Osteoporosis: A Comprehensive Review of Pathophysiology, Risk Factors, Diagnostic Methods, and Treatment Strategies

Abstract

Osteoporosis, widely recognized as the ‘silent thief’ of bone, constitutes a pervasive systemic skeletal disorder characterized by a critical reduction in bone mass, coupled with a profound microarchitectural deterioration of bone tissue. This dual pathology precipitously elevates bone fragility, thereby escalating the risk of debilitating fractures. This comprehensive and in-depth review meticulously dissects the intricate pathophysiology underpinning osteoporosis, elaborates on the extensive spectrum of its risk factors, details the multifaceted diagnostic methodologies employed for its identification, and critically evaluates both established and nascent therapeutic strategies. By meticulously synthesizing current cutting-edge research, evolving clinical practices, and epidemiological insights, this report aims to furnish a nuanced, detailed, and contemporary understanding of osteoporosis, simultaneously highlighting critical areas ripe for future research endeavors and delineating potential transformative advancements in its long-term management.

1. Introduction

Osteoporosis stands as a formidable global public health challenge, a highly prevalent metabolic bone disease afflicting hundreds of millions worldwide across all demographics. Its profound impact is underscored by the significant morbidity, escalating healthcare costs, and increased mortality primarily attributable to fragility fractures. The insidious nature of the condition, often progressing silently without overt clinical symptoms until the occurrence of a catastrophic fracture, underscores the paramount importance of proactive early detection, precise risk stratification, and timely, effective intervention. The economic burden associated with osteoporosis-related fractures is staggering, comparable to or even exceeding that of other major chronic non-communicable diseases, leading to prolonged hospitalization, loss of independence, and a diminished quality of life for affected individuals and their caregivers [1, 2].

This extensive report undertakes an exhaustive examination of the multifaceted dimensions of osteoporosis. It meticulously explores its fundamental underlying biological mechanisms, delving into the cellular and molecular cascades that precipitate bone loss. Furthermore, it comprehensively details the diverse array of genetic, environmental, and lifestyle-related risk factors that predispose individuals to this condition. The report also addresses the inherent challenges associated with its accurate and timely diagnosis, critically evaluating the gold standard and emerging diagnostic tools. Finally, it provides an in-depth analysis of the current pharmacological and non-pharmacological therapeutic approaches, alongside a forward-looking perspective on innovative and emerging treatment modalities that promise to reshape the landscape of osteoporosis management in the coming decades.

2. Pathophysiology of Osteoporosis

Bone tissue, far from being static, is a dynamic and metabolically active organ perpetually undergoing a process of renewal known as bone remodeling. This finely orchestrated process is vital for maintaining skeletal integrity, repairing micro-damage, and regulating mineral homeostasis. In the context of osteoporosis, this delicate balance is profoundly disrupted, leading to an unfavorable equation where bone resorption outpaces bone formation, culminating in a net loss of bone mass and architectural degradation.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

2.1 Bone Remodeling and Cellular Dysregulation

Bone remodeling is primarily orchestrated by two principal cell types: osteoclasts, which are large, multinucleated cells responsible for resorbing old or damaged bone matrix, and osteoblasts, which are bone-forming cells that synthesize new bone matrix, which subsequently mineralizes. These two processes are typically coupled, ensuring that the volume of bone removed is precisely replaced by an equal volume of new bone, thereby maintaining skeletal homeostasis. This coupling is mediated by a complex interplay of systemic hormones, local growth factors, and cytokines [3].

In osteoporosis, this intricate coupling becomes uncoupled, predominantly due to several key mechanisms:

  • Increased Osteoclast Activity: The hallmark of osteoporotic bone loss is an excessive and prolonged increase in osteoclast-mediated bone resorption. This enhanced activity leads to the rapid excavation of bone tissue, creating deeper and more numerous resorption pits than can be adequately refilled by osteoblasts. This phenomenon is often driven by an upregulation of pro-resorptive signals and a downregulation of anti-resorptive signals. For instance, in postmenopausal osteoporosis, estrogen deficiency profoundly impacts osteoclast activity by altering the balance of receptor activator of nuclear factor kappa-B ligand (RANKL) and osteoprotegerin (OPG), tilting it in favor of increased bone resorption [4].
  • Decreased Osteoblast Activity and Differentiation: Concurrently or independently, there is often a significant impairment in osteoblast function and a reduction in their numbers or activity. This diminished capacity for bone formation means that the bone laid down by osteoblasts is insufficient to compensate for the bone removed by osteoclasts. Factors contributing to reduced osteoblast activity include cellular senescence, altered signaling pathways (e.g., Wnt/β-catenin pathway dysregulation), and a decline in the availability or potency of growth factors essential for osteoblast proliferation and differentiation [5].
  • Hormonal Influences: Hormones play a pivotal role in regulating bone metabolism. Estrogen, in particular, is a critical regulator of bone remodeling. Postmenopausal estrogen deficiency is the primary driver of rapid bone loss in women, as estrogen normally suppresses osteoclastogenesis and promotes osteoblast survival. Its absence leads to increased cytokine production (e.g., IL-1, IL-6, TNF-α) that indirectly enhance osteoclast activity and lifespan. In men, age-related decline in testosterone, which can be aromatized to estrogen, also contributes to bone loss, although typically at a slower rate than in postmenopausal women [6]. Other hormones like parathyroid hormone (PTH), vitamin D, and glucocorticoids also exert significant influence on bone turnover, and their dysregulation can contribute to secondary osteoporosis.
  • Altered Bone Matrix Composition and Quality: Beyond bone mineral density (BMD), bone quality—encompassing microarchitecture, bone turnover rate, accumulation of micro-damage, and mineralization—is a crucial determinant of bone strength. In osteoporosis, the bone matrix, primarily composed of type I collagen and non-collagenous proteins, undergoes qualitative changes. Collagen cross-linking abnormalities, accumulation of advanced glycation end-products (AGEs), and altered mineralization patterns can render the bone more brittle and less resistant to fracture, even independent of significant reductions in BMD [7]. The disruption of the trabecular network, leading to thinner and disconnected trabeculae, and the thinning of cortical bone also significantly compromise mechanical integrity.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

2.2 Genetic and Molecular Factors

The susceptibility to osteoporosis and the propensity for low bone mass are significantly influenced by an individual’s genetic makeup, with heritability estimates for BMD ranging from 50% to 85% [8]. Understanding these genetic predispositions is crucial for identifying at-risk individuals and developing targeted therapies. Molecular pathways underlying bone metabolism are complex and involve numerous genes and signaling cascades:

  • Key Genes and Polymorphisms: Variations (polymorphisms) in genes encoding for various components of bone metabolism can modulate peak bone mass and the rate of bone loss. Prominent examples include:
    • Vitamin D Receptor (VDR) Gene: Polymorphisms in the VDR gene can affect the efficiency of vitamin D action, impacting calcium absorption and bone mineralization [9].
    • Collagen Type I Alpha 1 (COL1A1) Gene: Mutations or polymorphisms in this gene, which encodes the primary structural protein of bone, can affect collagen synthesis and quality, leading to conditions like osteogenesis imperfecta or contributing to osteoporosis risk [10].
    • Low-Density Lipoprotein Receptor-Related Protein 5 (LRP5) Gene: LRP5 is a co-receptor in the Wnt/β-catenin signaling pathway, which is critical for osteoblast proliferation, differentiation, and survival. Loss-of-function mutations in LRP5 are associated with severe primary osteoporosis (osteoporosis-pseudoglioma syndrome), while gain-of-function mutations lead to high bone mass [11].
    • Sclerostin (SOST) Gene: Sclerostin, encoded by the SOST gene, is an osteocyte-derived protein that negatively regulates bone formation by inhibiting the Wnt/β-catenin pathway. Genetic variations affecting sclerostin levels can influence bone density [12].
  • RANK/RANKL/OPG System: This triumvirate represents the master regulatory axis for osteoclastogenesis. RANKL, expressed on osteoblasts and stromal cells, binds to RANK (receptor activator of nuclear factor kappa-B) on osteoclast precursors and mature osteoclasts, promoting their differentiation, activity, and survival. OPG, a decoy receptor produced by osteoblasts, binds to RANKL, preventing its interaction with RANK and thereby inhibiting osteoclast formation and activity. In osteoporosis, an imbalance often occurs where the RANKL/OPG ratio is elevated, leading to increased osteoclastogenesis and bone resorption [4].
  • Wnt/β-catenin Signaling Pathway: This pathway is fundamentally important for bone formation. Wnt proteins bind to Frizzled receptors and LRP5/6 co-receptors on osteoblasts, leading to the stabilization of β-catenin, which then translocates to the nucleus to activate genes involved in osteoblast differentiation and bone matrix synthesis. Inhibition of this pathway by factors like sclerostin and DKK1 (Dickkopf-1) suppresses bone formation. Therapies targeting sclerostin aim to unleash this anabolic pathway [11].
  • Cytokines and Growth Factors: A host of other molecular mediators significantly influence bone remodeling. Pro-inflammatory cytokines such as Interleukin-1 (IL-1), Interleukin-6 (IL-6), and Tumor Necrosis Factor-alpha (TNF-α) are potent stimulators of bone resorption and are often elevated in inflammatory conditions associated with secondary osteoporosis [13]. Conversely, growth factors like Transforming Growth Factor-beta (TGF-β), Insulin-like Growth Factor-1 (IGF-1), and Bone Morphogenetic Proteins (BMPs) play crucial roles in promoting osteoblast differentiation and bone formation.
  • Epigenetic Factors: Emerging research suggests that epigenetic mechanisms, including DNA methylation, histone modification, and non-coding RNAs (e.g., microRNAs), can modulate gene expression involved in bone metabolism without altering the underlying DNA sequence. These epigenetic changes can be influenced by environmental factors and may represent another layer of complexity in osteoporosis pathogenesis [14].

3. Risk Factors for Osteoporosis

Osteoporosis is a multifactorial disease, resulting from a complex interplay of non-modifiable and modifiable risk factors. Understanding these factors is critical for effective risk assessment, prevention, and targeted intervention strategies.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

3.1 Non-Modifiable Risk Factors

These are inherent characteristics that cannot be changed but serve as crucial indicators of an individual’s predisposition to osteoporosis:

  • Age: Bone density naturally declines with advancing age, primarily after the age of 30-40 years. This age-related bone loss is multifactorial, involving a decrease in osteoblast proliferation and activity, an accumulation of senescent osteocytes, reduced physical activity, impaired nutrient absorption, and age-related changes in hormonal profiles. The elderly also experience a higher prevalence of falls, further increasing fracture risk [15].
  • Sex: Women are at a substantially higher lifetime risk of developing osteoporosis compared to men. This disparity is attributed to several factors: women generally achieve a lower peak bone mass than men; the accelerated bone loss associated with menopause due to estrogen deficiency is a major contributor; and women typically have a longer life expectancy, increasing their exposure to age-related bone loss [6].
  • Genetics and Family History: A strong family history of osteoporosis, particularly a maternal history of hip fracture, significantly elevates an individual’s risk. This indicates a genetic predisposition to lower peak bone mass or a higher rate of bone loss. Heritability of BMD is substantial, with multiple genes contributing to this complex trait [8].
  • Ethnicity: Ethnic background influences osteoporosis risk. Individuals of Caucasian and Asian descent generally have a higher incidence of osteoporosis and fractures compared to African Americans, who tend to achieve higher peak bone mass and experience slower rates of age-related bone loss. This difference is thought to be related to variations in bone geometry, body composition, and vitamin D metabolism across different ethnic groups [16].
  • Prior Fragility Fracture: A history of a fragility fracture (a fracture occurring from a fall from standing height or less, excluding severe trauma) is one of the strongest predictors of future fractures, irrespective of BMD. This indicates compromised bone strength and underlying osteoporosis [17].
  • Small Body Frame/Low Body Weight: Individuals with a naturally slender build or low body weight (Body Mass Index < 18.5 kg/m²) tend to have lower peak bone mass and reduced mechanical loading on bones, increasing their susceptibility to osteoporosis.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

3.2 Modifiable Risk Factors

These factors can be influenced through lifestyle modifications, medical interventions, or behavioral changes, offering opportunities for prevention and management:

  • Dietary Factors: Chronic insufficient intake of essential bone-building nutrients critically impairs bone health.
    • Calcium Deficiency: Calcium is the primary mineral component of bone. Inadequate dietary calcium intake forces the body to draw calcium from bones to maintain serum calcium levels, leading to progressive bone demineralization [18].
    • Vitamin D Deficiency: Vitamin D is crucial for efficient intestinal absorption of calcium and phosphorus, and for maintaining optimal bone mineralization. Chronic deficiency results in impaired calcium absorption, secondary hyperparathyroidism, and increased bone turnover [19].
    • Other Nutritional Deficiencies: Inadequate protein intake can impair bone matrix synthesis. Deficiencies in magnesium, phosphorus, potassium, and vitamins K and B12 also play roles in bone health, albeit less direct than calcium and vitamin D.
    • Malabsorption Syndromes: Conditions like celiac disease, inflammatory bowel disease (IBD), bariatric surgery, or chronic liver disease can lead to malabsorption of fat-soluble vitamins (including D and K) and calcium, contributing to bone loss.
  • Physical Activity: A sedentary lifestyle significantly contributes to bone loss. Bones respond to mechanical stress by becoming stronger; conversely, lack of weight-bearing and muscle-strengthening activities leads to decreased bone density and muscle atrophy, increasing fall risk [20].
  • Lifestyle Choices: Harmful habits exert detrimental effects on bone health.
    • Smoking: Both active and passive smoking are associated with lower BMD and increased fracture risk. Nicotine, cadmium, and other toxins in tobacco smoke directly inhibit osteoblast activity, promote osteoclast formation, alter hormone metabolism, and impair calcium absorption [21].
    • Excessive Alcohol Consumption: Chronic heavy alcohol intake (typically >3 units/day) is linked to reduced bone density and increased fracture risk. Alcohol can directly suppress osteoblast function, impair calcium and vitamin D metabolism, affect hormone levels, and increase the likelihood of falls [22].
    • Excessive Caffeine Intake: While moderate caffeine intake is generally not detrimental, very high consumption, particularly when accompanied by low calcium intake, may slightly increase bone loss.
  • Medical Conditions (Secondary Osteoporosis): Numerous chronic diseases can significantly impact bone metabolism.
    • Endocrine Disorders: Hyperthyroidism (accelerated bone turnover), hyperparathyroidism (excessive PTH leading to bone resorption), Cushing’s syndrome (excess glucocorticoids), hypogonadism (low sex hormones in both men and women), diabetes mellitus (especially type 1) [23].
    • Gastrointestinal Disorders: Chronic liver disease (impaired vitamin D metabolism, altered calcium absorption), celiac disease, inflammatory bowel disease, malabsorption syndromes.
    • Hematologic Disorders: Multiple myeloma, leukemia, lymphoma (direct bone destruction or cytokine effects).
    • Rheumatologic Conditions: Rheumatoid arthritis, ankylosing spondylitis, systemic lupus erythematosus (chronic inflammation, corticosteroid use, immobloization) [24].
    • Chronic Kidney Disease (CKD): Renal osteodystrophy, characterized by complex bone abnormalities due to altered mineral and hormone metabolism (e.g., hyperparathyroidism, vitamin D deficiency, hyperphosphatemia), is a significant cause of bone fragility in CKD patients.
    • Organ Transplantation: Immunosuppressive medications (especially corticosteroids) and pre-existing bone disease contribute to rapid bone loss post-transplantation.
  • Medications: Long-term use of various pharmacological agents can profoundly affect bone density and quality.
    • Glucocorticoids: The most common medication-induced cause of osteoporosis. They directly inhibit osteoblast function, promote osteoclast activity, reduce intestinal calcium absorption, and increase renal calcium excretion. The risk is dose and duration-dependent [25].
    • Anticonvulsants: Certain antiepileptic drugs (e.g., phenytoin, phenobarbital, carbamazepine) accelerate vitamin D metabolism, leading to reduced calcium absorption.
    • Proton Pump Inhibitors (PPIs): Long-term use may impair calcium absorption by reducing gastric acidity, though the evidence is somewhat mixed and the effect is generally small [26].
    • Selective Serotonin Reuptake Inhibitors (SSRIs): Some studies suggest an association with reduced BMD and increased fracture risk, possibly via effects on serotonin signaling in bone [27].
    • Thiazolidinediones (TZDs): Used for type 2 diabetes, these drugs (e.g., rosiglitazone, pioglitone) promote adipogenesis over osteoblastogenesis in bone marrow, leading to reduced bone formation.
    • Heparin: Long-term use of unfractionated heparin can cause osteoporosis.
    • Gonadotropin-Releasing Hormone (GnRH) Agonists: Used in prostate cancer or endometriosis, these drugs induce hypogonadism, leading to rapid bone loss similar to menopause.

4. Clinical Manifestations and Diagnosis

Osteoporosis is often termed a ‘silent disease’ because bone loss typically occurs without symptoms. The first overt clinical manifestation is frequently a fragility fracture, which then unveils the underlying skeletal fragility. Early detection before a fracture occurs is paramount to preventing subsequent morbidity and mortality.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

4.1 Clinical Manifestations

  • Fractures: The hallmark and most devastating clinical manifestation of osteoporosis. These are typically low-trauma fractures, meaning they result from a fall from standing height or less, or even spontaneously, and are disproportionate to the force applied. The most common sites include:
    • Vertebral Fractures: Occur predominantly in the thoracic and lumbar spine. They can be symptomatic, causing acute or chronic back pain, or asymptomatic (up to two-thirds are silent). Over time, multiple vertebral fractures lead to height loss, progressive kyphosis (a stooped or ‘dowager’s hump’ posture), and reduced mobility. Severe kyphosis can impact respiratory and gastrointestinal function [28].
    • Hip Fractures: One of the most severe consequences of osteoporosis, leading to significant morbidity, loss of independence, and increased mortality. Approximately 20-30% of patients die within one year following a hip fracture, and many more lose the ability to live independently [29].
    • Wrist Fractures (Colles’ fracture): Often occur as an initial warning sign, especially in younger postmenopausal women, typically from a fall onto an outstretched hand.
    • Other Sites: Fractures can also occur in the humerus, pelvis, and ribs.
  • Back Pain: While vertebral fractures can be silent, acute vertebral compression fractures often present with sudden, severe, localized back pain that worsens with movement and subsides over several weeks to months. Chronic back pain may result from multiple microfractures, muscle fatigue due to altered posture, or spinal stenosis secondary to vertebral deformities.
  • Postural Changes and Loss of Height: Progressive vertebral compression fractures lead to a gradual loss of height (often >1 inch or 2.5 cm per decade) and the development of thoracic kyphosis (a ‘hunchback’ appearance), which can significantly affect body image, balance, and organ function.
  • Reduced Quality of Life: Beyond the physical pain and disability, osteoporosis and its associated fractures can lead to psychological distress, fear of falling, social isolation, and a significant reduction in overall quality of life.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

4.2 Diagnostic Methods

The accurate diagnosis of osteoporosis involves a combination of clinical risk factor assessment, bone mineral density measurement, and, where appropriate, biochemical markers.

  • Bone Mineral Density (BMD) Testing: Dual-Energy X-ray Absorptiometry (DXA):
    • Gold Standard: DXA is the current gold standard for diagnosing osteoporosis and assessing fracture risk. It is a non-invasive, low-radiation imaging technique that measures the areal BMD (g/cm²) of specific skeletal sites, typically the lumbar spine, femoral neck, and total hip [30].
    • T-score and Z-score: BMD results are reported as T-scores and Z-scores.
      • T-score: Compares an individual’s BMD to the average peak bone mass of a healthy young adult (20-29 years old) of the same sex. According to the World Health Organization (WHO) criteria, osteoporosis is defined by a T-score of -2.5 standard deviations (SD) or lower at any central site (spine, hip). Osteopenia (low bone mass) is defined by a T-score between -1.0 and -2.5 SD. A T-score of -1.0 SD or above is considered normal. The T-score is primarily used for postmenopausal women and men aged 50 and older [31].
      • Z-score: Compares an individual’s BMD to the average BMD of an age- and sex-matched population. A Z-score of -2.0 SD or lower is considered ‘below the expected range for age’ and warrants investigation for secondary causes of osteoporosis, particularly in premenopausal women, men younger than 50, and children [31].
    • Limitations: DXA is a 2D projection, so it cannot fully assess bone microarchitecture or distinguish cortical from trabecular bone. It can be confounded by degenerative changes (e.g., osteoarthritis, scoliosis) in the spine, which may artificially elevate BMD readings. Accessibility and cost can also be barriers in some regions.
    • Other BMD Techniques: Peripheral DXA (pDXA) can measure BMD at peripheral sites (wrist, heel) but is generally used for screening and not for definitive diagnosis or monitoring. Quantitative Computed Tomography (QCT) and high-resolution peripheral Quantitative Computed Tomography (HR-pQCT) offer 3D assessment of bone density and microarchitecture, providing more detailed information on bone quality, but are typically used in research settings due to higher cost and radiation exposure [32].
  • Fracture Risk Assessment Tool (FRAX):
    • Algorithm: FRAX is a computer-based algorithm developed by the WHO that estimates the 10-year probability of a major osteoporotic fracture (clinical spine, hip, forearm, or humerus) and hip fracture specifically. It integrates clinical risk factors (age, sex, BMI, prior fragility fracture, parental hip fracture, glucocorticoid use, secondary osteoporosis, current smoking, alcohol intake, rheumatoid arthritis) with or without femoral neck BMD [33].
    • Utility: FRAX helps clinicians make treatment decisions, especially for individuals with osteopenia or those who do not meet the DXA criteria for osteoporosis but have multiple risk factors. Treatment thresholds vary by country.
    • Limitations: FRAX does not account for dose-response relationships of some risk factors (e.g., cumulative glucocorticoid dose), does not consider falls risk directly, and is validated only for untreated individuals. It also does not include all known risk factors (e.g., vitamin D deficiency) and is less precise for younger individuals or those with certain secondary causes.
  • Biochemical Markers of Bone Turnover (BTMs):
    • Utility: BTMs are substances released during bone formation or resorption that can be measured in blood or urine. While not typically used for initial diagnosis of osteoporosis, they can provide additional information on bone metabolism, help monitor treatment effectiveness (especially for anabolic agents), assess medication adherence, and potentially predict fracture risk [34].
    • Formation Markers: Reflect osteoblast activity and include:
      • Procollagen Type I N-terminal Propeptide (P1NP): A highly sensitive and specific marker of bone formation.
      • Osteocalcin (OC): A protein synthesized by osteoblasts and incorporated into bone matrix.
    • Resorption Markers: Reflect osteoclast activity and include:
      • C-terminal Telopeptide of Type I Collagen (CTX): A fragment released during collagen breakdown, widely used.
      • N-terminal Telopeptide of Type I Collagen (NTX): Another collagen breakdown product.
    • Variability: BTMs exhibit significant diurnal and intra-individual variation, necessitating standardized collection protocols and careful interpretation.
  • Other Laboratory Tests: A comprehensive workup for osteoporosis, especially when secondary causes are suspected, often includes:
    • Serum calcium, phosphate, albumin, creatinine (to assess kidney function).
    • 25-hydroxyvitamin D [25(OH)D] levels (to assess vitamin D status).
    • Parathyroid hormone (PTH) levels (to rule out primary or secondary hyperparathyroidism).
    • Thyroid-stimulating hormone (TSH) (to rule out thyroid disorders).
    • Liver function tests, complete blood count, and inflammatory markers (e.g., ESR, CRP) as clinically indicated.
  • Imaging (X-rays): While DXA is for BMD, plain X-rays are crucial for confirming the presence of fractures, assessing their severity, and excluding other pathologies (e.g., tumors, infections) that might mimic osteoporotic fractures. MRI can be useful for distinguishing acute from old vertebral fractures or identifying bone marrow edema.

5. Treatment Strategies

The management of osteoporosis is a multi-pronged approach integrating lifestyle modifications, nutritional support, and pharmacological interventions, with the overarching goal of preventing fractures and preserving bone strength and quality of life.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

5.1 Non-Pharmacological Interventions

These foundational strategies are crucial for all individuals at risk for or diagnosed with osteoporosis, serving as primary prevention and adjuncts to pharmacological therapy.

  • Nutrition for Bone Health: A well-balanced diet rich in bone-supporting nutrients is fundamental.
    • Calcium: The recommended daily intake (RDI) for calcium typically ranges from 1000-1200 mg for adults, depending on age and sex [18]. Dietary sources are preferred over supplements whenever possible. Excellent sources include dairy products (milk, yogurt, cheese), fortified plant-based milks, dark leafy green vegetables (kale, broccoli), fortified cereals, and certain fish (sardines, salmon with bones). If dietary intake is insufficient, calcium supplements (calcium carbonate or citrate) may be necessary, usually limited to 500-600 mg per single dose for optimal absorption [35].
    • Vitamin D: RDI for vitamin D is typically 600-800 IU/day for adults, with some guidelines suggesting higher doses for older adults (800-1000 IU/day) [19]. Many individuals, especially older adults, have insufficient vitamin D levels and require supplementation. Optimal serum 25(OH)D levels are generally considered to be between 30-50 ng/mL (75-125 nmol/L). Sources include fatty fish (salmon, mackerel, tuna), fortified foods, and sunlight exposure. Vitamin D3 (cholecalciferol) is generally preferred over D2 (ergocalciferol) due to its greater efficacy in raising serum 25(OH)D levels.
    • Protein: Adequate protein intake is essential for bone matrix formation and overall muscle health, which supports balance and reduces fall risk. Older adults, in particular, may benefit from higher protein intake to mitigate sarcopenia [36].
    • Other Nutrients: Magnesium, vitamin K (K1 and K2), phosphorus, and zinc also contribute to bone health and should be obtained through a varied diet.
  • Exercise and Physical Activity: Regular weight-bearing and muscle-strengthening exercises are critical for maintaining bone density and improving balance and coordination, thereby reducing the risk of falls and fractures [20].
    • Weight-Bearing Exercises: Activities that involve working against gravity, such as walking, jogging, stair climbing, dancing, and high-impact aerobics (if appropriate for the individual’s fracture risk), stimulate osteoblasts and promote bone formation.
    • Resistance Exercises: Strength training with weights or resistance bands helps build muscle mass and strengthen bones. Targeting major muscle groups multiple times a week is recommended.
    • Balance and Posture Training: Exercises like Tai Chi, yoga, and specific balance drills can significantly improve stability and reduce fall risk, especially important for older adults or those with previous fractures [37].
    • Caution: Exercise programs should be tailored to individual fitness levels, comorbidities, and fracture risk. High-impact exercises may be contraindicated in individuals with severe osteoporosis or spinal fractures.
  • Fall Prevention Strategies: A significant proportion of osteoporotic fractures are due to falls. Comprehensive fall prevention is crucial.
    • Environmental Modifications: Removing tripping hazards (loose rugs, clutter), improving lighting, installing grab bars in bathrooms, and securing handrails can reduce fall risk at home.
    • Vision Correction: Regular eye examinations and updated prescriptions can improve vision and reduce falls.
    • Medication Review: Certain medications (e.g., sedatives, hypnotics, antihypertensives) can cause dizziness or drowsiness, increasing fall risk. Regular review and optimization of medication regimens by a healthcare professional are important.
    • Assistive Devices: Use of walkers or canes if needed for stability.
    • Footwear: Wearing supportive, non-slip footwear.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

5.2 Pharmacological Treatments

Pharmacological therapies are typically recommended for individuals diagnosed with osteoporosis (T-score ≤ -2.5), those with osteopenia and a high FRAX score, or those with a history of fragility fractures. These agents work by inhibiting bone resorption (anti-resorptive) or stimulating bone formation (anabolic).

  • Anti-Resorptive Agents:
    • Bisphosphonates: These are the most commonly prescribed drugs for osteoporosis due to their efficacy in reducing fracture risk at multiple sites (vertebral, hip, non-vertebral). They work by binding to bone mineral at sites of active remodeling and are then internalized by osteoclasts, leading to osteoclast apoptosis and inhibition of bone resorption. Available agents include:
      • Alendronate (oral, daily or weekly): Highly effective in increasing BMD and reducing fracture risk.
      • Risedronate (oral, daily, weekly, or monthly): Similar efficacy to alendronate.
      • Ibandronate (oral, monthly, or intravenous, quarterly): Proven to reduce vertebral fracture risk, but evidence for hip fracture reduction is less robust.
      • Zoledronic Acid (intravenous, annual): Potent bisphosphonate, often used for patients with adherence issues to oral medications or those who cannot tolerate oral forms. Highly effective at reducing all major fracture types [38].
      • Side Effects and Considerations: Common side effects include gastrointestinal upset (oral forms). Rare but serious side effects include osteonecrosis of the jaw (ONJ) and atypical femoral fractures (AFF), which are extremely rare but important considerations, particularly with prolonged use [39]. A ‘drug holiday’ may be considered after 3-5 years of therapy for some patients, depending on their risk profile.
    • Selective Estrogen Receptor Modulators (SERMs): These agents act as estrogen agonists in some tissues (bone) and antagonists in others (breast, uterus).
      • Raloxifene: Mimics estrogen’s protective effects on bone, decreasing bone resorption and reducing vertebral fracture risk in postmenopausal women. It also offers the benefit of reducing the risk of invasive breast cancer. However, it does not reduce hip fracture risk and is associated with an increased risk of venous thromboembolism (VTE) and hot flashes [40].
    • Denosumab: A fully human monoclonal antibody that targets and inhibits RANKL, thereby preventing its interaction with RANK on osteoclast precursors and mature osteoclasts. This leads to a rapid and profound suppression of osteoclast activity and bone resorption. Administered as a subcutaneous injection every 6 months, it significantly reduces vertebral, hip, and non-vertebral fracture risk [41].
      • Side Effects and Considerations: Generally well-tolerated. Risks include hypocalcemia (especially in patients with renal impairment or vitamin D deficiency), ONJ, and AFF (rare). A critical consideration is the potential for rapid bone loss and increased vertebral fracture risk upon discontinuation, necessitating a plan for transition to another anti-osteoporotic agent if stopping treatment.
  • Anabolic Agents (Bone-Forming Agents):
    • Parathyroid Hormone (PTH) Analogs: These agents stimulate new bone formation by transiently activating osteoblasts when administered intermittently. They are typically reserved for patients with severe osteoporosis, very high fracture risk, or those who have failed or are intolerant to anti-resorptive therapies.
      • Teriparatide: A recombinant human PTH (1-34) fragment, administered daily via subcutaneous injection for a maximum of 2 years. It significantly increases BMD and reduces vertebral and non-vertebral fracture risk [42].
      • Abaloparatide: A synthetic analog of human PTH-related protein (PTHrP), also administered daily subcutaneously for a maximum of 2 years. It has been shown to reduce vertebral, non-vertebral, and major osteoporotic fracture risk, potentially with less hypercalcemia than teriparatide [43].
      • Side Effects and Considerations: Common side effects include hypercalcemia, dizziness, and nausea. Both are associated with a theoretical risk of osteosarcoma (based on rat studies), hence the 2-year treatment limit.
    • Romosozumab: A monoclonal antibody that inhibits sclerostin, an osteocyte-derived protein that negatively regulates bone formation. By blocking sclerostin, romosozumab promotes osteoblast activity (bone formation) and, to a lesser extent, inhibits osteoclast activity (bone resorption), thus exhibiting a dual effect. Administered monthly as a subcutaneous injection for 12 months, it rapidly and significantly increases BMD and reduces vertebral, hip, and non-vertebral fracture risk, often used for severe osteoporosis [44].
      • Side Effects and Considerations: Concerns regarding cardiovascular safety (increased risk of major adverse cardiovascular events like myocardial infarction and stroke) have been raised, leading to a black box warning. Like other anabolic agents, it is typically followed by an anti-resorptive agent to maintain the gained BMD.
  • Hormone Replacement Therapy (HRT): Estrogen therapy (with progestin for women with a uterus) can prevent bone loss and reduce fracture risk in postmenopausal women. However, due to concerns regarding cardiovascular events, breast cancer, and stroke raised by the Women’s Health Initiative (WHI) study, HRT is generally no longer considered a first-line treatment specifically for osteoporosis in postmenopausal women. It may be considered for symptom management of menopause in younger postmenopausal women, with bone benefits being an additional advantage [45].
  • Calcitonin: A hormone that inhibits osteoclast activity. While approved for osteoporosis, its fracture efficacy is modest (only for vertebral fractures) and its use is limited due to emerging concerns about a potential association with malignancy and the availability of more effective agents.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

5.3 Emerging and Investigational Therapies

The field of osteoporosis research continues to evolve, with novel therapeutic targets and modalities under investigation:

  • Cathepsin K Inhibitors: Cathepsin K is a protease highly expressed by osteoclasts and crucial for bone matrix degradation. Inhibitors of cathepsin K aimed to reduce bone resorption without affecting osteoclast viability. While some early candidates like Odanacatib showed promise in clinical trials by reducing fracture risk, development was halted due to safety concerns (e.g., stroke risk, skin reactions), highlighting the challenges of targeting specific pathways [46].
  • Activin A Pathway Inhibitors: Activin A is a member of the TGF-β superfamily that negatively regulates bone formation by inhibiting osteoblast differentiation. Inhibitors targeting this pathway, such as activin A antibodies, are being explored for their potential anabolic effects on bone [47].
  • Sustained-Release Parathyroid Hormone: Efforts are underway to develop sustained-release formulations of PTH or its analogs to reduce the frequency of injections and improve patient adherence.
  • Gene Therapy: Still in early experimental stages, gene therapy approaches aim to introduce genes that enhance bone formation (e.g., LRP5, BMPs) or inhibit bone resorption (e.g., OPG) into skeletal cells. This holds long-term promise for refractory cases or genetic forms of osteoporosis but faces significant challenges regarding delivery, specificity, and safety [48].
  • Targeting Senescence: Cellular senescence (the irreversible arrest of cell division) in osteocytes and other bone cells contributes to age-related bone loss. Senolytics, drugs that selectively remove senescent cells, are being investigated for their potential to rejuvenate the bone microenvironment and improve bone health [49].
  • Osteoimmunology-Based Therapies: A growing understanding of the intricate crosstalk between the immune system and bone (osteoimmunology) is opening new avenues for therapeutic intervention, targeting inflammatory pathways that influence bone remodeling.

6. Challenges and Future Directions

Despite significant advancements in understanding and managing osteoporosis, several challenges persist, necessitating ongoing research and innovative solutions to optimize patient care and public health outcomes.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

6.1 Diagnostic Challenges

  • Underdiagnosis and Undertreatment: A substantial proportion of individuals at high risk for fracture, or even those who have already experienced a fragility fracture, remain undiagnosed and untreated. This ‘care gap’ is multifactorial, including a lack of awareness among both the public and healthcare providers, limited access to DXA scanning (especially in rural or low-resource settings) due to high costs and infrastructure requirements, and a failure to act on fracture events as a sentinel for osteoporosis [50].
  • Accessibility and Cost of DXA: The capital cost of DXA machines, maintenance, and the need for trained personnel can limit their widespread availability, leading to geographical disparities in access to diagnostic services.
  • Beyond BMD: While DXA is the gold standard, it provides only a 2D measure of bone density and does not fully capture bone quality, microarchitecture, or localized bone strength. There is a pressing need for more comprehensive, accessible, and potentially cheaper diagnostic tools that can assess these additional dimensions of bone strength. Emerging technologies, such as advanced imaging techniques (HR-pQCT, quantitative ultrasound) and finite element analysis derived from CT scans, offer promise for a more detailed assessment of bone microarchitecture and biomechanical properties, though they are mostly confined to research at present [32].
  • Opportunistic Screening: Efforts are ongoing to leverage existing imaging studies, such as abdominal CT scans performed for other indications, or dental X-rays, to opportunistically assess vertebral BMD or cortical thickness, potentially identifying at-risk individuals who might otherwise be missed [51]. The integration of artificial intelligence (AI) and machine learning algorithms to automate and enhance bone assessment from these routine images is an exciting future direction.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

6.2 Treatment Adherence and Long-Term Management

  • Suboptimal Adherence: Adherence to pharmacological treatments for osteoporosis is often suboptimal, particularly for oral daily or weekly bisphosphonates, with discontinuation rates as high as 50% within the first year [52]. Reasons for non-adherence are diverse and include: a lack of immediate symptomatic relief (the ‘silent’ nature of the disease), perceived or actual side effects (e.g., gastrointestinal upset, fear of rare but serious adverse events like ONJ or AFF), complex dosing regimens, polypharmacy, and insufficient patient education regarding the chronic nature of the disease and the importance of long-term therapy.
  • Strategies to Improve Adherence: Improving patient adherence is critical for realizing the full benefits of treatment and requires a multi-faceted approach:
    • Patient Education: Comprehensive counseling on the disease, treatment benefits, potential side effects, and the importance of adherence.
    • Simplified Dosing Regimens: Less frequent dosing (e.g., weekly, monthly oral, or quarterly/annual intravenous/subcutaneous injections) can improve convenience and adherence.
    • Shared Decision-Making: Involving patients in treatment choices, considering their preferences and concerns.
    • Monitoring and Follow-up: Regular follow-up appointments, bone turnover marker monitoring, and patient support programs (e.g., nurse-led clinics, reminder systems) can reinforce adherence.
    • Sequential and Combination Therapy: Optimizing the sequence of different drug classes (e.g., anabolic followed by anti-resorptive) or exploring combination therapies for very high-risk individuals is an active area of research to maximize BMD gains and fracture prevention while managing side effects effectively [53].

Many thanks to our sponsor Esdebe who helped us prepare this research report.

6.3 Personalized Medicine

The future of osteoporosis management is increasingly moving towards a personalized approach, tailoring prevention and treatment strategies to individual patient profiles.

  • Pharmacogenomics: Research into pharmacogenomics aims to identify genetic markers that predict an individual’s response to specific osteoporosis medications or their susceptibility to adverse drug reactions. This could enable clinicians to select the most effective and safest treatment for each patient [54].
  • Advanced Risk Stratification: Beyond FRAX, integrating a wider array of risk factors, including genetic predispositions, detailed bone microarchitecture parameters, and dynamic biomarkers, into more sophisticated algorithms could provide highly individualized fracture risk predictions. AI and machine learning could play a significant role in developing these advanced models.
  • Biomarker-Guided Therapy: Utilizing bone turnover markers more effectively to monitor treatment response, predict fracture risk, and guide adjustments in therapy could lead to more personalized and dynamic management strategies [34].
  • Integration of Multi-Omics Data: Combining genomic, proteomic, metabolomic, and microbiomic data could provide a holistic understanding of an individual’s unique biological pathways influencing bone health, potentially revealing novel therapeutic targets and personalized nutritional interventions.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

6.4 Global Health Disparities and Public Health Initiatives

  • Addressing Disparities: Significant disparities exist in osteoporosis care globally, influenced by socioeconomic status, access to healthcare, cultural beliefs, and national healthcare policies. Efforts are needed to ensure equitable access to diagnostic tools and treatments, particularly in low- and middle-income countries where the burden of fractures is growing rapidly with aging populations.
  • Public Health Campaigns: Large-scale public health campaigns are crucial for raising awareness about osteoporosis, promoting bone-healthy lifestyles from childhood, and encouraging early screening and intervention. Implementing fracture liaison services (FLS) where patients presenting with fragility fractures are systematically identified, investigated, and treated for underlying osteoporosis, has proven highly effective in reducing secondary fractures and improving care coordination [55].

7. Conclusion

Osteoporosis represents a substantial and growing global health challenge, characterized by its insidious nature and the devastating consequences of fragility fractures. A profound and nuanced understanding of its complex pathophysiology, encompassing the intricate balance of bone remodeling and the molecular pathways that dictate it, is paramount. Furthermore, identifying and mitigating the diverse array of non-modifiable and modifiable risk factors forms the bedrock of effective prevention.

Effective management necessitates a comprehensive, multidisciplinary approach that integrates robust lifestyle modifications, meticulous nutritional planning, and appropriately timed pharmacological interventions. This holistic strategy is critical in mitigating the profound impact of this disease on individuals’ quality of life, independence, and overall survival. Despite significant progress, challenges persist in terms of widespread early diagnosis, consistent patient adherence to long-term therapies, and equitable access to care.

However, the horizon of osteoporosis research is bright, with ongoing and accelerated investigations into novel therapeutic targets, advanced diagnostic modalities, and personalized medicine approaches. The promise of emerging therapies, such as sclerostin inhibitors and potentially gene-based interventions, coupled with the increasing integration of sophisticated data analytics and artificial intelligence, holds immense potential for revolutionizing patient outcomes. By continuing to foster interdisciplinary collaboration and investing in innovative research, the global health community can strive towards a future where osteoporosis is effectively prevented, accurately diagnosed, and comprehensively managed, ultimately improving the bone health and quality of life for millions worldwide.

References

  1. Johnell, O., & Kanis, J. A. (2006). An estimate of the global prevalence and disability associated with osteoporosis. Osteoporosis International, 17(12), 1726-1733.
  2. Burge, R., Dawson-Hughes, B., Solomon, D. H., Wong, J. B., King, A., & Tosteson, A. (2007). Incidence and economic burden of osteoporosis-related fractures in the United States, 2005-2025. Journal of Bone and Mineral Research, 22(3), 465-475.
  3. Rachner, T. D., Khosla, S., & Hofbauer, L. C. (2011). Osteoporosis: Now and the future. The Lancet, 377(9773), 1276-1287.
  4. Hofbauer, L. C., Schoppet, P., & Rauner, M. (2004). The OPG-RANKL-RANK system in bone and vascular biology. Journal of Bone and Mineral Research, 19(8), 1256-1262.
  5. Riggs, B. L., & Khosla, S. (2007). Pathogenesis of senile osteoporosis: A perspective. Journal of Bone and Mineral Research, 22(8), 1149-1151.
  6. Riggs, B. L., Khosla, S., & Melton, L. J. (2002). Sex differences in bone density in the adult. Journal of Bone and Mineral Research, 17(8), 1485-1490.
  7. Bellido, T., Saini, V., & Pajevic, P. D. (2013). Osteocyte signaling: The mechanical control of bone remodeling and beyond. Annual Review of Physiology, 75, 59-81.
  8. Ralston, S. H., & Uitterlinden, A. G. (2010). Genetics of osteoporosis. Endocrine Reviews, 31(5), 629-662.
  9. Morrison, N. A., Qi, J. C., Tokita, A., Kelly, P. J., Crofts, L., Nguyen, T. V., … & Eisman, J. A. (1994). Prediction of bone density by vitamin D receptor allelic variants. Nature, 367(6460), 284-287.
  10. Grant, S. F. A., Reid, D. M., Blake, G., Herd, R., Fogelman, S., & Ralston, S. H. (1996). Reduced bone density and osteoporosis associated with a polymorphism of the type I collagen gene. Nature Genetics, 14(2), 203-205.
  11. Krishnan, V., Bryant, H. U., & Macdougald, O. A. (2006). Regulation of bone mass by Wnt signalling. Journal of Clinical Investigation, 116(5), 1202-1209.
  12. Moester, M. J., Papapoulos, S. E., & Löwik, C. W. (2010). Sclerostin: A novel target for the treatment of osteoporosis. Expert Opinion on Therapeutic Targets, 14(11), 1187-1196.
  13. Lorenzo, J. A., Horowitz, M. C., & Choi, Y. (2008). Osteoimmunology: The growing role of immunology in the study of bone. Immunological Reviews, 223(1), 1-13.
  14. Xiao, S., & Li, R. (2019). The Role of Epigenetics in Osteoporosis. Frontiers in Endocrinology, 10, 163.
  15. Reid, I. R. (2015). Age-related bone loss: Mechanisms and management. Clinical Endocrinology, 83(2), 170-181.
  16. Looker, A. C., Melton, L. J., 3rd, & Borrud, M. J. (2008). Racial/ethnic differences in hip fracture incidence and osteoporosis prevalence in older US adults. Osteoporosis International, 19(9), 1289-1296.
  17. Klotzbuecher, E. T., Ross, P. D., Landsman, P. A., Abbott, T. A., & Berger, M. (2000). Patients with prior fractures have an increased risk of future fractures: A meta-analysis. Journal of Bone and Mineral Research, 15(4), 721-729.
  18. NIH Osteoporosis and Related Bone Diseases National Resource Center. (2022). Calcium and Vitamin D: Important for Bone Health. Retrieved from https://www.bones.nih.gov/health-info/bone/bone-health/nutrition/calcium-and-vitamin-d
  19. Holick, M. F. (2007). Vitamin D deficiency. New England Journal of Medicine, 357(3), 266-281.
  20. Kemmler, W., & Stengel, S. v. (2017). Osteoporosis and exercise—Effects on bone mineral density, muscle strength and balance. Osteoporosis International, 28(6), 1787-1798.
  21. Law, M. R., & Hackshaw, A. K. (1997). A meta-analysis of cigarette smoking, bone mineral density and risk of hip fracture: Recognition of a major public health problem. BMJ, 315(7112), 841-846.
  22. Sampson, H. W. (1998). Alcohol and osteoporosis. Journal of Bone and Mineral Metabolism, 16(2), 111-125.
  23. Adami, S., & Zamberlan, N. (2002). Secondary osteoporosis. Best Practice & Research Clinical Rheumatology, 16(4), 627-642.
  24. Haugeberg, G., & van der Heijde, D. (2011). Bone loss in rheumatoid arthritis. Current Opinion in Rheumatology, 23(4), 382-387.
  25. Van Staa, T. P., Leufkens, H. G., Abenhaim, L., Zhang, B., & Cooper, C. (2000). Use of oral corticosteroids and risk of fractures. Journal of Bone and Mineral Research, 15(5), 993-1002.
  26. Park, M., & Kim, M. (2018). The effect of proton pump inhibitors on bone mineral density: a meta-analysis. Osteoporosis International, 29(1), 185-195.
  27. Caleyachetty, R., & et al. (2018). Association of selective serotonin reuptake inhibitors with the risk of fracture: A systematic review and meta-analysis. Osteoporosis International, 29(7), 1499-1509.
  28. Cooper, C., Atkinson, E. J., O’Fallon, W. M., & Melton, L. J. (1992). Incidence of clinically diagnosed vertebral fractures: A population-based study in Rochester, Minnesota, 1985-1989. Journal of Bone and Mineral Research, 7(2), 221-227.
  29. Brauer, C. A., Coca-Perraillon, M., Cutler, D. M., & Rosen, A. B. (2009). Incidence and mortality of hip fractures in the United States. JAMA, 302(14), 1573-1579.
  30. Blake, G. M., & Fogelman, I. (2007). DXA in the diagnosis and management of osteoporosis. Osteoporosis International, 18(11), 1391-1407.
  31. WHO Scientific Group on the Assessment of Osteoporosis at the Primary Health Care Level. (2004). Assessment of osteoporosis at the primary health care level. World Health Organization.
  32. Engelke, K., & Adams, J. E. (2007). Quantitative computed tomography and high-resolution peripheral quantitative computed tomography. Current Osteoporosis Reports, 5(2), 86-97.
  33. Kanis, J. A. (2008). Assessment of fracture risk and its application to screening for postmenopausal osteoporosis: Synopsis of a WHO report. Osteoporosis International, 19(5), 585-592.
  34. Vasikaran, S. D., Eastell, R., & et al. (2011). International Federation of Clinical Chemistry and Laboratory Medicine (IFCC) and International Osteoporosis Foundation (IOF) Joint Committee for Bone Metabolism. Clinical Chemistry, 57(1), 118-124.
  35. Weaver, C. M., & et al. (2016). Calcium and vitamin D: dietary reference intakes for bone health and osteoporosis. Osteoporosis International, 27(1), 1-4.
  36. Houston, D. K., Nicklas, B. J., & et al. (2011). Dietary protein intake and bone mineral density in older adults: the Health, Aging, and Body Composition Study. The American Journal of Clinical Nutrition, 93(6), 1314-1320.
  37. Sherrington, C., & et al. (2008). Exercise to prevent falls in older adults: An updated meta-analysis and economic evaluation. The Lancet, 371(9629), 1729-1736.
  38. Black, D. M., & et al. (2007). Once-yearly zoledronic acid for treatment of postmenopausal osteoporosis. New England Journal of Medicine, 356(18), 1809-1822.
  39. Shane, E., & et al. (2014). Atypical Femur Fractures and Bisphosphonate Use: A Systematic Review. Journal of Bone and Mineral Research, 29(1), 22-44.
  40. Ettinger, B., & et al. (1998). Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. JAMA, 280(7), 603-607.
  41. Cummings, S. R., & et al. (2009). Denosumab for prevention of fractures in postmenopausal women with osteoporosis. New England Journal of Medicine, 361(8), 756-765.
  42. Neer, R. M., & et al. (2001). Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis. New England Journal of Medicine, 344(19), 1434-1441.
  43. Miller, P. D., & et al. (2016). Abaloparatide versus teriparatide in postmenopausal women with osteoporosis: a 24-week randomised, controlled clinical trial. The Lancet, 387(10019), 747-755.
  44. Cosman, F., & et al. (2017). Romosozumab or Alendronate for Fracture Prevention in Women with Osteoporosis. New England Journal of Medicine, 377(15), 1417-1427.
  45. Rossouw, J. E., & et al. (2002). Risks and benefits of estrogen plus progestin in healthy postmenopausal women: Principal results From the Women’s Health Initiative randomized controlled trial. JAMA, 288(3), 321-333.
  46. McClung, M. R., & et al. (2017). Odanacatib in the treatment of postmenopausal osteoporosis: a randomized, double-blind, placebo-controlled trial. The Lancet Diabetes & Endocrinology, 5(7), 541-552.
  47. Grgurevic, L., & et al. (2018). Bone morphogenetic proteins, activins and their antagonists in bone physiology and pathophysiology. Cytokine & Growth Factor Reviews, 39, 13-26.
  48. Shen, P., & et al. (2020). Advances in Gene Therapy for Osteoporosis. Frontiers in Pharmacology, 11, 570997.
  49. Farr, J. N., & et al. (2017). Targeting cellular senescence in bone and muscle. Molecular and Cellular Endocrinology, 455, 147-152.
  50. Harvey, N., & et al. (2017). The Anabolic/Catabolic Challenge of Osteoporosis Treatment. Journal of Bone and Mineral Research, 32(11), 2244-2248.
  51. Pickhardt, P. J., & et al. (2019). Opportunistic Screening for Osteoporosis Using Abdominal CT Scans. Radiology, 290(3), 643-652.
  52. Compston, J., & et al. (2017). Osteoporosis and fracture prevention. The Lancet, 390(10103), 1537-1549.
  53. Tsai, J. N., & et al. (2013). Anabolic and anti-catabolic treatments for osteoporosis. Bone, 53(1), 47-51.
  54. Ralston, S. H., & Uitterlinden, A. G. (2010). Genetics of osteoporosis. Endocrine Reviews, 31(5), 629-662.
  55. Akesson, K., & et al. (2013). Capture the Fracture: A global campaign to break the cycle of fragility fractures. Osteoporosis International, 24(7), 1779-1785.

Be the first to comment

Leave a Reply

Your email address will not be published.


*