600
Review Article
Calcitonin: A useful old friend
Akash Srinivasan*, Felyx K Wong*, Dimitrios Karponis
Imperial College London School of Medicine, UK
*contributed equally
Background
Calcitonin, in various preparations, has been used to
treat metabolic bone disease for over forty years since its
discovery in 1961 as a blood-calcium lowering hormone
1
.
Salmon calcitonin, in particular, has been effective in
treating postmenopausal osteoporosis, Paget’s disease and
hypercalcaemia
2,3
. Due to its ability to inhibit osteoclast
activity, calcitonin reduces the risk of vertebral re-fracture,
and it is also a powerful analgesic agent with proven efficacy
in managing acute back pain caused by recent vertebral
compression fractures
4,5
.
By 1992, world sales of therapeutic calcitonin had
exceeded 900 million US dollars
6
. However, the rise of
bisphosphonates pushed calcitonin to the side; since the
1960s, etidronate has been utilised as a therapy, primarily
for hypercalcaemia and Paget’s disease, and in 1995,
alendronate received approval by the US Food and Drug
Administration (FDA) for the treatment of postmenopausal
osteoporosis
7
. Since 2007, zoledronic acid has also been
licensed for the treatment of postmenopausal osteoporosis,
following evidence on its beneficial effects on bone mineral
density (BMD), bone metabolism markers and a reduction
in vertebral, hip and other fractures
8
. Multiple trials have
demonstrated superior efficacy in bisphosphonates and
alternative treatment options, which have consequently led
to decreased use of calcitonin.
Although bisphosphonates possess multiple effects
and are potent medications, there are significant adverse
effects associated with long-term use, such as atypical
femoral fractures and osteonecrosis of the jaw
7
. Therefore,
it is interesting to see the extent to which these drugs
have superseded calcitonin. This review aims to explore
the reasons behind the decline of calcitonin and discuss its
potential role in the years to come.
Biochemistry and pharmacology
Calcitonin is a single-chain polypeptide hormone which is
made up of 32 amino acids. An N-terminal disulfide bridge
between the cysteine residues at positions 1 and 7 create
a 7-amino acid ring structure and there is also a C-terminal
amidated proline
9
. The physiological effects of calcitonin are
Abstract
Calcitonin regulates blood calcium levels and possesses certain clinically useful anti-fracture properties. Specifically,
it reduces vertebral fractures in postmenopausal osteoporotic women significantly compared to a placebo. Nevertheless,
the use of calcitonin has declined over the years and salmon calcitonin is no longer the first-line treatment for many
of its indications. Commercial calcitonin only exists in intranasal or injectable preparations, which are less preferable
for patients. Efficacy of a potential oral formulation has been under investigation but achieving adequate bioavailability
remains a conundrum and the latest phase III trials have not shown promising evidence justifying its use. Associations with
cancer have also derailed this treatment option. Furthermore, the rise of bisphosphonates and, more recently, monoclonal
antibodies (such as denosumab), has revolutionised the treatment of osteoporotic fractures. Therefore, we are posed with
an interesting question: is calcitonin a treatment of the past? This review aims to explore the reasons behind this paradigm
shift and outline the potential role of calcitonin in the management of fractures and other conditions in the years to come.
Keywords: Analgesia, Calcitonin, Formulations, Fracture, Osteoporosis
The authors have no conflict of interest.
Corresponding author: Dimitrios Karponis, South Kensington, London, SW7
2AZ, UK
E-mail: dimitriskarponis@gmail.com
Edited by: G. Lyritis
Accepted 11 March 2020
Journal of Musculoskeletal
and Neuronal Interactions
Published under Creative Common License CC BY-NC-SA 4.0 (Attribution-Non Commercial-ShareAlike)
J Musculoskelet Neuronal Interact 2020; 20(4):600-609
601http://www.ismni.org
A. Srinivasan et al.: Remembering calcitonin
known to occur through receptor-mediated processes, and
interactions involving the N-terminal ring and the C-terminus
appear to be involved in receptor binding and signal
transduction
9,10
.
In humans, calcitonin is secreted by the para-follicular
or C cells of the thyroid gland in response to an increase in
serum calcium concentration
11
. Primarily, calcitonin targets
the bone, where it profoundly inhibits osteoclast action and
bone resorption. Actively resorbing osteoclasts secrete acid
and acid hydrolases via their ruffled borders to degrade bone.
Calcitonin promotes the internalisation of the osteoclasts
ruffled border proteins into intracellular vesicles, thereby
thwarting acid release and preventing the demineralisation
of bone matrix
12
. Calcitonin also acts via the kidneys, where
it reduces the reabsorption of calcium, along with sodium,
potassium, chloride and phosphate. Furthermore, the
hormone works on the central nervous system to induce
analgesia, stomach acid secretion and anorexia
1
.
The exact mechanism behind the analgesic effects of
calcitonin remains elusive, yet several theories have been
proposed. A 2016 study on rats discovered that calcitonin
decreases the number of serotonin transporters, whilst
increasing the expression of thalamic serotonin receptors
13
.
Other studies have proposed that nerve injuries activate a
calcitonin-dependent signal, which reduces transcription
of the sodium channel in the neurons of the dorsal root
ganglion
14
.
Calcitonin has been studied in numerous species
including pig, rat, salmon and eel. Subtle structural
differences massively affect their respective affinities for
calcitonin receptors. For example, salmon calcitonin has
a greater affinity to calcitonin receptors in all species,
compared to mammalian calcitonin and therefore, its higher
potency combined with its longer half-life has made salmon
calcitonin the standard form used to treat bone disorders
9,12
.
Although clinical resistance from circulating antibodies
can form against non-human calcitonin, the use of human
calcitonin is limited due to its susceptibility to precipitating
as insoluble fibrils
9
.
Salmon calcitonin has been commercially available in
an injectable form and as a nasal spray, but developing an
oral formulation of a peptide hormone, which can survive
gastric enzymatic digestion and subsequently penetrate
the intestinal mucosa, has been a challenge
2
. However, oral
delivery of calcitonin has been shown to be feasible by linking
salmon calcitonin with various additives, including permeation
enhancers (e.g. a caprylic acid derivative), enzyme inhibitors
and particulate systems
2,15,16
.
The history of calcitonin
Parenteral calcitonin was the first FDA approved
formulation, available as an intramuscular or subcutaneous
injection. However, its use was associated with poor
patient compliance and tolerability due to its extensive
side effects, the most notable of which was gastrointestinal
disturbance
17-20
. Numerous studies reported nausea, loss of
appetite, abdominal pain and diarrhoea. In addition, patients
also experienced flushing in the face and peripheries; local
inflammatory reactions were also common at the site of
administration
18,19
. Data on the efficacy of the injectable
formulation is scarce. In a retrospective cohort study
conducted by Kanis et al., a significant risk reduction in
hip fractures (RR: 0.69, 95% CI: 0.51-0.92) was observed
following a year of daily calcitonin injections, compared to
control
21
. However, a similar magnitude of risk reduction was
observed in patients receiving daily calcium supplementation
(RR: 0.75, 95% CI: 0.60-0.94). It is important to note that,
due to its extensive side effect profile, the use of injectable
calcitonin has been mostly replaced by the later developed
intranasal formulation.
The intranasal formulation has been the preferred method
of administration over the parenteral route due to multiple
factors. Firstly, it is more convenient and less invasive
for the patient. Moreover, common side effects resulting
from salmon calcitonin administration, such as nausea and
vomiting, occur less frequently in studies using the intranasal
formulation
4,22
. Collectively, these factors promote better
drug tolerability and patient compliance. However, the nasal
formulation has been reported to have lower bioavailability
and slower absorption than the injectable formulation
23
. Mild
nasal symptoms have also been observed in multiple studies,
including nasal irritation, rhinitis and rhinorrhoea
4,18,22
.
One of the important studies which led to the 1995 FDA
approval of intranasal salmon calcitonin was the PROOF
study (Prevent Recurrence of Osteoporotic Fracture)
4
.
The PROOF study was an international, multi-centre trial
which demonstrated that a 5-year daily 200IU dose of
intranasal calcitonin, along with daily vitamin D and calcium
supplementation, was able to reduce vertebral fracture
risk by 36% compared to placebo in postmenopausal
women living with osteoporosis. A significant but modest
improvement in lumbar BMD was seen in all treatment arms
(100 IU, 200 IU, and 400 IU), while serum bone resorption
markers were also significantly reduced. This was the first
large-scale, prospective study looking into the anti-fracture
efficacy of calcitonin. Prior to this, studies demonstrating
salmon calcitonin’s anti-fracture properties were mostly
retrospective or prospective with small sample sizes
21,24-26
.
Interestingly, the efficacy of intranasal calcitonin was not
dose-dependent. Patients receiving the daily 200 IU dose
were able to benefit from a greater vertebral fracture risk
reduction than patients on the daily 400 IU and 100 IU
regimens. It is important to acknowledge that the PROOF study
has been criticised for its high dropout rate of 59% by the end
of the 5-year follow up. Although some of the main findings
from this study, including its potential in increasing lumbar
spine BMD and suppression of serum resorption markers,
were consistent with the literature, evidence surrounding
nasal calcitonin’s vertebral anti-fracture efficacy have been
conflicting
24,26-30
. Additionally, the effect of nasal calcitonin
on non-vertebral fracture risk and BMD is unclear
31, 32
. Aside
from its effects on the vertebrae, nasal calcitonin has been
602http://www.ismni.org
A. Srinivasan et al.: Remembering calcitonin
shown to have a significant analgesic effect during the early
stages of treating distal radius fractures
33
.
Oral calcitonin has raised interest as a result of a few
factors. Firstly, bisphosphonates have received increasing
levels of concern around their long-term, albeit rare, side
effects, including osteonecrosis of the jaw and atypical
femoral fractures
34,35
. This, therefore, renewed the interests
in alternative therapies, including older treatment options
such as calcitonin. Secondly, intranasal and injectable
salmon calcitonin have been associated with suboptimal
patient compliance
4
. Thus, a potentially more convenient
and accepting method of delivery was explored. However,
due to the peptide nature of calcitonin, achieving adequate
bioavailability from the oral route has been the main challenge
to tackle to date
36
.
At the time of composing this review, evidence on oral
calcitonin is limited and data from the latest phase III
trials have not shown promising results, to the best of our
knowledge. The most recent phase III trial, conducted by
Henriksen et al., investigated the anti-fracture efficacy of
oral salmon calcitonin in 4665 postmenopausal women
over 3 years and is the only study hitherto to directly
measure the anti-fracture efficacy of the oral formulation
37
.
Achieving adequate bioavailability was a challenge and
pharmacokinetic analysis demonstrated suboptimal
drug exposure in subjects who were administered
calcitonin. Overall, daily oral salmon calcitonin (0.8
mg/d) with calcium and vitamin D supplementation did not
significantly alter new vertebral fracture and non-vertebral
fracture incidence when compared to control (p=0.94). No
significant differences in cumulative fracture risk were
seen between the treatment and control groups across
the 36-month study period. However, participants in the
treatment arm had a statistically longer duration between
baseline and the time of first hip fracture compared to
placebo. The increase in lumbar spine BMD following
oral calcitonin treatment was significantly greater in
the treatment group than placebo (treatment: 1.02%,
control: 0.18%, p<0.0001). Two studies conducted by
Binkley et al. have demonstrated significant but mild
improvement in lumbar spine BMD improvement and
moderate suppression of bone resorption markers
38,39
.
Anti-fracture efficacy was not assessed in these trials.
Firstly, the ORACAL trial was a phase III study which
showed that daily 0.2 mg oral formulation over 48 weeks
induced a significantly greater improvement in lumbar
spine BMD than the intranasal formulation (p=0.027) and
placebo (p=0.010) groups. Yet, it is important to note
that the absolute differences between the change in BMD
of the oral, intranasal and placebo arms were modest
(1.5%±3.2%, 0.78%±2.9%, 0.5%±3.2%, respectively).
Similarly, the second and more recent trial conducted by
Binkley et al. demonstrated a significant yet mild effect
on lumbar spine BMD (1.03%, p<0.001) following 54-
Table 1. Trials on the anti-fracture efficacy and analgesic effects of intranasal and oral calcitonin.
Reference N Intervention Outcome
Chestnut et al.
(2000)
4
1255
(511 completed
full 5-year
follow up)
Daily nasal salmon
calcitonin (100, 200
or 400 IU) vs placebo
over 5 years
Daily 200 IU nasal calcitonin significantly reduced vertebral fracture risk.
The 100 IU nasal calcitonin group experienced significantly fewer non-
vertebral fractures compared to placebo.
All dosages significantly increased vertebral BMD compared to placebo.
Significant reductions in bone resorption markers were observed in 200 IU
and 400 IU groups.
Henriksen et al.
(2016)
37
4665
Daily 0.8mg oral
salmon calcitonin
vs placebo over 36
months
Oral salmon calcitonin did not significantly reduce vertebral and non-vertebral
fractures risk compared to placebo.
The treatment group experienced a significantly greater increase in lumbar
spine BMD than placebo but not in total hip or femoral neck BMD.
Bone resorption markers were significantly lower in oral calcitonin arm than
placebo arm at 12 and 24 months but not at 36 months.
Lyritis et al.
(1991)
42
56
Daily calcitonin 100 IU
vs placebo injections
for osteoporotic
vertebral fractures,
over 14 days
Calcitonin 100IU yielded significant reductions in pain (p<0.001) compared
to placebo. These were apparent as early as day 2 of the treatment period.
Urinary hydroxyproline and urinary calcium were significantly lower in the
calcitonin group.
Lyritis et al.
(1999)
43
40
Daily 200 IU calcitonin
suppositories vs
placebo over 28 days
Daily calcitonin suppositories demonstrated significant analgesic efficacy
on VAS scores compared to placebo in patients with recent osteoporotic
vertebral fractures.
Karponis et al.
(2015)
33
41
Daily 200 IU
intranasal calcitonin
vs placebo over 3
months
Nasal calcitonin demonstrates a statistically significant analgesic efficacy
after distal radius fractures when compared to placebo.
603http://www.ismni.org
A. Srinivasan et al.: Remembering calcitonin
week administration of daily 0.2mg recombinant salmon
calcitonin. At non-vertebral sites, changes in BMD at
the hip, femoral neck and trochanter sites were not
statistically different between oral, nasal calcitonin and
placebo groups in the ORACAL trial. At non-vertebral sites,
Henriksen et al. observed a reduction in hip and femoral
neck BMD in both treatment and placebo groups, but the
reduction was greater in the placebo group. In all studies,
a significant decrease in bone resorption markers was
observed, including C-terminal telopeptides of collagen
types I, II (CTX-I, CTX-II respectively) and N-terminal
cross-linked telopeptide of type I collagen (NTx-I)
37-39
. In
the ORACAL study, a significantly greater reduction in
CTx-I was reported in the oral calcitonin group than the
nasal calcitonin group. Henriksen et al. demonstrated that
bone resorption markers in the oral calcitonin group were
significantly lower than placebo at 6, 12 and 24 months,
but not at 36 months. Whether differences in biochemistry
translate to clinical significance, though, is a different
question and, frankly, the one that should be asked.
To date, most studies have been conducted on
postmenopausal women and very few have looked into
calcitonin’s therapeutic efficacy in other patient populations.
A trial conducted by Trovas et al. assessed the efficacy of
daily 200IU nasal calcitonin in 28 males with idiopathic
osteoporosis over 12 months. Nasal calcitonin was able to
significantly increase vertebral BMD compared to placebo
(mean ± standard error of the mean (SEM): 7.1±1.7%
and 2.4±1.5%, respectively, p<0.05). However, similar
improvements in BMD were not observed in the femoral
neck, trochanter and ward’s triangle following calcitonin
administration. Bone resorption markers, including CTX-1,
NTX-1 and urinary deoxypyridinoline, were also suppressed
and the reduction was significantly greater compared to
placebo. This study was not powered to analyse nasal
calcitonin’s anti-fracture efficacy
40
.
In the management of corticosteroid-induced
osteoporosis, a Cochrane review conducted by Cranney et al.
(including 9 trials) demonstrated that calcitonin was effective
in improving vertebral BMD at 12 months, with a weighted
mean difference of 3.2% (95% CI: 0.3 to 6.1) compared to
placebo. However, no significant difference was observed
at 24 months. Similar results were observed at the distal
radius, where calcitonin exerted a significant improvement in
BMD compared to placebo at 6 months only. No statistically
significant difference in BMD was observed at the femoral
neck compared to placebo. Additionally, no significant
difference in relative risk for vertebral and non-vertebral
fractures was observed in the treatment groups when
compared to placebo
41
. Table 1 summarizes key findings
from trials on the efficacy of calcitonin.
Current Indications
In the British National Formulary (BNF), calcitonin is
indicated for use in hypercalcaemia of malignancy, Paget’s
disease of bone, and the prevention of acute bone loss due
to sudden immobility. However, it is contraindicated in
hypocalcaemia, and factors such as heart failure, a history
of allergies and the risk of malignancy need to be taken
into account.
Although calcitonin is not approved by the European
Medicines Agency (EMA) for the treatment of postmenopausal
osteoporosis, it is FDA-approved for managing patients
who are at least 5 years postmenopausal and when the
alternatives are contraindicated. This was largely based on
the PROOF study in 2000 which showed a 30% reduction
in vertebral fracture occurrence in participants with prior
vertebral fractures
4
. However, due to meta-analyses
reporting a potential, albeit non-definitive, link between
salmon calcitonin and malignancy, it is no longer considered
to be the first-line treatment and should only be used
when the alternatives are contraindicated
44-46
. Calcitonin
may still be preferred for acute osteoporotic fractures as
several studies have observed significant analgesic effects
in the acute setting
33,42,43,46
. In this instance, calcitonin is
recommended for use until the pain resolves, at which point
it should be substituted with a more effective long-term drug.
Calcitonin is EMA and FDA-approved in the treatment of
hypercalcaemic emergencies. It is used due to its fast-acting
calcium-lowering effect which is useful when calcium levels
need to be lowered rapidly. After rehydrating the patient with
saline, calcitonin is co-administered with a bisphosphonate
and other calcium-lowering drugs such as loop diuretics.
The osteoclast-inhibiting effect of calcitonin administration
typically fades after 24-48 hours, but this coincides with
when bisphosphonates’ activity increases; as a result,
co-administration produces a rapid fall in calcium due to
calcitonin, and a sustained decrease over a few days from the
bisphosphonate.
For Paget’s disease of bone, calcitonin is authorised by
the EMA for short-term use and it is the FDA-approved
second-line treatment which should be administrated
when the treatment of choice, zoledronic acid, is not
tolerated or prompt surgery on the bone affected by the
disease is necessary. A study involving 85 participants
found that in the initial months of salmon calcitonin
therapy for Paget’s disease, the main markers of bone
remodelling and turnover (alkaline phosphatase and
urine hydroxyproline) decreased by approximately 50%.
However, 22 of these patients eventually returned to pre-
treatment levels despite continued treatment, and 19 of
those 22 had high titres of anti-calcitonin antibodies
47
.
This development of tolerance makes calcitonin a less
viable long-term option in the treatment of Paget’s disease
compared to bisphosphonates, which are not vulnerable to
antibodies. When administering calcitonin, serum alkaline
phosphatase needs to be measured every 3 to 6 months
until it normalises, after which it can be measured every 6
months; if levels rise again, antibody formation should be
suspected.
604http://www.ismni.org
A. Srinivasan et al.: Remembering calcitonin
Competitors in the treatment of postmenopausal
osteoporosis
In comparison to calcitonin, the literature surrounding the
anti-fracture efficacy of bisphosphonates has demonstrated
more promising data. Bisphosphonates are pyrophosphate
analogues, which exert their therapeutic effect by attaching
to bone. Active resorption of these areas of bone leads
to osteoclast inhibition via intracellular pathways
48,49
.
Alendronate and risedronate are the two most commonly
used bisphosphonates for postmenopausal osteoporosis
in the UK. Due to the long half-life of bisphosphonates,
convenient extended-interval dosing regimens, such as once
weekly or once monthly options, are available on the market
and some patient populations may also be eligible to undergo
drug holidays
50
. Numerous studies have demonstrated anti-
fracture efficacy in bisphosphonate use at both vertebral and
non-vertebral sites
51-54
. Specifically, the Fracture Intervention
Trial (FIT) highlighted that a daily alendronate regimen (5
mg/d for first 24 months, followed by 10 mg/d until 36
months) was able to induce a significant 50% reduction
in vertebral fractures and 30% reduction in wrist and hip
fractures in postmenopausal women with osteoporosis with
at least one previous vertebral fracture
51
. Furthermore,
femoral neck and lumbar spine BMD were also increased by
4.1% and 6.2%, respectively, throughout the study.
In the 3-year Vertebral Efficacy with Risedronate Therapy
(VERT) trial, Risedronate (5mg/d) was able to reduce
fracture incidence at both vertebral and non-vertebral sites
by 41 % (95% CI: 18%-58%) and 39% (95% CI: 6%-61%),
respectively, compared to placebo. Significant improvements
in BMD were observed at the lumbar spine (5.4%), femoral
neck (1.6%) and trochanter (3.3%) compared to placebo
(1.1%, -1.2%, -0.7%, respectively)
53
. In the Hip Intervention
Program (HIP) study, McClung et al. demonstrated that daily
risedronate over 3 years was able to significantly lower hip
fracture risk in elderly female patients with pre-diagnosed
osteoporosis, compared to placebo. (RR: 0.6; 95% CI:0.4 to
0.9; P=0.009)
54
.
Zoledronic acid is available as a convenient, once-
yearly IV infusion which may be used in cases where
oral bisphosphonates are contraindicated. In the Health
Outcomes and Reduced Incidence with Zoledronic Acid
Once Yearly (HORIZON) Pivotal Fracture Trial, annual
administration of zoledronic acid over 3-years demonstrated
both vertebral and hip anti-fracture efficacy when compared
to placebo (RR: 0.30; 95% CI: 0.24-0.38, HR: 0.59 95%
CI: 0.42-0.83, respectively)
8
. Additionally, in the HORIZON
Recurrent Fracture Trial, it was able to lower re-fracture
incidence in patients with previous hip fractures (HR:0.65
95% CI 0.50-0.84)
55
.
Ibandronate has been shown to exhibit vertebral anti-
fracture efficacy compared to placebo
1
but there is a lack of
strong evidence justifying its use for hip and non-vertebral
fractures. The BONE study (Ibandronate Osteoporosis
Vertebral Fracture Trial in North America and Europe)
was a 3-year trial conducted on 2946 postmenopausal
women which demonstrated that oral daily (2.5 mg/d) and
intermittent ibandronate (20 mg every other day for 12 doses
every 3 months) were able to reduce vertebral fracture rates
by 62% (p=0.0001) and 50% (p=0.0006), respectively,
compared to placebo. Significant improvements in vertebral
and hip BMD were also observed
56
.
Evidence directly comparing the efficacy of
bisphosphonates with calcitonin is limited. A study which
directly compared alendronate with salmon calcitonin
demonstrated a significantly greater increase in lumbar
spine (p<0.001), trochanter (p<0.001) and femoral neck
BMD (p<0.001) in patients who were administered the
bisphosphonate compared to salmon calcitonin over 12
months
57
. Moreover, a significantly greater reduction in
bone resorption markers within the alendronate group
was observed compared to calcitonin following 12 months
administration (p<0.001).
Denosumab, a fully human monoclonal antibody that
inhibits RANKL, is not regarded as the standard first-line
treatment for postmenopausal osteoporosis
58
. It has been
shown to be very powerful at countering bone resorption,
reducing fracture rates, increasing BMD and reducing serum
bone resorption markers
59,60
. Notably, the FREEDOM study
(Fracture Reduction Evaluation of Denosumab in Osteoporosis
Every 6 Months) was an international, multi-centre trial
which compared the efficacy of denosumab against placebo
in 7868 women with postmenopausal osteoporosis over
3 years
59
. The study showed that denosumab was able to
significantly reduce vertebral (RR: 0.32 95% CI: 0.26-
0.41) and hip (hazard ratio (HR): 0.60 95% CI: 0.37-0.97)
fracture risk, increase lumbar and hip BMD and suppress
bone resorption markers. An extension of the trial allowed
the monitoring of an additional 7 years for 4550 participants
from the FREEDOM study which demonstrated the long-
term maintenance of low fracture incidence and continued
rise in vertebral and non-vertebral BMD
60
. Yet, denosumab
discontinuation is associated with ‘rebound’ bone resorption
and anti-resorptive agents are often prescribed following
withdrawal to minimise this effect
61
.
Unlike the aforementioned anti-resorptive agents,
different “anabolic” options are also available on the market,
including teriparatide (PTH analogue) and abaloparatide
(PTHrP analogue). PTH has two contrasting functions on bone
turnover. Despite its net effect of bone resorption during
continuous administration, it promotes bone formation when
delivered intermittently
62
. In the UK, teriparatide is indicated
for the treatment for postmenopausal osteoporosis and in
males at high risk of fractures and corticosteroid-induced
osteoporosis. The use of abaloparatide is approved by the
FDA for the management of postmenopausal osteoporosis in
the US. However, evidence surrounding its use was deemed
insufficient by the EMA in 2017. In general, anabolic agents
are not first-line for the treatment of osteoporosis, owing to a
plethora of factors, such as the greater cost and inconvenient
administration (subcutaneous injection) compared to most
bisphosphonates
63
. Intriguingly, treatment withdrawal
605http://www.ismni.org
A. Srinivasan et al.: Remembering calcitonin
often results in a progressive decline in BMD which requires
management by anti-resorptive agents; this further limits
the use of anabolic agents
64
.
Teriparatide has been shown in numerous studies to
possess anti-fracture efficacy at vertebral and non-vertebral
sites
65-67
. Moreover, the Fracture Prevention Trial was an
international, multi-centre randomized control trial which
demonstrated that a daily regimen of 20 μg or 40 μg injection
over 21 months considerably reduced new vertebral fracture
risk by 65% (RR: 0.35 95%CI: 0.22-0.55) and 69% (RR:
0.31 95%CI: 0.19-0.50) compared to placebo, respectively
65
.
Non-vertebral fracture RR in the 20 μg and 40 μg arms were
0.47 (95%CI: 0.25-0.88) and 0.46 (95%CI: 0.25-0.861),
respectively. Daily 20 μg and 40 μg teriparatide were able
to induce a dose-dependent and statistically significant
increase in BMD measured at multiple sites including
the lumbar spine, hip and femoral neck. Studies directly
comparing teriparatide and calcitonin have shown a greater
increase in lumbar BMD in patients treated with teriparatide
compared to salmon calcitonin
68 -70
. However, data comparing
their efficacy on femoral neck and total hip BMD data has
been inconsistent. Specifically, trials have shown an increase
in serum bone formation markers including bone-specific
alkaline phosphatase and osteocalcin in participants taking
teriparatide but not in the calcitonin group
68 -70
. However, it is
important to question the external validity of these studies,
which were mostly conducted on Asian populations and
hence, a clinical benefit remains equivocal when generalising
these findings to other populations.
Discussion
The decline in the use of calcitonin can be attributed to a
variety of causes. There are issues intrinsic to the drug itself,
including an association with cancer, and the lack of a widely-
available oral formulation
2,4
. Additionally, there are extrinsic
factors: primarily, the development of alternative drugs with
greater efficacy.
Firstly, there is the potential risk of malignancy associated
with calcitonin use. This was first brought to attention
by the PROOF trial of nasal salmon calcitonin, where the
results showed a higher risk of cancer in the active group
(8.9%) compared to the placebo group (5.1%), with basal
cell carcinomas being the major finding
4
. At the time, no
action was taken by the FDA because the age and race of
the participants were confounding factors, but the potential
association was flagged up again after phase III trials for
an oral formulation of calcitonin (SMC021) which reported
cases of prostate cancer
71,72
. In addition to these findings, a
1994 study demonstrated a biological route through which
calcitonin might stimulate the growth of prostate cancer
cells
73
. Since then, the EMA commissioned a committee
to review all of the salmon calcitonin studies and, in July
2012, stated that salmon calcitonin should no longer be
recommended as a treatment option for postmenopausal
osteoporosis, citing an increased malignancy risk as one of
the paramount reasons. In March 2013, an advisory panel
to the FDA also recommended that calcitonin should not
be indicated for postmenopausal osteoporosis because the
safety risk outweighed the fracture reduction. Although the
FDA did not implement this recommendation, all calcitonin
nasal spray products were withdrawn by Health Canada in
October 2013 and two months later, the Taiwan FDA followed
suit. Furthermore, a case-control study involving 28222
osteoporotic patients in Taiwan found that using a calcitonin
nasal spray in women significantly elevated the risk of liver
cancer, although the risk of breast cancer was reduced
74
.
Another reason for the decline in use is that salmon
calcitonin carries significant side-effects, many of which
are related to the gastrointestinal tract such as abdominal
pain, diarrhoea, nausea and vomiting in addition to
arthralgia, musculoskeletal pain and flushing. Studies
investigating the short-term and long-term side-effects
of calcitonin have found that the unwanted effects of the
nasal spray preparation were a lot milder and had a lower
incidence compared to the subcutaneous injectable form of
calcitonin
17
. However, there were some side-effects specific
to the nasal spray including nasal irritation, sneezing and
rhinitis. As previously mentioned, randomised control
trials involving oral calcitonin found it to be well-tolerated
in general but it was still associated with hot flushes and
gastrointestinal symptoms like nausea and dyspepsia. In the
ORACAL trial, a higher incidence of nausea and dyspepsia
was reported in participants who took the oral calcitonin
than the nasal spray group
38
.
One of the most important factors which diverted
healthcare professionals away from the use of calcitonin
was the lack of strong evidence justifying its use, along
with the simultaneous presence of other treatment options
which demonstrated more promising data showing superior
efficacy in increasing multifocal BMD and anti-fracture
efficacy. Whilst the PROOF study was able to illustrate
calcitonin’s anti-fracture efficacy, many other studies did not
show consistent results. In most studies, the fracture rate
was either too low to allow meaningful statistical analysis
or that statistical analysis did not show a significant change
in vertebral fracture rates from calcitonin use. Although
calcitonin has been shown in various trials to increase lumbar
spine BMD and decrease bone resorption markers, including
CTX-I and CTX-II, the clinical utility of using BMD and serum
bone resorption markers in predicting fracture risks have
been questioned
75-77
.
Future direction
In the future, it is likely that calcitonin will continue to be
used in combination with bisphosphonates for the treatment
of emergency hypercalcaemia and hypercalcaemia of
malignancy due to its rapid action. It is unlikely that calcitonin
will become the first-line treatment for postmenopausal
osteoporotic fractures or Paget’s disease, mainly due to the
proven greater efficacy of bisphosphonates. However, its
606http://www.ismni.org
A. Srinivasan et al.: Remembering calcitonin
powerful analgesic effect should not be overlooked and if a
safe oral formulation that can achieve adequate bioavailability
is found, which would potentially be more beneficial and
acceptable to patients than the nasal spray, then there could
be renewed interest in this drug.
The efficacy of calcitonin in treating acute pain associated
with fractures has already been discussed, but calcitonin
may also be a useful alternative in the treatment of acute
and chronic neuropathic pain with recent studies having been
conducted to look at the potential mechanisms behind this.
This benefit of calcitonin has previously been demonstrated
in the treatment of phantom limb pain where a double-
blinded crossover study showed efficacy in the early post-
operative period
78
. Additionally, a case report by Visser et al.
illuminated a patient’s recovery from post-herpetic neuralgia
after being administered calcitonin following the failure of
traditional analgesics, and a 2011 case series highlighted
the role of calcitonin in treating acute neuropathic pain
associated with spinal cord injury
79,80
. Other reports which
have indicated potential uses of calcitonin in neuropathic pain
include diabetic neuropathy, lumbar spinal canal stenosis,
reflex sympathetic dystrophy, post-operative pain and
trigeminal neuralgia
81
.
Finally, in addition to calcitonin’s effects on reducing bone
resorption, a 2005 study found that an oral form of salmon
calcitonin significantly reduced the urinary excretion of CTx-II,
suggesting that it could decrease the degradation of cartilage
and act as a treatment for osteoarthritis
82
. Furthermore,
calcitonin has been shown to reduce the levels of rheumatoid
factor and interleukin-1b in patients with rheumatoid arthritis,
but ancillary work is necessary at this stage to establish it
in the armamentarium of anti-rheumatic medications
83,84
.
This is another example of how successfully creating a viable
oral formulation of calcitonin could be key to the future of
the drug, although achieving adequate concentrations in the
joints of interest may be another challenge.
Conclusion
Since its discovery by Douglas Harold Copp, calcitonin
has been a useful treatment for various metabolic bone
diseases. However, similar to many other medications, it has
been derailed by associations with cancer and superseded
by newer and more potent alternatives. There will still be a
role for calcitonin when patients are unsuitable candidates
for first-line therapies. Furthermore, the development of
an oral formulation could herald a new interest in the drug.
Although calcitonin will be used less for its original purpose
of increasing BMD and reducing fracture risk, its unique
analgesic efficacy means that there may still be a future for
this old friend.
References
1. Copp DH, Cameron EC. Demonstration of a
Hypocalcaemic Factor (Calcitonin) in Commercial
Parathyroid Extract. Science 1961;134(3495):2038.
2. Maricic MJ. Oral Calcitonin. Curr Osteoporos Rep 2012;
10:80 -85.
3. Azria M, Copp DH, Zanelli JM. 25 Years of salmon
calcitonin: From synthesis to therapeutic use. Calcif
Tissue Int 1995;57:405-408.
4. Chesnut CH, Silverman S, Andriano K, Genant H. A
randomized trial of nasal spray salmon calcitonin in
postmenopausal women with established osteoporosis:
the prevent recurrence of osteoporotic fractures study.
PROOF Study Group. Am J Med 2000;109(4):267-276.
5. Knopp-Sihota JA, Newburn-Cook CV, Homik J,
Cummings GG, Voaklander D. Calcitonin for Treating
Acute and Chronic Pain of Recent and Remote
Osteoporotic Vertebral Compression Fractures: A
Systematic Review and Meta-Analysis. Osteoporos Int
2012;23(1):17-38.
6. Copp DH. Calcitonin: Discovery, Development, and Clinical
Application. Clin Invest Med 1994;17(3):268-277.
7. Waalen J. Current and emerging therapies for the
treatment of osteoporosis. J Exp Pharmacol 2010;
2:121-134 .
8. Black DM, Delmas PD, Eastell R, et al. Once-Yearly
Zoledronic Acid for Treatment of Postmenopausal
Osteoporosis. N Engl J Med 2007;356(18):1809-1822.
9. Andreotti G, Méndez BL, Amodeo P, Morelli MAC,
Nakamuta H, Motta A. Structural Determinants of
Salmon Calcitonin Bioactivity-The Role of the Leu Based
Amphiphatic α-helix. J Biol Chem 2006;281:24193-
24203.
10. Green FR, Lynch B, Kaiser ET. Biological and physical
properties of a model calcitonin containing a glutamate
residue interrupting the hydrophobic face of the idealized
amphiphilic alpha-helical region. Proc Natl Acad Sci USA
1987;84:8340-8344.
11. Felsenfeld AJ, Levine BS. Calcitonin, the forgotten
hormone: does it deserve to be forgotten? Clin Kidney J
2015;8(2):180-187.
12. Horne WC, Shyu J, Chakraborty M, Baron R. Signal
transduction by calcitonin. Trends Endocrinol Metab
1994;5(10):395-401.
13. Yeh CB, Weng SJ, Chang KW, Chan JY, Huang SM, Chu
TH. Calcitonin Alleviates Hyperalgesia in Osteoporotic
Rats by Modulating Serotonin Transporter Activity.
Osteoporos Int 2016;27(11):3355-3364.
14. Terashima Y, Takebayashi T, Jimbo S, et al. Analgesic
effects of calcitonin on radicular pain in male rats. J Pain
Res 2019;12:223-230.
15. Karsal MA, Henriksen K, Bay-Jensen AC, Molloy
B, Arnold M, John MR. Lessons Learned From the
Development of Oral Calcitonin: The First Tablet
Formulation of a Protein in Phase III Clinical Trials. J
Clin Physiol 2011;51(4):460-471.
16. Buclin T, Rochat MC, Burckhardt P, Azria M, Attinger
M. Bioavailability and Biological Efficacy of a New Oral
Formulation of Salmon Calcitonin in Healthy Volunteers.
J Bone Miner Res 2002;17(8):1478-1485.
607http://www.ismni.org
A. Srinivasan et al.: Remembering calcitonin
17. Wimalawansa SJ. Long- and short-term side effects and
safety of calcitonin in man: a prospective study. Calcif
Tissue Int 1993;52(2):90-93.
18. Wüster C, Schurr W, Scharla S, Raue F, Minne HW, Ziegler
R. Superior local tolerability of human versus salmon
calcitonin preparations in young healthy volunteers. Eur
J C lin Ph ar ma col 1991;41(3):211-215.
19. Combe B, Cohen C, Aubin F. Equivalence of nasal spray
and subcutaneous formulations of salmon calcitonin.
Calcif Tissue Int 1997;61(1):10-5.
20. Reginster JY, Franchimont P. Side effects of synthetic
salmon calcitonin given by intranasal spray compared
with intramuscular injection. Clin Exp Rheumatol 1985;
3(2):155-157.
21. Kanis JA, Johnell O, Gullberg B, et al. Evidence
for efficacy of drugs affecting bone metabolism in
preventing hip fracture. BMJ 1992;305(6862):1124-8.
22. Tafazal SI, Ng L, Sell P. Randomised placebo-controlled
trial on the effectiveness of nasal salmon calcitonin in
the treatment of lumbar spinal stenosis. Eur Spine J
2007;16(2):207-212.
23. Bandeira L, Lewiecki EM, Bilezikian JP.
Pharmacodynamics and pharmacokinetics of oral
salmon calcitonin in the treatment of osteoporosis.
Expert Opin Drug Metab Toxicol 2016;12(6):681-9.
24. Thamsborg G, Jensen JE, Keollerup G, Hauge EM,
Melsen F, Sørensen OH. Effect of nasal salmon calcitonin
on bone remodelling and bone mass in postmenopausal
os te opo rosis. Bo ne 1996;18(2):207-212.
25. Overgaard K, Hansen MA, Jensen SB, Christiansen C.
Effect of salcatonin given intranasally on bone mass
and fracture rates in established osteoporosis: a dose-
response study. BMJ 1992;305(6853):556.
26. Reginster JY, Deroisy R, Lecart MP, et al. A double-
blind, placebo-controlled, dose-finding trial of
intermittent nasal salmon calcitonin for prevention of
postmenopausal lumbar spine bone loss. Am J Med
1995;98(5):452-8.
27. Kaskani E, Lyritis GP, Kosmidis C, et al. Effect of
intermittent administration of 200 IU intranasal
salmon calcitonin and low doses of 1alpha(OH) vitamin
D3 on bone mineral density of the lumbar spine and
hip region and biochemical bone markers in women
with postmenopausal osteoporosis: a pilot study. Clin
Rheumatol 2005;24(3):232-238.
28. Tascioglu F, Colak O, Armagan O, Alatas O, Oner C. The
treatment of osteoporosis in patients with rheumatoid
arthritis receiving glucocorticoids: a comparison
of alendronate and intranasal salmon calcitonin.
Rheumatol Int 2005;26(1):21-29.
29. Thamsborg G, Storm TL, Sykulski R, Brinch E, Nielsen HK,
Sørensen OH. Effect of different doses of nasal calcitonin
on bone mass. Calcif Tissue Int 1991;48(5):302-307.
30. Dursun N, Dursun E, Yalçin S. Comparison of alendronate,
calcitonin and calcium treatments in postmenopausal
osteoporosis. Int J Clin Pract 2001;55(8):505-509.
31. Ishida Y, Kawai S. Comparative efficacy of hormone
replacement therapy, etidronate, calcitonin, alfacalcidol,
and vitamin K in postmenopausal women with
osteoporosis: The Yamaguchi Osteoporosis Prevention
Study. Am J Med 2004;117(8):549-555.
32. Flicker L, Hopper JL, Larkins RG, Lichtenstein M, Buirski
G, Wark JD. Nandrolone decanoate and intranasal
calcitonin as therapy in established osteoporosis.
Oste opo ros I nt 1997;7(1):29-35.
33. Karponis A, Rizou S, Pallis D, Zafeiris CP, Georgiou DF,
Galanos A. Analgesic Effect of Nasal Salmon Calcitonin
During the Early Post-Fracture Period of the Distal
Radius Fracture. J Musculoskelet Neuronal Interact
2015;15(2):186-189.
34. Khan AA, Morrison A, Hanley DA, et al. Diagnosis and
management of osteonecrosis of the jaw: a systematic
review and international consensus. J Bone Miner Res
2015;30(1):3-23.
35. Gedmintas L, Solomon DH, Kim SC. Bisphosphonates
and risk of subtrochanteric, femoral shaft and atypical
femur fracture: a systematic review and meta-analysis.
J Bone Miner Res 2013;28(8):1729-1737.
36. Aguirre TAS, Aversa V, Rosa M, et al. Coated minispheres
of salmon calcitonin target rat intestinal regions to
achieve systemic bioavailability: Comparison between
intestinal instillation and oral gavage. J Control Release
2016;238:242-252.
37. Henriksen K, Byrjalsen I, Andersen JR, et al. A
randomized, double-blind, multicenter, placebo-
controlled study to evaluate the efficacy and safety of
oral salmon calcitonin in the treatment of osteoporosis
in postmenopausal women taking calcium and vitamin
D. B o n e 2016 ;91:122-129.
38. Binkley N, Bolognese M, Sidorowicz-Bialynicka A,
et al. A phase 3 trial of the efficacy and safety of
oral recombinant calcitonin: The oral calcitonin in
postmenopausal osteoporosis (ORACAL) trial. J Bone
Miner Res 2012;27(8):1821-1829.
39. Binkley N, Bone H, Gilligan JP, Krause DS. Efficacy
and safety of oral recombinant calcitonin tablets in
postmenopausal women with low bone mass and
increased fracture risk: a randomized, placebo-
controlled trial. Osteoporos Int 2014;25(11):2649-56.
40. Trovas GP, Lyritis GP, Galanos A, Raptou P, Constantelou
E. A randomized trial of nasal spray salmon calcitonin
in men with idiopathic osteoporosis: effects on bone
mineral density and bone markers. J Bone Miner Res
20 02;17(3):521-7.
41. Cranney A, Welch V, Adachi JD, Homik J, Shea B,
Suarez-Almazor ME et al. Calcitonin for the treatment
and prevention of corticosteroid-induced osteoporosis.
Cochrane Database Syst Rev 2000;(2):CD00198
42. Lyritis GP, Tsakalakos N, Magiasis B, Karachalios T,
Yiatzides A, Tsekoura M. Analgesic effect of salmon
calcitonin in osteoporotic vertebral fractures: a double-
blind placebo-controlled clinical study. Calcif Tissue Int
1991;49(6):369-372.
43. Lyritis GP, Ioannidis GV, Karachalios T, Roidis N,
608http://www.ismni.org
A. Srinivasan et al.: Remembering calcitonin
Kataxaki E, Papaioannou N, Kaloudis J, Galanos A.
Analgesic effect of salmon calcitonin suppositories
in patients with acute pain due to recent osteoporotic
vertebral crush fractures: a prospective double-blind,
randomized, placebo-controlled clinical study. Clin J
Pain 1999;15(4):284-289.
44. Cosman F, Beur SJ, LeBoff MS, et al. Clinician’s Guide to
Prevention and Treatment of Osteoporosis. Osteoporos
Int 2014;25(10):2359-2381.
45. Heep, M, Lesperance, S, Gasser, J, Chen, C, Aftring, R.
Calcitonin use and risk of malignancy: a meta-analysis of
17 RCTs in patients with osteoporosis. J Bone Miner Res
2012;27(S1): S 48 6.
46. Levernieux J, Bordier P, Julien D, Caulin F. The effect of
calcitonin on bone pain and acute resorption related to
recent osteoporotic fractures. Result of a double blind
and open study. In: Cecchettin M, Segre G, editors. 5th
International Congress on Calciotropic Hormones and
Calcium Metabolism. Venice: Elsevier Science Publishers
(Biomedical Division); 1985. p.171-178.
47. Singer FR, Fredericks RS, Minkin C. Salmon calcitonin
therapy for Paget’s disease of bone the problem of
acquired clinical resistance. Arthritis Rheumatol 1980;
2 3 (10 ) :114 8 -115 4 .
48. Nancollas GH, Tang R, Phipps RJ, et al. Novel insights
into actions of bisphosphonates on bone: differences
in interactions with hydroxyapatite. Bone 2006;
38(5):617-27.
49. Luckman SP, Hughes DE, Coxon FP, Graham R, Russell G,
Rogers MJ. Nitrogen-containing bisphosphonates inhibit
the mevalonate pathway and prevent post-translational
prenylation of GTP-binding proteins, including Ras. J
Bone Miner Res 1998;13(4):581-589.
50. Camacho PM, Petak SM, Binkley N, Clarke BL, Harris
ST, Hurley DL, et al. American Association of Clinical
Endocrinologists and American College of Endocrinology
Clinical Practice guidelines for the diagnosis and
treatment of postmenopausal osteoporosis - 2016.
Endocr Pract 2016;22(Suppl 4):1-42.
51. Black DM, Cummings SR, Karpf DB, et al. Randomised
trial of effect of alendronate on risk of fracture in women
with existing vertebral fractures. Fracture Intervention
Trial Research Group. Lancet 1996;348(9041):1535.
52. Black DM, Schwartz AV, Ensrud KE, et al. Effects of
continuing or stopping alendronate after 5 years of
treatment: the Fracture Intervention Trial Long-term
Extension (FLEX): a randomized trial. JAMA 2006;
296(24):2927-2938.
53. Harris ST, Watts NB, Genant HK, et al. Effects of
risedronate treatment on vertebral and nonvertebral
fractures in women with postmenopausal osteoporosis:
a randomized controlled trial. Vertebral Efficacy With
Risedronate Therapy (VERT) Study Group. JAMA 1999;
282(14):1344-52.
54. McClung MR, G,eusens P, Miller PD, et al. Effect of
risedronate on the risk of hip fracture in elderly women.
Hip Intervention Program Study Group. N Engl J Med
2001;344(5):333-340.
55. Lyles KW, Colón-Emeric CS, Magaziner JS, Adachi JD,
Pieper CF, Mautalen C et al. Zoledronic acid and clinical
fractures and mortality after hip fracture. N Engl J Med
2007;357(18):1799-809.
56. Chesnut CH 3
rd
, Skag A, Christiansen C, Recker
R, Stakkestad JA, Hoiseth A, et al. Effects of oral
ibandronate administered daily or intermittently on
fracture risk in postmenopausal osteoporosis. J Bone
Miner Res 2004;19(8):1241-9.
57. Downs RW Jr, Bell NH, Ettinger MP, et al. Comparison
of alendronate and intranasal calcitonin for treatment
of osteoporosis in postmenopausal women. J Clin
Endocrinol Metab 2000;85(5):1783-1788.
58. McClung MR, Lewiecki EM, Cohen SB, et al. Denosumab
in postmenopausal women with low bone mineral
density. N Engl J Med 2006;354(8):821-831.
59. Cummings SR, San Martin J, McClung MR, et
al. Denosumab for prevention of fractures in
postmenopausal women with osteoporosis. N Engl J
Med 2009;361(8):756-765.
60. Bone HG, Wagman RB, Brandi ML, et al. 10 years of
denosumab treatment in postmenopausal women with
osteoporosis: results from the phase 3 randomised
FREEDOM trial and open-label extension. Lancet
Diabetes Endocrinol 2017;5(7):513-523.
61. Miller PD, Bolognese MA, Lewiecki EM, McClung MR,
Ding B, Austin M et al. Effect of denosumab on bone
density and turnover in postmenopausal women with
low bone mass after long-term continued, discontinued,
and restarting of therapy: a randomized blinded phase 2
clinical trial. Bone 2008;43(2):222-9.
62. Etoh M, Yamaguchi A. Repetition of continuous PTH
treatments followed by periodic withdrawals exerts
anabolic effects on rat bone. J Bone Miner Res 2010;
28(6):641-649.
63. Liu H, Michaud K, Nayak S, Karpf DB, Owens DK, Garber
AM. The cost-effectiveness of therapy with teriparatide
and alendronate in women with severe osteoporosis.
Arch Intern Med 2006;166(11):1209-1217.
64. Black DM, Bilezikian JP, Ensrud KE, et al. One year of
alendronate after one year of parathyroid hormone (1-
84) for osteoporosis. N Engl J Med 2005;353(6):555-
565.
65. Neer RM, Arnaud CD, Zanchetta JR, et al. Effect of
parathyroid hormone (1-34) on fractures and bone
mineral density in postmenopausal women with
osteoporosis. N Engl J Med 2001;344(19):1434-1441.
66. Lindsay R, Scheele WH, Neer R, et al. Sustained vertebral
fracture risk reduction after withdrawal of teriparatide in
postmenopausal women with osteoporosis. Arch Intern
Med 2004;164(18):2024-2030.
67. Lindsay R, Nieves J, Formica C, et al. Randomised
controlled study of effect of parathyroid hormone
on vertebral-bone mass and fracture incidence
among postmenopausal women on oestrogen with
osteoporosis. Lancet 1997;350(9077):550-555.
609http://www.ismni.org
A. Srinivasan et al.: Remembering calcitonin
68. Kung AW, Pasion EG, Sofiyan M, et al. A comparison of
teriparatide and calcitonin therapy in postmenopausal
Asian women with osteoporosis: a 6-month study. Curr
Med Res Opin 2006;22(5):929-937.
69. Hwang JS, Tu ST, Yang TS, Chen JF, Wang CJ, Tsai KS.
Teriparatide vs. calcitonin in the treatment of Asian
postmenopausal women with established osteoporosis.
Osteoporos Int 2006;17(3):373-378.
70. Lu C, Chen Y, Zhang B, Chen Y, Bai F, Chen D. Response
to teriparatide in Chinese patients with established
osteoporosis: osteocalcin and lumbar spine bone-
mineral density changes from teriparatide Phase III
st u d y. Cl i n I n te r v Ag i n g 2017;12:1717-1723.
71. ClinicalTrials.gov [Internet]. Identifier NCT00486434.
Efficacy and Safety of Oral Salmon Calcitonin in Patients
With Knee Osteoarthritis. 2007 Available from: https://
clinicaltrials.gov/ct2/show/results/NCT00486434
72. ClinicalTrials.gov [Internet]. Identifier NCT00704847.
Efficacy and Safety of Oral Salmon Calcitonin in
Patients With Knee Osteoarthritis (OA 2 Study).
2008 Available from: https://clinicaltrials.gov/ct2/
show/results/NCT00704847
73. Shah GV, Rayford W, Noble MJ, et al. Calcitonin
stimulates growth of human prostate cancer cells
through receptor-mediated increase in cyclic adenosine
3,5’-monophosphates and cytoplasmic Ca2+ transients.
Endocrinology 1994;134(2):596-602.
74. Sun L, Lin M, Muo C, Liang J, Kao C. Calcitonin Nasal
Spray and Increased Cancer Risk: A Population-Based
Nested Case-Control Study. J Clin Endocrinol Metab
2014;99(11):4259-4264.
75. Leslie WD, Morin SN, Lix LM. Rate of bone density
change does not enhance fracture prediction in
routine clinical practice. J Clin Endocrinol Metab
2012; 97(4):1211-1218.
76. Berry SD, Samelson EJ, Pencina MJ, et al. Repeat bone
mineral density screening and prediction of hip and major
os te op o r oti c f ra c tu r e. JA M A 2013;310 (12):1256 -1262 .
77. Melton LJ 3rd, Crowson CS, O’Fallon WM, Wahner HW,
Riggs BL. Relative contributions of bone density, bone
turnover, and clinical risk factors to long-term fracture
prediction. J Bone Miner Res 2003;18(2):312-318.
78. Jaeger H, Maier C. Calcitonin in Phantom Limb Pain: A
Double-Blind Study. Pain 1992;48(1):21-27.
79. Visser EJ, Kwei PL. Salmon Calcitonin in the Treatment
of Post Herpetic Neuralgia. Anaesth Intensive Care
2006;34(5):668-671.
80. Humble SR. Calcitonin for Acute Neuropathic Pain
Associated With Spinal Cord Injury. Anaesth Intensive
Care 2011;39(4):682-686.
81. Yazdani J, Khiavi RK, Ghavimi MA, Mortazavi A, Hagh
EJ, Ahmadpour F. Calcitonin as an analgesic agent:
review of mechanisms of action and clinical applications.
Rev Bras Anestesiol 2019;69(6):594-604.
82. Bagger YZ, Tankó LB, Alexandersen P, et al. Oral salmon
calcitonin induced suppression of urinary collagen type II
degradation in postmenopausal women: a new potential
treatment of osteoarthritis. Bone 2005;37(3):425-430.
83. Karponis D. Rheumatoid arthritis: the journey in pursuit
of a cure. Rheumatol Adv Pract 2017;1(1):rkx008.
84. Aida S, Okawa-Takatsuji M, Aotsuka S, Shimoji
K, Yokohari R. Calcitonin inhibits production of
immunoglobulins, rheumatoid factor and interleukin-1
by mononuclear cells from patients with rheumatoid
arthritis. Ann Rheum Dis 1994;53(4):247-249.