail TP, Rubash HE, et al. (2015). Quantifying the Burden of
Revision Total Joint Arthroplasty for Periprosthetic Infection. J Arthroplasty 30: 1492–1497. doi:
10.1016/j.arth.2015.03.035 [PubMed: 25865815]
Karasneh JA, Al-Eryani K, Clark GT, & Sedghizadeh PP (2016). Modified protocol including topical
minocycline in orabase to manage medication-related osteonecrosis of the jaw cases. J Oral Pathol
Med 45: 718–720. doi: 10.1111/jop.12419 [PubMed: 26750149]
Kawano Y, Moschetta M, Manier S, Glavey S, Gorgun GT, Roccaro AM, et al. (2015). Targeting the
bone marrow microenvironment in multiple myeloma. Immunol Rev 263: 160–172. doi: 10.1111/
imr.12233 [PubMed: 25510276]
Keller DL (2014). Adverse cardiac effects of bisphosphonates. Mayo Clin Proc 89: 1025–1026. doi:
10.1016/j.mayocp.2014.04.012 [PubMed: 24996241]
Kempfle JS, Nguyen K, Hamadani C, Koen N, Edge AS, Kashemirov BA, et al. (2018).
Bisphosphonate-Linked TrkB Agonist: Cochlea-Targeted Delivery of a Neurotrophic Agent as
a Strategy for the Treatment of Hearing Loss. Bioconjug Chem 29: 1240–1250. doi: 10.1021/
acs.bioconjchem.8b00022 [PubMed: 29485861]
Kim BN, Kim ES, & Oh MD (2014). Oral antibiotic treatment of staphylococcal bone and joint
infections in adults. J Antimicrob Chemother 69: 309–322. doi: 10.1093/jac/dkt374 [PubMed:
24072167]
Kisgen JJ, Mansour H, Unger NR, & Childs LM (2014). Tedizolid: a new oxazolidinone antimicrobial.
Am J Health Syst Pharm 71: 621–633. doi: 10.2146/ajhp130482 [PubMed: 24688035]
Kowada T, Kikuta J, Kubo A, Ishii M, Maeda H, Mizukami S, et al. (2011). In vivo fluorescence
imaging of bone-resorbing osteoclasts. J Am Chem Soc 133: 17772–17776. doi: 10.1021/
ja2064582 [PubMed: 21939210]
Kuhn DJ, Berkova Z, Jones RJ, Woessner R, Bjorklund CC, Ma W, et al. (2012). Targeting the insulinlike growth factor-1 receptor to overcome bortezomib resistance in preclinical models of multiple
myeloma. Blood 120: 3260–3270. doi: 10.1182/blood-2011-10-386789 [PubMed: 22932796]
Lavrador P, Gaspar VM, & Mano JF (2018). Stimuli-responsive nanocarriers for delivery of
bone therapeutics - Barriers and progresses. J Control Release 273: 51–67. doi: 10.1016/
j.jconrel.2018.01.021 [PubMed: 29407678]
Lawson MA, McDonald MM, Kovacic N, Hua Khoo W, Terry RL, Down J, et al. (2015). Osteoclasts
control reactivation of dormant myeloma cells by remodelling the endosteal niche. Nat Commun 6:
8983. doi: 10.1038/ncomms9983 [PubMed: 26632274]
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 21
Author Manuscript
Author Manuscript
Author Manuscript
Author Manuscript
Lew DP, & Waldvogel FA (2004). Osteomyelitis. Lancet 364: 369–379. doi: 10.1016/
S0140-6736(04)16727-5 [PubMed: 15276398]
Li C, Chen S, Yue P, Deng X, Lonial S, Khuri FR, et al. (2010). Proteasome inhibitor PS-341
(bortezomib) induces calpain-dependent IkappaB(alpha) degradation. J Biol Chem 285: 16096–
16104. doi: 10.1074/jbc.M109.072694 [PubMed: 20335171]
Lin B, Caldwell C, Bhaduri S, Goel V, & Agarwal A (2017). Optimizing Vancomycin Release from
Calcium Phosphate-Based Cement by Carboxymethyl Cellulose for Prevention of Osteomyelitis.
Surg Infect (Larchmt) 18: 221–222. doi: 10.1089/sur.2016.189 [PubMed: 28009535]
Lonial S, Waller EK, Richardson PG, Jagannath S, Orlowski RZ, Giver CR, et al. (2005). Risk factors
and kinetics of thrombocytopenia associated with bortezomib for relapsed, refractory multiple
myeloma. Blood 106: 3777–3784. doi: 10.1182/blood-2005-03-1173 [PubMed: 16099887]
Mahindra A, Hideshima T, & Anderson KC (2010). Multiple myeloma: biology of the disease. Blood
Rev 24 Suppl 1: S5–11. doi: 10.1016/S0268-960X(10)70003-5 [PubMed: 21126636]
Manasanch EE, & Orlowski RZ (2017). Proteasome inhibitors in cancer therapy. Nat Rev Clin Oncol
14: 417–433. doi: 10.1038/nrclinonc.2016.206 [PubMed: 28117417]
McKenna CE, Haratipour P, Duro MVV, & Ebetino FH (2020). Chemistry of Bisphosphonates. In
Encyclopedia of Bone Biology. ed Zaidi M. Academic Press: Oxford, pp 551–564. DOI: 10.1016/
B978-0-12-801238-3.11260-7.
Mitra AK, Mukherjee UK, Harding T, Jang JS, Stessman H, Li Y, et al. (2016). Single-cell analysis
of targeted transcriptome predicts drug sensitivity of single cells within human myeloma tumors.
Leukemia 30: 1094–1102. doi: 10.1038/leu.2015.361 [PubMed: 26710886]
Montanari F, Lu M, Marcus S, Saran A, Malankar A, & Mazumder A (2014). A Phase II
Trial of Chloroquine in Combination with Bortezomib and Cyclophosphamide in Patients
with Relapsed and Refractory Multiple Myeloma. Blood 124: 5775–5775. doi: 10.1182/
blood.V124.21.5775.5775
Murai K, Kowata S, Shimoyama T, Yashima-Abo A, Fujishima Y, Ito S, et al. (2014). Bortezomib
induces thrombocytopenia by the inhibition of proplatelet formation of megakaryocytes. Eur J
Haematol 93: 290–296. doi: 10.1111/ejh.12342 [PubMed: 24750292]
Murray MY, Auger MJ, & Bowles KM (2014). Overcoming bortezomib resistance in multiple
myeloma. Biochem Soc Trans 42: 804–808. doi: 10.1042/BST20140126 [PubMed: 25109961]
Nagaratnam N, Chetiyawardana AD, & Rajiyah S (1978). Aplasia and leukaemia following
chloroquine therapy. Postgrad Med J 54: 108–112. doi: 10.1136/pgmj.54.628.108 [PubMed:
273209]
Niewerth D, Jansen G, Assaraf YG, Zweegman S, Kaspers GJ, & Cloos J (2015). Molecular basis of
resistance to proteasome inhibitors in hematological malignancies. Drug Resist Updat 18: 18–35.
doi: 10.1016/j.drup.2014.12.001 [PubMed: 25670156]
Obeng EA, Carlson LM, Gutman DM, Harrington WJ Jr., Lee KP, & Boise LH (2006). Proteasome
inhibitors induce a terminal unfolded protein response in multiple myeloma cells. Blood 107:
4907–4916. doi: 10.1182/blood-2005-08-3531 [PubMed: 16507771]
Painuly U, & Kumar S (2013). Efficacy of bortezomib as first-line treatment for patients with multiple
myeloma. Clin Med Insights Oncol 7: 53–73. doi: 10.4137/CMO.S7764 [PubMed: 23492937]
Paton-Hough J, Chantry AD, & Lawson MA (2015). A review of current murine models of multiple
myeloma used to assess the efficacy of therapeutic agents on tumour growth and bone disease.
Bone 77: 57–68. doi: 10.1016/j.bone.2015.04.004 [PubMed: 25868800]
Pazianas M, Cooper C, Ebetino FH, & Russell RG (2010). Long-term treatment with bisphosphonates
and their safety in postmenopausal osteoporosis. Ther Clin Risk Manag 6: 325–343. doi: 10.2147/
tcrm.s8054 [PubMed: 20668715]
Pennisi A, Li X, Ling W, Khan S, Zangari M, & Yaccoby S (2009). The proteasome inhibitor,
bortezomib suppresses primary myeloma and stimulates bone formation in myelomatous and
nonmyelomatous bones in vivo. Am J Hematol 84: 6–14. doi: 10.1002/ajh.21310 [PubMed:
18980173]
Pollak AN, Jones AL, Castillo RC, Bosse MJ, MacKenzie EJ, & Group LS (2010). The relationship
between time to surgical debridement and incidence of infection after open high-energy lower
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 22
Author Manuscript
Author Manuscript
Author Manuscript
Author Manuscript
extremity trauma. J Bone Joint Surg Am 92: 7–15. doi: 10.2106/JBJS.H.00984 [PubMed:
20048090]
Prabha A (1996). Hypoglycemic effect of chloroquine. J Assoc Physicians India 44: 149–150.
Reinholz MM, Zinnen SP, Dueck AC, Dingli D, Reinholz GG, Jonart LA, et al. (2010). A promising
approach for treatment of tumor-induced bone diseases: utilizing bisphosphonate derivatives
of nucleoside antimetabolites. Bone 47: 12–22. doi: 10.1016/j.bone.2010.03.006 [PubMed:
20233612]
Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, Irwin D, et al. (2003). A phase
2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med 348: 2609–2617. doi:
10.1056/NEJMoa030288 [PubMed: 12826635]
Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadtmauer EA, Facon T, et al. (2005).
Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Engl J Med 352:
2487–2498. doi: 10.1056/NEJMoa043445 [PubMed: 15958804]
Richardson PG, Xie W, Jagannath S, Jakubowiak A, Lonial S, Raje NS, et al. (2014). A phase 2 trial
of lenalidomide, bortezomib, and dexamethasone in patients with relapsed and relapsed/refractory
myeloma. Blood 123: 1461–1469. doi: 10.1182/blood-2013-07-517276 [PubMed: 24429336]
Robak T, Huang H, Jin J, Zhu J, Liu T, Samoilova O, et al. (2015). Bortezomib-based therapy
for newly diagnosed mantle-cell lymphoma. N Engl J Med 372: 944–953. doi: 10.1056/
NEJMoa1412096 [PubMed: 25738670]
Roy P, Sarkar UA, & Basak S (2018). The NF-kappaB Activating Pathways in Multiple Myeloma.
Biomedicines 6. doi: 10.3390/biomedicines6020059
Ruschak AM, Slassi M, Kay LE, & Schimmer AD (2011). Novel proteasome inhibitors to overcome
bortezomib resistance. J Natl Cancer Inst 103: 1007–1017. doi: 10.1093/jnci/djr160 [PubMed:
21606441]
Russell RG, Watts NB, Ebetino FH, & Rogers MJ (2008). Mechanisms of action of bisphosphonates:
similarities and differences and their potential influence on clinical efficacy. Osteoporos Int 19:
733–759. doi: 10.1007/s00198-007-0540-8 [PubMed: 18214569]
Scott EC, Maziarz RT, Spurgeon SE, Medvedova E, Gajewski J, Reasor-Heard S, et al. (2017).
Double autophagy stimulation using chemotherapy and mTOR inhibition combined with
hydroxychloroquine for autophagy modulation in patients with relapsed or refractory multiple
myeloma. Haematologica 102: e261–e265. doi: 10.3324/haematol.2016.162321 [PubMed:
28385778]
Sedghizadeh P, Ebetino FH, Sun S, Sadrerafi K, Junka A, Mahabady S, et al. (2017a). Bone targeted
bisphosphonate-antibiotic conjugates for the treatment of osteomyelitis biofilms. Journal of Bone
and Mineral Res 32: S285–S285.
Sedghizadeh P, Sodagar E, Garabedian R, Wadia J, Ebetino FH, Cherian P, et al. (2019). Design and
in vivo testing of novel bisphosphonate-fluoroquinolone conjugates chemisorbed to bone graft
material. Journal of Bone and Mineral Res 34: S367–S368.
Sedghizadeh PP (2019). Biofilm mediated infections in dentistry and the role of bisphosphonates in
oral diseases and therapeutics JBMR Plus 3: 48.
Sedghizadeh PP, Sun S, Junka AF, Richard E, Sadrerafi K, Mahabady S, et al. (2017b). Design,
Synthesis, and Antimicrobial Evaluation of a Novel Bone-Targeting Bisphosphonate-Ciprofloxacin
Conjugate for the Treatment of Osteomyelitis Biofilms. J Med Chem 60: 2326–2343. doi: 10.1021/
acs.jmedchem.6b01615 [PubMed: 28121436]
Sedghizadeh PP, Sun S, Jones AC, Sodagar E, Cherian P, Chen C, et al. (2021). Bisphosphonates
in Dentistry: Historical Perspectives, Adverse Effects, and Novel Applications. Bone, In press,
doi.org: 10.1016/j.bone.2021.115933.
Shain KH, Dalton WS, & Tao J (2015). The tumor microenvironment shapes hallmarks of mature
B-cell malignancies. Oncogene 34: 4673–4682. doi: 10.1038/onc.2014.403 [PubMed: 25639873]
Shi DS, Smith MC, Campbell RA, Zimmerman PW, Franks ZB, Kraemer BF, et al. (2014).
Proteasome function is required for platelet production. J Clin Invest 124: 3757–3766. doi:
10.1172/JCI75247 [PubMed: 25061876]
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 23
Author Manuscript
Author Manuscript
Author Manuscript
Author Manuscript
Silver IA, Murrills RJ, & Etherington DJ (1988). Microelectrode studies on the acid
microenvironment beneath adherent macrophages and osteoclasts. Exp Cell Res 175: 266–276.
doi: 10.1016/0014-4827(88)90191-7 [PubMed: 3360056]
Slane J, Gietman B, & Squire M (2018). Antibiotic elution from acrylic bone cement loaded with
high doses of tobramycin and vancomycin. J Orthop Res 36: 1078–1085. doi: 10.1002/jor.23722
[PubMed: 28876459]
Staats K, Sevelda F, Kaider A, Bohler C, Sigmund IK, Puchner SE, et al. (2017). The influence of
antibiotic-loaded cement spacers on the risk of reinfection after septic two-stage hip revision
surgery. Infection 45: 885–891. doi: 10.1007/s15010-017-1081-5 [PubMed: 29067626]
Stoodley P, Ehrlich GD, Sedghizadeh PP, Hall-Stoodley L, Baratz ME, Altman DT, et al.
(2011). Orthopaedic biofilm infections. Curr Orthop Pract 22: 558–563. doi: 10.1097/
BCO.0b013e318230efcf [PubMed: 22323927]
Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M, et al. (2013). Autophagy and chemotherapy
resistance: a promising therapeutic target for cancer treatment. Cell Death Dis 4: e838. doi:
10.1038/cddis.2013.350 [PubMed: 24113172]
Swami A, Reagan MR, Basto P, Mishima Y, Kamaly N, Glavey S, et al. (2014). Engineered
nanomedicine for myeloma and bone microenvironment targeting. Proc Natl Acad Sci U S A
111: 10287–10292. doi: 10.1073/pnas.1401337111 [PubMed: 24982170]
Tan SY, & Grimes S (2010). Paul Ehrlich (1854-1915): man with the magic bullet. Singapore Med J
51: 842–843. [PubMed: 21140107]
Tao J, Srinivasan V, Zhou X, Ebetino FH, Boeckman RK Jr. , Boyce BF, et al. (2018). Bone-targeting
Bortezomib significantly increases it efficacy in the treatment of human multiple myeloma in
vitro and in vivo in mice. Journal of Bone and Mineral Res 33: 45.
Terpos E, Ntanasis-Stathopoulos I, Gavriatopoulou M, & Dimopoulos MA (2018). Pathogenesis of
bone disease in multiple myeloma: from bench to bedside. Blood Cancer J 8: 7. doi: 10.1038/
s41408-017-0037-4 [PubMed: 29330358]
Tsujimoto Y, & Shimizu S (2005). Another way to die: autophagic programmed cell death. Cell Death
Differ 12 Suppl 2: 1528–1534. doi: 10.1038/sj.cdd.4401777 [PubMed: 16247500]
Uskokovic V (2015). Nanostructured platforms for the sustained and local delivery of antibiotics
in the treatment of osteomyelitis. Crit Rev Ther Drug Carr Syst 32: 1–59. doi: 10.1615/
CritRevTherDrugCarrierSyst.2014010920
Uy GL, Trivedi R, Peles S, Fisher NM, Zhang QJ, Tomasson MH, et al. (2007). Bortezomib inhibits
osteoclast activity in patients with multiple myeloma. Clin Lymphoma Myeloma 7: 587–589. doi:
10.3816/CLM.2007.n.045 [PubMed: 18186967]
Verbaanderd C, Maes H, Schaaf MB, Sukhatme VP, Pantziarka P, Sukhatme V, et al. (2017).
Repurposing Drugs in Oncology (ReDO)-chloroquine and hydroxychloroquine as anti-cancer
agents. Ecancermedicalscience 11: 781. doi: 10.3332/ecancer.2017.781 [PubMed: 29225688]
Vogl DT, Stadtmauer EA, Tan KS, Heitjan DF, Davis LE, Pontiggia L, et al. (2014). Combined
autophagy and proteasome inhibition: a phase 1 trial of hydroxychloroquine and bortezomib in
patients with relapsed/refractory myeloma. Autophagy 10: 1380–1390. doi: 10.4161/auto.29264
[PubMed: 24991834]
von Metzler I, Krebbel H, Hecht M, Manz RA, Fleissner C, Mieth M, et al. (2007). Bortezomib
inhibits human osteoclastogenesis. Leukemia 21: 2025–2034. doi: 10.1038/sj.leu.2404806
[PubMed: 17581612]
Vrabel D, Pour L, & Sevcikova S (2019). The impact of NF-kappaB signaling on pathogenesis
and current treatment strategies in multiple myeloma. Blood Rev 34: 56–66. doi: 10.1016/
j.blre.2018.11.003 [PubMed: 30501907]
Walker LC, Baker P, Holleyman R, & Deehan D (2017). Microbial resistance related to antibioticloaded bone cement: a historical review. Knee Surg Sports Traumatol Arthrosc 25: 3808–3817.
doi: 10.1007/s00167-016-4309-5 [PubMed: 27620468]
Wang H, Xiao L, Tao J, Srinivasan V, Boyce BF, Ebetino FH, et al. (2018). Synthesis of a BoneTargeted Bortezomib with In Vivo Anti-Myeloma Effects in Mice. Pharmaceutics 10. doi:
10.3390/pharmaceutics10030154
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 24
Author Manuscript
Author Manuscript
Author Manuscript
Author Manuscript
Wang H, Zhang H, Srinivasan V, Tao J, Sun W, Lin X, et al. (2020). Targeting Bortezomib to Bone
Increases Its Bone Anabolic Activity and Reduces Systemic Adverse Effects in Mice. J Bone
Miner Res 35: 343–356. doi: 10.1002/jbmr.3889 [PubMed: 31610066]
Wang M, Cai X, Yang J, Wang C, Tong L, Xiao J, et al. (2018). A Targeted and pH-Responsive
Bortezomib Nanomedicine in the Treatment of Metastatic Bone Tumors. ACS Appl Mater
Interfaces 10: 41003–41011. doi: 10.1021/acsami.8b07527 [PubMed: 30403331]
Xing L, Ebetino FH, Boeckman RK Jr., Srinivasan V, Tao J, Sawyer TK, et al. (2020).
Targeting anti-cancer agents to bone using bisphosphonates. Bone 138: 115492. doi: 10.1016/
j.bone.2020.115492 [PubMed: 32585321]
Xiu Y, Xu H, Zhao C, Li J, Morita Y, Yao Z, et al. (2014). Chloroquine reduces osteoclastogenesis
in murine osteoporosis by preventing TRAF3 degradation. J Clin Invest 124: 297–310. doi:
10.1172/JCI66947 [PubMed: 24316970]
Xu S, De Veirman K, De Becker A, Vanderkerken K, & Van Riet I (2018). Mesenchymal stem
cells in multiple myeloma: a therapeutical tool or target? Leukemia 32: 1500–1514. doi: 10.1038/
s41375-018-0061-9 [PubMed: 29535427]
Yao Z, Hou X, Lei W, Xiao L, Ebetino FH, Boeckman RK, et al. (2016). Bone targeted chloroquine
inhibits osteoclastogenesis and bone resorption more effectively than chloroquine. J Bone Miner
Res 31: S225.
Young RN, & Grynpas MD (2018). Targeting therapeutics to bone by conjugation with
bisphosphonates. Curr Opin Pharmacol 40: 87–94. doi: 10.1016/j.coph.2018.03.010 [PubMed:
29626715]
Yun Z, Zhichao J, Hao Y, Ou J, Ran Y, Wen D, et al. (2017). Targeting autophagy in multiple
myeloma. Leuk Res 59: 97–104. doi: 10.1016/j.leukres.2017.06.002 [PubMed: 28599191]
Zangari M, & Suva LJ (2016). The effects of proteasome inhibitors on bone remodeling in multiple
myeloma. Bone 86: 131–138. doi: 10.1016/j.bone.2016.02.019 [PubMed: 26947893]
Zavrski I, Krebbel H, Wildemann B, Heider U, Kaiser M, Possinger K, et al. (2005). Proteasome
inhibitors abrogate osteoclast differentiation and osteoclast function. Biochem Biophys Res
Commun 333: 200–205. doi: 10.1016/j.bbrc.2005.05.098 [PubMed: 15936724]
Zeng R, Chen Y, Zhao S, & Cui GH (2012). Autophagy counteracts apoptosis in human multiple
myeloma cells exposed to oridonin in vitro via regulating intracellular ROS and SIRT1. Acta
Pharmacol Sin 33: 91–100. doi: 10.1038/aps.2011.143 [PubMed: 22158107]
Zhang M, He J, Liu Z, Lu Y, Zheng Y, Li H, et al. (2015). Anti-beta(2)-microglobulin monoclonal
antibodies overcome bortezomib resistance in multiple myeloma by inhibiting autophagy.
Oncotarget 6: 8567–8578. doi: 10.18632/oncotarget.3251 [PubMed: 25895124]
Zhu J, Huo Q, Xu M, Yang F, Li Y, Shi H, et al. (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. doi: 10.1039/c8nr03899f [PubMed:
30256367]
Zinnen S, Rowinsky EK, Alexandrov A, Plekhova L, Roudas M, & Karpeisky A (2017). Phase 1
study of the bone-targeting cytotoxic conjugate, etidronate-cytosine arabinoside (MBC-11), in
cancer patients with bone metastases. Journal of Clinical Oncology 35: 2589–2589. doi: 10.1200/
JCO.2017.35.15_suppl.2589
Zinnen SP, Karpeisky A, Von Hoff DD, Plekhova L, & Alexandrov A (2019). First-in-Human Phase
I Study of MBC-11, a Novel Bone-Targeted Cytarabine-Etidronate Conjugate in Patients with
Cancer-Induced Bone Disease. Oncologist 24: 303–e102. doi: 10.1634/theoncologist.2018-0707
[PubMed: 30413669]
Zinszner H, Kuroda M, Wang X, Batchvarova N, Lightfoot RT, Remotti H, et al. (1998). CHOP
is implicated in programmed cell death in response to impaired function of the endoplasmic
reticulum. Genes Dev 12: 982–995. doi: 10.1101/gad.12.7.982 [PubMed: 9531536]
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 25
Author Manuscript
Author Manuscript
Author Manuscript
FIGURE 1.
Author Manuscript
(a)“Target-and-release”design of bisphosphonate-drug conjugates. (b) Structures of
bisphosphonate conjugates of bortezomib (BP-Btz), chloroquine (BP-CQ), and
hydroxychloroquine (BP-HCQ). (c–h) A combination of BP-Btz and BP-CQ kills more
Btz-resistant U266 human myeloma cells or primary myeloma cells from patients with
relapsed/refractory MM than a single agent. (c–f) Human U266 cells were treated with
various drugs for 72 h. Cell survival was determined using a CCK8 kit. The percentage of
survival was calculated using Veh-treated cells as 100%. Btz-sensitive/parental U266 cells
(U266.P) and Btz-resistant U266 cells (U266.BR) were treated with different doses of Btz
(c) or BP-Btz (d). U266. BR cells were treated with 6 nM Btz, 30 μM CQ, Btz + CQ
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 26
Author Manuscript
(e) or 30 μM BP, 6 nM BP-Btz, 30 μM BP-CQ, BP-Btz + BP-CQ (f). n = 2 repeats with
similar results. Bone marrow aspirates of Patients #1–3 with relapsed/refractory MM (g) and
Patients #4–5 with newly diagnosed MM (h) were used. CD138+ cells were isolated with
anti-CD138 antibody-conjugated magnetic beads via positive selection. Cells were treated
with various drugs for 24 h. The doses for cells from Patients #1, 2, and 4 were the same
as in (f), and the doses for patient #3 were 12 nM BP-Btz and 40 μM BP-CQ, and for
patient #5 were 6 nM BP-Btz, 20 μM BP-CQ, or BP-HCQ. *P<0.05, significantly different
as indicated. Figure adapted from Boyce, 2019; Tao et al., 2018 and Xing et al., 2020.
Author Manuscript
Author Manuscript
Author Manuscript
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 27
Author Manuscript
Author Manuscript
Author Manuscript
FIGURE 2.
Author Manuscript
(a) BP-antibiotic conjugates and parent antibiotics used in testing; (b) antimicrobial results
in a rat model of periprosthetic osteomyelitis. Data show efficacy of ciprofloxacin and
BCC for reducing bacterial load or mean CFU per g tissue (Y-axis). The greatest efficacy
was observed at a single high dose (10 mg•kg−1) of BCC where a 2-log reduction (99%
bactericidal activity) was seen, compared with the negative control (Figure 2b adapted from
Sedghizadeh, Sun, et al., 2017)
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 28
Author Manuscript
Author Manuscript
Author Manuscript
FIGURE 3.
(a, b) Preventative hydroxyapatite experiments with tested compounds against S. aureus and
Aa. (a) The BCC, BET, and BCX conjugates demonstrated statistically significant efficacy
for preventing the growth of S. aureus. (b) The BCX conjugate demonstrated statistically
significant efficacy for preventing the growth of Aa. (c, d) Eradication experiments with
test compounds against S. aureus and Aa biofilms on HA. (c) All tested compounds
except vancomycin were significantly efficacious in eradicating S. aureus, with the greatest
reduction seen with BCX. (d) The BCX eradicated a significant number of Aa cells. *
P<0.05, significantly different from control. Figure adapted from Ebetino et al., 2020;
Sedghizadeh et al., 2021)
Author Manuscript
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 29
Author Manuscript
Author Manuscript
Figure 4.
Author Manuscript
Author Manuscript
Ex vivo assays of rat femora saturated with antimicrobials against S. aureus (A, B) and
rat jaws saturated with antimicrobials against Aa (C, D). A. A picture of rat femora
saturated with antibiotics or conjugates after they are introduced to a solution of S. aureus
in our ex vivo preventative experimental setting. The more the bone appears red, the
more staphylococcal biofilms there are adhered to the surface in those regions. Femur 1
– negative control sample (no antimicrobial, no staphylococcus); 2 – positive control sample
(no antimicrobial, staphylococcal solution added); 3 – bone saturated with tedizolid and
introduced to Staphylococcal solution; 4 – bone saturated with ciprofloxacin and introduced
to staphylococcal solution; 5 – bone saturated with BET conjugate and introduced to
staphylococcal solution; 6 – bone saturated with BCC and introduced to staphylococcal
solution. B. In S. aureus biofilm prevention and eradication settings, conjugates BCC
and BET significantly prevented or eradicated biofilm growth on femurs ex vivo, as did
tedizolid alone, with greatest efficacy seen with the BET in both settings (K-W test, *
p<0.05 versus control). C. A picture of rat jaws saturated with antibiotics or conjugates after
they are introduced to a solution of Aa in our ex vivo preventative experimental setting.
The more the bone appears red, the more Aa biofilms there are adhered to the surface
in those regions. Jaw 1 – negative control sample (no antimicrobial, no Aa); 2 – positive
control sample (no antimicrobial, Aa solution added); 3 – bone saturated with BCC and
introduced to Aa solution; 4 – bone saturated with ciprofloxacin and introduced to Aa
solution; 5 – bone saturated with minocycline and introduced to Aa solution. D. In Aa
biofilm prevention and eradication settings, the BCC significantly prevented biofilm growth
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 30
Author Manuscript
Author Manuscript
on jaws ex vivo whereas ciprofloxacin and minocycline were less effective in both settings
(K-W test, * p<0.05; comparator=control). (Rats were killed by intraperitoneal application
of pentobarbital (40 mg/kg), the chest was surgically opened and the heart was removed. No
drugs or other procedures, which could potentially influence bone structure, were performed
during rat inbreeding or experiments. The jaws were removed postmortem from Wistar
male rats weighing 300-350g. To obtain osseous surfaces for biofilm development, the
soft tissues were surgically removed from their bones after resection. Subsequently, bones
were antiseptically rinsed using saline and then UV-irradiated to achieve sterilization. To
test bone sterility, 6 bones (3 jaws and 3 femora) were incubated in anaerobic conditions
in Thioglycolate Broth for 7 days according to microbiological procedures of anaerobic
organism culturing. Another 6 bones (3 jaws and 3 femora) were incubated for 3 days in
aerobic conditions in Brain-Heart Infusion (BHI) Broth. The sterilized bones were then
frozen at −80°C to prevent their decomposition and microbial contamination). (Figure
adapted from ref. (Ebetino, Sun, McKenna, Sadrerafi & Cherian, 2020; Sedghizadeh et
al., 2019; Sedghizadeh, 2019; Sedghizadeh et al., 2021)).
Author Manuscript
Author Manuscript
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.
Sun et al.
Page 31
Table 1.
Author Manuscript
Antimicrobial susceptibility results for experimental pathogens in polystyrene wells.
Antimicrobials [µg/mL]
M
C
X
T
V
BCC
BCX
BCT
Microbial
pathogens
MIC
MBEC
MIC
MBEC
MIC
MBEC
MIC
MBEC
MIC
MBEC
MIC
MBEC
MIC
MBEC
MIC
MBEC
M
S. aureus
0.25
16
0.5
32
0.05
0.5
0.5
8
1
8
8
64
4
16
16
32
4
2
8
0.5
8
0.05
0.1
*
*
*
*
16
250
2
4
*
*
*
Aa
M – Minocycline; C – Ciprofloxacin; X – Moxifloxacin; T – Tedizolid; V – Vancomycin
*
spectrum of antimicrobial activity does not cover this specific pathogen and not supported for use against specific bacteria. For experimental
purposes, the following reference strains were used: S. aureus ATCC 6538 and Aggregatibacter actinomycetemcomitans (Aa) D7S1. Moreover,
27 clinical strains of S. aureus were screened for their susceptibility to tested antibiotics (adapted from ref. (Ebetino, Sun, McKenna, Sadrerafi &
Cherian, 2020; Sedghizadeh et al., 2020)).
Author Manuscript
Author Manuscript
Author Manuscript
Br J Pharmacol. Author manuscript; available in PMC 2021 October 14.