Osteoporosis

Back

Practice Essentials

Osteoporosis (see the image below) is the most common metabolic bone disease in the United States and can result in devastating physical, psychosocial, and economic consequences. It is often overlooked and undertreated, however, in large part because it is clinically silent before manifesting as fracture.



View Image

Osteoporosis of the spine. Observe the considerable reduction in overall vertebral bone density and note the lateral wedge fracture of L2.

See Menopause: Changes and Challenges, a Critical Images slideshow, to help identify comorbidities and diseases in the postmenopausal population.

Signs and symptoms

Osteoporosis generally does not become clinically apparent until a fracture occurs. Two thirds of vertebral fractures are painless. Typical findings in patients with painful vertebral fractures may include the following:

Patients who have sustained a hip fracture may experience the following:

On physical examination, patients with vertebral compression fractures may demonstrate the following:

Patients with hip fractures may demonstrate the following:

Patients with Colles fractures may have the following:

Patients with pubic and sacral fractures may have the following:

Balance difficulties may be evident, especially in patients with an altered center of gravity from severe kyphosis.[1] Patients may have difficulty performing tandem gait and performing single limb stance.

See Presentation for more detail.

Diagnosis

Baseline laboratory studies include the following:

Bone mineral density (BMD) measurement is recommended in the following patients[2] :

Dual-energy x-ray absorptiometry (DXA) is currently the criterion standard for the evaluation of BMD.[3, 4] Peripheral DXA is used to measure BMD at the wrist; it may be most useful in identifying patients at very low fracture risk who require no further workup.

DXA provides the patient’s T-score, which is the BMD value compared with that of control subjects who are at their peak BMD.[5, 6, 7, 8] World Health Organization (WHO) criteria define a normal T-score value as within 1 standard deviation (SD) of the mean BMD value in a healthy young adult. Values lying farther from the mean are stratified as follows[7] :

DXA also provides the patient’s Z-score, which reflects a value compared with that of persons matched for age and sex. Z-scores adjusted for ethnicity or race should be used in the following patients:

Z-score values of –2.0 SD or lower are defined as "below the expected range for age" and those above –2.0 SD as "within the expected range for age." The diagnosis of osteoporosis in these groups should not be based on densitometric criteria alone.

Quantitative calcaneal ultrasonography offers the following benefits[9] :

However, no diagnostic criteria based on quantitative ultrasonography or a combination of quantitative ultrasonography and DXA have been defined.

The National Osteoporosis Foundation (NOF) recommends vertebral imaging for the following patients[2] :

Vertebral imaging is also recommended for postmenopausal women and men age 50 and older with the following specific risk factors:

If bone density testing is not available, vertebral imaging may be considered based on age alone.

Other plain radiography features and recommendations are as follows:

See Workup for more detail.

Management

Lifestyle modification for prevention of osteoporotic fractures includes the following[10] :

The NOF recommends that pharmacologic therapy should be reserved for postmenopausal women and men aged 50 years or older who present with the following[2] :

Guidelines from the American Association of Clinical Endocrinologists include the following recommendations for choosing drugs to treat osteoporosis[11] :

Guidelines from the American College of Rheumatology for the treatment of glucocorticoid- induced osteoporosis include the following[12] :

Other approved agents include the following:

Medical care also includes the identification and treatment of potentially treatable underlying causes of osteoporosis such as hyperparathyroidism and hyperthyroidism. Surgical care in selected patients may include vertebroplasty and kyphoplasty, which are minimally invasive spine procedures used for the management of painful osteoporotic vertebral compression fractures.

See Treatment and Medication for more detail.

Background

Osteoporosis, a chronic, progressive disease of multifactorial etiology (see Etiology), is the most common metabolic bone disease in the United States. It has been most frequently recognized in elderly white women, although it does occur in both sexes, all races, and all age groups. Screening at-risk populations is essential (see Workup).

Osteoporosis is a systemic skeletal disease characterized by low bone mass and microarchitectural deterioration of bone tissue, with a consequent increase in bone fragility.[13] The disease often does not become clinically apparent until a fracture occurs.

Osteoporosis represents an increasingly serious health and economic problem in the United States and around the world.[14] Many individuals, male and female, experience pain, disability, and diminished quality of life as a result of having this condition.

Despite the adverse effects of osteoporosis, it is a condition that is often overlooked and undertreated, in large part because it is so often clinically silent before manifesting in the form of fracture. For example, a Gallup survey performed by the National Osteoporosis Foundation revealed that 86% of women with osteoporosis had never discussed its prevention with their physicians.[15] Failure to identify at-risk patients, to educate them, and to implement preventive measures may lead to tragic consequences.

Medical care includes calcium, vitamin D, and antiresorptive agents such as bisphosphonates, the selective estrogen receptor modulator (SERM) raloxifene, calcitonin, and denosumab. One anabolic agent, teriparatide (see Medication), is available as well. Surgical care includes vertebroplasty and kyphoplasty (see Treatment).

Osteoporosis is a preventable disease that can result in devastating physical, psychosocial, and economic consequences. Prevention and recognition of the secondary causes of osteoporosis are first-line measures to lessen the impact of this condition (see the images below).



View Image

Osteoporosis. Lateral radiograph demonstrates multiple osteoporotic vertebral compression fractures. Kyphoplasty has been performed at one level.



View Image

Osteoporosis of the spine. Note the lateral wedge fracture in L3 and the central burst fracture in L5. The patient had suffered a recent fall.

WHO definition of osteoporosis

Bone mineral density (BMD) in a patient is related to peak bone mass and, subsequently, bone loss. Whereas the T-score is the patient’s bone density compared with the BMD of control subjects who are at their peak BMD, the Z-score reflects a bone density compared with that of patients matched for age and sex.[5, 6, 7, 8]

The World Health Organization’s (WHO) definitions of osteoporosis based on BMD measurements in white women are summarized in Table 1, below.[7, 8] For each standard deviation (SD) reduction in BMD, the relative fracture risk is increased 1.5-3 times.

The WHO definition applies to postmenopausal women and men aged 50 years or older. Although these definitions are necessary to establish the prevalence of osteoporosis, they should not be used as the sole determinant of treatment decisions. This diagnostic classification should not be applied to premenopausal women, men younger than 50 years, or children.

Table 1. WHO Definition of Osteoporosis Based on BMD Measurements by DXA



View Table

See Table

Z-scores should be used in premenopausal women, men younger than 50 years, and children. Z-scores adjusted for ethnicity or race should be used, with Z-scores of –2.0 or lower defined as "below the expected range for age" and with Z-scores above –2.0 being defined as "within the expected range for age." The diagnosis of osteoporosis in these groups should not be based on densitometric criteria alone.

For more information, see the following:

Pathophysiology

It is increasingly being recognized that multiple pathogenetic mechanisms interact in the development of the osteoporotic state. Understanding the pathogenesis of osteoporosis starts with knowing how bone formation and remodeling occur.

Normal bone formation and remodeling

Bone is continually remodeled throughout our lives in response to microtrauma. Bone remodeling occurs at discrete sites within the skeleton and proceeds in an orderly fashion, and bone resorption is always followed by bone formation, a phenomenon referred to as coupling.

Dense cortical bone and spongy trabecular or cancellous bone differ in their architecture but are similar in molecular composition. Both types of bone have an extracellular matrix with mineralized and nonmineralized components. The composition and architecture of the extracellular matrix is what imparts mechanical properties to bone. Bone strength is determined by collagenous proteins (tensile strength) and mineralized osteoid (compressive strength).[17] The greater the concentration of calcium, the greater the compressive strength. In adults, approximately 25% of trabecular bone is resorbed and replaced each year, compared with only 3% of cortical bone.

Osteoclasts, derived from hematopoietic precursors, are responsible for bone resorption, whereas osteoblasts, from mesenchymal cells, are responsible for bone formation (see the images below). The 2 types of cells are dependent on each other for production and linked in the process of bone remodeling.

Osteoblasts not only secrete and mineralize osteoid but also appear to control the bone resorption carried out by osteoclasts. Osteocytes, which are terminally differentiated osteoblasts embedded in mineralized bone, direct the timing and location of bone remodeling. In osteoporosis, the coupling mechanism between osteoclasts and osteoblasts is thought to be unable to keep up with the constant microtrauma to trabecular bone. Osteoclasts require weeks to resorb bone, whereas osteoblasts need months to produce new bone. Therefore, any process that increases the rate of bone remodeling results in net bone loss over time.[18]



View Image

This image depicts bone remodeling with osteoclasts resorbing one side of a bony trabecula and osteoblasts depositing new bone on the other side.



View Image

Osteoclast, with bone below it. This image shows typical distinguishing characteristics of an osteoclast: a large cell with multiple nuclei and a "foa....



View Image

In this image, several osteoblasts display a prominent Golgi apparatus and are actively synthesizing osteoid. Two osteocytes can also be seen.

Furthermore, in periods of rapid remodeling (eg, after menopause), bone is at an increased risk for fracture because the newly produced bone is less densely mineralized, the resorption sites are temporarily unfilled, and the isomerization and maturation of collagen are impaired.[19]

The receptor activator of nuclear factor-kappa B ligand (RANKL)/receptor activator of nuclear factor-kappa B (RANK)/osteoprotegerin (OPG) system is the final common pathway for bone resorption. Osteoblasts and activated T cells in the bone marrow produce the RANKL cytokine. RANKL binds to RANK expressed by osteoclasts and osteoclast precursors to promote osteoclast differentiation. OPG is a soluble decoy receptor that inhibits RANK-RANKL by binding and sequestering RANKL.

Bone mass peaks around the third decade of life and slowly decreases afterward. A failure to attain optimal bone strength by this point is one factor that contributes to osteoporosis, which explains why some young postmenopausal women have low bone mineral density (BMD) and why some others have osteoporosis. Therefore, nutrition and physical activity are important during growth and development. Nevertheless, hereditary factors play the principal role in determining an individual's peak bone strength. In fact, genetics account for up to 80% of the variance in peak bone mass between individuals.[10, 20]

Alterations in bone formation and resorption

The hallmark of osteoporosis is a reduction in skeletal mass caused by an imbalance between bone resorption and bone formation. Under physiologic conditions, bone formation and resorption are in a fair balance. A change in either—that is, increased bone resorption or decreased bone formation—may result in osteoporosis.

Osteoporosis can be caused both by a failure to build bone and reach peak bone mass as a young adult and by bone loss later in life. Accelerated bone loss can be affected by hormonal status, as occurs in perimenopausal women; can impact elderly men and women; and can be secondary to various disease states and medications.

Aging and loss of gonadal function are the 2 most important factors contributing to the development of osteoporosis. Studies have shown that bone loss in women accelerates rapidly in the first years after menopause. The lack of gonadal hormones is thought to up-regulate osteoclast progenitor cells. Estrogen deficiency leads to increased expression of RANKL by osteoblasts and decreased release of OPG; increased RANKL results in recruitment of higher numbers of preosteoclasts as well as increased activity, vigor, and lifespan of mature osteoclasts.

Estrogen deficiency

Estrogen deficiency not only accelerates bone loss in postmenopausal women but also plays a role in bone loss in men. Estrogen deficiency can lead to excessive bone resorption accompanied by inadequate bone formation. Osteoblasts, osteocytes, and osteoclasts all express estrogen receptors. In addition, estrogen affects bones indirectly through cytokines and local growth factors. The estrogen-replete state may enhance osteoclast apoptosis via increased production of transforming growth factor (TGF)–beta.

In the absence of estrogen, T cells promote osteoclast recruitment, differentiation, and prolonged survival via interleukin-1 ( IL-1), IL-6, and tumor necrosis factor (TNF)–alpha. A murine study, in which either the mice's ovaries were removed or sham operations were performed, found that IL-6 and granulocyte-macrophage CFU levels were much higher in the ovariectomized mice.[21] This finding provided evidence that estrogen inhibits IL-6 secretion and that IL-6 contributes to the recruitment of osteoclasts from the monocyte cell line, thus contributing to osteoporosis.

IL-1 has also been shown to be involved in the production of osteoclasts. The production of IL-1 is increased in bone marrow mononuclear cells from ovariectomized rats. Administering IL-1 receptor antagonist to these animals prevents the late stages of bone loss induced by the loss of ovarian function, but it does not prevent the early stages of bone loss. The increase in the IL-1 in the bone marrow does not appear to be a triggered event but, rather, a result of removal of the inhibitory effect of sex steroids on IL-6 and other genes directly regulated by sex steroids.

T cells also inhibit osteoblast differentiation and activity and cause premature apoptosis of osteoblasts through cytokines such as IL-7. Finally, estrogen deficiency sensitizes bone to the effects of parathyroid hormone (PTH).

Aging

In contrast to postmenopausal bone loss, which is associated with excessive osteoclast activity, the bone loss that accompanies aging is associated with a progressive decline in the supply of osteoblasts in proportion to the demand. This demand is ultimately determined by the frequency with which new multicellular units are created and new cycles of remodeling are initiated.

After the third decade of life, bone resorption exceeds bone formation and leads to osteopenia and, in severe situations, osteoporosis. Women lose 30-40% of their cortical bone and 50% of their trabecular bone over their lifetime, as opposed to men, who lose 15-20% of their cortical bone and 25-30% of trabecular bone.

Calcium deficiency

Calcium, vitamin D, and PTH help maintain bone homeostasis. Insufficient dietary calcium or impaired intestinal absorption of calcium due to aging or disease can lead to secondary hyperparathyroidism. PTH is secreted in response to low serum calcium levels. It increases calcium resorption from bone, decreases renal calcium excretion, and increases renal production of 1,25-dihydroxyvitamin D (1,25[OH]2 D)—an active hormonal form of vitamin D that optimizes calcium and phosphorus absorption, inhibits PTH synthesis, and plays a minor role in bone resorption.

Vitamin D deficiency

Vitamin D deficiency can result in secondary hyperparathyroidism via decreased intestinal calcium absorption.

Osteoporotic fractures

Osteoporotic fractures represent the clinical significance of these derangements in bone. They can result both from low-energy trauma, such as falls from a sitting or standing position, and from high-energy trauma, such as a pedestrian struck in a motor vehicle accident. Fragility fractures, which occur secondary to low-energy trauma, are characteristic of osteoporosis.

Nearly all osteoporotic hip fractures are related to falls.[22] The frequency and direction of falls can influence the likelihood and severity of fractures. The risk of falling may be amplified by neuromuscular impairment due to vitamin D deficiency with secondary hyperparathyroidism or to corticosteroid therapy.

Vertebral bodies are composed primarily of cancellous bone with interconnected horizontal and vertical trabeculae. Osteoporosis not only reduces bone mass in vertebrae but also decreases interconnectivity in their internal scaffolding.[17] Therefore, minor loads can lead to vertebral compression fractures.

An understanding of the biomechanics of bone provides greater appreciation as to why bone may be susceptible to an increased risk of fracture. In bones that sustain vertical loads, such as tibial and femoral metaphyses and vertebral bodies, resistance to lateral bowing and fractures is provided by a horizontal trabecular cross-bracing system that helps support the vertical elements. Disruption of such trabecular connections is known to occur preferentially in patients with osteoporosis, particularly in postmenopausal women, making females more at risk than males for vertebral compression fractures (see the images below).



View Image

Osteoporosis is defined as a loss of bone mass below the threshold of fracture. This slide (methylmethacrylate embedded and stained with Masson's tric....



View Image

The bone loss of osteoporosis can be severe enough to create separate bone "buttons" with no connection to the surrounding bone. This easily leads to ....

Rosen and Tenenhouse studied the unsupported trabeculae and their susceptibility to fracture within each vertebral body and found an extraordinarily high prevalence of trabecular fracture callus sites within vertebral bodies examined at autopsy—typically, 200-450 healing or healed fractures per vertebral body.[23] These horizontal trabecular fractures are asymptomatic, and their accumulation reflects the impact of lost trabecular bone and greatly weakens the cancellous structure of the vertebral body.

The reason for preferential osteoclastic severance of horizontal trabeculae is unknown. Some authors have attributed this phenomenon to overaggressive osteoclastic resorption.

Osteoporosis versus osteomalacia

Osteoporosis may be confused with osteomalacia. The normal human skeleton is composed of a mineral component, calcium hydroxyapatite (60%), and organic material, mainly collagen (40%). In osteoporosis, the bones are porous and brittle, whereas in osteomalacia, the bones are soft. This difference in bone consistency is related to the mineral-to-organic material ratio. In osteoporosis, the mineral-to-collagen ratio is within the reference range, whereas in osteomalacia, the proportion of mineral composition is reduced relative to organic material content.

The Wnt signaling pathway and bone

The Wnt family is a highly conserved group of proteins that were initially studied in relationship with cancer initiation and progression due to their involvement in intercellular communication.[24] Subsequently, the Wnt signaling cascade was recognized as a critical regulator of bone metabolism.

Wnt signaling plays a key role in the fate of mesenchymal stem cells (MSCs), which are the progenitor cells of mature bone-forming osteoblasts.[25] MSCs have the capability to differentiate into adipocytes, chondrocytes, neurons, and muscle cells, as well as into osteoblasts.[26] Certain Wnt signaling pathways promote the differentiation of MSCs along the osteoblast lineage. The emerging details about the specific molecules involved in the Wnt pathway have improved the understanding of bone metabolism and led to the development of new therapeutic targets for metabolic bone diseases.

Wnt signal activation may progress along one of three pathways, with the “canonical” pathway involving β-catenin being most relevant to bone metabolism. The canonical Wnt signaling pathway is initiated by the binding of a Wnt protein to an extracellular co-receptor complex consisting of “Frizzled” (Fr) and low density lipoprotein receptor–related protein–5 or –6 (LRP5, LRP6).[27] This activation recruits another protein, “Disheveled” (Dvl) to the intracellular segment of the Fz/Dvl co-receptor.[28] This is where β-catenin comes into play.

β-Catenin is an important intracellular signaling molecule and normally exists in a phosphorylated state targeted for ubiquination and subsequent degradation within intracellular lysosomes. Activation of the Wnt pathway leads to dephosphorylation and stabilization of intracellular β-catenin and rising cytosolic concentrations of β-catenin. As the concentration of β-catenin reaches a critical level, β-catenin travels to the nucleus, where it activates the transcription of Wnt target genes. Ultimately, canonical Wnt signaling inhibits the expression of transcription factors important in the differentiation of MSCs such as peroxisome proliferator-activated receptor gamma (PPAR-γ) and promotes survival of osteoblast lineage cells.[29]

Several human bone abnormalities have been linked to the Wnt pathway. For example, a single amino acid substitution in the LRP5 receptor gene has been associated with high bone mass phenotypes in humans; specifically, the mutant LRP5 receptor had an impaired interaction with the Wnt signal inhibitor Dickkopf-1 (Dkk-1).[30] Similarly, other missense mutations of LRP5 have been implicated in other high bone mass diseases such as Van Buchem disease and osteopetrosis.[31] Conversely, loss-of-function mutations of LRP5 have resulted in a rare but severe congenital osteoporosis in humans.[32]

There are also several antagonists to the Wnt pathway. Two of the most well-known are Dkk-1 and sclerostin. Dkk-1 is secreted by MSCs[33] and binds to LRP-5 and LRP-6,[34] thereby competitively inhibiting Wnt signaling. Interestingly, serum levels of Dkk-1 positively correlate with the extent of lytic bone lesions in patients with multiple myeloma.[35]  In animal models, anti-Dkk1 monoclonal antibody accelerates bone formation and increases bone mineral density, and anti-Dkk antibody is under development as a bone-anabolic agent.[36]

Similarly, sclerostin, a product of osteocytes,[37] has also been found to antagonize the Wnt signaling pathway by binding to LRP5 and LRP6.[38] Romosozumab, a monoclonal antibody that binds with and inhibits sclerostin, and thus both increases bone formation and decreases bone resorption, has been approved for treatment of osteoporosis in postmenopausal women who are at high risk for fracture.[39]

Additional factors and conditions

Endocrinologic conditions or medications that lead to bone loss (eg, glucocorticoids) can cause osteoporosis. Corticosteroids inhibit osteoblast function and enhance osteoblast apoptosis.[40] Polymorphisms of IL-1, IL-6 and TNF-alpha, as well as their receptors, have been found to influence bone mass.

Other factors implicated in the pathogenesis of osteoporosis include the following[18] :

Epigenetics

Prenatal and postnatal factors contribute to adult bone mass. In one study, the health of the mother in pregnancy, the infant’s birth weight, and the child’s weight at age 1 year were predictive of adult bone mass in the seventh decade for men and women.[41] It is postulated that growth in the first year of life programs growth hormone secretion, and that this programming is maintained into the seventh decade.[42] Higher birth weight and rapid growth in the first year of life predicted increased bone mass in adults aged 65-75 years.

Etiology

Etiologically, osteoporosis is categorized as primary or secondary.

Primary osteoporosis

Primary osteoporosis is divided into juvenile and idiopathic osteoporosis. Idiopathic osteoporosis can be further subdivided into postmenopausal (type I) and age-associated or senile (type II) osteoporosis. Postmenopausal osteoporosis is primarily due to estrogen deficiency. Senile osteoporosis is primarily due to an aging skeleton and calcium deficiency. SeeTable 2, below.

Table 2. Types of Primary Osteoporosis



View Table

See Table

Secondary osteoporosis

Secondary osteoporosis occurs when an underlying disease, deficiency, or drug causes osteoporosis (see Table 3, below). Up to one third of postmenopausal women, as well as many men and premenopausal women, have a coexisting cause of bone loss,[43, 44] of which renal hypercalciuria is one of the most important secondary causes of osteoporosis and treatable with thiazide diuretics.[45]

Table 3. Causes of Secondary Osteoporosis in Adults



View Table

See Table

Risk factors

Risk factors for osteoporosis, such as advanced age and reduced bone mineral density (BMD), have been established by virtue of their direct and strong relationship to the incidence of fractures; however, many other factors have been considered risk factors based on their relationship to BMD as a surrogate indicator of osteoporosis.

Risk factors for osteoporosis include the following[52, 53, 54] :

A potentially useful mnemonic for osteoporotic risk factors is OSTEOPOROSIS, as follows:

Exposure to the antibacterial agent triclosan may increase risk for osteoporosis in women. Analysis of data on 1848 women from the 2005-2010 US National Health and Nutrition Examination Survey (NHANES) showed that women in the highest tertile of urinary triclosan level had lower BMD in the total femur, intertrochanter, and lumbar spine; compared with women in the lowest tertile, those in the highest tertile were more likely to have increased prevalence of intertrochanter osteoporosis (odds ratio (OR)=2.464, 95% CI = 1.190, 5.105).[57]

In animal studies, triclosan has been found to disrupt hormone activity, and in vitro studies have shown that it can cause interstitial collagen accumulation and an increase in trabecular bone. The US Food & Drug Administration has banned triclosan from consumer hand sanitizers and other antiseptics, but it continues to be used in a variety of consumer products—including clothing, kitchenware, furniture, and toys—to prevent bacterial contamination.[57]

Epidemiology

According to the National Osteoporosis Foundation (NOF), in the United States in 2010 more than 10 million adults age 50 years and older had osteoporosis and more than 43 million had low bone mineral density (BMD). In the United States in 2015, as many as 2 million Medicare beneficiaries sustained 2.3 million osteoporotic fractures. Within 12 months of experiencing a new osteoporotic fracture, approximately 15% of patients suffered one or more subsequent fractures and nearly 20% died. Mortality was highest in those with hip fracture, with 30% dying within 12 months.[58]

Most studies assessing the prevalence and incidence of osteoporosis use the rate of fracture as a marker for the presence of this disorder, although BMD also relates to risk of disease and fracture. The risk of new vertebral fractures increases by a factor of 2-2.4 for each standard deviation (SD) decrease of BMD measurement. Women and men with metabolic disorders associated with secondary osteoporosis have a 2- to 3-fold higher risk of hip and vertebral fractures.

Globally, osteoporosis is by far the most common metabolic bone disease, estimated to affect over 200 million people worldwide.[59] An estimated 75 million people in Europe, the United States, and Japan have osteoporosis.[60]

Age- and sex-related demographics

Risk for osteoporosis increases with age as BMD declines. Senile osteoporosis is most common in persons aged 70 years or older. Secondary osteoporosis, however, can occur in persons of any age. Although bone loss in women begins slowly, it speeds up around the time of menopause, typically at about age 50 years or later. The frequency of postmenopausal osteoporosis is highest in women aged 50-70 years.

The number of osteoporotic fractures increases with age. Wrist fractures typically occur first, when individuals are aged approximately 50-59 years.

Vertebral fractures occur more often in the seventh decade of life. Jensen et al studied Danish women aged 70 years and found a 21% prevalence of vertebral fractures.[61] Melton et al reported that 27% of women in their study had evidence of vertebral fractures by age 65 years.[62]

Ninety percent of hip fractures occur in persons aged 50 years or older, occurring most often in the eighth decade of life.[63]

Women are at a significantly higher risk for osteoporosis. Half of all postmenopausal women will have an osteoporosis-related fracture during their lifetime; 25% of these women will develop a vertebral deformity, and 15% will experience a hip fracture.[64] Risk factors for hip fracture are similar in different ethnic groups.[65]

Men have a higher prevalence of secondary osteoporosis, with an estimated 45-60% of cases being a consequence of hypogonadism, alcoholism, or glucocorticoid excess.[49] Only 35-40% of osteoporosis diagnosed in men is considered primary in nature. Overall, osteoporosis has a female-to-male ratio of 4:1.

Although loss of BMD is typically associated with postmenopausal women, a study to assess the likelihood of low BMD and related risk factors for osteoporosis in men and women aged 35 to 50 years found higher rates of osteopenia in men: 28% of men and 26% of women had osteopenia at the femoral neck region, and 6% and 2%, respectively, had osteoporosis of the lumbar spine. Of the 173 study subjects, 92 (53%) were women and 162 (94%) were white; none had previous known health issues or were taking medications that can affect BMD.[66]

Fifty percent of all women and 21% of all men older than 50 years experience one or more osteoporosis-related fractures in their lifetime.[67] Eighty percent of hip fractures occur in women.[63] Women have a two-fold increase in the number of fractures resulting from nontraumatic causes, as compared with men of the same age.

Racial demographics

Osteoporosis can occur in persons of all races and ethnicities. In general, however, whites (especially of northern European descent) and Asians are at increased risk. In particular, non-Hispanic white women and Asian women are at higher risk for osteoporosis. In the most recent government census, 178 million Chinese were over age 60 years in 2009, a number that the United Nations estimates may reach 437 million—one-third of the population—by 2050.[68]

These numbers suggest that approximately 50% of all hip fractures will occur in Asia in the next century. In fact, although age-standardized incidence rates of fragility fractures, particularly of the hip and forearm, have been noted to be decreasing in many countries over the last decade, that is not the case in Asia.[69]

Table 4, below, summarizes some osteoporosis prevalence statistics among racial/ethnic groups. Note that this disease is under-recognized and undertreated in white and black women. Relative to other racial/ethnic groups, the risk of developing osteoporosis is increasing fastest among Hispanic women.

Table 4. Prevalence of Osteoporosis Among Racial and Ethnic Groups



View Table

See Table

Melton et al reported that the prevalence of hip fractures is higher in white populations, regardless of geographic location.[70] Another study indicated that, in the United States and South Africa, the incidence of hip fractures was lower in black persons than in age-matched white persons. Cauley et al found that the absolute fracture incidence across BMD distribution was 30-40% lower in black women than in white women. This lower fracture risk was independent of BMD and other risk factors.[71]

Prognosis

The prognosis for osteoporosis is good if bone loss is detected in the early phases and proper intervention is undertaken. Patients can increase bone mineral density (BMD) and decrease fracture risk with the appropriate anti-osteoporotic medication. In addition, patients can decrease their risk of falls by participating in a multifaceted approach that includes rehabilitation and environmental modifications. Worsening of medical status can be prevented by providing appropriate pain management and, if indicated, orthotic devices.

Effect of fractures on prognosis

Many individuals experience morbidity associated with the pain, disability, and diminished quality of life caused by osteoporosis-related fractures. According to a 2004 Surgeon General's report, osteoporosis and other bone diseases are responsible for about 1.5 million fractures per year. Osteoporosis-related fractures result in annual direct care expenditures of $12.2 billion to $17.9 billion.[72] In 2005, over 2 million osteoporosis-related fractures occurred in the United States.[73]

Osteoporosis is the leading cause of fractures in the elderly. Women aged 50 years have about a 50% lifetime fracture rate as a result of osteoporosis. Osteoporosis is associated with 80% of all the fractures in people aged 50 years or older. Approximately 33% of women who live to age 90 years will suffer a hip fracture, which is associated with functional decline, nursing home placement, and death.[74]

Researchers analyzed data from a subgroup of the Study of Osteoporotic Fractures, a prospective cohort study that began recruiting in 1986. The study had 1528 participants, all of whom were women, with a mean (SD) age of 84.1 (3.4) years. During follow-up, 125 (8.0%) women experienced a hip fracture and 287 (18.8%) died before experiencing this event. Five-year mortality probability was 24.9% (95% CI, 21.8-28.1) among women with osteoporosis and 19.4% (95% CI, 16.6-22.3) among women without osteoporosis but at high fracture risk. In both groups, mortality probability similarly increased with more comorbidities and poorer prognosis. In contrast, 5-year hip fracture probability was 13.0% (95% CI, 10.7-15.5) among women with osteoporosis and 4.0% (95% CI, 2.8-5.6) among women without osteoporosis but at high fracture risk. The difference was most pronounced among women with more comorbidities or worse prognosis. For example, among women with 3 or more comorbid conditions, hip fracture probability was 18.1% (95% CI, 12.3-24.9) among women with osteoporosis vs 2.5% (95% CI, 1.3-4.2) among women without osteoporosis but at high fracture risk.[75]

If full recovery is not achieved, osteoporotic fractures may lead to chronic pain, disability, and, in some cases, death. This is particularly true of vertebral and hip fractures.

Vertebral fractures

Vertebral compression fractures (see the images below) are associated with increased morbidity and mortality rates. In addition, the impact of vertebral fractures increases as they increase in number. As posture worsens and kyphosis progresses, patients experience difficulty with balance, back pain, respiratory compromise, and an increased risk of pneumonia. Overall function declines, and patients may lose their ability to live independently.



View Image

Osteoporosis. Lateral radiograph demonstrates multiple osteoporotic vertebral compression fractures. Kyphoplasty has been performed at one level.



View Image

Osteoporosis. Lateral radiograph of the patient seen in the previous image following kyphoplasty performed at 3 additional levels.

In one study, Cooper et al found that vertebral fractures increased the 5-year risk of mortality by 15%.[76] In a subsequent study, Kado et al[77] demonstrated that women with one or more fractures had a 1.23-fold increased age-adjusted mortality rate and that women with 5 or more vertebral fractures had a 2.3-fold increased age-adjusted mortality rate.

Furthermore, the mortality rate correlated with number of vertebral fractures: there were 19 deaths per 1000 woman-years in women with no fracture, versus 44 per 1000 woman-years in women with five or more fractures. Vertebral fractures related to risk of subsequent cancer and pulmonary death, and severe kyphosis was further correlated with pulmonary deaths.

Symptoms of vertebral fracture may include back pain, height loss, and disabling kyphosis. Compression deformities can lead to restrictive lung disease, abdominal pain, and early satiety.

Hip fractures

More than 250,000 hip fractures are attributed to osteoporosis each year. Like vertebral fractures, they are associated with significantly increased morbidity and mortality rates in men and women. In the year following hip fracture, excess mortality rates can be as high as 20%.[76, 78] Men have higher mortality rates following hip fracture than do women.

Patients with hip fractures incur decreased independence and a diminished quality of life. Of all patients with hip fracture, approximately 20% require long-term nursing care.[2] Among women who sustain a hip fracture, 50% spend time in a nursing home while recovering. Approximately 50% of previously independent individuals become partially dependent, and one third become completely dependent.[79] Only one third of patients return to their prefracture level of function.[80]

Secondary complications of hip fractures include nosocomial infections and pulmonary thromboembolism.

Additional fractures

Patients who have sustained one osteoporotic fracture are at increased risk for developing additional osteoporotic fractures.[60] For example, the presence of at least one vertebral fracture results in a 5-fold increased risk of developing another vertebral fracture. One in 5 postmenopausal women with a new vertebral fracture incurs another vertebral fracture within one year.[81]

Patients with previous hip fracture have a two-fold[82] to 10-fold increased risk of sustaining a second hip fracture. In addition, patients with ankle, knee, olecranon, and lumbar spine fractures have a 1.5-, 3.5-, 4.1-, and 4.8-fold increased risk of subsequent hip fracture, respectively. Site of prior fracture impacts on future risk of osteoporotic fractures independent of BMD such that in postmenopausal women, prior fractures of the spine, humerus, patella, and pelvis are more predictive of future osteoporotic fractures than fractures at other sites.[83]

WHO fracture-risk algorithm

The World Health Organization fracture-risk algorithm (FRAX) was developed to calculate the 10-year probability of a hip fracture and the 10-year probability of any major osteoporotic fracture (defined as clinical spine, hip, forearm, or humerus fracture) in a given patient. These calculations account for femoral neck BMD and other clinical risk factors, as follows[84] :

The National Osteoporosis Foundation (NOF) recommends osteoporosis treatment in patients with low bone mass in whom a US-adapted WHO 10-year probability of a hip fracture is 3% or more or in whom the risk for a major osteoporosis-related fracture is 20% or more.[2] Note that osteoporosis is, by definition, present in those with a fragility fracture, irrespective of their T-score.

Algorithms such as the FRAX algorithm are useful in identifying patients with low bone mass (T-scores in the osteopenic range) who are most likely to benefit from treatment. A study by Leslie et al demonstrated the effects of including a patient's 10-year fracture risk along with DXA results in Manitoba, Canada.[85] The authors found an overall reduction in dispensation of osteoporosis medications as more women were reclassified into lower fracture risk categories.

Although type 2 diabetes mellitus (DM) is associated with a higher BMD, a study by Schwartz et al concluded that for a given T score and age or for a given FRAX score, the risk of fracture is higher in patients with type 2 DM than in those without type 2 DM. The study conclusions were based on data from three prospective observational studies, statistics from self-reported incidence of fractures in 9449 women and 7436 men in the United States.[86]

The FRAX tool has a low sensitivity for predicting fracture risk in perimenopausal and early-menopausal women. In a study by Trémollieres et al, FRAX had 50% sensitivity in the 30% of women in the study who were at the highest risk.[87] FRAX also does not include risk of falls; 90% of hip fractures[88] and the majority of Colles fractures are associated with falls.[89]  

The Garvan fracture risk tool is not as widely used as the FRAX but is another validated fracture prediction tool that does account for falls, and may be a better tool for use in men.[90]

Complications

Vertebral compression fractures often occur with minimal stress, such as coughing, lifting, or bending. The vertebrae of the middle and lower thoracic spine and upper lumbar spine are involved most frequently. In many patients, vertebral fracture can occur slowly and without symptoms. Only one third of people with radiographic vertebral fractures are diagnosed clinically.[91]

Hip fractures are the most devastating and occur most commonly at the femoral neck and intertrochanteric regions (see the image below). Hip fractures are associated with falls. The likelihood of sustaining a hip fracture during a fall is related to the direction of the fall. Fractures are more likely to occur in falls to the side, because less subcutaneous tissue is available to dissipate the impact. Secondary complications of hip fractures include nosocomial infections and pulmonary thromboembolism.



View Image

Stable intertrochanteric fracture of the femur.

Fractures can cause further complications, including chronic pain from vertebral compression fractures and increased morbidity and mortality secondary to vertebral compression fractures and hip fractures. Patients with multiple fractures have significant pain, which leads to functional decline and a poor quality of life (QOL).[92] They are also at risk for the complications associated with immobility, including deep vein thrombosis (DVT) and pressure ulcers. Respiratory compromise can occur in patients with multiple vertebral fractures that result in severe kyphosis.

Patients with osteoporosis develop spinal deformities and a dowager's hump, and they may lose 1-2 inches of height by their seventh decade of life. These patients can lose their self-esteem and are at increased risk for depression.

Patient Education

Patient education is paramount in the treatment of osteoporosis. Many patients are unaware of the serious consequences of osteoporosis, including increased morbidity and mortality, and only become concerned when osteoporosis manifests in the form of fracture; accordingly, it is important to educate them regarding these consequences. Early prevention and treatment are essential in the appropriate management of osteoporosis.

The focus of patient education is on the prevention of osteoporosis. Prevention has 2 components, behavior modification and pharmacologic interventions. Appropriate preventive measures may include adequate calcium and vitamin D intake, exercise, cessation of smoking, and moderation of alcohol consumption.

Patients should be educated about the risk factors for osteoporosis, with a special emphasis on family history and the effects of menopause. Patients also need to be educated about the benefits of calcium and vitamin D supplements, as well as strategies to prevent falls in the elderly (see Primary Care–Relevant Interventions to Prevent Falling in Older Adults: A Systematic Evidence Review for the US Preventive Services Task Force [USPSTF]).

All postmenopausal women older than 65 years should be offered bone densitometry, as well as some younger women and men. These patients should understand the benefits of bone density monitoring. Society at large also should be educated about the benefits of exercise with regard to osteoporosis.

For patient education information, see the Osteoporosis Health Center, as well as the Anorexia Nervosa Health Center, Inflammatory Bowel Disease Health Center, and Menopause Health Center.

History

Keep in mind that osteoporosis occurs in many people who have few or no risk factors for this condition. Often, patients who have not sustained a fracture do not report symptoms that would alert the clinician to suspect a diagnosis of osteoporosis; thus, this disease is a "silent thief" that generally does not become clinically apparent until a fracture occurs.

Screening at-risk populations is, therefore, essential; unfortunately, many women do not receive proper screening or treatment for osteoporosis, which, in turn, may result in improper management of this disease and its related complications.[93] For example, the  presence of a disorder known to cause secondary osteoporosis—such as rheumatoid arthritis, celiac disease, or Crohn disease—should increase a clinician’s suspicion that the patient may have osteoporosis and that screening may be indicated.

Multiple risk factors exist for osteoporosis. The National Osteoporosis Foundation (NOF) has categorized these risk factors into two categories: nonmodifiable and modifiable. Nonmodifiable risk factors include the following:

Potentially modifiable risk factors include the following:

Assessment of fracture risk

A thorough history should be obtained to screen for and identify the presence of known risk factors for osteoporosis and osteoporotic fracture. Specifically, the history should focus on the following[94, 95] :

FRAX tool

Although the USPSTF did not find any studies that assessed effects of the use of risk prediction instruments on patient outcomes, either alone or in combination with bone measurement tests, there are many validated instruments for predicting the risk for low bone mineral density (BMD) in postmenopausal women; few of these, however, have been validated for use in men.[95]

The Fracture Risk Assessment (FRAX) tool, accessible to healthcare providers and patients, is a validated instrument used to estimate 10-year risks for fractures, including those for black, Asian, and Hispanic women.[84] A 65-year-old white woman with no other risk factors has a 9.3% 10-year risk for any osteoporotic fracture. Generally, estimated fracture risks in nonwhite women are lower than those for white women of the same age.

White women between the ages of 50 and 64 years with ≥10-year fracture risks based on specific risk factors include the following persons[95] :

Differentiating fracture types by history

Patients with acute insufficiency fractures may report a history of minimal or no trauma resulting in pain. They may report a fall from a standing or sitting position. Patients with compression fractures resulting in thoracic kyphosis may report iliocostal friction with associated abdominal protrusion, decreased tolerance for oral intake, and breathing difficulties. Patients with hip, pelvic, or sacral fractures may report pain that is worsened with weight-bearing.

Patients who have sustained a vertebral compression fracture may note progressive kyphosis with loss of height. They may also present with an episode of acute back pain after bending, lifting, or coughing. It should be noted, however, that two thirds of vertebral fractures are asymptomatic.

With respect to those vertebral fractures that are painful, typical subjective information may include the following:

Patients who have sustained a hip fracture may experience the following:

Patients with osteoporosis may report lactose intolerance and celiac sprue. Celiac sprue has been shown to be associated with osteoporosis in approximately 5% of cases.

Physical Examination

Patients with suspected osteoporosis should undergo a comprehensive physical examination. The physical examination should begin with an inspection of the patient. Height measurement with a stadiometer at each visit may be useful. Examination of active and passive range of motion (ROM) assists in determining whether spine, hip, wrist, or other osseous pathology may be present. A thorough neurologic examination is essential to rule out spinal cord and/or peripheral nerve compromise.

The examination may elicit pain, or the patient may be pain free. Thoracic kyphosis may be present secondary to vertebral compression fractures, a dowager hump, and a history of loss of height. Patients may have an associated scoliosis.

Areas of concern include the following:

A 10-year longitudinal study assessed the Timed Up and Go (TUG) test performance (a validated predictor of falling) and hip area BMD (bone mineral density calculated using bone area as opposed to bone volume). Using data from 1126 women (mean age, 75 years), the study noted that risks of nonvertebral fracture and hip fracture were significantly higher among those who had slow TUG test performance and normal hip BMD or both slow TUG test performance and low hip BMD. These results suggest that the TUG test is an independent risk factor for incident nonvertebral fracture; this inexpensive physical assessment may be beneficial in screening patients with increased risk of fracture.[96]

Signs of fracture

Patients with vertebral compression fractures may demonstrate a thoracic kyphosis with an exaggerated cervical lordosis (dowager hump). This is followed by a loss of lumbar lordosis. After each episode of vertebral compression fracture and progressive kyphosis, the patient's height may decrease by 2-3 cm.

Patients with acute vertebral fractures may have point tenderness over the involved vertebrae. Palpation of the spinous processes often does not aid the examiner in localizing point tenderness, but percussion may be helpful in acute or subacute vertebral compression fractures.

Patients with hip fractures may have severe pain with ambulation. A FABER (ie, flexion in abduction and external rotation) hip joint test may reveal limited ROM with end-range pain. Patients with hip fractures may show decreased weight-bearing on the fractured side or an antalgic gait pattern.

Patients with pubic and sacral fractures may report marked pain with ambulation and tenderness to palpation, percussion, or both. Furthermore, patients with sacral fractures may have pain with physical examination techniques used to assess the sacroiliac joint, such as the FABER, Gaenslen, or squish test.

Fractures in other parts of the body, including the distal radius and humerus, are typically painful and result in limited range of motion of the involved joint.

Signs of collagen defects

Patients with osteoporosis may have physical findings consistent with subtle collagen defects. These include a short fifth digit, dentinogenesis imperfecta, hyperlaxity, hearing loss, pes planus, bunions, and blue sclerae.

Balance difficulties

Patients with osteoporosis are known to have decreased balance, possibly secondary to differences in balance control strategies and sway amplitude. Patients may have difficulty performing tandem gait and performing single limb stance. Poor balance may be noted particularly in patients with severe kyphosis resulting from vertebral compression fractures because their altered center of gravity makes ambulation with a stable base of support difficult for them.[1]

Fractures are the most common and serious complication of osteoporosis. Patients with osteoporosis are at high risk for recurrent fractures of the hips, vertebrae, ribs, and wrists.

Screening in Men

Although routinely screening men for osteoporosis is not as widespread a practice as that of screening in women, the US Preventive Services Task Force (USPSTF) indicates that using bone measurement tests in men may not only help to detect this disease but also prevent its associated burden of fractures and fracture-related illness.[9]

In weighing the risk versus benefit of screening men, clinicians should consider that osteoporosis is a preventable condition and that the potential harm in screening is likely to be small and mostly due to cost, such as that required to increase the number of dual-energy x-ray absorptiometry (DXA) scanners available for screening.[9] In addition, assuming that the risk versus benefit of therapy for osteoporosis is similar for men and women, the men who are most likely to benefit from screening would be those with a 10-year risk for osteoporotic fracture that is equal to or greater than that for 65-year-old white women who have no additional risk factors.[9] It is important to note, however, that there is insufficient current evidence for assessing the risk versus benefit of screening for osteoporosis in men.

The American College of Physicians (ACP) has similar recommendations for screening of osteoporosis in men, including periodic evaluation of risk factors in older men before age 65 years (particularly men who do not choose to be screened) and obtaining DXA for men at increased risk for the disease who are candidates for drug therapy.[97] Risk factors for osteoporosis in men include the following[97] :

Like the USPSTF, the ACP recommends further research to assess osteoporosis screening tests in men, to determine whether risk factors for osteoporosis in women also apply to men.[97]

Approach Considerations

Workup consists of appropriate laboratory studies to establish baseline values and to look for potential secondary causes of osteoporosis, along with measurement of bone mineral density (BMD) to assess bone loss and estimate the risk of fracture. Bone biopsy may be indicated in specific situations.

Conventional radiography is used for the qualitative and semiquantitative evaluation of osteoporosis; morphometry assesses the presence of fractures.[98] Quantitative imaging methods commonly used are dual-energy x-ray absorptiometry (DXA) and quantitative computed tomography (QCT) scanning.[98] In the United States, current diagnostic and treatment criteria for osteoporosis are based solely on QCT hip and DXA spine or hip T-score measurements.[99, 100]

BMD should be measured at both the posteroanterior (PA) spine and hip in all patients undergoing DXA.[3] Forearm BMD should be measured under the following circumstances:

For information on American College of Radiology (ACR) recommendations for evaluating the appropriateness of BMD measurement tests for osteoporosis in patients at risk of developing this condition, see ACR Appropriateness Criteria for osteoporosis and bone mineral density.

Children and adolescents

The official position of the International Society for Clinical Densitometry (ISCD) is that “fracture prediction should primarily identify children at risk of clinically significant fractures” (eg, fracture of lower-extremity long bones, vertebral compression fractures, or two or more upper-extremity long-bone fractures).[3] However, densitometric criteria alone should not be used to diagnose osteoporosis in children and adolescents. Rather, such a diagnosis in this population must be based on a low bone mineral content (BMC) or BMD in conjunction with a clinically significant fracture history. Fractures are considered clinically significant[3] if one or more of the following are present:

Laboratory Studies

Laboratory studies are used to establish baseline conditions or to exclude secondary causes of osteoporosis. They are summarized in Tables 5 and 6, below.

Table 5. Baseline Studies for Baseline Conditions in Osteoporosis



View Table

See Table

An important study by Tannenbaum evaluated 173 healthy women (ages 46-87 years) for secondary causes of osteoporosis and found that 55 (32%) had a previously undiagnosed disorder of bone or mineral metabolism.[103] Given that occult disorders are so common in patients with osteoporosis, minimal laboratory screening is indicated in all patients who present with decreased bone mass.

Table 6. Tests for Secondary Causes of Osteoporosis



View Table

See Table

Biochemical Markers of Bone Turnover

Biochemical markers of bone turnover reflect bone formation or bone resorption. These markers (both formation and resorption) may be elevated in high-bone-turnover states (eg, early postmenopausal osteoporosis) and may be useful in some patients for monitoring early response to therapy.

Currently available serum markers of bone formation (osteoblast products) include the following:

Currently available urinary markers of bone resorption (osteoclast products) include the following:

Currently available serum markers of bone resorption include cross-linked C-telopeptide of type I collagen (ICTP) and tartrate-resistant acid phosphatase, as well as NTx and CTx. Of all the biochemical markers of bone turnover mentioned above, the ones most commonly used in clinical practice are BSAP, OC, urinary NTx, and serum CTx.

BSAP can be mildly elevated in patients with fractures. In addition, patients with hyperparathyroidism, Paget disease, or osteomalacia can have elevations of BSAP. Serum OC levels, if high, indicate a high-turnover type of osteoporosis.[104] Elevation of urinary NTx (>40 nmol bone collagen equivalent per mmol urinary creatine) indicates a high turnover state.

Significant controversy exists regarding the use of these biochemical markers, and concerns have been raised about intra-assay and interassay variability. Further study is needed to determine the clinical utility of these markers in osteoporosis management.

For more information, see the Medscape Reference article Bone Markers in Osteoporosis.

Plain Radiography

Plain radiography is recommended to assess overall skeletal integrity. In particular, in the workup for osteoporosis, plain radiography may be indicated if a fracture is already suspected or if patients have lost more than 1.5 inches of height (see the following image).



View Image

Asymmetric loss in vertebral body height, without evidence of an acute fracture, can develop in patients with osteoporosis. These patients become prog....

Obtain radiographs of the affected area in symptomatic patients. Lateral spine radiography can be performed in asymptomatic patients in whom a vertebral fracture is suspected, in those with height loss in the absence of other symptoms, or in those with pain in the thoracic or upper lumbar spine (see the following image). A scoliosis series is useful for detecting occult vertebral fractures.



View Image

Severe osteoporosis. This radiograph shows multiple vertebral crush fractures. Source: Government of Western Australia Department of Health.

Radiographic findings can suggest the presence of osteopenia, or bone loss, but cannot be used to diagnose osteoporosis. Osteopenia is suggested by a cortical width that is less than the medullary width. Radiographs may also show fractures or other conditions, such as osteoarthritis, disk disease, or spondylolisthesis.

Plain radiography is not as accurate as BMD testing. Because osteoporosis predominantly affects trabecular bone rather than cortical bone, radiography does not reveal osteoporotic changes until they affect the cortical bone. Cortical bone is not affected by osteoporosis until more than 30% of bone loss has occurred. Approximately 30-80% of bone mineral must be lost before radiographic lucency becomes apparent on radiographs.[105] Thus, plain radiography is an insensitive tool for diagnosing osteoporosis.

Dual-Energy X-Ray Absorptiometry (DXA)

Several large prospective studies have shown that BMD measurements of the distal and proximal femur and the vertebral bodies can predict the development of the major types of osteoporotic fractures. BMD has been shown to be the best indicator of fracture risk. According to the National Osteoporosis Foundation (NOF), evaluating BMD on a periodic basis is the best way to monitor bone mass and future fracture risk,[2, 4] although there is controversy about how frequently to measure this.[106]

DXA is currently the criterion standard for the evaluation of BMD.[3, 4] Compared with other screening tools (eg, calcaneal quantitative ultrasonography, the Simple Calculated Osteoporosis Risk Estimation [SCORE]), DXA has been found to be efficacious and cost-effective.[107] DXA is not as sensitive as quantitative computed tomography (QCT) scanning for detecting early trabecular bone loss, but it provides comparable costs,[107] it is done on an outpatient basis,[2] and there are no special requirements for performing it. Also, radiation exposure is kept to a minimum.

DXA is used to calculate BMD at the lumbar spine, hip, and proximal femur (see the images below). Densitometric spine imaging can be performed at the time of DXA scanning to detect vertebral fractures. Vertebral fracture assessment (VFA) is not available with all DXA machines. When available, VFA should be considered when the results may influence clinical management of the patient.[108] Peripheral DXA is used to measure BMD at the wrist; it may be most useful in identifying patients at very low fracture risk who require no further workup.



View Image

Example of a dual energy x-ray absorption (DXA) scan. This image is of the left hip bone. Source: Government of Western Australia Department of Health....



View Image

Example of a dual energy x-ray absorption (DXA) scan. This image is of the lumbar spine. Source: Government of Western Australia Department of Health.....

Although measurement of BMD at any site can be used to assess overall fracture risk, measurement at a particular site is the best predictor of fracture risk at that site. Whenever possible, the same technologist should perform subsequent measurements on the same patient using the same machine. This method can be used in both adults and children. Factors that may result in a falsely high BMD determination include spinal fractures, osteophytosis, scoliosis, and extraspinal (eg, aortic) calcification.

Bone density data from a DXA are reported as T-scores and Z-scores. The T-score is the value compared to that of control subjects who are at their peak BMD, whereas the Z-score reflects a value compared to that of patients matched for age and sex.[5, 6, 7, 8]

WHO T-score and Z-score criteria

World Health Organization (WHO) criteria define a normal T-score value as within 1 standard deviation (SD) of the mean BMD value in a healthy young adult. Values lying farther from the mean are stratified as follows[7] :

For each SD reduction in BMD, the relative fracture risk is increased 1.5-3 times. Of note, about half of osteoporotic fractures occur in women with a T-score greater than –2.5, and the other half occur in those with a T-score lower than –2.5, the WHO’s cutoff for DXA-based diagnosis of osteoporosis.

This diagnostic classification should not be applied to premenopausal women, men younger than 50 years, or children. Instead, Z-scores adjusted for ethnicity or race should be used, with values of –2.0 SD or lower defined as "below the expected range for age" and those above –2.0 SD being "within the expected range for age." The diagnosis of osteoporosis in these groups should not be based on densitometric criteria alone.

International Society of Clinical Densitometry (ISCD) positions

The following are the official positions of the ISCD on peripheral DXA (pDXA)[3] :

Measurements of bone strength

Measurements of bone strength that can be obtained by DXA, with appropriate software, include hip structural analysis (HSA) and trabecular bone scores (TBS). An additional technique for measurement of bone strength, finite element analysis (FEA), can be based on either computed tomography (CT) or DXA .

Hip structural analysis

HSA uses data obtained from DXA examinations to estimate structural properties of the femur. Specifically, estimated parameters such as femoral neck cross-sectional moment of inertia, hip axis length, and cross sectional area parameters can be combined with clinical characteristics (eg, age, weight, ethnicity) to calculate the femoral strength index, which is ultimately an estimate of how well the femur can withstand a direct impact to the greater trochanter.[109]

HSA measurements have been found to correlate well with other measures of bone density such as DXA-derived BMD and quantitative CT[110] but have not consistently demonstrated superiority.[111] Furthermore, the ability of HSA to determine bending strength is limited to the specific plane of the image; comparisons require consistent and correct positioning, which has proven a hindrance to widespread clinical use.[112, 113]

Trabecular bone scores

TBS are another DXA-derived measurement of bone strength. TBS uses “gray-level texture measurements” from two-dimensional DXA scans of the lumbar spine to derive bone strength parameters.[114] TBS may complement standard measures of BMD.[115]

Finite element analysis

Traditional methods of measuring bone density (eg, DXA), while inexpensive and easy to perform, do not account for all the material properties of bone. In fact, many females, and even more males, who sustain hip fractures have BMDs above the osteoporotic threshold.[116] FEA is a relatively new technique for measuring fracture risk that is emerging as a highly accurate tool to evaluate bone strength.

With FEA, three-dimensional renderings of bone created using CT images[117] or DXA[118] are subjected to computer-simulated loads until fracture. This technique for measuring bone strength is mostly used in research settings, but has the potential for future clinical use.[119]

Quantitative Computed Tomography

Quantitative computed tomography (QCT) is another method employed to measure spinal BMD. At the spine, it measures BMD as a true volume density in g/cm3, which is not influenced by bone size. This technique can be used in both adults and children. QCT scanning of the spine is the most sensitive method for diagnosing osteoporosis, because it measures trabecular bone within the vertebral body. At the hip, QCT produces DXA-equivalent T-scores and BMD measures in g/cm2.[99, 100]

QCT scanning may be useful in identifying fractures. It can be used to identify not only the fracture line but also areas of callus formation and sclerosis, consistent with healing fracture. It may also be used for evaluation of metastatic bone disease.

Compared with DXA scanning, QCT has a comparable cost[107] and precision.[99, 120, 121, 122] In addition, as with DXA, no dye injection should be used.[123] QCT is a very sensitive technique when repeated measurements are needed to detect small changes in BMD, and modern three-dimensional (3D) QCT acquisition has a scan time less than 10 seconds for the lumbar spine or proximal femur, and there is no interference by osteophytes.[124, 125] However, QCT requires a higher radiation dose.[125]

Nonetheless, QCT scanning is less commonly used than DXA; based on Medicare data, about 5% of all BMD assessments are done with QCT scanning. Smaller, rural hospitals may favor QCT scanning, as they often already have a CT scanner for trauma cases and may not be able to afford a DXA machine as well.

International Society of Clinical Densitometry positions

The following are the official positions of the International Society of Clinical Densitometry (ISCD) on QCT of the lumbar spine (ref31)

The following are the official positions of the ISCD on pQCT of the radius[3] :

Single-Photon Emission CT

Single-photon emission computed tomography (SPECT) scanning represents a tomographic (CT-like) bone imaging technique that offers better image contrast and more accurate lesion localization than planar bone scanning. It increases the sensitivity and specificity of bone scanning for detection of lumbar spine lesions by 20-50% over planar techniques.

SPECT scanning is helpful when accurate localization of skeletal lesions within large and/or anatomically complex bony structures is required. This localization is possible because SPECT can visualize bony structures that would overlap on planar images (eg, separating vertebral body, facet and pars interarticularis lesions).

Quantitative Ultrasonography

Quantitative ultrasonography (QUS) of the calcaneus is a low-cost portable screening tool. It has the advantage of not involving radiation, but it is not as accurate as other imaging methods. Ultrasonography cannot be used for monitoring skeletal changes over time, nor can it be used to monitor the response to treatment, because of its lack of precision.

International Society of Clinical Densitometry positions

The following are the official positions of the ISCD on QUS[3] :

Magnetic Resonance Imaging

Magnetic resonance imaging (MRI) may be useful in identifying fractures and in the assessment of metabolic bone disease. Using fat-suppression sequences, marrow edema consistent with fracture may be noted as areas of hypointensity on T1-weighted images in association with corresponding areas of hyperintensity on T2-weighted images. MRI is a very sensitive modality and is believed by some to be the diagnostic imaging method of choice in the detection of acute fractures, such as sacral fractures.

MRI can be used to discriminate between acute and chronic fractures of the vertebrae and occult stress fractures of the proximal femur. These osteoporotic fractures demonstrate characteristic changes in the bone marrow that distinguish them from other uninvolved parts of the skeleton and the adjacent vertebrae.

Bone Scanning

Bone scanning assesses the function and tissue metabolism of organs by using a radionuclide (technetium-99m [99m Tc]) that emits radiation in proportion to its attachment to a target structure. This technique detects an increase in osteoblastic activity (as seen in compression fractures).

Images may be obtained in three phases of the bone scanning process (immediate-flow study, immediate static blood pool study, and delayed static study). Acute fractures are visible in all phases of bone scanning and may remain beyond the reference range for up to 2 years.

Bone Biopsy and Histologic Features

Bone biopsy can help to exclude underlying pathologic conditions, such as multiple myeloma, that may be responsible for presumed osteoporotic fracture. Typically, iliac crest biopsy is performed either in the minor procedure suite or in the operating room.

Tetracycline double labeling is a process used to calculate data on bone turnover. In this procedure, patients are given tetracycline, which binds to newly formed bone. This appears on biopsy samples as linear fluorescence. A second dose of tetracycline is given 11-14 days after the first dose; this appears on a biopsy sample as a second line of fluorescence. The distance between the two fluorescent labels can be measured to calculate the amount of bone formed during that interval, which may potentially indicate that too little bone formation or too much bone resorption is the cause of osteoporosis in a patient. Tetracycline labeling may also help clinicians to test potential therapy (ie, did the treatment slow bone resorption, increase bone formation, or both?) and study other metabolic bone responses.

One may also perform a vertebral body bone biopsy when performing a therapeutic procedure such as kyphoplasty (see the images below) or vertebroplasty for fixation of a vertebral compression fracture.



View Image

In kyphoplasty, a KyphX inflatable bone tamp is percutaneously advanced into the collapsed vertebral body (A). It is then inflated, (B) elevating the ....



View Image

Osteoporosis. Lateral radiograph demonstrates multiple osteoporotic vertebral compression fractures. Kyphoplasty has been performed at one level.



View Image

Osteoporosis. Lateral radiograph of the patient seen in the previous image following kyphoplasty performed at 3 additional levels.



View Image

Percutaneous vertebroplasty, transpedicular approach.

Histologic examination of osteoporotic bone may reveal generalized thinning of trabeculae and irregular perforation of trabeculae, reflecting unbalanced osteoclast-mediated bone resorption.[63] The following images are of histologic specimens from patients with osteoporosis.



View Image

Osteoporosis is defined as a loss of bone mass below the threshold of fracture. This slide (methylmethacrylate embedded and stained with Masson's tric....



View Image

The bone loss of osteoporosis can be severe enough to create separate bone "buttons" with no connection to the surrounding bone. This easily leads to ....



View Image

Inactive osteoporosis is the most common form and manifests itself without active osteoid formation.



View Image

Osteoporosis that is active contains osteoid seams (red here in the Masson's trichrome).



View Image

Woven bone arising directly from surrounding mesenchymal tissue.



View Image

This image depicts bone remodeling with osteoclasts resorbing one side of a bony trabecula and osteoblasts depositing new bone on the other side.



View Image

Osteoclast, with bone below it. This image shows typical distinguishing characteristics of an osteoclast: a large cell with multiple nuclei and a "foa....



View Image

In this image, several osteoblasts display a prominent Golgi apparatus and are actively synthesizing osteoid. Two osteocytes can also be seen.

Approach Considerations

A clinical practice guideline from the American College of Physicians on treatment to prevent fractures in men and women with low bone density or osteoporosis.includes six recommendations: two strong recommendations, based on high- or moderate-quality evidence, and four weak ones, based on low-quality evidence.[126] The two strong recommendations are as follows:

The four weak recommendations are as follows:

Nonpharmacologic preventive measures include modification of general lifestyle factors, such as increasing weight-bearing and muscle-strengthening exercise, which epidemiologic studies have linked to lower fracture rates, and ensuring optimum calcium and vitamin D intake as adjunct to active antifracture therapy.[10] In addition, potentially treatable underlying causes of osteoporosis such as hyperparathyroidism and hyperthyroidism should be ruled out or treated if detected.

Surgical care includes vertebroplasty and kyphoplasty, which are minimally invasive spine procedures used for the management of painful osteoporotic vertebral compression fractures. However, there may be an increased risk of fractures involving adjacent vertebrae after these procedures.[127, 128]

A 2008 literature review suggested that the use of "reminders plus education targeted to physicians and patients" can lead to increased BMD testing and greater use of osteoporosis medications.[129] In addition, a physician reminder in conjunction with a patient risk assessment strategy apparently can result in a reduction in patient fractures and an increase in osteoporosis therapy. The authors concluded that multicomponent tools aimed at doctors and patients may support clinical decision making in the management of osteoporosis.

A 2009 study indicated that the use of a case manager for the treatment of patients with hip fractures can lead to more frequent use of appropriate osteoporosis treatment and may result in fewer fractures, increased life expectancy, and significant health-care cost savings.[14]

In patients who have experienced an osteoporotic fracture, the first goal of rehabilitation is to control pain. Spinal compression fractures can be extremely painful and can cause short- and long-term morbidity. Oral analgesics on a regular schedule can be implemented. Pain-relieving modalities such as moist hot packs and transcutaneous electrical nerve stimulation should also be considered. During this period, monitoring the patient carefully for signs of constipation, urinary retention, and respiratory depression, which can occur with the use of narcotic analgesics, is essential.

A comfortable mechanical support for the spine and, in some cases, a thoracic orthosis may need to be prescribed. The primary reason for the application of a thoracic orthosis is to limit motion in the spine. The length of time a patient should wear a rigid spinal orthosis is undetermined. What is well known is that immobilization contributes to bone demineralization. During the early mobilization period, deep breathing exercises, pectoral and intercostal muscle strengthening, and back conservation techniques need to be implemented.

Pharmacologic Therapy

Currently, no treatment can completely reverse established osteoporosis. Early intervention can prevent osteoporosis in most people. For patients with established osteoporosis, medical intervention can halt its progression. If secondary osteoporosis is present, treatment for the primary disorder should be provided. Therapy should be individualized based on each patient’s clinical scenario, with the risks and benefits of treatment discussed between the clinician and patient.[9, 130]

Patients identified as at risk for osteoporosis (including children and adolescents) should undergo preventive measures, including adequate calcium intake, vitamin D intake, and exercise. Patients should be counseled to avoid tobacco and excessive alcohol use.

Protective measures should be taken in patients who must take glucocorticoids for other medical conditions. These include using the minimum effective dose, discontinuing the drug as soon as possible, and supplementing with calcium and vitamin D.

Bisphosphonates

Bisphosphonates are the most commonly used agents for osteoporosis. They have been employed for both treatment and prevention. Oral and intravenous options are available.

Alendronate (Fosamax) is approved for the treatment of osteoporosis in men, in postmenopausal women, and in patients with glucocorticoid-induced osteoporosis. It has been shown to increase spinal and hip mineral density in postmenopausal women. Well-conducted controlled clinical trials indicate that alendronate reduces the rate of fracture at the spine, hip, and wrist by 50% in patients with osteoporosis.

The treatment dose of alendronate is 70 mg/wk, to be taken sitting upright with a large glass of water at least 30 minutes before eating in the morning. Alendronate is also available in combination with cholecalciferol (vitamin D3). The combination of alendronate with vitamin D3 (Fosamax Plus D) is indicated for the treatment of osteoporosis in men to increase bone mass.

The results of a population-based, national register–based, open cohort study of 38,088 patients suggest that elderly patients who use proton pump inhibitors in conjunction with alendronate have a dose-dependent loss of protection against hip fracture.[131]

Other oral bisphosphonates include risedronate (Actonel) or risedronate delayed-release (Atelvia), given daily, weekly, or monthly. It is also available as a combination product with calcium as risedronate/calcium carbonate (Actonel with Calcium). Risedronate reduced vertebral fractures by 41% and nonvertebral fractures by 39% over 3 years.[132]

Ibandronate (Boniva) is another bisphosphonate that can be given orally once a month and is also available as an intravenous formulation that is given every 3 months. Intravenous bisphosphonates are excellent choices for patients intolerant of oral bisphosphonates or for those in whom adherence is an issue.  However, ibandronate has not shown efficacy in nonvertebral fractures in clinical trials.

Zoledronic acid

Zoledronic acid (Reclast) is a once-yearly intravenous infusion approved for the treatment of osteoporosis in men, in postmenopausal women, and in patients with glucocorticoid-induced osteoporosis.[133]  Zoledronic acid is the most potent bisphosphonate available. It increases BMD at the spine by 4.3-5.1% and the hip by 3.1-3.5%, as compared with placebo. Over 3 years, it reduces the incidence of spine fractures by 70%, hip fractures by 41%, and nonvertebral fractures by 25%. A similar effect on vertebral fractures has been shown in men. A 2012 randomized, 2-year trial of men with osteoporosis found that once-yearly zoledronic acid infusions significantly decreased the risk of new morphometric vertebral fractures by 67%.[134]

A randomized, placebo-controlled, double-blind trial suggested that a once-yearly 5-mg dose of IV zoledronic acid increases bone mass in men within 90 days of hip fracture repair; similar increases were noted in women.[135]

In September 2011, the US Food and Drug Administration (FDA) updated the prescribing information for zoledronic acid to provide improved information regarding the risk of kidney failure. Acute renal failure requiring dialysis and fatal outcomes have been reported to the FDA following the use of zoledronic acid. The drug is contraindicated in patients with moderate to severe renal impairment (ie, creatinine clearance < 35 mL/min) or evidence of acute renal impairment. Risk for renal impairment is also increased by the following[136] :

In March 2012, the FDA made safety labeling changes for zoledronic acid to warn against atypical subtrochanteric and diaphyseal femoral fractures in patients who have received bisphosphonate therapy including zoledronic acid. Fractures occur with minimal or no trauma and may present as groin pain weeks to months after fracture[137]

In November 2012, the FDA made safety labeling changes for zoledronic acid to warn against the following adverse reactions[137] :

Bisphosphonates and bone turnover

Over time, bisphosphonate therapy decreases bone turnover and, at very high levels in animals, decreases bone strength and resilience. Some limited reports, including that by Odvina et al, describe patients on long-term bisphosphonate therapy developing transverse stress fractures; biopsy specimens of these individuals have suggested extremely low turnover states in some of them.[138]

Alendronate and risedronate inhibit bone resorption at doses 10-fold lower than those reducing osteoclast number. Thus, suppression of bone resorption with these agents is independent of their effects on apoptosis.[139]

Treatment interval and complications with bisphosphonate therapy

The limited trial data available regarding long-term treatment with bisphosphonates has raised questions about the optimal length of treatment with these medications.[140] This issue has become more important, given newly recognized complications of bisphosphonate use, including osteonecrosis of the jaw[141] and atypical (subtrochanteric or femoral shaft) femur fractures (see the images below).



View Image

Normal femoral anatomy.



View Image

Stable intertrochanteric fracture of the femur.

Some studies have sought to clarify the true risks of complications in patients receiving bisphosphonates. A Canadian study by Park-Willie et al found the estimated absolute risk of a subtrochanteric or femoral shaft fracture to be low in 52,595 women with at least 5 years of bisphosphonate therapy (0.13% during the subsequent year and 0.22% within 2 years).[142] Overall, a patient’s risk of fracture can be used to help guide length of treatment. Patients at high risk may be continued on bisphosphonates after 5 years; however, in some patients, especially those with a lower risk of fracture, bisphosphonate treatment may be stopped.[143]

The AACE recommends that if osteoporosis is mild, clinicians should consider a drug holiday after 4-5 years of bisphosphonate treatment; if fracture risk is high, a drug holiday of 1-2 years may be considered after 10 years of treatment.[11] BMD and bone turnover markers should be monitored during the drug holiday, and treatment should be restarted if density declines substantially, bone turnover markers increase, or a fracture occurs.[11]

In 2016, the American Society for Bone and Mineral Research published guidelines on long-term bisphosphonate treatment that included the following recommendations[144] :

Selective estrogen receptor modulators

SERMs are considered to provide the beneficial effects of estrogen without the potentially adverse outcomes. Raloxifene (Evista) is a SERM indicated for the treatment and prevention of osteoporosis in postmenopausal women. The usual dose is 60 mg given orally daily. It can also be given in combination with calcium and vitamin D. It is the first SERM studied for breast cancer prevention, and it decreases bone resorption through actions on estrogen receptors.

Raloxifene has been shown to prevent bone loss, and data in women with osteoporosis have demonstrated that this agent causes a 35% reduction in the risk of vertebral fractures. It has also been shown to reduce the prevalence of invasive breast cancer. However, it has been shown to increase the incidence of deep vein thrombosis, stroke, and hot flashes.

Raloxifene may be most useful in younger postmenopausal women without severe osteoporosis. In 601 postmenopausal women who had daily therapy with raloxifene, BMD was increased, serum concentrations of total low-density lipoprotein cholesterol were lowered, and the endometrium was not stimulated.

Pooled mortality data from large clinical trials of raloxifene (60 mg/day) were analyzed by Grady et al in 2010. When compared with placebo, all-cause mortality was 10% lower in older postmenopausal women receiving raloxifene. The primary reduction was in noncardiovascular, noncancer deaths.[145]

The combination product of bazedoxifene, a SERM, and conjugated estrogens (CEs) was approved by the FDA in October 2013 for prevention of osteoporosis and treatment of vasomotor symptoms in postmenopausal women. Combining a SERM with CEs lowers the risk of uterine hyperplasia caused by estrogens. This eliminates the need for a progestin and its associated risks (eg, breast cancer, myocardial infarction, venous thromboembolism).

In clinical trials, this combination decreased bone turnover and bone loss in postmenopausal women at risk for osteoporosis. Bone mineral density increased significantly more with all bazedoxifene/CE doses compared with placebo at the lumbar spine and total hip and with most bazedoxifene/CE doses compared with raloxifene at the lumbar spine.[146]

Parathyroid hormone

Teriparatide

Teriparatide (Forteo) is a recombinant human parathyroid hormone (1-34) (PTH [1-34]) that acts as an anabolic agent for the treatment of osteoporosis. It is indicated for the treatment of women with postmenopausal osteoporosis who are at high risk of fracture, who have been intolerant of previous osteoporosis therapy, or in whom osteoporosis treatment has failed to increase bone mass. It is indicated in men with idiopathic or hypogonadal osteoporosis who are at high risk of fracture, who have been intolerant of previous osteoporosis therapy, or in whom osteoporosis therapy has failed. Teriparatide is also approved for the treatment of patients with glucocorticoid-induced osteoporosis.

Before treatment with teriparatide, levels of serum calcium, PTH, and 25(OH)D need to be monitored. Teriparatide cannot be given for more than 2 years. Contraindications include the following:

When PTH is given continuously, it is associated with increased osteoclastic and osteoblastic turnover, leading to a net loss of bone. However, in an intermittent subcutaneous administration of 20 mcg/day, PTH has been demonstrated to lead to a very active anabolic phase, with bone mass increasing up to 13% over 2 years in the spine and to a lesser degree in the hip.[149, 150, 151]

Indications for PTH in men and women are a bone density decline while on bisphosphonate therapy, bone density stabilization while on extremely low-level bisphosphonate therapy, a fracture occurring while on bisphosphonate therapy, or a very low initial bone turnover rate for which an anabolic effect is clearly warranted. Teriparatide should be considered in younger and older postmenopausal women with severe osteoporosis.

Most studies with PTH have been performed on women. The medication decreases the risk of vertebral and nonvertebral fractures to the same extent as bisphosphonates. Teriparatide is given for a maximum of 2 years, after which time the gains in BMD achieved with PTH are secure and can even be augmented with bisphosphonate therapy; otherwise, the BMD slowly deteriorates to pretreatment levels.[152]

According to Finkelstein et al, initial studies using a combination of concurrent PTH and bisphosphonate therapy showed decreased benefit compared with therapy with either agent alone; therefore, the general recommendation is that these drugs be given separately and in sequence.[153]

A study by Cosman and colleagues challenged this conclusion by giving 3-month-on, 3-month-off pulses of teriparatide while the patients were on weekly alendronate; BMD in the spine increased above that of the alendronate-only arm.[154] This pulsed regimen appears to take advantage of the 3- to 4-month so-called anabolic window, in which the markers of bone formation rise more quickly than the markers of bone resorption.

Studies by Deal et al and Ste-Marie et al on women have shown that the concurrent use of estrogen or raloxifene can enhance the bone-forming effects of teriparatide.[155, 156] Data on the use of PTH in men are much more limited, but they appear to have relatively comparable efficacy.

In a randomized, controlled trial, 94 postmenopausal women with weak bones who took teriparatide and denosumab in combination had increased BMD after 12 months. The 12-month changes in posterior-anterior lumbar spine, femoral-neck, and total-hip BMD in the combination-therapy group (9.1%, 4.2% and 4.9%, respectively) were greater than those in the groups receiving only teriparatide (6.2%, 0.8% and 0.7%, respectively) or only denosumab (5.5%, 2.1% and 2.5%, respectively).[157]

In a retrospective analysis of the data from the Fracture Prevention Trial and the Multiple Outcomes of Raloxifene Evaluation trial, Bouxsein et al found that teriparatide reduced fracture risk to a greater extent than raloxifene in postmenopausal osteoporotic women. Compared with placebo, teriparatide reduced the risk of any new fractures by 72%, new adjacent fractures by 75%, and new nonadjacent vertebral fractures by 70%. Raloxifene reduced the risks by 54%, 54%, and 53%, respectively.[158]

A double-blind, double-dummy trial by Kendler et al found that in postmenopausal women with severe osteoporosis, the risk of new vertebral and clinical fractures was significantly lower in patients receiving teriparatide than in those receiving risedronate. Study subjects had experienced at least two moderate or one severe vertebral fracture and had a bone mineral density T score of -1.50 or less. They were randomly assigned to receive teriparatide (daily injections of 20 μg) plus oral weekly placebo or risedronate (35 mg orally once weekly) plus daily injections of placebo for 24 months.[159]

At 24 months, new vertebral fractures occurred in 28 (5.4%) of 680 patients in the teriparatide group and 64 (12.0%) of 680 patients in the risedronate group (hazard ratio [HR] 0.44, 95% confidence index [CI] 0.29-0.68; P < 0·0001). Clinical fractures occurred in 30 (4.8%) of 680 patients in the teriparatide group compared with 61 (9.8%) of 680 in the risedronate group (hazard ratio 0.48, 95% CI 0.32-0.74; P=0.0009).[159]

A study performed by an Austrian group using PTH 1-84 to treat pelvic fractures in postmenopausal women with osteoporosis demonstrated that this anabolic agent has the ability to both increase the rate of union and enhance the speed of the process. In addition to improved fracture healing, treatment with PTH 1-84 was also associated with a significant decrease of pain and improved function over the placebo arm. This clinical study supports the extensive animal data that predicted a clear role for PTH in fracture repair.[160]

Use of teriparatide for atypical fractures has not been studied in large randomized controlled trials due to the small numbers of atypical fractures. Gomberg et al reported a case of successful healing of bilateral subtrochanteric stress fractures in a postmenopausal woman who had used a bisphosphonate for 13 years.[161] A small case series showed that teriparatide may have potential in promoting healing of atypical fractures with radiolucent line in an incomplete fracture.[162]

For osteonecrosis of the jaw (ONJ), teriparatide may be a useful adjunctive therapy. Three cases have been reported in which teriparatide has been used successfully in patients with bisphosphonate-associated ONJ.[163, 164, 165]

Abaloparatide

Another PTH analogue, abaloparatide (Tymlos), was approved by the FDA in April 2017. Approval was based on results at 18 months from the Abaloparatide Comparator Trial in Vertebral Endpoints (ACTIVE) trial[166] and first 6 months of the ACTIVExtend trial.[167]

In the ACTIVE trial of more than 2000 women, subcutaneous abaloparatide was associated with significant reductions in the relative risk for new vertebral fractures (86% reduction) and nonvertebral fractures (43% reduction) compared with placebo. The absolute risk reductions were 3.6% and 2.0%, respectively. The benefits were evident regardless of age, years since menopause, presence or absence of prior fracture (vertebral or nonvertebral), and bone mineral density (BMD) at baseline.[166]

In the ACTIVExtend trial, roughly 1100 patients who completed 18 months of abaloparatide or placebo in ACTIVE received open-label alendronate for up to 24 additional months. Data from the first 6 months of ACTIVExtend showed improved BMD and reduced fracture risk throughout the skeleton.[167]

Romosozumab

Romosozumab (Evenity) is a monoclonal antibody that binds with and inhibits sclerostin, and thus both increases bone formation and decreases bone resorption. It was approved by the FDA in 2019 for treatment of osteoporosis in postmenopausal women who are at high risk for fracture. It has been shown to reduce vertebral fracture rates in postmenopausal women with osteoporosis.[168, 169, 170, 171]

In the Fracture Study in Postmenopausal Women With Osteoporosis (FRAME), a phase 3 randomized trial in 7180 postmenopausal women who had a T score of -2.5 to -3.5 at the total hip or femoral neck, 1 year of treatment with romosozumab reduced vertebral fracture rates by 73% compared with placebo. A further reduction in vertebral fracture risk occurred in the second year following transition to denosumab.[169, 171]

Romosozumab is also being investigated in men with osteoporosis. In the BRIDGE clinical trial, spine and hip BMD significantly improved following romosozumab for 12 months compared with placebo.[172]

Denosumab

Denosumab (Prolia) is a humanized monoclonal antibody directed against the receptor activator of the nuclear factor–kappa B ligand (RANKL), which is a key mediator of the resorptive phase of bone remodeling.[156] It decreases bone resorption by inhibiting osteoclast activity. Denosumab was approved by the US Food and Drug Administration in June 2010. It has been studied in cancer patients, patients with postmenopausal osteoporosis, and men with low bone mineral density (BMD).[173, 174, 175]  The approved dosage is 60 mg given subcutaneously every 6 months.

Denosumab may be considered in certain patients with renal insufficiency, as impaired renal function does not significantly affect the metabolism or excretion of the drug.[176]

In May 2018, denosumab was approved for treatment of glucocorticoid-induced osteoporosis (GIOP) in men and women at high risk of fracture who are either initiating or continuing systemic glucocorticoids in a daily dosage equivalent to 7.5 mg or greater of prednisone and who are expected to remain on glucocorticoids for at least 6 months. Patients are considered at high risk of fracture if they have a history of osteoporotic fracture, multiple risk factors for fracture, or failed or cannot tolerate other available osteoporosis therapy.

Approval for use in GIOP was based on an international phase 3 study in which denosumab proved more effective than the bisphosphonate risedronate for increasing BMD at the lumbar spine. In patients who had been on steroids for at least 3 months (n=505), the change from baseline in BMD at 12 months was 4.4% with denosumab versus 2.3% with risedronate (P < 0.0001); in those who had started steroids less than 3 months previously (n=290), the change was 3.8% vs 0.8% (P < 0.0001).[177, 178]

In postmenopausal women with osteoporosis, denosumab reduces the incidence of vertebral, nonvertebral, and hip fractures.[179]  Denosumab also increases bone mass in men at high risk for fracture who are receiving androgen deprivation therapy for nonmetastatic prostate cancer,[180]  and in women at high risk for fracture receiving adjuvant aromatase inhibitor therapy for breast cancer. 

A meta-analysis of 10 randomized, controlled trials concluded that denosumab improved BMD significantly more than bisphosphonate treatment at the lumbar spine, total hip, and femoral neck at 12 and 24 months. However, only 1 of the 10 studies demonstrated greater osteoporotic fracture reduction with denosumab treatment.[181]

In patients with multiple myeloma or bone metastases from breast cancer, a single subcutaneous dose of denosumab decreases bone turnover markers within 1 day, and this effect is sustained through 84 days at higher doses.[173] Denosumab has been shown to increase BMD and decrease bone resorption in postmenopausal women with osteoporosis over a 12-month period.

Because the overactivity of RANKL is a major factor in bone loss in patients with autoimmune and inflammatory disorders, such as ulcerative colitis, denosumab may become first-line therapy for these patients.[182]

Denosumab in combination with teriparatide has been shown to increase BMD more than either drug alone.[183]

Calcitonin

Calcitonin-salmon (Fortical, Miacalcin) is a hormone that decreases osteoclast activity, thereby impeding postmenopausal bone loss. It is indicated for the treatment of women who are more than 5 years post menopause and have low bone mass relative to healthy premenopausal women. Calcitonin-salmon should be reserved for patients who refuse or cannot tolerate estrogens or in whom estrogens are contraindicated. It is recommended in conjunction with adequate calcium and vitamin D intake to prevent the progressive loss of bone mass. It is available as an injection and as an intranasal spray. The intranasal spray is delivered as a single daily spray that provides 200 IU of the drug. The drug can be delivered subcutaneously, but this route is rarely used.

Results from a single controlled clinical trial indicate that calcitonin may decrease osteoporotic vertebral fractures by approximately 30%. In the first 2 years, calcitonin has been found to increase spinal bone mineral density (BMD) by approximately 2%. Calcitonin also has an analgesic property that makes it ideally suited for the treatment of acute vertebral fractures.

In 2013, an FDA post-marketing review was prompted after studies showed increased risk of malignancies in calcitonin-treated patients. In a meta-analysis of 21 randomized, controlled clinical trials with calcitonin-salmon (nasal spray or investigational oral formulations), the incidence of malignances in calcitonin-treated patients was 4.1%, versus 2.9% in placebo-treated patients. The data were not sufficient for further analysis by specific malignancies and a definitive causal relationship between calcitonin use and malignancies could not be established. The FDA has recommended that health care professionals assess a patient’s need for osteoporosis therapy as well as benefits and risk of available treatments.[184]

Calcitonin is no longer widely used for treatment of osteoporosis.[126]  Nevertheless, it remains an option for patients who are not candidates for other available osteoporosis treatments. Common side effects of nasally administered calcitonin include nasal discomfort, rhinitis, irritation of nasal mucosa, and occasional epistaxis. Nausea, local inflammatory reactions at the injection site, sweating, and flushing are side effects noted with parenteral use.

Hormone replacement therapy

Hormone replacement therapy (HRT) was once considered a first-line therapy for the prevention and treatment of osteoporosis in women. Although HRT is not currently recommended for the treatment of osteoporosis, it is important to mention because many osteoporosis patients in a typical practice still use it for controlling postmenopausal symptoms.

The American College of Physicians concluded that moderate-quality evidence showed that estrogen treatment did not reduce fracture rates in postmenopausal women with established osteoporosis.[126] Data from the Women's Health Initiative indicated that HRT can reduce fractures.[185] However, the results of the Women's Health Initiative were distressing with respect to the adverse outcomes associated with combined estrogen and progesterone therapy (eg, risks for breast cancer, myocardial infarction, stroke, and venous thromboembolic events) and estrogen alone (eg, risks for stroke and venous thromboembolic events).

Other agents

Strontium ranelate is approved for the treatment of osteoporosis in some countries in Europe. It reduces the risk of both spine and nonvertebral fractures.[186, 187] Strontium is not approved for the treatment of osteoporosis in the United States.

Vertebroplasty and Kyphoplasty

The goals of surgical treatment of osteoporotic fractures include rapid mobilization and return to normal function and activities. Traditional operative management of vertebral compression fractures is uncommon and is usually reserved for gross spinal deformity or for threatened or existing neurologic impairment.

Operative interventions include anterior and posterior decompression and stabilization with placement of such internal fixation devices as screws, plates, cages, or rods. Bone grafting is routinely performed to achieve bony union. The failure rate of spinal arthrodesis is significant because achieving adequate fixation of hardware in osteoporotic bone is difficult. Moreover, patients who are elderly have a reduced osteogenic potential.

Vertebroplasty and balloon kyphoplasty are indicated in patients with incapacitating and persistent severe focal back pain related to vertebral collapse. Both procedures involve the injection of acrylic cement (methylmethacrylate) into the fractured vertebral body. The procedures are performed under local anesthesia and either fluoroscopic or CT guidance. Both procedures relieve pain; however, in kyphoplasty a balloon tamp is inflated within and between the fracture fragments before the cement is infused, in order to restore vertebral body height (see the images below).



View Image

In kyphoplasty, a KyphX inflatable bone tamp is percutaneously advanced into the collapsed vertebral body (A). It is then inflated, (B) elevating the ....



View Image

Osteoporosis. Lateral radiograph demonstrates multiple osteoporotic vertebral compression fractures. Kyphoplasty has been performed at one level.



View Image

Osteoporosis. Lateral radiograph of the patient seen in the previous image following kyphoplasty performed at 3 additional levels.

There is still considerable controversy about the efficacy and safety of these procedures.[188] For more information, see Percutaneous Vertebroplasty and Kyphoplasty.

Dietary Measures

Adequate calcium and vitamin D intake are important in persons of any age, particularly in childhood as the bones are maturing, and are essential in the prevention and treatment of osteoporosis. Vitamin D is increasingly being recognized as a key element in overall bone health, calcium absorption, balance (eg, reduction in risk of falls),[189] and muscle performance. Patients who ingest inadequate amounts of vitamin D and calcium should receive oral supplementation.

Good dietary sources of calcium include dairy products, sardines, nuts, sunflower seeds, tofu, vegetables such as turnip greens, and fortified food such as orange juice. Good dietary sources of vitamin D include eggs, liver, butter, fatty fish, and fortified food such as milk and orange juice. See the National Osteoporosis Foundation Website for further calcium and vitamin D recommendations.

The goal of the current recommendations for daily calcium intake is to ensure that individuals maintain an adequate calcium balance. Current recommendations from the American Association of Clinical Endocrinologists (AACE) for daily calcium intake are as follows[11] :

The Institute of Medicine (IOM) has issued recommendations for calcium and vitamin D daily intake in older adults. For women older than 50 years, the IOM recommended 1200 mg/day of calcium. The IOM recommended 1000 mg/day of calcium for men 51-70 years of age and 1200 mg/day for men over 70. For both sexes, the recommended upper level was 2000 mg/day.[190]

For both women and men, the recommended daily dietary allowance of vitamin D was 600 IU from age 51-70 and 800 IU for after age 70, with a recommended maximum of 4000 IU. Amid considerable controversy, the IOM also stated that the evidence supported a role for vitamin D and calcium in bone health, but not in other conditions.[190]

The minimum daily requirement of vitamin D in patients with osteoporosis is 800 IU of cholecalciferol. Many patients require higher levels (continuously or for a short period) to be considered vitamin D replete, which is defined as a serum 25-hydroxyvitamin D level of at least 32 ng/mL.

Alcohol and anorexia nervosa can interfere with nutrition. Excessive alcohol intake can interfere with calcium balance by increasing PTH production and by inhibiting the enzymes that convert inactive vitamin D to its active form; in addition, alcohol can result in hormonal deficiencies and can increase the tendency for falls. Poor nutritional states, such as in anorexia nervosa,[191] have been strongly associated with bone loss. Nutritional and endocrine factors contribute to bone loss; in particular, low estrogen states, which result from low body weight, result in significant bone loss.

Calcium and vitamin D supplementation

Commonly used calcium supplements include calcium carbonate and calcium citrate. Calcium carbonate is generally less expensive and is recommended as a first choice option. Calcium carbonate has better absorption with food, as opposed to calcium citrate, which is better absorbed in the fasting state. Also, fewer tablets are needed with calcium carbonate than with calcium citrate.

Vitamin D is available as ergocalciferol (vitamin D2) and cholecalciferol (vitamin D3). Vitamin D is metabolized to active metabolites. These metabolites promote the active absorption of calcium and phosphorus by the small intestine, elevating serum calcium and phosphate levels sufficiently to permit bone mineralization. Ergocalciferol, the form mostly used in supplements and fortified foods, is absorbed with similar efficiency to cholecalciferol, the main dietary form, with similar effects on suppression of PTH.

Calcium and vitamin D studies

Several large studies have demonstrated that supplementation with a combination of calcium and vitamin D can reduce fracture risk.[192] A meta-analysis of 12 double-blind, randomized, controlled trials for nonvertebral fractures and eight trials for hip fractures found that nonvertebral fracture prevention with vitamin D was dose dependent and that a higher dose reduced fractures by at least 20% for individuals aged 65 years or older.[193] However, a longitudinal and prospective cohort study concluded that gradual increases in dietary calcium intake did not further reduce fracture risk or osteoporosis in women.[194]

Another meta-analysis concluded that vitamin D alone is not effective in preventing fractures, but vitamin D given together with calcium reduced hip fractures and total fractures, and possibly vertebral fractures.[195] The conclusions were based on seven large studies that were randomized with at least one intervention arm in which vitamin D was given and included analysis of fractures as an outcome and at least 1000 participants.

More information is needed regarding risks associated with long-term calcium supplementation. Bolland et al and Li et al found an increased risk for myocardial infarction associated with calcium supplements.[196, 197] Others have not found an association.[198, 199] Dietary calcium intake has not been associated with an increased risk of cardiovascular events.[197] Additionally, some[200] but not all[201] studies suggest an increased risk of nephrolithiasis with calcium and vitamin D supplements, but not with calcium ingested in the diet.[202]

Physical and Occupational Therapy

Physical therapy

Physical therapy focuses on improving a patient's strength, flexibility, posture, and balance to prevent falls and maximize physical function.[203, 204] Postural retraining is key in this population. Spinal bone mineral density (BMD) is directly correlated with the strength of the back extensors; therefore, maintaining and strengthening the back extensors should be emphasized.[205] In studies by Sinaki and colleagues, strengthening the back extensor muscles reduced kyphosis and decreased the risk of sustaining vertebral compression fractures.[206, 207]

As soon as the course of therapy allows, weight-bearing exercises should be initiated. Regular weight-bearing exercises are essential for the maintenance of bone mass[55] and should be encouraged in all patients, including children and adolescents (to strengthen the skeleton during the maturation process). Exercise also improves agility and balance, thereby reducing the risk of falls.

Occupational therapy

Training in the performance of activities of daily living (ADLs) and in the proper use of adaptive equipment are essential to the prevention of future falls.[204] Home modification focuses on reducing the risk of falling by installing handrails and grab bars in hallways, stairs, and bathrooms. The use of a shower chair, tub bench, and adaptive bathing devices also can be beneficial. The application of nonskid tape to steps (indoors and outdoors), as well as the removal of throw rugs, greatly improves home safety.

Exercise

Aerobic low-impact exercises, such as walking and bicycling, generally are recommended. During these activities, ensure that the patient maintains an upright spinal alignment. Sinaki and Mikkelsen showed that exercises that place flexion forces on the vertebrae tend to cause an increase in the number of vertebral fractures in patients.[205]

Proper therapy for osteoporosis includes 3-5 sessions per week of weight-bearing exercises, such as walking or jogging, with each session lasting 45-60 minutes. The patient should be instructed in a home-exercise program that incorporates the necessary elements for improving posture and overall physical fitness.

In postmenopausal women, impact exercises can increase BMD in the hip and spine. Chien et al examined the efficacy of a 24-week aerobic exercise program consisting of treadmill walking followed by stepping exercises in osteopenic postmenopausal women aged 48-65 years. Women who exercised had increased bone mineral density in L2-L4 and the femoral neck, as well as improved quadriceps strength, muscular endurance, and peak exercise oxygen consumption (VO2 max), whereas values in the control group declined.[208]

Snow et al found increased BMD of the femoral neck, trochanter, and total hip in 18 postmenopausal women (average age, 64 years) who wore weighted vests and participated in jumping exercises 3 times per week for 32 weeks a year for 5 years.[209]

The results of a Cochrane Database of Systematic Reviews study found that exercise may help prevent bone loss and fractures in postmenopausal women. The most effective type of exercise on BMD for the neck of the femur was found to be non–weight-bearing, high-force exercise, such as lower limb resistance strength training; combination exercise programs were most effective for BMD at the spine.[210]

Although swimming is not a weight-bearing exercise that will improve BMD, it does provide chest expansion, spinal extension, and low-impact cardiopulmonary fitness. Isometric exercises should also be used to strengthen abdominal muscles, aiding in the prevention of a kyphosis.

The physical therapist must address balance training, because fall prevention is important in eliminating the complication of fracture. Improving one's balance can significantly lower the risk of falling. Balance training incorporates the strengthening of various parts of the body (eg, trunk, legs), proprioception, and vestibular input. Several different exercises have been shown to be beneficial in patients with osteoporosis.[211, 212, 213, 214]

Tai chi chuan and specific physical therapy programs have been shown to be particularly effective in improving balance and reducing falls. Wolf et al monitored 200 elderly community dwellers who received tai chi and computerized balance training. After a 15-week intervention, the authors documented decreased fear of falling responses. In addition, tai chi was shown to reduce the risk of multiple falls by 47.5%.[215]

Campbell et al monitored 233 elderly community dwellers randomized to an individually tailored physical therapy program in the home compared with usual care and an equal number of social visits. The authors found that after one year, the mean rate of falls was lower in the exercise group than in the control group (0.87 vs 1.34, respectively). In addition, after 6 months, persons in the exercise group had improved balance.

Other types of exercise training programs may also positively impact balance and strength. Carter et al demonstrated that osteoporotic women aged 65-75 years who underwent a 10-week community-based physical activity intervention program improved their static balance, dynamic balance, and knee extension strength, although they did not benefit from a significant reduction in fall risk factors.[216]

Prevention of Osteoporosis

Primary prevention of osteoporosis starts in childhood. Patients require adequate calcium intake, vitamin D intake, and weight-bearing exercise. Beyond this, prevention of osteoporosis has two components: behavior modification and pharmacologic interventions.

The National Osteoporosis Foundation specifies that the following behaviors should be modified to reduce the risk of developing osteoporosis[2] :

Patients should be counseled on smoking cessation and moderation of alcohol intake. Regular weight-bearing exercise and back extensor strengthening help delay bone loss. In a study that found osteopenia in over a quarter of men and women in early middle age, there was a negative correlation between exercise and BMD in the men despite relatively high levels of exercise—but the majority of men in the study reported cycling as their preferred exercise, rather than weight-bearing activities such as walking or running.[66]

Patients who have disorders or take medications that can cause or accelerate bone loss should receive calcium and vitamin D supplementation and, in some cases, pharmacologic treatment.[217]  Pharmacologic prevention methods include calcium supplementation and administration of raloxifene or bisphosphonates (alendronate or risedronate). Raloxifene and bisphosphonates should be considered as first-line agents for the prevention of osteoporosis.[218]

In 2010, the American College of Rheumatology published revised recommendations for the prevention and treatment of glucocorticoid-induced osteoporosis. Recommendations included the categorization of patients by fracture risk (using the FRAX score) and initiation of treatment in appropriate patients including alendronate, risedronate, zoledronic acid, and teriparatide (in those patients at highest risk).[12]

Estrogen-progestin therapy is no longer considered a first-line approach for the treatment of osteoporosis in postmenopausal women, because it has been linked to an increased risk for breast cancer, stroke, venous thromboembolism, and perhaps coronary disease. Estrogen is now only recommended if patients are also seeking relief of postmenopausal symptoms.

Regular monitoring may be helpful. Periodic bone densitometry helps in diagnosing osteoporosis in the early phase and aids in preventing fractures. According to the NOF, evaluating BMD on a periodic basis is the best way to monitor bone density and future fracture risk.[2] However, there is considerable controversy about how often to repeat BMD measurements, particularly in postmenopausal women with a normal baseline BMD.[219]

Consultations

For a patient with osteoporosis in the diagnostic and therapeutic phases, the most important consultation is with a rheumatologist or an endocrinologist. These specialists can help obtain the proper laboratory tests and imaging studies needed to rule out causes of secondary osteoporosis. In patients with uncontrolled pain that does not respond to conventional therapies, an invasive pain specialist may be consulted for proper interventional procedures.

A rheumatologist may also provide useful assistance with management and determination of underlying etiologies in complex cases.

Consultations can include discussions of nonmedical/nonpharmacologic management of osteoporosis.[220, 221] Consult an orthopedist to assist with fracture management. Consultation with a spine surgeon is appropriate for patients with intractable, severe, function-limiting symptomatology that has not been relieved by noninterventional techniques. Consultation with a nonsurgical spine specialist is appropriate for a patient who is not a surgical candidate or whose symptoms persist despite surgical fixation.

Long-Term Monitoring

The US Preventive Services Task Force (USPSTF) 2011 recommendations state that evidence is lacking regarding optimal intervals for repeated screening by dual-energy x-ray absorptiometry (DXA) for individuals with osteoporosis, as well as regarding whether a woman with a normal BMD requires repeated screening. The USPSTF noted that, “a minimum of 2 years may be needed to reliably measure a change in BMD; however, longer intervals may be necessary to improve fracture risk prediction.”[9]

According to a study funded by the National Institutes of Health, osteoporosis will develop in fewer than 10% of older, postmenopausal women during the following rescreening intervals[222] :

Orthotics

Orthotics are used to decrease the flexion forces to prevent the worsening of kyphosis and to reduce the pressure on the fracture site in the acute phase of disease.[223, 224] Common orthotics used include the following:

Screening

The following organizations have issued recommendations on measurement of bone mineral density (BMD) for osteoporosis screening:

National Osteoporosis Foundation

The 2014 NOF guidelines recommend BMD measurement in the following patients[2] :

International Society for Clinical Densitometry

The 2015 ISCD Official Positions recommend bone density testing in the following patients[3] :

Women discontinuing estrogen should be considered for bone density testing according to the indications listed above.

US Preventive Services Task Force

The USPSTF recommends measuring BMD in the following patients[226, 225]

Suggested risk assessment tools include the following:

In contrast to the NOF and ISCD, the USPSTF concludes that the current evidence is insufficient to assess the balance of benefits and harms of screening to prevent osteoporotic fractures in men.

Diagnosis

According to National Osteoporosis Foundation guidelines, a clinical diagnosis of osteoporosis may be made in a postmenopausal woman or in a man over age 50 years who is at an elevated risk for fracture, as indicated by any of the following[227] :

Treatment

The National Osteoporosis Foundation (NOF) recommends that pharmacologic therapy should be reserved for postmenopausal women and men aged 50 years or older who present with the following[2] :

A clinical practice guideline from the American College of Physicians (ACP) recommends offering pharmacologic treatment to women with known osteoporosis, to reduce the risk of hip and vertebral fractures.[126] The ACP recommends use of any of the following agents:

The ACP recommends against the use of estrogen or estrogen plus progestogen or raloxifene for the treatment of osteoporosis in postmenopausal women. Additional recommendations, based on low-quality evidence, include the following:

Guidelines from the American Association of Clinical Endocrinologists (AACE), published in 2010, include the following recommendations for choosing drugs to treat osteoporosis[11] :

Combination therapy with two or more agents has not been shown to have a greater effect on fracture reduction than single therapy. The AACE guidelines advise against the use of combination therapy, until the effect of combination therapy on fracture is better understood.

In June 2013, the National Osteoporosis Guideline Group (NOGG) updated its guidelines on the diagnosis and management of osteoporosis in men and postmenopausal women, aged 50 years or older, in the United Kingdom. Recommendations include the following[228] :

A 2019 guideline from the Endocrine Society on pharmacological management of osteoporosis in postmenopausal women includes the following recommendations[229] :

Medication Summary

Pharmacologic therapy for osteoporosis includes the use of antiresorptive agents to decrease bone resorption, such as bisphosphonates, the selective estrogen-receptor modulator (SERM) raloxifene, romosozumab, and denosumab. In addition, there are parathyroid hormone analogues that promote bone formation in patients with osteoporosis (eg, teriparatide, abaloparatide). All therapies should be given with calcium and vitamin D supplementation.

Alendronate (Fosamax); alendronate sodium/cholecalciferol (Fosamax Plus D)

Clinical Context:  Alendronate inhibits osteoclast activity and bone resorption. By binding to calcium salts, alendronate blocks the transformation of calcium phosphate into hydroxyapatite and inhibits the formation, aggregation, and dissolution of hydroxyapatite crystals in bone. Alendronate increases bone mineral density (BMD) at the spine by 8% and the hip by 3.5%. It reduces the incidence of vertebral fractures by 47% and nonvertebral fractures by 50% over 3 years. Alendronate is approved for the treatment and prevention of postmenopausal osteoporosis, male osteoporosis, and glucocorticoid-induced osteoporosis.

Risedronate (Actonel, Atelvia)

Clinical Context:  Risedronate is a potent antiresorptive agent that does not affect bone mineralization. The inclusion of an amino group within the heterocyclic ring makes risedronate one of the most potent antiresorptive bisphosphonates. As with other bisphosphonates, risedronate inhibits osteoclast formation and activity. Risedronate increases BMD at the spine by 5.4% and the hip by 1.6%. It reduces vertebral fractures by 41% and nonvertebral fractures by 39% over 3 years. It is approved for the treatment and prevention of postmenopausal osteoporosis, male osteoporosis, and glucocorticoid-induced osteoporosis.

Calcitonin salmon (Miacalcin, Fortical)

Clinical Context:  Calcitonin is used for the treatment of postmenopausal osteoporosis in women more than 5 years post menopause with low bone mass relative to healthy premenopausal females. Calcitonin-salmon injection should be reserved for patients who refuse or cannot tolerate estrogens or in whom estrogens are contraindicated. Use of calcitonin-salmon injection is recommended in conjunction with adequate calcium and vitamin D intake to prevent the progressive loss of bone mass. It inhibits osteoclastic bone resorption and has some analgesic effects in patients with fractures.

Although no research data support the idea that the use of intranasal calcitonin reduces the incidence of fractures, studies do show an increase in BMD with the use of calcitonin. Calcitonin increases BMD at the lumbar spine by 1-1.5%. It reduces the incidence of spine fracture by 33% in groups receiving 200 IU/day. It is available in parenteral and intranasal forms; however, the intranasal form is more convenient and better tolerated.

Ibandronate (Boniva)

Clinical Context:  Ibandronate increases BMD and reduces the incidence of vertebral fractures. Ibandronate increases BMD at the spine by 5.7-6.5% and the hip by 2.4-2.8%. It reduces vertebral fractures by 50% with intermittent (nondaily) dosing over 3 years; it has no effects on reduction of nonvertebral fractures. Ibandronate is approved for the treatment and prevention of postmenopausal osteoporosis. It is available as a 150-mg oral tablet and intravenous solution.

Zoledronic acid (Reclast)

Clinical Context:  Zoledronic acid inhibits bone resorption by altering osteoclast activity and by inhibiting normal endogenous, as well as tumor induced, mediators of bone degradation. Like other bisphosphonates, zoledronic acid binds to hydroxyapatite crystals in mineralized bone matrix. The binding to calcium phosphates slows the dissolution of hydroxyapatite crystals, as well as inhibits the formation and aggregation of these crystals. It increases BMD at the spine by 4.3-5.1% and at the hip by 3.1-3.5%, as compared with placebo. It reduces the incidence of spine fractures by 70%, hip fractures by 41%, and nonvertebral fractures by 25% over 3 years. Zoledronic acid is approved for the treatment and prevention of postmenopausal osteoporosis, glucocorticoid-induced osteoporosis, osteoporosis in men, and Paget disease of bone. It is contraindicated in patients with severe renal failure.

Class Summary

Calcium metabolism modifiers such as bisphosphonates are stable analogues of inorganic pyrophosphate. Bisphosphonates have a high affinity for hydroxyapatite crystals, and by binding at sites of active bone resorption, these agents can inhibit osteoclastic resorption. All oral bisphosphonates have poor absorption and have a bioavailability of less than 5%. Bone uptake is 20-80%, with the remainder being rapidly excreted through the kidneys.[230]

Bisphosphonates are approved in the United States for the prevention and treatment of postmenopausal osteoporosis, osteoporosis in males, and glucocorticoid-induced osteoporosis. Their major pharmacologic action is the inhibition of bone resorption.

Teriparatide (Forteo, Bonsity)

Clinical Context:  Teriparatide is recombinant human PTH 1-34, which has identical sequence to the 34 N-terminal amino acids (the biologically active region) of 84-amino acid human PTH. This anabolic agent acts as endogenous PTH, thus regulating calcium and phosphate metabolism in bone and kidneys. It works primarily to stimulate new bone by increasing number and activity of osteoblasts (bone-forming cells).

Additional physiological actions include regulation of bone metabolism, renal tubular reabsorption of calcium and phosphate, and intestinal calcium absorption. Teriparatide increases BMD at the lumbar spine by 9-13% and the hip by 3-6% compared with placebo. When given intermittently, PTH increases bone remodeling with the net effect of increased bone mass and improved skeletal microarchitecture. (This is in contrast to continuous exposure to PTH, which increases bone resorption with a net effect of decreased trabecular bone volume). PTH promotes new bone formation, leading to increased BMD. It reduces the risk of spine fractures by 65% and nonspinal fractures by 54% in patients after an average of 18 mo of therapy. Teriparatide is approved for men or women at high risk of fracture due to primary or hypogonadal osteoporosis or postmenopausal osteoporosis, respectively.

Abaloparatide (Tymlos)

Clinical Context:  Synthetic peptide analog of human parathyroid hormone-related protein (hPTHrP); hPTHrP is a naturally occurring hormone that, among other functions, regulates bone formation. It elicits anabolic effect on bone, demonstrated by increases in bone mineral density and content that correlated with increases in bone strength at vertebral and/or nonvertebral sites. Daily SC administration for 18 mo was associated with significant reductions in the relative risk for new vertebral fractures (86% reduction) and nonvertebral fractures (43% reduction) compared with placebo. The absolute risk reductions were 3.6% and 2.0%, respectively.

It is indicated for the treatment of postmenopausal women with osteoporosis at high risk for fracture, defined as a history of osteoporotic fracture, multiple risk factors for fracture, or patients who have failed or are intolerant to other available osteoporosis therapy.

Class Summary

Parathyroid hormone (PTH) is the primary regulator of calcium and phosphate metabolism in bone and kidneys.

Raloxifene (Evista)

Clinical Context:  The biological actions of raloxifene are largely mediated through binding to estrogen receptors, which results in activation of estrogenic pathways in some tissues (agonism) and blockade of estrogenic pathways in others (antagonism). Raloxifene increases BMD at the spine and the hip. It reduces the incidence of spine fractures by 30-55% over 3 years. Raloxifene is approved for the prevention and treatment of postmenopausal osteoporosis in women. It is available as 60 mg tablets that are given orally daily. Adverse reactions commonly seen include hot flashes, leg cramps, peripheral edema, flu syndrome, arthralgia, and sweating.

Class Summary

Selective estrogen receptor modulators (SERMs) affect some of the receptors stimulated by estrogen but can selectively act as an antagonist or agonist, depending on the organ system. Like estrogen, these are antiresorptive agents. However, because of their selective receptor-modulating property, they provide the beneficial effects of estrogens without the adverse effects.

Denosumab (Prolia)

Clinical Context:  Denosumab binds to the receptor activator of nuclear factor-kappa B ligand (RANKL), a transmembrane or soluble protein essential for the formation, function, and survival of osteoclasts, which are the cells that are responsible for bone resorption. It is indicated to increase bone mass in men and postmenopausal women with osteoporosis at high risk for fracture. It is also indicated for men at high risk for fracture who are receiving androgen deprivation therapy for nonmetastatic prostate cancer. Additionally, it is indicated for glucocorticoid-induced osteoporosis in men and women at high risk of fracture. The general dosage is 60 mg every 6 months as an SC injection in the upper arm, upper thigh, or abdomen. Patients should be instructed to take 1000 mg of calcium daily and at least 400 IU of vitamin D daily.

Romosozumab (Evenity)

Clinical Context:  Monoclonal antibody (IgG2) that binds sclerostin, a regulatory factor in bone metabolism. Sclerostin inhibition increases bone formation and, to a lesser extent, decreases bone resorption. It is indicated for osteoporosis treatment in postmenopausal women at high risk for fracture, defined as a history of osteoporotic fracture or multiple risk factors for fracture. It is also indicated for patients who have failed or are intolerant to other available osteoporosis therapy.

Class Summary

Monoclonal antibodies such as denosumab (Prolia) inhibit osteoclast formation, decrease bone resorption, increase BMD, and reduce the risk of fracture. Another monoclonal antibody, romosozumab, binds sclerostin, a regulatory factor in bone metabolism.

Calcium citrate (Cal-Citrate, Cal-Cee, Cal-C-Caps)

Clinical Context:  Calcium is the primary component of skeletal tissue, providing structural integrity and support for individual growth. Bone undergoes constant remodeling and turnover. A combination of supplemental calcium and vitamin D can potentially lower the incidence of fractures. Calcium citrate is absorbed equally well when taken with or without food.

Calcium carbonate (Caltrate 600, Calcarb 600, Oysco 500, Super Calcium 600, Tums Ultra)

Clinical Context:  Calcium intake is essential in the prevention and treatment of osteoporosis. Calcium carbonate is generally more inexpensive and requires fewer tablets. Because of its dependence on stomach acid for absorption, calcium carbonate is absorbed most efficiently when taken with food.

Class Summary

Calcium and vitamin D are essential to increase bone density. Vitamin D repletion is essential for calcium absorption. Calcium supplements are used to increase calcium levels.[231] Adequate calcium intake is essential to attain peak bone mass and for continued maintenance of bone health.

Conjugated estrogens (Premarin)

Clinical Context:  Conjugated estrogens (Premarin)

Estrogens can directly affect bone mass through estrogen receptors in bone, reducing bone turnover and bone loss. Estrogens can also indirectly increase intestinal calcium absorption and renal calcium conservation and, therefore, improve calcium balance. When prescribing solely for the prevention of postmenopausal osteoporosis, therapy should be considered only for women at significant risk of osteoporosis and for whom nonestrogen medications need to be carefully considered.

Estradiol (Estrace, Estraderm, Menostar, Vivelle-Dot, Climara, Estraderm, Alora)

Clinical Context:  Estradiol restores estrogen levels to concentrations that induce negative feedback at gonadotropic regulatory centers; this, in turn, reduces the release of gonadotropins from the pituitary. Estradiol increases the synthesis of DNA, RNA, and many proteins in target tissues; it also inhibits osteoclastic activity and delays bone loss. In addition, evidence suggests a reduced incidence of fractures.

Estropipate

Clinical Context:  Estropipate is indicated for the prevention of osteoporosis. The results of a double-blind, placebo-controlled 2-year study have shown that treatment with 1 tablet of estropipate, 0.75 mg daily for 25 days (of a 31-day cycle per month), prevents vertebral bone mass loss in postmenopausal women. When estrogen therapy is discontinued, bone mass declines at a rate comparable to that of the immediate postmenopausal period. There is no evidence that estrogen replacement therapy restores bone mass to premenopausal levels.

Class Summary

Estrogen derivatives are approved for the prevention of osteoporosis and relief of menopause-associated vasomotor symptoms and vulvovaginal atrophy. They are used to increase the serum estrogen level, which, in turn, decreases the rate of bone resorption.[232] The lowest effective dose at the shortest duration necessary should be used. Estrogen therapy reduces bone resorption and retards or halts postmenopausal bone loss. Estrogen therapy is no longer a first-line approach for the treatment of osteoporosis in postmenopausal women because of increased risk of breast cancer, stroke, venous thromboembolism, and coronary disease. The FDA recommends that other approved nonestrogen treatments be considered first for osteoporosis prevention.

Vitamin D (Calciferol, Drisdol)

Clinical Context:  Vitamin D refers to both ergocalciferol (D2) and cholecalciferol (D3). Following exposure to UV light, 7-dehydrocholesterol (provitamin D3) is converted to cholecalciferol, which is then converted by the liver to calcifediol and then again by the kidney to calcitriol. Vitamin D is available in various forms, including tablets, oral liquid, and capsules. It is commonly coadministered with calcium supplements in patients with osteoporosis.

Class Summary

Vitamin D is a fat-soluble sterol compound that includes ergocalciferol (vitamin D2) and cholecalciferol (vitamin D3). It can be obtained from food and produced by skin when exposed to sunlight of sufficient intensity. When activated in the liver and then the kidney, vitamin D promotes calcium absorption and bone mass. Vitamin D replacement also increases muscle strength and lowers the risk of falling.

Norethindrone/ethinylestradiol (Femhrt)

Clinical Context:  Ethinyl estradiol with norethindrone is used to prevent osteoporosis associated with menopause. When prescribing it solely for the prevention of postmenopausal osteoporosis, therapy should be considered only for women at significant risk of osteoporosis.

Estradiol/norethindrone acetate (Activella, Mimvey, CombiPatch)

Clinical Context:  Ethinyl estradiol with norethindrone is used to prevent osteoporosis associated with menopause. When prescribing it solely for the prevention of postmenopausal osteoporosis, therapy should be considered only for women at significant risk of osteoporosis.

Class Summary

Estrogen derivatives are approved for the prevention of osteoporosis and relief of menopause-associated vasomotor symptoms and vulvovaginal atrophy. They are used to increase the serum estrogen level, which, in turn, decreases the rate of bone resorption.[232] The lowest effective dose at the shortest duration necessary should be used. Estrogen therapy reduces bone resorption and retards or halts postmenopausal bone loss. Estrogen therapy is no longer a first-line approach for the treatment of osteoporosis in postmenopausal women because of increased risk of breast cancer, stroke, venous thromboembolism, and coronary disease. The FDA recommends that other approved nonestrogen treatments be considered first for osteoporosis prevention.

Conjugated estrogens/medroxyprogesterone acetate (Prempro, Premphase)

Clinical Context:  The combination of conjugated estrogens and medroxyprogesterone reduces bone resorption and retards or halts postmenopausal bone loss.

Estradiol/levonorgestrel (Climara Pro)

Clinical Context:  Estradiol/levonorgestrel transdermal system releases both estradiol and levonorgestrel continuously upon application to skin. It is approved for the prevention of postmenopausal osteoporosis.

Estradiol/norgestimate (Prefest)

Clinical Context:  Estradiol/norgestimate is approved for the prevention of postmenopausal osteoporosis. It is available as a combination of estradiol 1 mg/norgestimate 0.09 mg.

Class Summary

The combination of conjugated estrogens and medroxyprogesterone reduces bone resorption and retards or halts postmenopausal bone loss.

What is osteoporosis?What are the signs and symptoms of vertebral fractures due to osteoporosis?What are the signs and symptoms of hip fracture due to osteoporosis?What are the physical findings of vertebral compression fractures due to osteoporosis?What are physical findings of hip fractures in patients with osteoporosis?What are signs of Colles fractures in patients with osteoporosis?What are the signs of pubic and sacral fractures in patients with osteoporosis?Which lab studies are included in the workup of osteoporosis?When is bone mineral density (BMD) measurement indicated for screening and diagnosis of osteoporosis?What is a dual-energy x-ray absorptiometry (DXA) T-score and how is it defined?What is the role of dual-energy x-ray absorptiometry (DXA) Z-scores in the diagnosis of osteoporosis?What are the benefits of performing quantitative calcaneal ultrasonography during the workup of osteoporosis?What are the NOF recommendations for vertebral imaging for osteoporosis?What is the role of radiography in the diagnosis of osteoporosis?Which lifestyle modifications prevent osteoporotic fractures?What are the NOF recommendations for pharmacologic therapy in the treatment of osteoporosis?What are the AACE recommendations for drug selection in the treatment of osteoporosis?What are the American College of Rheumatology (ACR) guidelines for the treatment of glucocorticoid-induced osteoporosis?How are the underlying causes of osteoporosis treated?How does the WHO define osteoporosis?How common is osteoporosis and who is at highest risk?What causes osteoporosis?What is the effect of osteoporosis on quality of life?Why is osteoporosis often overlooked and undertreated?What are the treatment options for osteoporosis?What steps can be taken to lessen the impact of osteoporosis?What is bone mineral density (BMD)?What is known regarding the pathophysiology of osteoporosis?How does bone remodeling and formation normally occur?What is the difference between dense cortical bone and spongy trabecular or cancellous bone?What is the role of osteoclasts in the pathogenesis of osteoporosis?Why does rapid bone remodeling result in an increased risk of osteoporosis and fracture?What is the role of receptor activator of nuclear factor-kappa B ligand (RANKL) and receptor activator of nuclear factor-kappa B (RANK) in the pathogenesis of osteoporosis?Which factors affect bone mass and bone strength?What is the pathogenesis of osteoporosis?What is the role of estrogen deficiency in the pathogenesis of osteoporosis?What is the role of aging in age the pathogenesis of osteoporosis?What is the role of calcium and vitamin D deficiency in the pathogenesis of osteoporosis?How do osteoporotic fractures occur?How does osteoporosis affect vertebral bodies?What causes trabecular fracture in patients with osteoporosis?What is the difference between osteoporosis and osteomalacia?What is the role of Wnt signaling in the pathogenesis of osteoporosis?Which endocrinologic conditions or medications may cause osteoporosis?What are lesser known factors in the pathogenesis of osteoporosis?Which prenatal and postnatal factors increase the risk for osteoporosis in older adults?How is osteoporosis classified?What are the types of primary osteoporosis?What causes secondary osteoporosis?What are the risk factors for osteoporosis?What is the mnemonic for osteoporotic risk factors?What is the prevalence of osteoporosis in the US?What is the global prevalence of osteoporosis?How does the incidence of osteoporosis differ among age groups?How does the prevalence of osteoporosis differ between males and females?How does the incidence of osteoporosis differ among racial groups?What is the prognosis for osteoporosis?What is the prognosis for osteoporosis-related fractures?What is the prognosis for vertebral compression fractures caused by osteoporosis?What is the prognosis for hip fractures caused by osteoporosis?Which factor increases the risk for multiple fractures in patients with osteoporosis?What is the WHO fracture-risk algorithm (FRAX)?How is the WHO fracture-risk algorithm (FRAX) used in the treatment of osteoporosis?What is the effect of type 2 diabetes mellitus on the prognosis for osteoporosis?What is the sensitivity of the WHO fracture-risk algorithm (FRAX) for perimenopausal and early-menopausal women?What are the possible complications of osteoporosis?How should patients be educated about osteoporosis?Screening for osteoporosis is indicated in patients with a history of which disorders?What are the modifiable and nonmodifiable risk factors for osteoporosis identified by the NOF?Which risk factors for osteoporosis and osteoporotic fractures should be identified in the patient history?What is the effect of risk prediction instruments on patient outcomes for osteoporosis?What is the Fracture Risk Assessment (FRAX) tool for osteoporosis?Which individuals are at highest risk for osteoporotic fractures?Which history suggests osteoporotic fractures?Which history suggests a sustained vertebral compression fracture due to osteoporosis?How is pain characterized in vertebral fractures caused by osteoporosis?What are the signs and symptoms of hip fracture in a patient with osteoporosis?Which GI disorders may be reported in the history of patients with osteoporosis report?Which physical exams are indicated in suspected osteoporosis?Which physical findings should raise concern in patients with suspected osteoporosis?What is the Timed Up and Go (TUG) test?Which physical findings suggest vertebral compression fractures caused by osteoporosis?Which are physical findings suggestive of acute vertebral fractures caused by osteoporosis?Which physical findings suggest hip fractures caused by osteoporosis?Which physical findings suggest pubic and sacral fractures caused by osteoporosis?Which physical findings suggest osteoporotic fractures?Which physical findings are signs of collagen defects in osteoporosis?Which findings of balance difficulties suggest osteoporosis?What is the most common and serious complication of osteoporosis?What is the USPSTF guideline for routine screening of men for osteoporosis?What should be considered when weighing the risk versus benefit of screening men for osteoporosis?What are the ACP recommendations for screening men for osteoporosis?What role does reduced bone density have in the differential diagnosis of osteoporosis?Which disorders should be considered in the differential diagnoses of an atraumatic osteoporotic compression fracture?How is osteoporosis differentiated from osteomalacia?What does the presence of a fracture indicate in patients with osteoporosis?Which conditions should be included in the differential diagnoses of osteoporosis?What are the differential diagnoses for Osteoporosis?Which tests should be performed in the workup of osteoporosis?What is the role of radiography in the workup of osteoporosis?How should bone mineral density (BMD) be measured in the workup of osteoporosis?What are the ISCD recommendations for diagnosing osteoporosis in children and adolescents?What is the role of lab studies in the workup of osteoporosis?What is the role of biochemical markers of bone turnover in the workup of osteoporosis?Which serum markers of bone formation are used in the diagnosis of osteoporosis?Which urinary markers of bone resorption are used in the diagnosis of osteoporosis?Which serum markers of bone resorption are used in the diagnosis of osteoporosis?What do high elevations of bone-specific alkaline phosphatase (BSAP) suggest in the workup of osteoporosis?What is the role of biochemical markers in the treatment of osteoporosis?When is plain radiography indicated in the workup of osteoporosis?When is lateral spine radiography indicated in the workup of osteoporosis?Which radiographic findings suggest osteoporosis?What is the accuracy of plain radiography for the diagnosis of osteoporosis?Which test results best indicate the risk of osteoporotic fractures?What is the criterion standard for the evaluation of bone mineral density (BMD) in the workup of osteoporosis?What is the role of dual-energy x-ray absorptiometry (DXA) in the workup of osteoporosis?Which factors may result in a falsely high bone mineral density (BMD) in the workup of osteoporosis?How are bone density data from dual-energy x-ray absorptiometry (DXA) reported in the workup of osteoporosis?What are the WHO criteria for diagnosing osteoporosis?How much is the risk of fracture increased for each standard deviation (SD) reduction in bone mineral density (BMD) in patients with osteoporosis?What are the ISCD guidelines for use of peripheral dual-energy x-ray absorptiometry (pDXA) in the workup of osteoporosis?Which techniques for measurement of bone strength are used in the workup of osteoporosis?What is the role of hip structural analysis (HSA) in the workup of osteoporosis?What is the role of trabecular bone scores (TBS) in the workup of osteoporosis?What is the role of finite element analysis (FEA) in the workup of osteoporosis?What is the role of quantitative CT (QCT) in the workup of osteoporosis?When is quantitative CT (QCT) scanning indicated in the workup of osteoporosis?How does dual-energy x-ray absorptiometry (DXA) scanning compare to quantitative CT (QCT) in the diagnosis of osteoporosis?What are the ISCD recommendations for quantitative CT (QCT) in the workup of osteoporosis?What are the ISCD guidelines on the use of peripheral quantitative CT (pQCT) in the workup of osteoporosis?What is the role of SPECT scanning in the workup of osteoporosis?What is the role of quantitative ultrasonography (QUS) in the workup of osteoporosis?What are the ISCD recommendations on quantitative ultrasonography (QUS) in the workup of osteoporosis?What is the role of MRI in the workup of osteoporosis?What is the role of bone scanning in the workup of osteoporosis?What is the role of bone biopsy in the workup of osteoporosis?What is the role of tetracycline double labeling in the workup of osteoporosis?When is vertebral body bone biopsy performed in the workup of osteoporosis?What are the histologic findings of osteoporotic bone?What are the ACP guidelines for prevention of fractures in patients with osteoporosis?Which lifestyle modifications may prevent osteoporosis?What is the surgical treatment of osteoporosis?Which measures may lead to a reduction in fractures and an increase in osteoporosis therapy?What are the goals of rehabilitation in osteoporotic fracture?How can progression of osteoporosis be halted?What are preventive measures for patients at risk for osteoporosis?What protective measures should be taken for patients taking glucocorticoids who are at risk for osteoporosis?What are the most commonly used agents for treatment and prevention of osteoporosis?What is the role of alendronate (Fosamax) in the treatment of osteoporosis?What is the dosage regimen for alendronate (Fosamax) in the treatment of osteoporosis?Which bisphosphonates are used for the treatment of osteoporosis?What is the role of zoledronic acid (Reclast) in the treatment of osteoporosis?What are the possible risks of zoledronic acid (Reclast) for the treatment of osteoporosis?What warnings have been issued by the FDA regarding the use of zoledronic acid (Reclast) for the treatment of osteoporosis?How does bisphosphonate therapy affect bone turnover in patients with osteoporosis?What is the optimal length of treatment with bisphosphonates for osteoporosis?Which risk factors should be considered in determining the duration of bisphosphonate therapy for osteoporosis?What are the recommendations for pharmacologic therapy if osteoporosis is mild or if fracture risk is high?What are the AACE recommendations for long-term bisphosphonate therapy for osteoporosis?What is the role of selective estrogen receptor modulators (SERMs) in the treatment of osteoporosis?What is the role of raloxifene (Evista) in the treatment of osteoporosis?Which combination therapy has been approved for prevention of osteoporosis?What is the role of teriparatide (Forteo) in the treatment of osteoporosis?What are the contraindications for use of teriparatide (Forteo) in the treatment of osteoporosis?What are the possible complications of parathyroid hormone (PTH) for the treatment of osteoporosis?What is the role of parathyroid hormone (PTH) in the treatment of osteoporosis?Is parathyroid hormone (PTH) effective in the treatment of osteoporosis?Is combination therapy with parathyroid hormone (PTH) and bisphosphonate effective in the treatment of osteoporosis?When is teriparatide (Forteo) indicated in the treatment of osteoporosis?What is the role of abaloparatide (Tymlos) in the treatment of osteoporosis?What is the role of calcitonin-salmon (Fortical, Miacalcin) in the treatment of osteoporosis?What is the efficacy of calcitonin-salmon (Fortical, Miacalcin) in the treatment of osteoporosis?Does calcitonin-salmon (Fortical, Miacalcin) increase the risk of malignancy in patients with osteoporosis?When is calcitonin-salmon (Fortical, Miacalcin) indicated in the treatment of osteoporosis?What is the role of denosumab (Prolia) in the treatment of osteoporosis?Is denosumab (Prolia) effective in the treatment of osteoporosis?In which patients should denosumab (Prolia) be considered as first-line therapy for osteoporosis?Which combination therapy increases bone mineral density (BMD) in patients with osteoporosis?What is the role of hormone replacement therapy (HRT) in the treatment of osteoporosis?What is the role of romosozumab in the treatment of osteoporosis?What is the role of strontium ranelate in the treatment of osteoporosis?What are the goals of surgical treatment of osteoporotic fractures?When are vertebroplasty and balloon kyphoplasty indicated for the treatment of osteoporosis?Which dietary measures are important for the prevention of osteoporosis?Which dietary sources of calcium and vitamin D should be considered for the prevention of osteoporosis?What are the AACE recommendations for daily calcium intake for the prevention of osteoporosis?What are the Institute of Medicine (IOM) (Health and Medicine Division) recommendations for calcium and vitamin D daily intake in older adults for the prevention of osteoporosis?Which factors may interfere with nutrition and what is their effect on the risk for developing osteoporosis?Which calcium supplements are used for the prevention of osteoporosis?Which vitamin D supplements are used for the prevention of osteoporosis?What is the efficacy of calcium and vitamin D supplements in reducing the risk of fracture in patients with osteoporosis?What is the focus of physical therapy for osteoporosis?What is the focus of occupational therapy for osteoporosis?What exercises are recommended for patients with osteoporosis?What does exercise therapy for osteoporosis include?Are impact exercises an effective treatment of osteoporosis?What is the efficacy of swimming for osteoporosis?What is the role of balance training in the treatment of osteoporosis?How is osteoporosis prevented?What are the NOF recommendations for lifestyle modifications to reduce the risk of developing osteoporosis?Which pharmacologic agents are used in the prevention of osteoporosis?What is the role of estrogen-progestin therapy in the treatment of osteoporosis?What monitoring should be performed for prevention of osteoporosis?Which specialist consultations are needed for a patient with osteoporosis?What should be considered in consultations for osteoporosis?What are USPSTF recommendations for repeated screening of osteoporosis?What is the role of osteopenia in the development of osteoporosis in older, postmenopausal women during rescreening intervals?What is the role of orthotics in the treatment of osteoporosis?What are the guidelines for screening patients for osteoporosis with bone mineral density (BMD) measurement?What are the NOF recommendations for pharmacologic therapy of osteoporosis?What are the ACP recommendations for pharmacologic treatment of osteoporosis?According to ACP guidelines, which agents are contraindicated for the treatment of osteoporosis?What are the AACE recommendations on choosing medications to treat osteoporosis?What are the AACE recommendations for combination therapy for osteoporosis?What are the NOGG recommendations for the diagnosis and management of osteoporosis?Which agents are used to treat osteoporosis?Which medications in the drug class Estrogens/Progestins-HRT are used in the treatment of Osteoporosis?Which medications in the drug class Estrogens/Progestins are used in the treatment of Osteoporosis?Which medications in the drug class Vitamins, Fat-Soluble are used in the treatment of Osteoporosis?Which medications in the drug class Estrogen Derivatives are used in the treatment of Osteoporosis?Which medications in the drug class Calcium Salts are used in the treatment of Osteoporosis?Which medications in the drug class Monoclonal Antibodies, Endocrine are used in the treatment of Osteoporosis?Which medications in the drug class Selective Estrogen Receptor Modulator are used in the treatment of Osteoporosis?Which medications in the drug class Parathyroid Hormone Analogs are used in the treatment of Osteoporosis?Which medications in the drug class Calcium Metabolism Modifiers are used in the treatment of Osteoporosis?

Author

Monique Bethel, MD, Resident Physician, Department of Internal Medicine, Georgia Regents University

Disclosure: Nothing to disclose.

Coauthor(s)

Kristine M Lohr, MD, MS, Professor, Department of Internal Medicine, Interim Chief, Division of Rheumatology, Director, Rheumatology Training Program, University of Kentucky College of Medicine

Disclosure: Nothing to disclose.

Laura D Carbone, MD, MS, FACP, Professor, Department of Internal Medicine, Section Chief of Rheumatology, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center

Disclosure: Nothing to disclose.

Wambui Machua, MD, Fellow, Department of Internal Medicine, Division of Rheumatology, Georgia Regents University

Disclosure: Nothing to disclose.

Chief Editor

Herbert S Diamond, MD, Visiting Professor of Medicine, Division of Rheumatology, State University of New York Downstate Medical Center; Chairman Emeritus, Department of Internal Medicine, Western Pennsylvania Hospital

Disclosure: Nothing to disclose.

Acknowledgements

Michael T Andary, MD, MS Professor, Residency Program Director, Department of Physical Medicine and Rehabilitation, Michigan State University College of Osteopathic Medicine

Michael T Andary, MD, MS is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation, American Association of Neuromuscular and Electrodiagnostic Medicine, American Medical Association, and Association of Academic Physiatrists

Disclosure: Allergan Honoraria Speaking and teaching; Pfizer Honoraria Speaking and teaching

Harris Gellman, MD Consulting Surgeon, Broward Hand Center; Voluntary Clinical Professor of Orthopedic Surgery and Plastic Surgery, Departments of Orthopedic Surgery and Surgery, University of Miami, Leonard M Miller School of Medicine

Harris Gellman, MD is a member of the following medical societies: American Academy of Medical Acupuncture, American Academy of Orthopaedic Surgeons, American Orthopaedic Association, American Society for Surgery of the Hand, and Arkansas Medical Society

Disclosure: Nothing to disclose.

Elliot Goldberg, MD Dean of the Western Pennsylvania Clinical Campus, Professor, Department of Medicine, Temple University School of Medicine

Elliot Goldberg, MD is a member of the following medical societies: Alpha Omega Alpha, American College of Physicians, and American College of Rheumatology

Disclosure: Nothing to disclose.

Coburn Hobar, MD Clinician in Rheumatology, Hobar Health and Wellness, and Anti-Aging & Wellness Center of Sarasota

Coburn Hobar, MD is a member of the following medical societies: American Academy of Anti-Aging Medicine and American College of Rheumatology

Disclosure: Nothing to disclose.

Dana Jacobs-Kosmin, MD, FACP Attending Physician, Department of Medicine, Division of Rheumatology, Einstein Medical Center; Clinical Assistant Professor of Medicine, Jefferson Medical College of Thomas Jefferson University

Dana Jacobs-Kosmin, MD, FACP is a member of the following medical societies: American College of Physicians, American College of Rheumatology, and American Medical Association

Disclosure: Nothing to disclose.

Robert J Kaplan, MD James E Van Zandt VA Medical Center, Staff Physician, Department of Rehabilitation Medicine

Robert J Kaplan, MD is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation

Disclosure: Nothing to disclose.

Joseph M Lane, MD Professor of Orthopedic Surgery, Weill Medical College of Cornell University; Chief, Metabolic Bone Disease Service, Hospital for Special Surgery

Joseph M Lane, MD is a member of the following medical societies: American Academy of Orthopaedic Surgeons, American Association of University Professors, American Federation for Aging Research, American Orthopaedic Association, American Society for Bone and Mineral Research, Association of Bone and Joint Surgeons, Medical Society of the State of New York, Musculoskeletal Tumor Society, National Osteoporosis Foundation, North American Spine Society, and Orthopaedic Research Society

Disclosure: Lilly; Aventis; Novartis; Warner Chilcott; Biomimetics; Zimmer; DFine; Innovative Solutions; Honoraria Speaking and teaching; Graftys; Bone Technologies SA; CollPlant Consulting fee Consulting

David Lenrow, MD Vice Chair of Clinical Services, Medical Director, Erdman Clinic; Associate Professor, Department of Rehabilitation Medicine, University of Pennsylvania at Philadelphia

David Lenrow, MD is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation and American Medical Association

Disclosure: Nothing to disclose.

Julie Lin, MD Assistant Professor, Department of Rehabilitation Medicine, Weill Medical College of Cornell University; Assistant Attending Physiatrist, Physiatry Department, Hospital for Special Surgery

Julie Lin, MD is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation, American Association of Neuromuscular and Electrodiagnostic Medicine, American Medical Association, North American Spine Society, and Physiatric Association of Spine, Sports and Occupational Rehabilitation

Disclosure: Nothing to disclose.

Elizabeth A Moberg-Wolff, MD Associate Professor, Department of Physical Medicine and Rehabilitation, Children's Hospital of Wisconsin, Medical College of Wisconsin

Elizabeth A Moberg-Wolff, MD is a member of the following medical societies: American Academy for Cerebral Palsy and Developmental Medicine and American Academy of Physical Medicine and Rehabilitation

Disclosure: Medtronic Neurological Grant/research funds Speaking and teaching

Srinivas R Nalamachu, MD Clinical Assistant Professor, Department of Internal Medicine, Kansas City University of Medicine and Biosciences; President and Medical Director, Internation Clinical Research Institute, Inc; Medical Director, Pain Management Institute

Srinivas R Nalamachu, MD is a member of the following medical societies: International Association for the Study of Pain

Disclosure: Nothing to disclose.

Richard Salcido, MD Chairman, Erdman Professor of Rehabilitation, Department of Physical Medicine and Rehabilitation, University of Pennsylvania School of Medicine

Richard Salcido, MD is a member of the following medical societies: American Academy of Pain Medicine, American Academy of Physical Medicine and Rehabilitation, American College of Physician Executives, American Medical Association, and American Paraplegia Society

Disclosure: Nothing to disclose.

Miguel A Schmitz, MD Consulting Surgeon, Department of Orthopedics, Klamath Orthopedic and Sports Medicine Clinic

Miguel A Schmitz, MD is a member of the following medical societies: American Academy of Orthopaedic Surgeons, American Orthopaedic Society for Sports Medicine, Arthroscopy Association of North America, and North American Spine Society

Disclosure: Nothing to disclose.

Alana C Serota, MD Fellow in Metabolic Bone Disease and Osteoporosis, Department of Orthopedics, Hospital for Special Surgery

Alana C Serota, MD is a member of the following medical societies: American Academy of Family Physicians

Disclosure: Nothing to disclose.

Sucharitha Shanmugam, MD Consulting Physician, PMA Medical Specialists, Limerick, PA

Sucharitha Shanmugam, MD is a member of the following medical societies: American College of Rheumatology

Disclosure: Nothing to disclose.

Curtis W Slipman, MD Director, University of Pennsylvania Spine Center; Associate Professor, Department of Physical Medicine and Rehabilitation, University of Pennsylvania Medical Center

Curtis W Slipman, MD is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation, Association of Academic Physiatrists, International Association for the Study of Pain, and North American Spine Society

Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Medscape Salary Employment

Shireesha Vuppalanchi, MD Consulting Staff, Methodist Hospital, Indianapolis; Hospitalist, Respiratory and Critical Care Consultants, PC

Disclosure: Nothing to disclose.

William S Whyte II, MD Director of Interventional Spine and Pain Management, Louisiana Pain Physicians

William S Whyte II, MD is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation, American Medical Association, Association of Academic Physiatrists, North American Spine Society, Physiatric Association of Spine, Sports and Occupational Rehabilitation, and Southern Medical Association

Disclosure: Nothing to disclose.

Jerome D Wiedel, MD Chair, Professor, Department of Orthopedics, University of Colorado Health Sciences Center

Disclosure: Nothing to disclose.

Authors' Disclaimer

This work does not reflect the views of the Veterans Health Administration or the United States government.

References

  1. Lynn SG, Sinaki M, Westerlind KC. Balance characteristics of persons with osteoporosis. Arch Phys Med Rehabil. 1997 Mar. 78(3):273-7. [View Abstract]
  2. [Guideline] Cosman F, de Beur SJ, LeBoff MS, Lewiecki EM, Tanner B, Randall S, et al. Clinician's Guide to Prevention and Treatment of Osteoporosis. Osteoporos Int. 2014 Oct. 25 (10):2359-81. [View Abstract]
  3. [Guideline] Schousboe JT, Shepherd JA, Bilezikian JP, Baim S. Executive summary of the 2013 International Society for Clinical Densitometry Position Development Conference on bone densitometry. J Clin Densitom. 2013 Oct-Dec. 16(4):455-66. [View Abstract]
  4. Gosfield E 3rd, Bonner FJ Jr. Evaluating bone mineral density in osteoporosis. Am J Phys Med Rehabil. 2000 May-Jun. 79(3):283-91. [View Abstract]
  5. Kanis JA, McCloskey EV, Johansson H, Oden A, Melton LJ 3rd, Khaltaev N. A reference standard for the description of osteoporosis. Bone. 2008 Mar. 42(3):467-75. [View Abstract]
  6. Silverman SL. Selecting patients for osteoporosis therapy. Ann N Y Acad Sci. 2007 Nov. 1117:264-72. [View Abstract]
  7. Czerwinski E, Badurski JE, Marcinowska-Suchowierska E, Osieleniec J. Current understanding of osteoporosis according to the position of the World Health Organization (WHO) and International Osteoporosis Foundation. Ortop Traumatol Rehabil. 2007 Jul-Aug. 9(4):337-56. [View Abstract]
  8. Kanis JA. Assessment of fracture risk and its application to screening for postmenopausal osteoporosis: synopsis of a WHO report. WHO Study Group. Osteoporos Int. 1994 Nov. 4(6):368-81. [View Abstract]
  9. Nayak S, Roberts MS, Greenspan SL. Cost-effectiveness of different screening strategies for osteoporosis in postmenopausal women. Ann Intern Med. 2011 Dec 6. 155(11):751-61. [View Abstract]
  10. Sandhu SK, Hampson G. The pathogenesis, diagnosis, investigation and management of osteoporosis. J Clin Pathol. 2011 Dec. 64(12):1042-50. [View Abstract]
  11. [Guideline] 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 Sep 2. 22 (Suppl 4):1-42. [View Abstract]
  12. [Guideline] Buckley L, Guyatt G, Fink HA, Cannon M, Grossman J, Hansen KE, et al. 2017 American College of Rheumatology Guideline for the Prevention and Treatment of Glucocorticoid-Induced Osteoporosis. Arthritis Care Res (Hoboken). 2017 Aug. 69 (8):1095-1110. [View Abstract]
  13. Ahmed SF, Elmantaser M. Secondary osteoporosis. Endocr Dev. 2009. 16:170-90. [View Abstract]
  14. Majumdar SR, Lier DA, Beaupre LA, Hanley DA, Maksymowych WP, Juby AG, et al. Osteoporosis case manager for patients with hip fractures: results of a cost-effectiveness analysis conducted alongside a randomized trial. Arch Intern Med. 2009 Jan 12. 169(1):25-31. [View Abstract]
  15. Maybury K. Strengthening Communication on Bone Health. Gallup. Available at http://www.gallup.com/poll/5851/Strengthening-Communication-Bone-Health.aspx?g_source=osteoporosis&g_medium=search&g_campaign=tiles. April 23, 2002; Accessed: June 15, 2019.
  16. World Health Organization. WHO scientific group on the assessment of osteoporosis at primary health care level: summary meeting report. Available at http://www.who.int/chp/topics/Osteoporosis.pdf. 2007; Accessed: June 15, 2019.
  17. Bono CM, Einhorn TA. Overview of osteoporosis: pathophysiology and determinants of bone strength. Eur Spine J. 2003 Oct. 12 Suppl 2:S90-6. [View Abstract]
  18. Raisz LG. Pathogenesis of osteoporosis: concepts, conflicts, and prospects. J Clin Invest. 2005 Dec. 115(12):3318-25. [View Abstract]
  19. Seeman E, Delmas PD. Bone quality--the material and structural basis of bone strength and fragility. N Engl J Med. 2006 May 25. 354(21):2250-61. [View Abstract]
  20. Mora S, Gilsanz V. Establishment of peak bone mass. Endocrinol Metab Clin North Am. 2003 Mar. 32(1):39-63. [View Abstract]
  21. Jilka RL, Hangoc G, Girasole G, Passeri G, Williams DC, Abrams JS, et al. Increased osteoclast development after estrogen loss: mediation by interleukin-6. Science. 1992 Jul 3. 257(5066):88-91. [View Abstract]
  22. Cummings SR, Melton LJ. Epidemiology and outcomes of osteoporotic fractures. Lancet. 2002 May 18. 359(9319):1761-7. [View Abstract]
  23. Rosen CJ, Tenenhouse A. Biochemical markers of bone turnover. A look at laboratory tests that reflect bone status. Postgrad Med. Oct 1998. 104(4):101-2, 107-10.
  24. Nusse R. Wnt signaling in disease and in development. Cell Res. 2005 Jan. 15(1):28-32. [View Abstract]
  25. Ling L, Nurcombe V, Cool SM. Wnt signaling controls the fate of mesenchymal stem cells. Gene. 2009 Mar 15. 433(1-2):1-7. [View Abstract]
  26. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999 Apr 2. 284(5411):143-7. [View Abstract]
  27. Rao TP, Kühl M. An updated overview on Wnt signaling pathways: a prelude for more. Circ Res. 2010 Jun 25. 106(12):1798-806. [View Abstract]
  28. Clevers H, Nusse R. Wnt/ß-catenin signaling and disease. Cell. 2012 Jun 8. 149(6):1192-205. [View Abstract]
  29. Krishnan V, Bryant HU, Macdougald OA. Regulation of bone mass by Wnt signaling. J Clin Invest. 2006 May. 116(5):1202-9. [View Abstract]
  30. Boyden LM, Mao J, Belsky J, Mitzner L, Farhi A, Mitnick MA, et al. High bone density due to a mutation in LDL-receptor-related protein 5. N Engl J Med. 2002 May 16. 346(20):1513-21. [View Abstract]
  31. Van Wesenbeeck L, Cleiren E, Gram J, Beals RK, Bénichou O, Scopelliti D, et al. Six novel missense mutations in the LDL receptor-related protein 5 (LRP5) gene in different conditions with an increased bone density. Am J Hum Genet. 2003 Mar. 72(3):763-71. [View Abstract]
  32. Levasseur R, Lacombe D, de Vernejoul MC. LRP5 mutations in osteoporosis-pseudoglioma syndrome and high-bone-mass disorders. Joint Bone Spine. 2005 May. 72(3):207-14. [View Abstract]
  33. Zhu Y, Sun Z, Han Q, Liao L, Wang J, Bian C, et al. Human mesenchymal stem cells inhibit cancer cell proliferation by secreting DKK-1. Leukemia. 2009 May. 23(5):925-33. [View Abstract]
  34. Kawano Y, Kypta R. Secreted antagonists of the Wnt signalling pathway. J Cell Sci. 2003 Jul 1. 116:2627-34. [View Abstract]
  35. Kaiser M, Mieth M, Liebisch P, Oberländer R, Rademacher J, Jakob C, et al. Serum concentrations of DKK-1 correlate with the extent of bone disease in patients with multiple myeloma. Eur J Haematol. 2008 Jun. 80(6):490-4. [View Abstract]
  36. Tai N, Inoue D. [Anti-Dickkopf1 (Dkk1) antibody as a bone anabolic agent for the treatment of osteoporosis]. Clin Calcium. 2014 Jan. 24 (1):75-83. [View Abstract]
  37. Bonewald LF. The amazing osteocyte. J Bone Miner Res. 2011 Feb. 26(2):229-38. [View Abstract]
  38. Li X, Zhang Y, Kang H, Liu W, Liu P, Zhang J, et al. Sclerostin binds to LRP5/6 and antagonizes canonical Wnt signaling. J Biol Chem. 2005 May 20. 280(20):19883-7. [View Abstract]
  39. FDA approves new treatment for osteoporosis in postmenopausal women at high risk of fracture. U.S. Food & Drug Administration. Available at https://www.fda.gov/news-events/press-announcements/fda-approves-new-treatment-osteoporosis-postmenopausal-women-high-risk-fracture. April 09, 2019; Accessed: June 20, 2019.
  40. Ringe JD, Farahmand P. Advances in the management of corticosteroid-induced osteoporosis with bisphosphonates. Clin Rheumatol. 2007 Apr. 26(4):474-84. [View Abstract]
  41. Dennison EM, Syddall HE, Sayer AA, Gilbody HJ, Cooper C. Birth weight and weight at 1 year are independent determinants of bone mass in the seventh decade: the Hertfordshire cohort study. Pediatr Res. 2005 Apr. 57(4):582-6. [View Abstract]
  42. Fall C, Hindmarsh P, Dennison E, Kellingray S, Barker D, Cooper C. Programming of growth hormone secretion and bone mineral density in elderly men: a hypothesis. J Clin Endocrinol Metab. 1998 Jan. 83(1):135-9. [View Abstract]
  43. [Guideline] American Association of Clinical Endocrinologists medical guidelines for clinical practice for the prevention and treatment of postmenopausal osteoporosis: 2001 edition, with selected updates for 2003. Endocr Pract. 2003 Nov-Dec. 9(6):544-64. [View Abstract]
  44. Kelman A, Lane NE. The management of secondary osteoporosis. Best Pract Res Clin Rheumatol. 2005 Dec. 19(6):1021-37. [View Abstract]
  45. Adams JS, Song CF, Kantorovich V. Rapid recovery of bone mass in hypercalciuric, osteoporotic men treated with hydrochlorothiazide. Ann Intern Med. 1999 Apr 20. 130(8):658-60. [View Abstract]
  46. Mann GB, Kang YC, Brand C, Ebeling PR, Miller JA. Secondary causes of low bone mass in patients with breast cancer: a need for greater vigilance. J Clin Oncol. 2009 Aug 1. 27(22):3605-10. [View Abstract]
  47. Holick MF. Vitamin D deficiency. N Engl J Med. 2007 Jul 19. 357(3):266-81. [View Abstract]
  48. di Munno O, Mazzantini M, Sinigaglia L, Bianchi G, Minisola G, Muratore M, et al. Effect of low dose methotrexate on bone density in women with rheumatoid arthritis: results from a multicenter cross-sectional study. J Rheumatol. 2004 Jul. 31(7):1305-9. [View Abstract]
  49. Migliaccio S, Brama M, Malavolta N. Management of glucocorticoids-induced osteoporosis: role of teriparatide. Ther Clin Risk Manag. 2009 Apr. 5(2):305-10. [View Abstract]
  50. van Staa TP, Leufkens HG, Cooper C. The epidemiology of corticosteroid-induced osteoporosis: a meta-analysis. Osteoporos Int. 2002 Oct. 13(10):777-87. [View Abstract]
  51. Licata AA. Challenges of Estimating Fracture Risk with DXA: Changing Concepts About Bone Strength and Bone Density. Aerosp Med Hum Perform. 2015 Jul. 86 (7):628-32. [View Abstract]
  52. Assessment of fracture risk and its application to screening for postmenopausal osteoporosis. Report of a WHO Study Group. World Health Organ Tech Rep Ser. 1994. 843:1-129. [View Abstract]
  53. Lyles KW, Schenck AP, Colón-Emeric CS. Hip and other osteoporotic fractures increase the risk of subsequent fractures in nursing home residents. Osteoporos Int. 2008 Aug. 19(8):1225-33. [View Abstract]
  54. Fink HA, Kuskowski MA, Taylor BC, Schousboe JT, Orwoll ES, Ensrud KE. Association of Parkinson's disease with accelerated bone loss, fractures and mortality in older men: the Osteoporotic Fractures in Men (MrOS) study. Osteoporos Int. 2008 Sep. 19(9):1277-82. [View Abstract]
  55. Sinaki M. Exercise and osteoporosis. Arch Phys Med Rehabil. 1989 Mar. 70(3):220-9. [View Abstract]
  56. Yaturu S, DjeDjos S, Alferos G, Deprisco C. Bone mineral density changes on androgen deprivation therapy for prostate cancer and response to antiresorptive therapy. Prostate Cancer Prostatic Dis. 2006. 9(1):35-8. [View Abstract]
  57. Busko M. Chemical in Toothpaste Tied to Osteoporosis in Women. Medscape Medical News. Available at https://www.medscape.com/viewarticle/914888. June 26, 2019; Accessed: June 27, 2019.
  58. Hansen D, Bazell C, Pelizzari P, Pyenson B. Medicare cost of osteoporotic fractures: The clinical and cost burden of an important consequence of osteoporosis. Available at https://static1.squarespace.com/static/5c0860aff793924efe2230f3/t/5d76b949deb7e9086ee3d7dd/1568061771769/Medicare+Cost+of+Osteoporotic+Fractures+20190827.pdf. August 2019; Accessed: September 26, 2019.
  59. Cooper C, Campion G, Melton LJ 3rd. Hip fractures in the elderly: a world-wide projection. Osteoporos Int. 1992 Nov. 2(6):285-9. [View Abstract]
  60. Who are candidates for prevention and treatment for osteoporosis?. Osteoporos Int. 1997. 7(1):1-6. [View Abstract]
  61. Jensen GF, Christiansen C, Boesen J, Hegedüs V, Transbøl I. Epidemiology of postmenopausal spinal and long bone fractures. A unifying approach to postmenopausal osteoporosis. Clin Orthop Relat Res. 1982 Jun. 75-81. [View Abstract]
  62. Melton LJ 3rd, Kan SH, Frye MA, Wahner HW, O'Fallon WM, Riggs BL. Epidemiology of vertebral fractures in women. Am J Epidemiol. 1989 May. 129(5):1000-11. [View Abstract]
  63. Smith R, Wordsworth P. Osteoporosis. Clinical and Biochemical Disorders of the Skeleton. 2005. 123.
  64. Chon KS, Sartoris DJ, Brown SA, Clopton P. Alcoholism-associated spinal and femoral bone loss in abstinent male alcoholics, as measured by dual X-ray absorptiometry. Skeletal Radiol. 1992. 21(7):431-6. [View Abstract]
  65. Robbins J, Aragaki AK, Kooperberg C, Watts N, Wactawski-Wende J, Jackson RD, et al. Factors associated with 5-year risk of hip fracture in postmenopausal women. JAMA. 2007 Nov 28. 298(20):2389-98. [View Abstract]
  66. Bass MA, Sharma A, Nahar VK, Chelf S, Zeller B, Pham L, et al. Bone Mineral Density Among Men and Women Aged 35 to 50 Years. J Am Osteopath Assoc. 2019 Jun 1. 119 (6):357-363. [View Abstract]
  67. Woolf AD, Pfleger B. Burden of major musculoskeletal conditions. Bull World Health Organ. 2003. 81(9):646-56. [View Abstract]
  68. Balfour F. China’s "Demographic Tsunami." Bloomberg Businessweek. January 5, 2012. Available at http://www.businessweek.com/magazine/chinas-demographic-tsunami-01052012.html. Accessed: February 16, 2012.
  69. Morin SN, Lix LM, Majumdar SR, Leslie WD. Temporal trends in the incidence of osteoporotic fractures. Curr Osteoporos Rep. 2013 Dec. 11(4):263-9. [View Abstract]
  70. Melton LJ 3rd, Sampson JM, Morrey BF, Ilstrup DM. Epidemiologic features of pelvic fractures. Clin Orthop Relat Res. 1981 Mar-Apr. 43-7. [View Abstract]
  71. Cauley JA, Lui LY, Ensrud KE, Zmuda JM, Stone KL, Hochberg MC, et al. Bone mineral density and the risk of incident nonspinal fractures in black and white women. JAMA. 2005 May 4. 293(17):2102-8. [View Abstract]
  72. Bone Health and Osteoporosis: A Report of the Surgeon General. Washington, DC: Department of Health and Human Services; 2004.
  73. Burge R, Dawson-Hughes B, Solomon DH, et al. Incidence and economic burden of osteoporosis-related fractures in the United States, 2005-2025. J Bone Miner Res. 2007 Mar. 22(3):465-75. [View Abstract]
  74. Hackethal V. Osteoporosis Drugs May Help Oldest, Sickest Women Most. Medscape Medical News. June 18, 2019. Available at https://www.medscape.com/viewarticle/914573#vp_2
  75. Ensrud KE, Kats AM, Boyd CM, Diem SJ, Schousboe JT, Taylor BC, et al. Association of Disease Definition, Comorbidity Burden, and Prognosis With Hip Fracture Probability Among Late-Life Women. JAMA Intern Med. 2019 Jun 17. [View Abstract]
  76. Cooper C, Atkinson EJ, Jacobsen SJ, et al. Population-based study of survival after osteoporotic fractures. Am J Epidemiol. 1993 May 1. 137(9):1001-5. [View Abstract]
  77. Kado DM, Browner WS, Palermo L, Nevitt MC, Genant HK, Cummings SR. Vertebral fractures and mortality in older women: a prospective study. Study of Osteoporotic Fractures Research Group. Arch Intern Med. 1999 Jun 14. 159(11):1215-20. [View Abstract]
  78. Vestergaard P, Rejnmark L, Mosekilde L. Increased mortality in patients with a hip fracture-effect of pre-morbid conditions and post-fracture complications. Osteoporos Int. 2007 Dec. 18(12):1583-93. [View Abstract]
  79. Trombetti A, Herrmann F, Hoffmeyer P, Schurch MA, Bonjour JP, Rizzoli R. Survival and potential years of life lost after hip fracture in men and age-matched women. Osteoporos Int. 2002 Sep. 13(9):731-7. [View Abstract]
  80. Michel JP, Hoffmeyer P, Klopfenstein C, et al. Prognosis of functional recovery 1 year after hip fracture: typical patient profiles through cluster analysis. J Gerontol A Biol Sci Med Sci. 2000 Sep. 55(9):M508-15. [View Abstract]
  81. Lindsay R, Silverman SL, Cooper C, et al. Risk of new vertebral fracture in the year following a fracture. JAMA. 2001 Jan 17. 285(3):320-3. [View Abstract]
  82. Klotzbuecher CM, Ross PD, Landsman PB, et al. Patients with prior fractures have an increased risk of future fractures: a summary of the literature and statistical synthesis. J Bone Miner Res. 2000 Apr. 15(4):721-39. [View Abstract]
  83. Morin SN, Lix LM, Leslie WD. The importance of previous fracture site on osteoporosis diagnosis and incident fractures in women. J Bone Miner Res. 2014 Jul. 29(7):1675-80. [View Abstract]
  84. The World Health Organization Fracture Risk Assessment Tool. Available at http://www.shef.ac.uk/FRAX/. Accessed: May 5, 2008.
  85. Leslie WD, Morin S, Lix LM. Before-and-After Study of Fracture Risk Reporting and Osteoporosis Treatment Initiation. Ann Intern Med. Nov 2 2010. 153(9):580-6.
  86. Schwartz AV, Vittinghoff E, Bauer DC, et al. Association of BMD and FRAX score with risk of fracture in older adults with type 2 diabetes. JAMA. 2011 Jun 1. 305(21):2184-92. [View Abstract]
  87. Tremollieres FA, Pouilles JM, Drewniak N, Laparra J, Ribot CA, Dargent-Molina P. Fracture risk prediction using BMD and clinical risk factors in early postmenopausal women: sensitivity of the WHO FRAX tool. J Bone Miner Res. 2010 May. 25(5):1002-9. [View Abstract]
  88. Grisso JA, Kelsey JL, Strom BL, Chiu GY, Maislin G, O'Brien LA, et al. Risk factors for falls as a cause of hip fracture in women. The Northeast Hip Fracture Study Group. N Engl J Med. 1991 May 9. 324(19):1326-31. [View Abstract]
  89. Nellans KW, Kowalski E, Chung KC. The epidemiology of distal radius fractures. Hand Clin. 2012 May. 28(2):113-25. [View Abstract]
  90. Sandhu SK, Nguyen ND, Center JR, Pocock NA, Eisman JA, Nguyen TV. Prognosis of fracture: evaluation of predictive accuracy of the FRAX algorithm and Garvan nomogram. Osteoporos Int. 2010 May. 21(5):863-71. [View Abstract]
  91. Cooper C, Atkinson EJ, O'Fallon WM, et al. Incidence of clinically diagnosed vertebral fractures: a population-based study in Rochester, Minnesota, 1985-1989. J Bone Miner Res. 1992 Feb. 7(2):221-7. [View Abstract]
  92. Cook DJ, Guyatt GH, Adachi JD, Clifton J, Griffith LE, Epstein RS, et al. Quality of life issues in women with vertebral fractures due to osteoporosis. Arthritis Rheum. 1993 Jun. 36(6):750-6. [View Abstract]
  93. Schnatz PF, Marakovits KA, Dubois M, O'Sullivan DM. Osteoporosis screening and treatment guidelines: are they being followed?. Menopause. 2011 Oct. 18(10):1072-8. [View Abstract]
  94. Geusens P, Dumitrescu B, van Geel T, van Helden S, Vanhoof J, Dinant GJ. Impact of systematic implementation of a clinical case finding strategy on diagnosis and therapy of postmenopausal osteoporosis. J Bone Miner Res. 2008 Jun. 23(6):812-8. [View Abstract]
  95. [Guideline] Screening for osteoporosis: U.S. preventive services task force recommendation statement. Ann Intern Med. 2011 Mar 1. 154(5):356-64. [View Abstract]
  96. Zhu K, Devine A, Lewis JR, Dhaliwal SS, Prince RL. Timed up and go test and bone mineral density measurement for fracture prediction. Arch Intern Med. 2011 Oct 10. 171(18):1655-61. [View Abstract]
  97. [Guideline] Liu H, Paige NM, Goldzweig CL, Wong E, Zhou A, Suttorp MJ, et al. Screening for osteoporosis in men: a systematic review for an American College of Physicians guideline. Ann Intern Med. 2008 May 6. 148(9):685-701. [View Abstract]
  98. Guglielmi G, Muscarella S, Bazzocchi A. Integrated imaging approach to osteoporosis: state-of-the-art review and update. Radiographics. 2011 Sep-Oct. 31(5):1343-64. [View Abstract]
  99. Khoo BC, Brown K, Cann C, et al. Comparison of QCT-derived and DXA-derived areal bone mineral density and T scores. Osteoporos Int. 2009 Sep. 20(9):1539-45. [View Abstract]
  100. Link TM. Axial and peripheral QCT. Guglielmi G, ed. Osteoporosis and Bone Densitometry Measurements. New York, NY: Springer Heidelberg; 2013. 123-32.
  101. Paunier L. Effect of magnesium on phosphorus and calcium metabolism. Monatsschr Kinderheilkd. 1992 Sep. 140(9 Suppl 1):S17-20. [View Abstract]
  102. Lee WY, Oh KW, Rhee EJ, Jung CH, Kim SW, Yun EJ, et al. Relationship between subclinical thyroid dysfunction and femoral neck bone mineral density in women. Arch Med Res. 2006 May. 37(4):511-6. [View Abstract]
  103. Tannenbaum C, Clark J, Schwartzman K, Wallenstein S, Lapinski R, Meier D. Yield of laboratory testing to identify secondary contributors to osteoporosis in otherwise healthy women. J Clin Endocrinol Metab. 2002 Oct. 87(10):4431-7. [View Abstract]
  104. Liu JM, Zhao HY, Ning G, Chen Y, Zhang LZ, Sun LH, et al. IGF-1 as an early marker for low bone mass or osteoporosis in premenopausal and postmenopausal women. J Bone Miner Metab. 2008. 26(2):159-64. [View Abstract]
  105. Resnick D, Kransdorf M. Osteoporosis. Bone and Joint Imaging. Third Edition. 2005. 551.
  106. Hillier TA, Stone KL, Bauer DC, Rizzo JH, Pedula KL, Cauley JA, et al. Evaluating the value of repeat bone mineral density measurement and prediction of fractures in older women: the study of osteoporotic fractures. Arch Intern Med. 2007 Jan 22. 167(2):155-60. [View Abstract]
  107. Nayak S, Roberts MS, Greenspan SL. Cost-effectiveness of different screening strategies for osteoporosis in postmenopausal women. Ann Intern Med. 2011 Dec 6. 155(11):751-61. [View Abstract]
  108. Lin JT, Lane JM. Bisphosphonates. J Am Acad Orthop Surg. 2003 Jan-Feb. 11(1):1-4. [View Abstract]
  109. Faulkner KG, Wacker WK, Barden HS, Simonelli C, Burke PK, Ragi S, et al. Femur strength index predicts hip fracture independent of bone density and hip axis length. Osteoporos Int. 2006. 17(4):593-9. [View Abstract]
  110. Prevrhal S, Shepherd JA, Faulkner KG, Gaither KW, Black DM, Lang TF. Comparison of DXA hip structural analysis with volumetric QCT. J Clin Densitom. 2008 Apr-Jun. 11(2):232-6. [View Abstract]
  111. Kaptoge S, Beck TJ, Reeve J, Stone KL, Hillier TA, Cauley JA, et al. Prediction of incident hip fracture risk by femur geometry variables measured by hip structural analysis in the study of osteoporotic fractures. J Bone Miner Res. 2008 Dec. 23(12):1892-904. [View Abstract]
  112. Beck TJ. Extending DXA beyond bone mineral density: understanding hip structure analysis. Curr Osteoporos Rep. 2007 Jun. 5(2):49-55. [View Abstract]
  113. Hans D, Barthe N, Boutroy S, Pothuaud L, Winzenrieth R, Krieg MA. Correlations between trabecular bone score, measured using anteroposterior dual-energy X-ray absorptiometry acquisition, and 3-dimensional parameters of bone microarchitecture: an experimental study on human cadaver vertebrae. J Clin Densitom. 2011 Jul-Sep. 14(3):302-12. [View Abstract]
  114. Pothuaud L, Barthe N, Krieg MA, Mehsen N, Carceller P, Hans D. Evaluation of the potential use of trabecular bone score to complement bone mineral density in the diagnosis of osteoporosis: a preliminary spine BMD-matched, case-control study. J Clin Densitom. 2009 Apr-Jun. 12(2):170-6. [View Abstract]
  115. Silva BC, Leslie WD, Resch H, Lamy O, Lesnyak O, Binkley N, et al. Trabecular bone score: a noninvasive analytical method based upon the DXA image. J Bone Miner Res. 2014 Mar. 29(3):518-30. [View Abstract]
  116. Schuit SC, van der Klift M, Weel AE, de Laet CE, Burger H, Seeman E, et al. Fracture incidence and association with bone mineral density in elderly men and women: the Rotterdam Study. Bone. 2004 Jan. 34(1):195-202. [View Abstract]
  117. Imai K. Recent methods for assessing osteoporosis and fracture risk. Recent Pat Endocr Metab Immune Drug Discov. 2014 Jan. 8(1):48-59. [View Abstract]
  118. Yang L, Palermo L, Black DM, Eastell R. Prediction of incident hip fracture with the estimated femoral strength by finite element analysis of DXA Scans in the study of osteoporotic fractures. J Bone Miner Res. 2014 Dec. 29(12):2594-600. [View Abstract]
  119. Torres-del-Pliego E, Vilaplana L, Güerri-Fernández R, Diez-Pérez A. Measuring bone quality. Curr Rheumatol Rep. 2013 Nov. 15(11):373. [View Abstract]
  120. Prior JC, Vigna YM, Wark JD, et al. Premenopausal ovariectomy-related bone loss: a randomized, double-blind, one-year trial of conjugated estrogen or medroxyprogesterone acetate. J Bone Miner Res. 1997 Nov. 12(11):1851-63. [View Abstract]
  121. Henzell S, Dhaliwal S, Pontifex R, et al. Precision error of fan-beam dual X-ray absorptiometry scans at the spine, hip, and forearm. J Clin Densitom. 2000 Winter. 3(4):359-64. [View Abstract]
  122. White J, Harris SS, Dallal GE, Dawson-Hughes B. Precision of single vs bilateral hip bone mineral density scans. J Clin Densitom. 2003 Summer. 6(2):159-62. [View Abstract]
  123. Bauer JS, Henning TD, Müeller D, Lu Y, Majumdar S, Link TM. Volumetric quantitative CT of the spine and hip derived from contrast-enhanced MDCT: conversion factors. AJR Am J Roentgenol. 2007 May. 188(5):1294-301. [View Abstract]
  124. Engelke K, Adams JE, Armbrecht G, et al. Clinical use of quantitative computed tomography and peripheral quantitative computed tomography in the management of osteoporosis in adults: the 2007 ISCD Official Positions. J Clin Densitom. 2008 Jan-Mar. 11(1):123-62. [View Abstract]
  125. Liu G, Peacock M, Eilam O, Dorulla G, Braunstein E, Johnston CC. Effect of osteoarthritis in the lumbar spine and hip on bone mineral density and diagnosis of osteoporosis in elderly men and women. Osteoporos Int. 1997. 7(6):564-9. [View Abstract]
  126. [Guideline] Qaseem A, Forciea MA, McLean RM, Denberg TD, for the Clinical Guidelines Committee of the American College of Physicians. Treatment of Low Bone Density or Osteoporosis to Prevent Fractures in Men and Women: A Clinical Practice Guideline Update from the American College of Physicians. Ann Intern Med. 9 May 2017.
  127. Fribourg D, Tang C, Sra P, Delamarter R, Bae H. Incidence of subsequent vertebral fracture after kyphoplasty. Spine (Phila Pa 1976). 2004 Oct 15. 29(20):2270-6; discussion 2277. [View Abstract]
  128. Movrin I, Vengust R, Komadina R. Adjacent vertebral fractures after percutaneous vertebral augmentation of osteoporotic vertebral compression fracture: a comparison of balloon kyphoplasty and vertebroplasty. Arch Orthop Trauma Surg. 2010 Sep. 130(9):1157-66. [View Abstract]
  129. Kastner M, Straus SE. Clinical decision support tools for osteoporosis disease management: a systematic review of randomized controlled trials. J Gen Intern Med. 2008 Dec. 23(12):2095-105. [View Abstract]
  130. [Guideline] Eastell R, Rosen CJ, Black DM, Cheung AM, Murad MH, Shoback D. Pharmacological Management of Osteoporosis in Postmenopausal Women: An Endocrine Society* Clinical Practice Guideline. J Clin Endocrinol Metab. 2019 May 1. 104 (5):1595-1622. [View Abstract]
  131. Abrahamsen B, Eiken P, Eastell R. Proton pump inhibitor use and the antifracture efficacy of alendronate. Arch Intern Med. 2011 Jun 13. 171(11):998-1004. [View Abstract]
  132. Harris ST, Watts NB, Genant HK, McKeever CD, Hangartner T, Keller M, 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 Oct 13. 282 (14):1344-52. [View Abstract]
  133. Black DM, Delmas PD, Eastell R, et al. Once-yearly zoledronic acid for treatment of postmenopausal osteoporosis. N Engl J Med. 2007 May 3. 356(18):1809-22. [View Abstract]
  134. Boonen S, Reginster JY, Kaufman JM, Lippuner K, Zanchetta J, Langdahl B, et al. Fracture risk and zoledronic acid therapy in men with osteoporosis. N Engl J Med. 2012 Nov. 367(18):1714-23. [View Abstract]
  135. Boonen S, Orwoll E, Magaziner J, Colón-Emeric CS, Adachi JD, Bucci-Rechtweg C, et al. Once-yearly zoledronic acid in older men compared with women with recent hip fracture. J Am Geriatr Soc. 2011 Nov. 59(11):2084-90. [View Abstract]
  136. US Food and Drug Administration. FDA drug safety communication: New contraindication and updated warning on kidney impairment for Reclast (zoledronic acid). Available at http://www.fda.gov/Drugs/DrugSafety/ucm270199.htm. September 1, 2011; Accessed: June 19, 2019.
  137. Safety Information: Zometa (zoledronic acid) for injection. U.S. Food and Drug Administration. Available at https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/021817s022lbl.pdf. April 2016; Accessed: June 15, 2019.
  138. Odvina CV, Zerwekh JE, Rao DS, Maalouf N, Gottschalk FA, Pak CY. Severely suppressed bone turnover: a potential complication of alendronate therapy. J Clin Endocrinol Metab. 2005 Mar. 90(3):1294-301. [View Abstract]
  139. Halasy-Nagy JM, Rodan GA, Reszka AA. Inhibition of bone resorption by alendronate and risedronate does not require osteoclast apoptosis. Bone. 2001 Dec. 29(6):553-9. [View Abstract]
  140. Strampel W, Emkey R, Civitelli R. Safety considerations with bisphosphonates for the treatment of osteoporosis. Drug Saf. 2007. 30(9):755-63. [View Abstract]
  141. Zebic L, Patel V. Preventing medication-related osteonecrosis of the jaw. BMJ. 2019 May 8. 365:l1733. [View Abstract]
  142. Park-Wyllie LY, Mamdani MM, Juurlink DN, Hawker GA, Gunraj N, Austin PC, et al. Bisphosphonate use and the risk of subtrochanteric or femoral shaft fractures in older women. JAMA. 2011 Feb 23. 305(8):783-9. [View Abstract]
  143. Geusens P. Bisphosphonates for postmenopausal osteoporosis: determining duration of treatment. Curr Osteoporos Rep. 2009 Mar. 7(1):12-7. [View Abstract]
  144. [Guideline] Adler RA, El-Hajj Fuleihan G, Bauer DC, Camacho PM, Clarke BL, Clines GA, et al. Managing Osteoporosis in Patients on Long-Term Bisphosphonate Treatment: Report of a Task Force of the American Society for Bone and Mineral Research. J Bone Miner Res. 2016 Jan. 31 (1):16-35. [View Abstract]
  145. Grady D, Cauley JA, Stock JL, Cox DA, Mitlak BH, Song J, et al. Effect of Raloxifene on all-cause mortality. Am J Med. 2010 May. 123(5):469.e1-7. [View Abstract]
  146. Lindsay R, Gallagher JC, Kagan R, Pickar JH, Constantine G. Efficacy of tissue-selective estrogen complex of bazedoxifene/conjugated estrogens for osteoporosis prevention in at-risk postmenopausal women. Fertil Steril. 2009 Sep. 92(3):1045-52. [View Abstract]
  147. Quattrocchi E, Kourlas H. Teriparatide: a review. Clin Ther. 2004 Jun. 26(6):841-54. [View Abstract]
  148. Koski AM, Sikiö A, Forslund T. Teriparatide treatment complicated by malignant myeloma. BMJ Case Rep. 2010 Aug 13. 2010:[View Abstract]
  149. Body JJ, Gaich GA, Scheele WH, Kulkarni PM, Miller PD, Peretz A. A randomized double-blind trial to compare the efficacy of teriparatide [recombinant human parathyroid hormone (1-34)] with alendronate in postmenopausal women with osteoporosis. J Clin Endocrinol Metab. 2002 Oct. 87(10):4528-35. [View Abstract]
  150. Dempster DW, Cosman F, Kurland ES, Zhou H, Nieves J, Woelfert L, et al. Effects of daily treatment with parathyroid hormone on bone microarchitecture and turnover in patients with osteoporosis: a paired biopsy study. J Bone Miner Res. 2001 Oct. 16(10):1846-53. [View Abstract]
  151. Neer RM, Arnaud CD, Zanchetta JR, Prince R, Gaich GA, Reginster JY, et al. Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med. 2001 May 10. 344(19):1434-41. [View Abstract]
  152. Kurland ES, Heller SL, Diamond B, McMahon DJ, Cosman F, Bilezikian JP. The importance of bisphosphonate therapy in maintaining bone mass in men after therapy with teriparatide [human parathyroid hormone(1-34)]. Osteoporos Int. 2004 Dec. 15(12):992-7. [View Abstract]
  153. Finkelstein JS, Hayes A, Hunzelman JL, Wyland JJ, Lee H, Neer RM. The effects of parathyroid hormone, alendronate, or both in men with osteoporosis. N Engl J Med. 2003 Sep 25. 349(13):1216-26. [View Abstract]
  154. Cosman F, Nieves J, Zion M, Woelfert L, Luckey M, Lindsay R. Daily and cyclic parathyroid hormone in women receiving alendronate. N Engl J Med. 2005 Aug 11. 353(6):566-75. [View Abstract]
  155. Deal C, Omizo M, Schwartz EN, Eriksen EF, Cantor P, Wang J, et al. Combination teriparatide and raloxifene therapy for postmenopausal osteoporosis: results from a 6-month double-blind placebo-controlled trial. J Bone Miner Res. 2005 Nov. 20(11):1905-11. [View Abstract]
  156. Ste-Marie LG, Schwartz SL, Hossain A, Desaiah D, Gaich GA. Effect of teriparatide [rhPTH(1-34)] on BMD when given to postmenopausal women receiving hormone replacement therapy. J Bone Miner Res. 2006 Feb. 21(2):283-91. [View Abstract]
  157. Tsai JN, Uihlein AV, Lee H, Kumbhani R, Siwila-Sackman E, McKay EA, et al. Teriparatide and denosumab, alone or combined, in women with postmenopausal osteoporosis: the DATA study randomised trial. Lancet. 2013 Jul 6. 382(9886):50-6. [View Abstract]
  158. Bouxsein ML, Chen P, Glass EV, Kallmes DF, Delmas PD, Mitlak BH. Teriparatide and raloxifene reduce the risk of new adjacent vertebral fractures in postmenopausal women with osteoporosis. Results from two randomized controlled trials. J Bone Joint Surg Am. 2009 Jun. 91(6):1329-38. [View Abstract]
  159. Kendler DL, Marin F, Zerbini CAF, Russo LA, Greenspan SL, Zikan V, et al. Effects of teriparatide and risedronate on new fractures in post-menopausal women with severe osteoporosis (VERO): a multicentre, double-blind, double-dummy, randomised controlled trial. Lancet. 2017 Nov 9. [View Abstract]
  160. Peichl P, Holzer LA, Maier R, Holzer G. Parathyroid hormone 1-84 accelerates fracture-healing in pubic bones of elderly osteoporotic women. J Bone Joint Surg Am. 2011 Sep 7. 93(17):1583-7. [View Abstract]
  161. Gomberg SJ, Wustrack RL, Napoli N, Arnaud CD, Black DM. Teriparatide, vitamin D, and calcium healed bilateral subtrochanteric stress fractures in a postmenopausal woman with a 13-year history of continuous alendronate therapy. J Clin Endocrinol Metab. 2011 Jun. 96(6):1627-32. [View Abstract]
  162. Saleh A, Hegde VV, Potty AG, Schneider R, Cornell CN, Lane JM. Management strategy for symptomatic bisphosphonate-associated incomplete atypical femoral fractures. HSS J. 2012 Jul. 8(2):103-10. [View Abstract]
  163. Lau AN, Adachi JD. Resolution of osteonecrosis of the jaw after teriparatide [recombinant human PTH-(1-34)] therapy. J Rheumatol. 2009 Aug. 36(8):1835-7. [View Abstract]
  164. Cheung A, Seeman E. Teriparatide therapy for alendronate-associated osteonecrosis of the jaw. N Engl J Med. 2010 Dec 16. 363(25):2473-4. [View Abstract]
  165. Narongroeknawin P, Danila MI, Humphreys LG Jr, Barasch A, Curtis JR. Bisphosphonate-associated osteonecrosis of the jaw, with healing after teriparatide: a review of the literature and a case report. Spec Care Dentist. 2010 Mar-Apr. 30(2):77-82. [View Abstract]
  166. Miller PD, Hattersley G, Riis BJ, Williams GC, Lau E, Russo LA, et al. Effect of Abaloparatide vs Placebo on New Vertebral Fractures in Postmenopausal Women With Osteoporosis: A Randomized Clinical Trial. JAMA. 2016 Aug 16. 316 (7):722-33. [View Abstract]
  167. Cosman F, Miller PD, Williams GC, Hattersley G, Hu MY, Valter I, et al. Eighteen Months of Treatment With Subcutaneous Abaloparatide Followed by 6 Months of Treatment With Alendronate in Postmenopausal Women With Osteoporosis: Results of the ACTIVExtend Trial. Mayo Clin Proc. 2017 Feb. 92 (2):200-210. [View Abstract]
  168. Harrison P. FRAME: Romosozumab Rapidly Reduces Fracture Risk in Osteoporosis. Medscape Medical News. Available at http://www.medscape.com/viewarticle/868923. September 19, 2016; Accessed: June 15, 2019.
  169. Cosman F, Crittenden DB, Adachi JD, Binkley N, Czerwinski E, Ferrari S, et al. Romosozumab Treatment in Postmenopausal Women with Osteoporosis. N Engl J Med. 2016 Sep 18. [View Abstract]
  170. Saag KG, Petersen J, Brandi ML, Karaplis AC, Lorentzon M, Thomas T, et al. Romosozumab or Alendronate for Fracture Prevention in Women with Osteoporosis. N Engl J Med. 2017 Oct 12. 377 (15):1417-1427. [View Abstract]
  171. Lewiecki EM, Dinavahi RV, Lazaretti-Castro M, Ebeling PR, Adachi JD, Miyauchi A, et al. One Year of Romosozumab Followed by Two Years of Denosumab Maintains Fracture Risk Reductions: Results of the FRAME Extension Study. J Bone Miner Res. 2019 Mar. 34 (3):419-428. [View Abstract]
  172. Lewiecki EM, Blicharski T, Goemaere S, Lippuner K, Meisner PD, Miller PD, et al. A Phase III Randomized Placebo-Controlled Trial to Evaluate Efficacy and Safety of Romosozumab in Men With Osteoporosis. J Clin Endocrinol Metab. 2018 Sep 1. 103 (9):3183-3193. [View Abstract]
  173. Body JJ, Facon T, Coleman RE, Lipton A, Geurs F, Fan M, et al. A study of the biological receptor activator of nuclear factor-kappaB ligand inhibitor, denosumab, in patients with multiple myeloma or bone metastases from breast cancer. Clin Cancer Res. 2006 Feb 15. 12(4):1221-8. [View Abstract]
  174. McClung MR, Lewiecki EM, Cohen SB, Bolognese MA, Woodson GC, Moffett AH, et al. Denosumab in postmenopausal women with low bone mineral density. N Engl J Med. 2006 Feb 23. 354(8):821-31. [View Abstract]
  175. Orwoll E, Teglbjærg CS, Langdahl BL, Chapurlat R, Czerwinski E, Kendler DL, et al. A randomized, placebo-controlled study of the effects of denosumab for the treatment of men with low bone mineral density. J Clin Endocrinol Metab. 2012 Sep. 97(9):3161-9. [View Abstract]
  176. Block GA, Bone HG, Fang L, Lee E, Padhi D. A single-dose study of denosumab in patients with various degrees of renal impairment. J Bone Miner Res. 2012 Jul. 27(7):1471-9. [View Abstract]
  177. Saag KG, Wagman RB, Geusens P, Adachi JD, Messina OD, Emkey R, et al. Denosumab versus risedronate in glucocorticoid-induced osteoporosis: a multicentre, randomised, double-blind, active-controlled, double-dummy, non-inferiority study. Lancet Diabetes Endocrinol. 2018 Apr 6. [View Abstract]
  178. Harrison P. Steroid-Induced Bone Loss Better Treated With Denosumab?. Medscape Medical News. Available at https://www.medscape.com/viewarticle/894997. April 10, 2018; Accessed: June 15, 2019.
  179. Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, et al. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009 Aug 20. 361(8):756-65. [View Abstract]
  180. Smith MR, Egerdie B, Hernández Toriz N, Feldman R, Tammela TL, Saad F, et al. Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med. 2009 Aug 20. 361(8):745-55. [View Abstract]
  181. Lyu H, Jundi B, Xu C, Tedeschi SK, Yoshida K, Zhao S, et al. Comparison of Denosumab and Bisphosphonates in Patients With Osteoporosis: A Meta-Analysis of Randomized Controlled Trials. J Clin Endocrinol Metab. 2019 May 1. 104 (5):1753-1765. [View Abstract]
  182. Schwarz EM, Ritchlin CT. Clinical development of anti-RANKL therapy. Arthritis Res Ther. 2007. 9 Suppl 1:S7. [View Abstract]
  183. Tsai JN, Uihlein AV, Lee H, Kumbhani R, Siwila-Sackman E, McKay EA, et al. Teriparatide and denosumab, alone or combined, in women with postmenopausal osteoporosis: the DATA study randomised trial. Lancet. 2013 May 14. [View Abstract]
  184. Postmark Drug Safety Information for Patients and Providers-Questions and Answers: Changes to the Indicated Population for Miacalcin (calcitonin-salmon). U.S. Food and Drug Administration. Available at http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/ucm388641.htm. September 1, 2015; Accessed: June 15, 2019.
  185. Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. JAMA. 2002 Jul 17. 288(3):321-33. [View Abstract]
  186. Blake GM, Fogelman I. Long-term effect of strontium ranelate treatment on BMD. J Bone Miner Res. 2005 Nov. 20(11):1901-4. [View Abstract]
  187. Burlet N, Reginster JY. Strontium ranelate: the first dual acting treatment for postmenopausal osteoporosis. Clin Orthop Relat Res. 2006 Feb. 443:55-60. [View Abstract]
  188. Bauer DC. Vertebral augmentation vs nonsurgical therapy: improved symptoms, improved survival, or neither?. JAMA Intern Med. 2013 Sep 9. 173(16):1522-3. [View Abstract]
  189. Bischoff-Ferrari HA, Dawson-Hughes B, Willett WC, Staehelin HB, Bazemore MG, Zee RY, et al. Effect of Vitamin D on falls: a meta-analysis. JAMA. 2004 Apr 28. 291(16):1999-2006. [View Abstract]
  190. Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, Clinton SK, et al. The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab. 2011 Jan. 96(1):53-8. [View Abstract]
  191. Bruni V, Dei M, Filicetti MF, Balzi D, Pasqua A. Predictors of bone loss in young women with restrictive eating disorders. Pediatr Endocrinol Rev. 2006 Jan. 3 Suppl 1:219-21. [View Abstract]
  192. Tang BM, Eslick GD, Nowson C, Smith C, Bensoussan A. Use of calcium or calcium in combination with vitamin D supplementation to prevent fractures and bone loss in people aged 50 years and older: a meta-analysis. Lancet. 2007 Aug 25. 370(9588):657-66. [View Abstract]
  193. Bischoff-Ferrari HA, Willett WC, Wong JB, Stuck AE, Staehelin HB, Orav EJ, et al. Prevention of nonvertebral fractures with oral vitamin D and dose dependency: a meta-analysis of randomized controlled trials. Arch Intern Med. 2009 Mar 23. 169(6):551-61. [View Abstract]
  194. Warensjö E, Byberg L, Melhus H, et al. Dietary calcium intake and risk of fracture and osteoporosis: prospective longitudinal cohort study. BMJ. 2011 May 24. 342:d1473. [View Abstract]
  195. DIPART (Vitamin D Individual Patient Analysis of Randomized Trials) Group. Patient level pooled analysis of 68 500 patients from seven major vitamin D fracture trials in US and Europe. BMJ. 2010 Jan 12. 340:b5463. [View Abstract]
  196. Bolland MJ, Grey A, Avenell A, Gamble GD, Reid IR. Calcium supplements with or without vitamin D and risk of cardiovascular events: reanalysis of the Women's Health Initiative limited access dataset and meta-analysis. BMJ. 2011 Apr 19. 342:d2040. [View Abstract]
  197. Li K, Kaaks R, Linseisen J, Rohrmann S. Associations of dietary calcium intake and calcium supplementation with myocardial infarction and stroke risk and overall cardiovascular mortality in the Heidelberg cohort of the European Prospective Investigation into Cancer and Nutrition study (EPIC-Heidelberg). Heart. 2012 Jun. 98(12):920-5. [View Abstract]
  198. Hsia J, Heiss G, Ren H, Allison M, Dolan NC, Greenland P, et al. Calcium/vitamin D supplementation and cardiovascular events. Circulation. 2007 Feb 20. 115(7):846-54. [View Abstract]
  199. Wang L, Manson JE, Song Y, Sesso HD. Systematic review: Vitamin D and calcium supplementation in prevention of cardiovascular events. Ann Intern Med. 2010 Mar 2. 152(5):315-23. [View Abstract]
  200. Curhan GC, Willett WC, Speizer FE, Spiegelman D, Stampfer MJ. Comparison of dietary calcium with supplemental calcium and other nutrients as factors affecting the risk for kidney stones in women. Ann Intern Med. 1997 Apr 1. 126(7):497-504. [View Abstract]
  201. Candelas G, Martinez-Lopez JA, Rosario MP, Carmona L, Loza E. Calcium supplementation and kidney stone risk in osteoporosis: a systematic literature review. Clin Exp Rheumatol. 2012 Nov-Dec. 30(6):954-61. [View Abstract]
  202. Favus MJ. The risk of kidney stone formation: the form of calcium matters. Am J Clin Nutr. 2011 Jul. 94(1):5-6. [View Abstract]
  203. Sinaki M. Postmenopausal spinal osteoporosis: physical therapy and rehabilitation principles. Mayo Clin Proc. 1982 Nov. 57(11):699-703. [View Abstract]
  204. Tinetti ME, Speechley M. Prevention of falls among the elderly. N Engl J Med. 1989 Apr 20. 320(16):1055-9. [View Abstract]
  205. Sinaki M, Mikkelsen BA. Postmenopausal spinal osteoporosis: flexion versus extension exercises. Arch Phys Med Rehabil. 1984 Oct. 65(10):593-6. [View Abstract]
  206. Sinaki M, Itoi E, Wahner HW, Wollan P, Gelzcer R, Mullan BP, et al. Stronger back muscles reduce the incidence of vertebral fractures: a prospective 10 year follow-up of postmenopausal women. Bone. 2002 Jun. 30(6):836-41. [View Abstract]
  207. Sinaki M, Itoi E, Rogers JW, Bergstralh EJ, Wahner HW. Correlation of back extensor strength with thoracic kyphosis and lumbar lordosis in estrogen-deficient women. Am J Phys Med Rehabil. 1996 Sep-Oct. 75(5):370-4. [View Abstract]
  208. Chien MY, Wu YT, Hsu AT, Yang RS, Lai JS. Efficacy of a 24-week aerobic exercise program for osteopenic postmenopausal women. Calcif Tissue Int. 2000 Dec. 67(6):443-8. [View Abstract]
  209. Snow CM, Shaw JM, Winters KM, Witzke KA. Long-term exercise using weighted vests prevents hip bone loss in postmenopausal women. J Gerontol A Biol Sci Med Sci. 2000 Sep. 55(9):M489-91. [View Abstract]
  210. Howe TE, Shea B, Dawson LJ, et al. Exercise for preventing and treating osteoporosis in postmenopausal women. Cochrane Database Syst Rev. 2011 Jul 6. CD000333. [View Abstract]
  211. Iwamoto J, Takeda T, Ichimura S. Effect of exercise training and detraining on bone mineral density in postmenopausal women with osteoporosis. J Orthop Sci. 2001. 6(2):128-32. [View Abstract]
  212. Kerschan-Shindl K, Uher E, Kainberger F, Kaider A, Ghanem AH, Preisinger E. Long-term home exercise program: effect in women at high risk of fracture. Arch Phys Med Rehabil. 2000 Mar. 81(3):319-23. [View Abstract]
  213. Robertson MC, Devlin N, Gardner MM, Campbell AJ. Effectiveness and economic evaluation of a nurse delivered home exercise programme to prevent falls. 1: Randomised controlled trial. BMJ. 2001 Mar 24. 322(7288):697-701. [View Abstract]
  214. Walker M, Klentrou P, Chow R, Plyley M. Longitudinal evaluation of supervised versus unsupervised exercise programs for the treatment of osteoporosis. Eur J Appl Physiol. 2000 Nov. 83(4 -5):349-55. [View Abstract]
  215. Wolf SL, Barnhart HX, Kutner NG, McNeely E, Coogler C, Xu T. Reducing frailty and falls in older persons: an investigation of Tai Chi and computerized balance training. Atlanta FICSIT Group. Frailty and Injuries: Cooperative Studies of Intervention Techniques. J Am Geriatr Soc. 1996 May. 44(5):489-97. [View Abstract]
  216. Carter ND, Khan KM, Petit MA, Heinonen A, Waterman C, Donaldson MG, et al. Results of a 10 week community based strength and balance training programme to reduce fall risk factors: a randomised controlled trial in 65-75 year old women with osteoporosis. Br J Sports Med. 2001 Oct. 35(5):348-51. [View Abstract]
  217. Riggs BL, Melton LJ 3rd. The prevention and treatment of osteoporosis. N Engl J Med. 1992 Aug 27. 327(9):620-7. [View Abstract]
  218. Iwamoto J, Sato Y, Uzawa M, Takeda T, Matsumoto H. Comparison of effects of alendronate and raloxifene on lumbar bone mineral density, bone turnover, and lipid metabolism in elderly women with osteoporosis. Yonsei Med J. 2008 Feb 29. 49(1):119-28. [View Abstract]
  219. Gourlay ML, Fine JP, Preisser JS, May RC, Li C, Lui LY, et al. Bone-density testing interval and transition to osteoporosis in older women. N Engl J Med. 2012 Jan 19. 366(3):225-33. [View Abstract]
  220. Schwab P, Klein RF. Nonpharmacological approaches to improve bone health and reduce osteoporosis. Curr Opin Rheumatol. 2008 Mar. 20(2):213-7. [View Abstract]
  221. Lin JT, Lane JM. Nonmedical management of osteoporosis. Curr Opin Rheumatol. 2002 Jul. 14(4):441-6. [View Abstract]
  222. Gourlay ML, Fine JP, Preisser JS, May RC, Li C, Lui LY, et al. Bone-density testing interval and transition to osteoporosis in older women. N Engl J Med. 2012 Jan 19. 366(3):225-33. [View Abstract]
  223. Kirshblum SC. Rehabilitation Medicine: Principles and Practice. DeLisa JA, Gans BM. Spinal and upper extremity orthotics. 3rd. Philadelphia, Pa: Lippincott-Raven; 1998. 635-50.
  224. Stillo JV. Low back orthoses. Phys Med Rehab Clin North Am. 1992. 3:57-94.
  225. [Guideline] Viswanathan M, Reddy S, Berkman N, Cullen K, Middleton JC, Nicholson WK, et al. Screening to Prevent Osteoporotic Fractures: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA. 2018 Jun 26. 319 (24):2532-2551. [View Abstract]
  226. Melville NA. Updated Osteoporosis Screening Guidelines Released by USPSTF. Medscape Medical News. Available at https://www.medscape.com/viewarticle/898590. June 27, 2018; Accessed: June 15, 2019.
  227. [Guideline] Updates to the Clinician’s Guide to the Prevention and Treatment of Osteoporosis. National Osteoporosis Foundation. Available at https://my.nof.org/bone-source/Incorporation-of-diagnostic-criteria-to-be-used-to-establish-a-clinical-diagnosis-of-osteoporosis. November 11, 2015; Accessed: June 21, 2019.
  228. Compston J, Bowring C, Cooper A, Cooper C, Davies C, Francis R, et al. Diagnosis and management of osteoporosis in postmenopausal women and older men in the UK: National Osteoporosis Guideline Group (NOGG) update 2013. Maturitas. 2013 Aug. 75(4):392-6. [View Abstract]
  229. [Guideline] Eastell R, Rosen CJ, Black DM, Cheung AM, Murad MH, Shoback D. Pharmacological Management of Osteoporosis in Postmenopausal Women: An Endocrine Society* Clinical Practice Guideline. J Clin Endocrinol Metab. 2019 Mar 25. [View Abstract]
  230. Keen R. Osteoporosis: strategies for prevention and management. Best Pract Res Clin Rheumatol. 2007 Feb. 21(1):109-22. [View Abstract]
  231. NIH Consensus conference. Optimal calcium intake. NIH Consensus Development Panel on Optimal Calcium Intake. JAMA. 1994 Dec 28. 272(24):1942-8. [View Abstract]
  232. Hulley SB, Grady D. The WHI estrogen-alone trial--do things look any better?. JAMA. 2004 Apr 14. 291(14):1769-71. [View Abstract]

Osteoporosis of the spine. Observe the considerable reduction in overall vertebral bone density and note the lateral wedge fracture of L2.

Osteoporosis. Lateral radiograph demonstrates multiple osteoporotic vertebral compression fractures. Kyphoplasty has been performed at one level.

Osteoporosis of the spine. Note the lateral wedge fracture in L3 and the central burst fracture in L5. The patient had suffered a recent fall.

This image depicts bone remodeling with osteoclasts resorbing one side of a bony trabecula and osteoblasts depositing new bone on the other side.

Osteoclast, with bone below it. This image shows typical distinguishing characteristics of an osteoclast: a large cell with multiple nuclei and a "foamy" cytosol.

In this image, several osteoblasts display a prominent Golgi apparatus and are actively synthesizing osteoid. Two osteocytes can also be seen.

Osteoporosis is defined as a loss of bone mass below the threshold of fracture. This slide (methylmethacrylate embedded and stained with Masson's trichrome) demonstrates the loss of connected trabecular bone.

The bone loss of osteoporosis can be severe enough to create separate bone "buttons" with no connection to the surrounding bone. This easily leads to insufficiency fractures.

Osteoporosis. Lateral radiograph demonstrates multiple osteoporotic vertebral compression fractures. Kyphoplasty has been performed at one level.

Osteoporosis. Lateral radiograph of the patient seen in the previous image following kyphoplasty performed at 3 additional levels.

Stable intertrochanteric fracture of the femur.

Asymmetric loss in vertebral body height, without evidence of an acute fracture, can develop in patients with osteoporosis. These patients become progressively kyphotic (as shown) over time, and the characteristic hunched-over posture of severe osteoporosis develops eventually.

Severe osteoporosis. This radiograph shows multiple vertebral crush fractures. Source: Government of Western Australia Department of Health.

Example of a dual energy x-ray absorption (DXA) scan. This image is of the left hip bone. Source: Government of Western Australia Department of Health.

Example of a dual energy x-ray absorption (DXA) scan. This image is of the lumbar spine. Source: Government of Western Australia Department of Health.

In kyphoplasty, a KyphX inflatable bone tamp is percutaneously advanced into the collapsed vertebral body (A). It is then inflated, (B) elevating the depressed endplate, creating a central cavity, and compacting the remaining trabeculae to the periphery. Once the balloon tamp is deflated and withdrawn, the cavity (C) is filled under low pressure with a viscous preparation of methylmethacrylate (D).

Osteoporosis. Lateral radiograph demonstrates multiple osteoporotic vertebral compression fractures. Kyphoplasty has been performed at one level.

Osteoporosis. Lateral radiograph of the patient seen in the previous image following kyphoplasty performed at 3 additional levels.

Percutaneous vertebroplasty, transpedicular approach.

Osteoporosis is defined as a loss of bone mass below the threshold of fracture. This slide (methylmethacrylate embedded and stained with Masson's trichrome) demonstrates the loss of connected trabecular bone.

The bone loss of osteoporosis can be severe enough to create separate bone "buttons" with no connection to the surrounding bone. This easily leads to insufficiency fractures.

Inactive osteoporosis is the most common form and manifests itself without active osteoid formation.

Osteoporosis that is active contains osteoid seams (red here in the Masson's trichrome).

Woven bone arising directly from surrounding mesenchymal tissue.

This image depicts bone remodeling with osteoclasts resorbing one side of a bony trabecula and osteoblasts depositing new bone on the other side.

Osteoclast, with bone below it. This image shows typical distinguishing characteristics of an osteoclast: a large cell with multiple nuclei and a "foamy" cytosol.

In this image, several osteoblasts display a prominent Golgi apparatus and are actively synthesizing osteoid. Two osteocytes can also be seen.

Normal femoral anatomy.

Stable intertrochanteric fracture of the femur.

In kyphoplasty, a KyphX inflatable bone tamp is percutaneously advanced into the collapsed vertebral body (A). It is then inflated, (B) elevating the depressed endplate, creating a central cavity, and compacting the remaining trabeculae to the periphery. Once the balloon tamp is deflated and withdrawn, the cavity (C) is filled under low pressure with a viscous preparation of methylmethacrylate (D).

Osteoporosis. Lateral radiograph demonstrates multiple osteoporotic vertebral compression fractures. Kyphoplasty has been performed at one level.

Osteoporosis. Lateral radiograph of the patient seen in the previous image following kyphoplasty performed at 3 additional levels.

Osteoporosis. Lateral radiograph demonstrates multiple osteoporotic vertebral compression fractures. Kyphoplasty has been performed at one level.

Osteoporosis. Lateral radiograph of the patient seen in the previous image following kyphoplasty performed at 3 additional levels.

Osteoporosis of the spine. Observe the considerable reduction in overall vertebral bone density and note the lateral wedge fracture of L2.

Osteoporosis of the spine. Note the lateral wedge fracture in L3 and the central burst fracture in L5. The patient had suffered a recent fall.

Normal femoral anatomy.

Stable intertrochanteric fracture of the femur.

Percutaneous vertebroplasty, transpedicular approach.

Asymmetric loss in vertebral body height, without evidence of an acute fracture, can develop in patients with osteoporosis. These patients become progressively kyphotic (as shown) over time, and the characteristic hunched-over posture of severe osteoporosis develops eventually.

In kyphoplasty, a KyphX inflatable bone tamp is percutaneously advanced into the collapsed vertebral body (A). It is then inflated, (B) elevating the depressed endplate, creating a central cavity, and compacting the remaining trabeculae to the periphery. Once the balloon tamp is deflated and withdrawn, the cavity (C) is filled under low pressure with a viscous preparation of methylmethacrylate (D).

Osteoporosis is defined as a loss of bone mass below the threshold of fracture. This slide (methylmethacrylate embedded and stained with Masson's trichrome) demonstrates the loss of connected trabecular bone.

The bone loss of osteoporosis can be severe enough to create separate bone "buttons" with no connection to the surrounding bone. This easily leads to insufficiency fractures.

Inactive osteoporosis is the most common form and manifests itself without active osteoid formation.

Osteoporosis that is active contains osteoid seams (red here in the Masson's trichrome).

Woven bone arising directly from surrounding mesenchymal tissue.

This image depicts bone remodeling with osteoclasts resorbing one side of a bony trabecula and osteoblasts depositing new bone on the other side.

Osteoclast, with bone below it. This image shows typical distinguishing characteristics of an osteoclast: a large cell with multiple nuclei and a "foamy" cytosol.

In this image, several osteoblasts display a prominent Golgi apparatus and are actively synthesizing osteoid. Two osteocytes can also be seen.

Severe osteoporosis. This radiograph shows multiple vertebral crush fractures. Source: Government of Western Australia Department of Health.

Lateral spine radiograph depicting osteoporotic wedge fractures of L1-L2. Source: Wikimedia Commons.

Dual-energy computed tomography (CT) scan in a patient with involutional osteoporosis. Insufficiency fractures of the sacrum and the pubic rami are seen on an isotopic bone scan as a characteristic H, or Honda, sign (arrows), which appears as intense radiopharmaceutical uptake at the fracture sites.

Schematic example of an early bone densitometer: the QDR-1000 System (spine scan). (From: Third National Health and Nutrition Examination Survey Bone Densitometry Manual. Rockville, Md: Westat, Inc; 1989 [revised].)

Bone density scanner. This machine measures bone density to check for osteoporosis in the elderly and other vulnerable subjects. Source: Wikimedia Commons.

Example of a dual energy x-ray absorption (DXA) scan. This image is of the left hip bone. Source: Government of Western Australia Department of Health.

Example of a dual energy x-ray absorption (DXA) scan. This image is of the lumbar spine. Source: Government of Western Australia Department of Health.

Definition Bone Mineral Density Measurement T-Score
NormalBMD within 1 SD of the mean bone density for young adult womenT-score ≥ –1
Low bone mass (osteopenia)BMD 1–2.5 SD below the mean for young-adult womenT-score between –1 and –2.5
OsteoporosisBMD ≥2.5 SD below the normal mean for young-adult womenT-score ≤ –2.5
Severe or “established” osteoporosisBMD ≥2.5 SD below the normal mean for young-adult women in a patient who has already experienced ≥1 fracturesT-score ≤ –2.5 (with fragility fracture[s])
Sources:



(1) World Health Organization (WHO). WHO scientific group on the assessment of osteoporosis at primary health care level: summary meeting report. Available at: http://www.who.int/chp/topics/Osteoporosis.pdf. Accessed February 23, 2015.[16]



(2) Kanis JA. Assessment of fracture risk and its application to screening for postmenopausal osteoporosis: synopsis of a WHO report. WHO Study Group. Osteoporos Int. Nov 1994;4(6):368-81.[8]



(3) Czerwinski E, Badurski JE, Marcinowska-Suchowierska E, Osieleniec J. Current understanding of osteoporosis according to the position of the World Health Organization (WHO) and International Osteoporosis Foundation. Ortop Traumatol Rehabil. Jul-Aug 2007;9(4):337-56.[7]



BMD = bone mineral density; DXA = dual x-ray absorptiometry; SD = standard deviation; T-score = a measurement expressed in SD units from a given mean that is equal to a patient's BMD measured by DXA minus the value in a young healthy person, divided by the SD measurement in the population.



Type of Primary Osteoporosis Characteristics
Juvenile osteoporosis
  • Usually occurs in children or young adults of both sexes
  • Normal gonadal function
  • Age of onset: usually 8-14 years
  • Hallmark characteristic: abrupt bone pain and/or a fracture following trauma
Idiopathic osteoporosis
  • Postmenopausal osteoporosis (type I osteoporosis)
  • Occurs in women with estrogen deficiency
  • Characterized by a phase of accelerated bone loss, primarily from trabecular bone
  • Fractures of the distal forearm and vertebral bodies common
  • Age-associated or senile osteoporosis (type II osteoporosis)
  • Occurs in women and men as BMD gradually declines with aging
  • Represents bone loss associated with aging
  • Fractures occur in cortical and trabecular bone
  • Wrist, vertebral, and hip fractures often seen 
Cause Examples
Genetic/congenital
  • Renal hypercalciuria – one of the most important secondary causes of osteoporosis; can be treated with thiazide diuretics
  • Cystic fibrosis
  • Ehlers-Danlos syndrome
  • Glycogen storage disease
  • Gaucher disease
  • Marfan syndrome
  • Menkes steely hair syndrome
  • Riley-Day syndrome
  • Osteogenesis imperfecta
  • Hemochromatosis
  • Homocystinuria
  • Hypophosphatasia
  • Idiopathic hypercalciuria
  • Porphyria
  • Hypogonadal states
Hypogonadal states
  • Androgen insensitivity
  • Anorexia nervosa/bulimia nervosa
  • Female athlete triad
  • Hyperprolactinemia
  • Panhypopituitarism
  • Premature menopause
  • Turner syndrome
  • Klinefelter syndrome
Endocrine disorders[46]
  • Cushing syndrome
  • Diabetes mellitus
  • Acromegaly
  • Adrenal insufficiency
  • Estrogen deficiency
  • Hyperparathyroidism
  • Hyperthyroidism
  • Hypogonadism
  • Pregnancy
  • Prolactinoma
Deficiency states
  • Calcium deficiency
  • Magnesium deficiency
  • Protein deficiency
  • Vitamin D deficiency[46, 47]
  • Bariatric surgery
  • Celiac disease
  • Gastrectomy
  • Malabsorption
  • Malnutrition
  • Parenteral nutrition
  • Primary biliary cirrhosis
Inflammatory diseases
  • Inflammatory bowel disease
  • Ankylosing spondylitis
  • Rheumatoid arthritis
  • Systemic lupus erythematosus
Hematologic and neoplastic disorders
  • Hemochromatosis
  • Hemophilia
  • Leukemia
  • Lymphoma
  • Multiple myeloma
  • Sickle cell anemia
  • Systemic mastocytosis
  • Thalassemia
  • Metastatic disease
Medications
  • Anticonvulsants
  • Antipsychotic drugs
  • Antiretroviral drugs
  • Aromatase inhibitors
  • Chemotherapeutic/transplant drugs: cyclosporine, tacrolimus, platinum compounds, cyclophosphamide, ifosfamide, high-dose methotrexate[48]
  • Furosemide
  • Glucocorticoids and corticotropin[49] : prednisone (≥5 mg/day for ≥3 mo)[50]
  • Heparin (long term)
  • Hormonal/endocrine therapies: gonadotropin-releasing hormone (GnRH) agonists, luteinizing hormone–releasing hormone (LHRH) analogues, depomedroxyprogesterone, excessive thyroxine
  • Lithium
  • Selective serotonin reuptake inhibitors (SSRIs)
Miscellaneous
  • Alcoholism
  • Amyloidosis
  • Chronic metabolic acidosis
  • Chronic heart failure
  • Depression
  • Emphysema
  • Chronic or end-stage renal disease
  • Chronic liver disease
  • HIV/AIDS
  • Idiopathic scoliosis
  • Immobility
  • Multiple sclerosis
  • Ochronosis
  • Organ transplantation
  • Pregnancy/lactation
  • Sarcoidosis
  • Weightlessness[51]
Sources:



(1) American Association of Clinical Endocrinologists medical guidelines for clinical practice for the prevention and treatment of postmenopausal osteoporosis: 2001 edition, with selected updates for 2003. Endocr Pract. Nov-Dec 2003;9(6):544-64.[43]



(2) Kelman A, Lane NE. The management of secondary osteoporosis. Best Pract Res Clin Rheumatol. Dec 2005;19(6):1021-37.[44]



Race/Ethnicity Sex (age ≥50 y) % Estimated to have osteoporosis % Estimated to have low bone mass
Non-Hispanic white; AsianWomen15.852.6
Men3.936
Non-Hispanic blackWomen7.736.2
Men1.321.3
HispanicWomen20.447.8
Men5.938.3
Source: Wright NC, Looker AC, Saag KG, Curtis JR, Delzell ES, Randall S, et al. The recent prevalence of osteoporosis and low bone mass in the United States based on bone mineral density at the femoral neck or lumbar spine. J Bone Miner Res. Nov 2014;29(11):2520-6.
Baseline test Disorder
Complete blood count (CBC)CBC results may reveal anemia, as in sickle cell disease (patients with anemia, particularly those older than 60 years, should also be evaluated for multiple myeloma), and may raise the suspicion for alcohol abuse (in conjunction with results from serum chemistry tests and liver function tests)
Serum chemistry levelsCalcium levels can reflect underlying disease states (eg, severe hypercalcemia may reflect underlying malignancy or hyperparathyroidism; hypocalcemia can contribute to osteoporosis)



levels of serum calcium, phosphate, and alkaline phosphatase are usually normal in persons with primary osteoporosis, although alkaline phosphatase levels may be elevated for several months after a fracture



levels of serum calcium, phosphate, alkaline phosphatase, and 25(OH) vitamin D may be obtained to assess osteomalacia



Creatinine levels may decrease with increasing parathyroid hormone (PTH) levels or may be elevated in patients with multiple myeloma



Creatinine levels are also used to estimate creatinine clearance, which may indicate reduced renal function in elderly patients



Magnesium is very important in calcium homeostasis[101] ; decreased levels of magnesium may affect calcium absorption and metabolism



Liver function testsIncreased levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transferase (GGT), bilirubin, and alkaline phosphatase may indicate alcohol abuse
Thyroid-stimulating hormone (TSH) levelThyroid dysfunction has been associated with osteoporosis and should therefore be ruled out[102]
25-Hydroxyvitamin D levelThis test assesses for vitamin D insufficiency; inadequate vitamin D levels can predispose persons to osteoporosis
Tests for Secondary Causes of Osteoporosis Disorder
24-Hour urine calcium levelThis study assesses for hypercalciuria and hypocalciuria
Parathyroid hormone (PTH) levelAn intact PTH result is essential in ruling out hyperparathyroidism; an elevated PTH level may be present in benign familial hypocalciuric hypercalcemia
Thyrotropin level (if on thyroid replacement)Experts are divided on whether to include thyrotropin testing, regardless of a history of thyroid disease or replacement; however, one study showed reduced femoral neck bone mineral density (BMD) in women with subclinical hypothyroidism and hyperthyroidism[102]
Testosterone and gonadotropin levels in younger men with low bone densitiesThese tests may help evaluate a sex hormone deficiency as a secondary cause of osteoporosis
Urinary free cortisol level and tests for adrenal hypersecretionThese tests are used to exclude Cushing syndrome, which, although uncommon, can lead to rapidly progressive osteoporosis when the condition is present; a urine free cortisol value or overnight dexamethasone suppression test should be ordered in suspected cases
Serum protein electrophoresis (SPEP) and urine protein electrophoresis (UPEP)These are used to identify multiple myeloma
Antigliadin and antiendomysial antibodiesThese tests can help identify celiac disease
Serum tryptase and urine N-methylhistamineThese tests help identify mastocytosis
Bone marrow biopsyThis study is obtained when a hematologic disorder is suspected