Osteosarcoma is a primary malignant neoplasm of bone that originates from osteoblasts, the cells responsible for bone formation, and represents the most common type of primary bone cancer, particularly in children and adolescents.[1] It typically arises in the metaphysis of long bones, such as the distal femur, proximal tibia, or proximal humerus, and is characterized by the production of malignant osteoid tissue by tumor cells.[2] Osteosarcoma has a bimodal age distribution, with peaks in adolescence (10-30 years) and older adulthood (>60 years), with a higher incidence in males than females, and accounts for approximately 35% of all primary bone sarcomas (as of 2022).[3][4]The etiology of osteosarcoma is multifactorial and not fully elucidated, but it is associated with disruptions in bone growth and development, often occurring during periods of rapid skeletal expansion in puberty.[2] Key risk factors include genetic predispositions, such as germline mutations in the TP53 gene seen in Li-Fraumeni syndrome or retinoblastoma susceptibility gene alterations, as well as prior exposure to ionizing radiation or alkylating chemotherapy agents used in treating other cancers.[5] Other contributing elements encompass tall stature, high birth weight, and rare hereditary conditions like Rothmund-Thomson syndrome, though the majority of cases are sporadic without identifiable environmental triggers.[6] Environmental exposures, including certain heavy metals or fluoridated water, have been investigated but lack conclusive causal links.[7]Common presenting symptoms of osteosarcoma include persistent bone or jointpain that may initially be attributed to growing pains or injury, swelling or a palpable mass near the affected site, and reduced range of motion in the involved limb.[8] In advanced cases, pathological fractures can occur due to bone weakening, and systemic signs such as fatigue or weight loss may emerge if the tumor metastasizes, most frequently to the lungs.[9]Diagnosis typically involves imaging modalities like X-rays, MRI, or CT scans to identify characteristic features such as a "sunburst" periosteal reaction, followed by biopsy confirmation of osteoid production by atypical cells.[2]Treatment for osteosarcoma is multimodal and aggressive, centered on neoadjuvant chemotherapy to shrink the tumor and eradicate micrometastases, followed by wide surgical resection—often limb-sparing procedures with endoprosthetic reconstruction rather than amputation— and adjuvant chemotherapy.[10] Radiation therapy is reserved for cases where surgery is infeasible or for palliation.[9] Prognosis varies by stage at diagnosis; for localized disease, the 5-year overall survival rate is approximately 60-75% (as of 2025), but drops to around 20-30% for metastatic cases at presentation, with ongoing research focusing on targeted therapies such as IGF-1R inhibitors to improve outcomes.[11]
Clinical Presentation
Signs and Symptoms
Osteosarcoma typically presents with persistent and worsening bone pain in the affected area, which is often the initial and most common symptom. This pain is frequently described as deep, aching, and nocturnal, exacerbated by physical activity, and may initially be mistaken for growing pains or sports-related injuries in adolescents. In many cases, the pain precedes other signs by weeks to months and does not respond well to standard analgesics.[12][8][13]Localized swelling, tenderness, or a palpable mass often develops near the tumor site, contributing to visible or palpable changes in the limb. Functional impairments such as limping, reduced range of motion in the joint, or even pathological fractures can occur due to bone weakening caused by the tumor's destructive growth. These local signs are particularly prominent in the extremities, with the most common sites being the distal femur (approximately 40-50% of cases), proximal tibia (20-30%), and proximal humerus (10-15%).[12][14][15]In advanced or metastatic disease, systemic symptoms may emerge, including fatigue, unintentional weight loss, and low-grade fever, often resulting from the body's response to widespread tumor burden or distant spread. The clinical presentation can vary by age: in pediatric and adolescent patients, who account for the majority of cases, symptoms are commonly attributed to rapid growth or trauma in the metaphyseal regions of long bones; in contrast, adult cases more frequently involve the axial skeleton, such as the pelvis or spine, and may present with more subtle or delayed symptoms due to lower bone turnover rates.[8][16][2]
Epidemiology
Osteosarcoma is the most common primary malignant bone tumor, accounting for approximately 20% of all primary bone sarcomas. Worldwide, its incidence is estimated at 3 to 5 cases per million individuals annually, with an age-standardized rate of about 3.4 cases per million people per year. In the United States, approximately 1,000 new cases are diagnosed each year, representing less than 1% of all new cancer diagnoses.[2][2][17]The disease exhibits a bimodal age distribution, with the primary peak occurring during adolescence (ages 10-20 years), accounting for roughly half of all cases, and a secondary peak in older adults over 60 years, comprising about 20% of cases. Incidence is slightly higher in males, with a male-to-female ratio of approximately 1.5:1, and is associated with taller stature, particularly among individuals experiencing rapid growth during pubertal spurts.[15][17]Geographically, incidence shows limited variation globally, with age-standardized rates ranging from 2 cases per million in Southern Asia to 4.2 per million in sub-Saharan Africa, though underreporting in low-resource regions may affect observed patterns. Rates in developed countries like North America and Europe are around 3 cases per million, potentially due to better diagnostic access. Historically, incidence has remained stable over decades, but survival rates have improved significantly—from about 20% five-year survival in the mid-20th century to 60-70% today—owing to advances in multi-agent chemotherapy and surgical techniques.[18][19][20]Socioeconomic factors influence outcomes, with lower socioeconomic status linked to delayed diagnosis and reduced survival, often due to barriers in access to specialized care and higher rates of metastatic presentation at diagnosis. For instance, patients in areas of high unemployment or language isolation face increased odds of advanced disease.[21][22]
Etiology
Risk Factors
Exposure to ionizing radiation is a well-established risk factor for developing osteosarcoma, particularly following therapeutic radiotherapy for other cancers, where the risk increases significantly (relative risks reported from 6- to 24-fold at doses around 20 Gy) compared to the general population.[23] This association is most evident after high doses exceeding 30 Gy, with a typical latency period of 5 to 20 years between exposure and tumor onset.[2] Such secondary osteosarcomas often arise within or near the previously irradiated field, as observed in survivors of childhood cancers like Hodgkin lymphoma.[23]Prior treatment with certain chemotherapeutic agents, especially alkylating agents such as cyclophosphamide, also elevates the risk of osteosarcoma, with the magnitude of risk rising linearly with cumulative dose.[24] This is particularly notable among survivors of retinoblastoma or acute leukemia, where combined exposure to alkylating agents and radiation synergistically heightens susceptibility. The mechanism likely involves DNA damage accumulation in bone progenitor cells, though the overall attributable risk remains low due to the rarity of the malignancy.[25]Rapid bone growth during adolescence has been associated with higher osteosarcoma incidence, with taller stature and pubertal growth spurts correlating to increased risk, though this link is not directly causal and may reflect underlying hormonal or growth-related factors.[26] Epidemiological studies indicate that individuals above average height face a modestly elevated odds ratio for the disease, aligning with its peak occurrence during the second decade of life when skeletal growth is most active.[27]Approximately 1% of patients with Paget's disease of bone develop osteosarcoma, representing a several-thousand-fold increase in risk compared to the general population.[28] This transformation typically occurs in long-standing, polyostotic disease affecting older adults, where pagetic bone undergoes sarcomatous change, often with a poorer prognosis than primary osteosarcoma.[29]Evidence linking environmental exposures to osteosarcoma is limited and inconsistent; potential associations have been suggested with fluoride in drinking water, asbestos, and heavy metals like cadmium, but large-scale studies have found no definitive causal relationship.[30] For instance, while some rodent models and early human data hinted at risks from high fluoride levels, subsequent epidemiological reviews classify the evidence as equivocal, with no clear dose-response pattern in humans.[7]Occupational risks for osteosarcoma are supported by sparse data, with limited evidence pointing to elevated incidence among farmers and industrial workers exposed to pesticides or chemicals, though confounding factors like overall exposure duration complicate attribution.[31]Cohort studies of agricultural workers have reported modestly increased rates of bone sarcomas, potentially tied to herbicide use, but these findings require further validation through larger prospective analyses.[32]
Genetic Predispositions
Osteosarcoma can arise in the context of several inherited genetic syndromes characterized by germline mutations that impair DNA repair, tumor suppression, or chromosomal stability, thereby increasing susceptibility to this malignancy. These predispositions are typically rare but account for a notable subset of cases, particularly in pediatric and young adult populations, where familial patterns may suggest screening for underlying syndromes. Autosomal dominant or recessive inheritance patterns predominate, with mechanisms often involving disrupted cell cycle regulation or genomic integrity.Li-Fraumeni syndrome (LFS), caused by germline mutations in the TP53 tumor suppressor gene on chromosome 17p13.1, is an autosomal dominant disorder that confers a high lifetime cancer risk, including osteosarcoma as a sentinel malignancy. These mutations lead to impaired DNA damage response and apoptosis, promoting tumorigenesis. LFS accounts for approximately 4% of pediatric osteosarcoma cases, with osteosarcoma occurring in about 12% of affected individuals.[33][34]Hereditary retinoblastoma, resulting from germline mutations in the RB1 gene on chromosome 13q14, disrupts retinoblastoma protein function essential for cell cycle control at the G1/S checkpoint. This autosomal dominant condition, often involving deletion of 13q14, predisposes survivors of retinoblastoma to secondary malignancies, with osteosarcoma developing in 5-10% of cases due to biallelic inactivation in osteoblasts.[35][36]Rothmund-Thomson syndrome (RTS), an autosomal recessive disorder primarily due to biallelic mutations in the RECQL4 gene encoding a DNA helicase involved in replication and repair, causes poikiloderma and skeletal abnormalities alongside heightened cancer risk. These mutations result in defective DNA repair, leading to genomic instability that predisposes to osteosarcoma, with a lifetime risk of approximately 30% in affected individuals.[35][37]Bloom syndrome, caused by biallelic mutations in the BLM gene on chromosome 15q26.1, which encodes a RecQ helicase critical for DNA unwinding and recombination, features chromosomal instability manifested as high sister chromatid exchange rates. This autosomal recessive syndrome elevates the overall cancer risk, including a higher-than-expected incidence of osteosarcoma compared to the general population.[35][38]Other rare syndromes also contribute to osteosarcoma predisposition. Werner syndrome, an autosomal recessive progeroid disorder from mutations in the WRN gene on chromosome 8p12 encoding another RecQ helicase, impairs DNA repair and telomere maintenance, resulting in premature aging and increased sarcoma risk, including osteosarcoma, which accounts for a significant portion of malignancies in affected patients. Diamond-Blackfan anemia, often linked to heterozygous mutations in ribosomal protein genes such as RPS19 on chromosome 19q13.2, involves defective ribosome biogenesis and p53 activation, leading to bone marrow failure and an elevated risk of solid tumors like osteosarcoma.[35][39][40]In addition to defined syndromes, 5-10% of osteosarcoma cases exhibit familial clustering without an identifiable underlying syndrome, suggesting polygenic or low-penetrance germline variants that contribute to heritable risk.[41][42]
Pathophysiology
Tumor Biology
Osteosarcoma is defined as a primary malignant tumor of mesenchymal origin in which the malignant cells directly produce osteoid, the unmineralized organic portion of bone matrix, distinguishing it from other bone tumors.[43] This osteoid production is a hallmark pathological feature required for diagnosis, reflecting the tumor's ability to recapitulate aspects of aberrant bone formation.[44]The histogenesis of osteosarcoma involves primitive mesenchymal cells that undergo abnormal differentiation toward osteoblasts, rather than arising from mature bone cells or osteoblasts themselves.[2] These progenitor cells, often mesenchymal stem cells or their committed precursors, give rise to the neoplastic population capable of osteoid synthesis, leading to disorganized bone production within the tumor mass.[45]Osteosarcoma exhibits aggressive growth patterns characterized by rapid local invasion into surrounding soft tissues and bone, facilitated by its high vascularity that supports nutrient delivery and tumor expansion.[46] At diagnosis, approximately 80% of patients harbor micrometastases to the lungs, which are often occult and undetectable by imaging, contributing to the tumor's poor prognosis if not addressed.[47]Microscopically, osteosarcoma displays pleomorphic tumor cells—ranging from spindle-shaped to round or epithelioid—with hyperchromatic nuclei, prominent nucleoli, and high mitotic activity indicative of rapid proliferation.[48] These cells produce lace-like patterns of osteoid, often interspersed with areas of necrosis due to the tumor's outpacing blood supply, creating a heterogeneous landscape of viable tumor, matrix, and dead tissue.[44]The tumor microenvironment in osteosarcoma plays a critical role in its progression, with osteoclasts recruited to resorb surrounding bone, enabling local invasion and creating space for tumor growth.[49]Angiogenesis is prominently driven by vascular endothelial growth factor (VEGF) secreted by tumor cells, promoting new vessel formation to sustain the highly metabolic neoplasm.[50] Additionally, the microenvironment facilitates immune evasion through immunosuppressive mechanisms, including the recruitment of regulatory T cells and myeloid-derived suppressor cells that dampen antitumor immune responses.[51]Metastatic behavior in osteosarcoma predominantly occurs via hematogenous spread, with the lungs serving as the primary site due to the tumor's access to pulmonary vasculature through bone marrow venous drainage.[52] Lymphatic dissemination is rare, occurring in less than 10% of cases, underscoring the hematogenous route as the dominant mechanism for distant dissemination.[53]In most cases, osteosarcoma shows osteoblastic differentiation dominance, though brief mentions of chondroblastic or fibroblastic variants highlight histological heterogeneity without altering core biology.[54]
Molecular Alterations
Osteosarcoma tumors exhibit a high degree of genomic instability, characterized by frequent somatic mutations in key tumor suppressor genes. Mutations in the TP53 gene, which encodes the p53 protein, are inactivated in approximately 70-90% of cases, resulting in impaired DNA damage response, increased genomic instability, and resistance to apoptosis, thereby facilitating tumor progression and metastasis.[55] Similarly, disruptions in the RB1 pathway, including mutations, deletions, or hypermethylation of the RB1 gene, occur in 30-60% of osteosarcomas, leading to deregulation of the G1/S cell cycle checkpoint and uncontrolled cellular proliferation.[56] These alterations often co-occur, with studies showing that up to 20% of tumors harbor both TP53 and RB1 pathway defects, underscoring their cooperative role in osteosarcoma oncogenesis.[57]Cytogenetic analyses reveal a complex karyotype in the majority of osteosarcoma cases, typically featuring near-triploidy with numerous structural and numerical chromosomal abnormalities. Recent studies have identified chromothripsis, a process of massive chromosome shattering and rearrangement, as a major driver of this genomic chaos, occurring subclonally in up to 74% of cases and enabling rapid tumor evolution.[55] Common chromosomal gains include regions 1q, 6p, and 8q, which harbor oncogenes promoting cell survival and growth, while losses frequently affect 3q, 10p, and 17p, encompassing tumor suppressor loci that further destabilize the genome.[58] This chaotic genomic landscape contributes to the aggressive phenotype of osteosarcoma by enabling rapid evolutionary adaptation of tumor cells. Additional genetic changes include amplification of the MDM2 gene in about 15% of cases, which inhibits p53 function and enhances cell survival independently of TP53 mutations.[56] Overexpression of HER2 (ERBB2) is observed in 20-30% of tumors, correlating with increased proliferation and poor prognosis through activation of downstream signaling pathways like PI3K/AKT.[59] Alterations in the ALK gene, such as fusions or rearrangements, are rare but have been reported in isolated cases, potentially driving tyrosine kinase signaling and offering opportunities for targeted inhibition.[60]Epigenetic modifications also play a critical role in osteosarcoma pathogenesis, with aberrant DNA methylation patterns frequently silencing tumor suppressor genes. Hypermethylation of the CDKN2A promoter, which encodes p16INK4a and p14ARF, disrupts cell cycle regulation and p53 pathway integrity, contributing to tumor initiation and progression in a subset of cases where genetic deletions are absent.[61]MicroRNA dysregulation further modulates oncogenesis, notably with upregulation of miR-21 in tumor tissues and plasma, which promotes invasion and metastasis by targeting genes like PTEN and PDCD4, while serving as a potential circulating biomarker for early detection and monitoring.[62] These epigenetic changes, often intertwined with genetic alterations, highlight the multifaceted molecular drivers in osteosarcoma.
Diagnosis
Imaging Techniques
Imaging of osteosarcoma relies on a multimodal approach to detect, characterize, and stage the tumor, with each modality offering complementary information on local extent, bone involvement, and metastatic spread. Plain radiography serves as the initial screening tool, typically revealing lytic bone destruction, aggressive periosteal reaction manifesting as a "sunburst" pattern or Codman's triangle, and an associated soft tissue mass, which are highly suggestive of the diagnosis in the appropriate clinical context.[63]Magnetic resonance imaging (MRI) is the gold standard for delineating local tumor extent, providing superior soft tissue contrast to assess tumor margins, bone marrow infiltration, neurovascular involvement, and skip metastases within the same bone. On T1-weighted sequences, the tumor often appears as low-signal-intensity marrow replacement, while T2-weighted and fat-suppressed images highlight high-signal-intensity areas of edema, necrosis, or hemorrhage, enabling precise preoperative planning.[64]Computed tomography (CT) excels in evaluating the mineralized osteoid matrix characteristic of osteosarcoma, quantifying cortical destruction, and identifying pulmonary metastases through dedicated chest protocols, with low-dose CT additionally guiding percutaneousbiopsy. It is particularly valuable for tumors in complex anatomical sites like the axial skeleton, where it offers detailed bone architecture assessment.[65]Bone scintigraphy, typically using technetium-99m-labeled diphosphonates, detects multifocal skeletal involvement or metastases with high sensitivity, often showing intense uptake at the primary site and any distant bony lesions, though it lacks specificity and requires correlation with other imaging.[13]Positron emission tomography-computed tomography (PET-CT) with 18F-fluorodeoxyglucose (FDG) assesses tumor metabolic activity, aiding in response evaluation after neoadjuvant chemotherapy via changes in standardized uptake value (SUV), and detects occult metastases, with SUV thresholds helping differentiate high-grade from low-grade lesions.[64]These imaging techniques are integrated into staging frameworks, such as the Enneking system for musculoskeletal sarcomas or the American Joint Committee on Cancer (AJCC) TNM classification, where MRI and CT primarily inform T-stage (tumor size, location, and extracompartmental extension), rare N-stage (nodal involvement), and M-stage (presence of metastases, especially pulmonary).[65]
Biopsy and Histopathology
Diagnosis of osteosarcoma requires tissue sampling through biopsy to confirm the presence of malignant cells and their characteristics. The preferred method is core needle biopsy, typically using a 14- to 18-gauge needle, which provides sufficient tissue for histopathologicalanalysis while minimizing invasiveness.[66] Open incisional biopsy is an alternative when core needle biopsy is inadequate, involving a small surgical incision to obtain a representative sample. Fine-needle aspiration is generally avoided due to its inadequacy in yielding enough material for accurate diagnosis and subtyping.[2][16]Biopsy procedures are ideally performed under image guidance, such as CT or MRI, to target the most representative area of the lesion and reduce sampling errors. Multidisciplinary planning, involving orthopedic surgeons, radiologists, and pathologists, is essential to position the biopsy tract in a way that facilitates subsequent tumor resection and avoids soft tissue contamination, which could complicate limb-sparing surgery.[63]Histopathological examination is the gold standard for confirming osteosarcoma, characterized by the production of osteoid matrix by malignant spindle-shaped or pleomorphic cells. These cells exhibit high nuclear atypia, mitoses, and necrosis, with the majority of cases classified as high-grade, accounting for approximately 90% of osteosarcomas. Grading follows systems like Enneking, where high-grade lesions (stage II) show aggressive features such as marked pleomorphism and rapid growth.[67][63][2]Immunohistochemistry supports the diagnosis by demonstrating vimentin positivity in neoplastic cells, reflecting their mesenchymal origin. Osteocalcin and S100 staining are variable, often positive in areas of osteoid production but not uniformly across the tumor, aiding in confirmation of osteogenic differentiation. These markers help rule out mimics like Ewing sarcoma, which typically shows strong CD99 positivity and lacks osteoid production.[68][69][70]Molecular testing, including fluorescence in situ hybridization (FISH) for HER2 amplification and next-generation sequencing (NGS) for identifying mutations in genes like TP53 or RB1, is employed when the subtype is unclear or to guide potential targeted therapies.[71][72]Diagnostic challenges include distinguishing osteosarcoma from reactive bone formation or chronic infection, which may show osteoid-like material but lack malignant cellular features. Diagnostic error rates for osteosarcoma biopsies are generally 5-10% with experienced pathologists and image guidance, but can be higher (up to 18%) with inadequate sampling or inexperienced teams.[70][63][73]
Subtypes
Osteosarcoma is classified into several histological subtypes based on microscopic appearance, location, and clinical behavior, with conventional osteosarcoma accounting for approximately 75-80% of all cases.[13] This subtype is high-grade and typically arises within the medullary cavity of long bones, most commonly around the knee in adolescents.[2] Within conventional osteosarcoma, further variants include osteoblastic (about 50% of cases, characterized by prominent mineralized osteoid matrix production), chondroblastic (25%, featuring cartilaginous differentiation), and fibroblastic (25%, dominated by spindle-shaped cells with less matrix).[74] All conventional subtypes share similar aggressive behavior and lack significant prognostic differences among themselves.[75]Telangiectatic osteosarcoma represents 3-5% of cases and is marked by cystic, blood-filled spaces resembling aneurysmal bone cysts, often presenting with a lytic appearance on imaging.[76][77] It is highly aggressive but shows a good response to neoadjuvant chemotherapy, often comparable or superior to conventional types.[77]Small cell osteosarcoma comprises 1-2% of cases and consists of round blue cells mimicking Ewing sarcoma, though Ewing-like translocations are rare.[76] These tumors typically occur in the same demographic and locations as conventional osteosarcoma but may show slightly worse outcomes due to diagnostic challenges.[2]Surface osteosarcomas arise from the bone surface and include parosteal (about 4% of cases, low-grade, often on the posterior distal femur with a better prognosis and 5-year survival exceeding 80%), periosteal (1%, intermediate-grade, commonly on the tibia with diaphyseal involvement), and high-grade surface (<1%, aggressive despite superficial location).[76][78] Low-grade surface subtypes generally have superior outcomes compared to intramedullary high-grade forms.[75]Rare variants include multifocal osteosarcoma (0.4-4%, involving multiple bone sites synchronously or metachronously without lung metastases) and extraskeletal osteosarcoma (1%, arising in soft tissues without bone attachment, often in older adults with poorer prognosis).[79][80][81] Other uncommon forms, such as gonadoblastoma-associated osteosarcoma, occur in the context of gonadal dysgenesis syndromes. Most subtypes share molecular alterations like TP53 mutations.[11]
Treatment
Standard Therapies
The standard treatment for osteosarcoma involves a multimodal approach combining neoadjuvant chemotherapy, surgical resection, and adjuvant chemotherapy, primarily guided by protocols from the Children's Oncology Group (COG) and the European and American Osteosarcoma Study Group (EURAMOS).[11][82]Neoadjuvant chemotherapy is administered prior to surgery to shrink the tumor, improve resectability, and assess treatment response through tumor necrosis. The MAP regimen, consisting of high-dose methotrexate, doxorubicin, and cisplatin, remains the cornerstone of this phase, typically delivered in two cycles over approximately 10 weeks.[83][84][85] This regimen is recommended by NCCN and ESMO guidelines for patients with localized high-grade osteosarcoma, with adjustments for age and renal function to mitigate toxicity.[84]Surgical resection follows neoadjuvant therapy and aims for wide margins to achieve local control while preserving function whenever possible. Limb-salvage surgery is feasible in 80-90% of extremity cases, utilizing endoprostheses, allografts, or rotationplasty to reconstruct the defect; amputation is reserved for unresectable tumors or cases with neurovascular involvement.[11][86] For low-grade subtypes, surgery alone may suffice without systemic therapy.[84]Adjuvant chemotherapy commences shortly after surgery, typically using the same MAP agents to eliminate microscopic disease, with the regimen tailored based on histologic response—defined as greater than 90% tumor necrosis indicating a favorable outcome.[83][87] An additional 4-6 cycles are standard, completing the total course in 6-12 months as per COG (e.g., AOST0331) and EURAMOS-1 protocols.[82][85]Radiation therapy is rarely used in resectable osteosarcoma due to its radioresistance but may be employed for unresectable primary tumors, positive margins, or palliation, with doses ranging from 50-70 Gy in conventional fractionation.[11][88]Supportive care is integral to manage treatment-related complications, including bisphosphonates such as zoledronic acid to alleviate bone pain and prevent pathologic fractures, and granulocyte colony-stimulating factors (e.g., filgrastim) to counteract chemotherapy-induced neutropenia.[89][90]
Emerging and Experimental Treatments
Emerging treatments for osteosarcoma are primarily focused on addressing the limitations of conventional chemotherapy and surgery, particularly in metastatic or recurrent disease, through targeted molecular approaches and immune modulation. Immunotherapies represent a major area of investigation, with chimeric antigen receptor T-cell (CAR-T) therapies targeting surface antigens such as HER2 and GD2 showing preliminary efficacy in early-phase trials. For instance, phase I/II studies of HER2-targeted CAR-T cells in patients with recurrent osteosarcoma have reported objective response rates of approximately 20-30% in small cohorts, often combined with lymphodepletion to enhance persistence, though challenges include on-target off-tumor toxicity and limited tumor infiltration. Similarly, GD2-directed CAR-T cells, adapted from neuroblastoma protocols, have demonstrated stable disease in up to 25% of osteosarcoma patients in phase I trials, with ongoing efforts to optimize constructs for sarcoma-specific microenvironments. Checkpoint inhibitors, particularly anti-PD-1 agents like pembrolizumab, have shown limited single-agent efficacy in osteosarcoma due to its immunologically "cold" tumor profile, with response rates below 10% in phase II trials; however, combinations with chemotherapy or other immunomodulators are under evaluation to overcome this barrier.[91]Targeted therapies aim to exploit specific molecular vulnerabilities identified in osteosarcoma subtypes. Tyrosine kinase inhibitors such as regorafenib have demonstrated modest activity in phase II trials for relapsed disease, with a progression-free survival of 3.6 months compared to 1.7 months with placebo, primarily through multi-kinase inhibition of VEGFR and PDGFR pathways that support tumor angiogenesis.[92]mTOR inhibitors like everolimus are being explored for chemotherapy-resistant cases, where preclinical models and phase I/II data indicate partial responses in 15-20% of patients with recurrent osteosarcoma harboring PI3K/AKT pathway alterations, often in combination with standard agents to sensitize tumors.Vaccine-based approaches seek to prevent recurrence following resection by eliciting antitumor immunity. The OST-HER2 vaccine, a DC-based therapy targeting HER2-overexpressing osteosarcoma, has advanced to phase IIb trials, showing a reduction in recurrence risk by up to 40% in high-risk resected patients through induction of HER2-specific T-cell responses, with durable immunity observed in follow-up data. As of October 2025, the phase IIb trial reported a statistically significant improvement in 2-year overall survival (66.6% vs. 40% historical control) in patients with resected pulmonary metastatic osteosarcoma.[93] Listeria-based vaccines, such as ADXS31-164 engineered to express HER2 epitopes, have entered phase I testing for HER2-expressing solid tumors including osteosarcoma, leveraging the bacterium's ability to stimulate innate and adaptive immunity, with preclinical and canine studies showing immune responses and stable disease.Gene therapy and oncolytic virotherapy offer innovative strategies for direct tumor modification and local control. CRISPR-Cas9 editing targeting TP53 and RB1 mutations, common in osteosarcoma, remains largely preclinical but has shown promise in restoring tumor suppressor function and reducing proliferation in patient-derived xenografts, with initial safety data from phase I vector delivery trials expected by 2026. Oncolytic viruses like talimogene laherparepvec (T-VEC), a GM-CSF-armed herpes simplex virus, are under investigation in sarcomas, including potential for unresectable osteosarcoma, with phase II studies in advanced sarcomas reporting objective responses and enhanced immune infiltration, particularly when combined with checkpoint blockade.[94]Among novel agents, denosumab, a RANKL inhibitor, is being repurposed to mitigate bone destruction and metastasis in osteosarcoma, with phase II trials indicating delayed skeletal-related events in advanced disease, achieving a 50% reduction in bone resorption markers without significant additive toxicity to chemotherapy. Bispecific antibodies, such as those engaging T-cells against GD2 or EGFR, are in early clinical development (phase I), demonstrating redirected cytotoxicity in preclinical osteosarcoma models and initial partial responses in basket trials for sarcomas.[91] As of 2025, initiatives like Break Through Cancer have accelerated collaborative trials integrating these agents, funding multi-arm studies that combine targeted therapies with immunotherapy to address osteosarcoma's heterogeneity.Clinical trials targeting metastatic osteosarcoma, which accounts for over 80% of treatment failures, number more than 50 active studies worldwide, registered on ClinicalTrials.gov. These trials highlight persistent challenges such as tumor heterogeneity, low mutational burden limiting neoantigen targets, and the need for biomarkers to predict responders, with adaptive designs increasingly employed to refine patient selection.
Prognosis
Survival Statistics
The overall 5-year survival rate for osteosarcoma patients with localized disease at diagnosis is approximately 60-70%, while for those with metastatic disease, it ranges from 20-30%. According to data from the Surveillance, Epidemiology, and End Results (SEER) program, the 5-year relative survival rate across all cases from 2013-2019 was 65.9%, with the latest SEER data (2015-2021) showing 68.5% for bone and joint cancers.[95][4] Recent data as of 2025 confirm stable survival rates for localized disease at 60-75%.[96]Survival outcomes vary by age, with adolescents and young adults experiencing higher rates of around 70% at 5 years, compared to approximately 50% for adults over 40 years.[97] The 1-year survival rate exceeds 80% for most patients, reflecting early treatment responses.[98]Site-specific survival differs significantly, with appendicular osteosarcoma (in limbs) showing higher 5-year rates of about 67%, versus approximately 34% for axial tumors (in the spine or pelvis), due to challenges in surgical resection.[99]Historically, survival was poor before the 1970s, with rates around 20% relying solely on surgery; the introduction of multi-agent chemotherapy improved this to current levels of 60-70% for localized cases.[16]In the United States, SEER data from 2018-2022 indicate an incidence of 1.0 per 100,000 population and a mortality rate of 0.5 per 100,000 for bone and joint cancers, predominantly osteosarcoma.[4]For metastatic disease, 5-year survival is approximately 30% when confined to the lungs, but drops below 10% with multi-site involvement.[100][101]
Prognostic Factors
Prognostic factors in osteosarcoma encompass a range of clinical, pathological, and treatment-related variables that modulate disease outcomes, with tumor response to therapy emerging as a primary determinant. The degree of histologic tumor necrosis following neoadjuvant chemotherapy serves as a critical predictor; patients achieving greater than 90% necrosis exhibit markedly improved survival, with 10-year overall survival rates of approximately 84%, compared to around 37% for those with less than 90% necrosis (poor responders). This threshold reflects the tumor's chemosensitivity and is widely recognized as an independent prognostic indicator in high-grade cases.[102]Stage at diagnosis significantly influences prognosis, with approximately 80% of osteosarcomas presenting as localized disease, which confers a more favorable outlook than the 20% of cases with synchronous metastases, where long-term survival drops below 30%. Tumor size exceeding 8 cm at presentation is associated with adverse outcomes, including higher rates of metastasis and reduced event-free survival, independent of other variables.[103][104]Tumor location further stratifies risk; axial or pelvic osteosarcomas demonstrate 20-30% lower overall survival rates relative to extremity-based tumors, attributable to challenges in achieving wide surgical margins and higher metastatic potential. Younger age at diagnosis correlates with better prognosis, particularly in pediatric patients under 18 years, while high-grade histology—prevalent in nearly all conventional cases—universally portends poorer outcomes due to aggressive biology.[105][106]Elevated serum lactate dehydrogenase (LDH) levels, especially exceeding twice the upper limit of normal, signal increased tumor burden and are linked to roughly 50% reduced survival, serving as an independent marker of poor prognosis. Similarly, raised alkaline phosphatase (ALP) levels indicate potential visceral metastases, such as to the liver, and correlate with diminished event-free survival.[107][108]Pathological features at resection, including positive surgical margins, skip lesions within the same bone, and vascular invasion, are additional adverse factors that heighten local recurrence risk and compromise long-term survival, often necessitating intensified adjuvant strategies.[109][110][111]
Osteosarcoma in Animals
In Dogs
Osteosarcoma is the most common primary bone tumor in dogs, accounting for approximately 85% of all malignant bone neoplasms.[112] It predominantly affects large and giant breed dogs, such as Rottweilers, Greyhounds, Irish Wolfhounds, Great Danes, and Saint Bernards, with a median age of diagnosis between 7 and 10 years, though a smaller peak occurs in young dogs aged 18 to 24 months.[112][113][114]The tumor most frequently arises in the appendicular skeleton, particularly the long bones of the limbs (about 90% of cases), leading to clinical presentation of progressive lameness, pain, and localized swelling or a firm mass at the affected site.[115][116][117] At diagnosis, approximately 90% of dogs have micrometastases, most commonly to the lungs, though overt metastasis may not be evident initially.[115][116]Key risk factors include increased body size and height, with giant breeds showing the highest incidence due to greater mechanical stress on long bones; breed predispositions are evident in lines like Rottweilers and Irish Wolfhounds, where genetic factors contribute alongside rapid growth.[118][119][120]Molecularly, canine osteosarcoma shares significant similarities with the human disease, including frequent mutations in TP53 (detected in 53-71% of cases) and RB1, which disrupt cell cycle control and promote tumorigenesis; these parallels position dogs as a spontaneous translational model for human trials, including immunotherapy approaches targeting immune pathways associated with TP53 alterations.[121][122][123]Standard treatment involves limb amputation combined with adjuvant chemotherapy using agents like carboplatin or doxorubicin, yielding a median survival of 10-12 months and approximately 20-35% one-year survival rate, compared to 4-6 months with amputation alone.[124][125][126] Recent veterinary advances include stereotactic body radiation therapy (SBRT) for limb-sparing and pain palliation, achieving pain relief in over 80% of cases with median response durations of 4-8 months, often combined with bisphosphonates like pamidronate to inhibit osteoclast activity and manage bone pain without surgery.[127][128][129]
In Cats
Osteosarcoma in cats is a rare malignancy, representing approximately 70-80% of primary bone tumors, which themselves occur at an incidence of 3.1 to 4.9 per 100,000 feline cases.[130][131] It predominantly affects older cats over 10 years of age, with no strong breed predisposition identified.[132] Unlike the appendicular predominance in dogs, feline osteosarcoma frequently involves the axial skeleton in about 40-50% of cases, such as the mandible, vertebrae, ribs, or pelvis, leading to presentations of chronic pain, lameness, pathologic fractures, or neurologic deficits depending on the site.[133][134]Metastasis in feline osteosarcoma is less common than in canine cases, occurring in roughly 10-40% of affected cats, most often to the lungs when it does develop.[135][136] Potential risk factors include prior radiation therapy, which has been linked to secondary osteosarcoma development in isolated reports, and possible associations with feline immunodeficiency virus (FIV) or feline leukemia virus (FeLV) infections, though evidence for the latter remains limited and indirect through general oncogenic effects.[137][138]Feline osteosarcoma tends to be less aggressive overall compared to the canine form, with lower metastatic potential contributing to this distinction.[136]Treatment focuses on surgical intervention as the primary modality, though amputation is less feasible for axial tumors, often limiting options to local excision, debulking, or palliative procedures.[139]Chemotherapy, typically with doxorubicin, is sometimes employed adjunctively but demonstrates limited efficacy in extending survival beyond surgery alone.[140] Histologically, feline osteosarcoma is predominantly osteoblastic, producing malignant osteoid, with sparse molecular data available; alterations in the p53tumor suppressor gene have been noted in some cases, similar to patterns observed in human conventional osteosarcoma.[141][142]Prognosis for localized disease can be more favorable than in dogs, with median survival times exceeding 1 year following appendicular amputation in select cases, but overall outcomes remain poor, with median survival of 4-6 months due to frequent late-stage detection, axial location challenges, and incomplete resection.[135][136][139]
In Dinosaurs
Paleopathological evidence of osteosarcoma in dinosaurs has been identified through analysis of fossilized bone lesions exhibiting characteristics analogous to the malignant bone tumors observed in modern vertebrates. The first systematic survey of tumors in dinosaurs, conducted in 2003, examined over 10,000 vertebrae from more than 700 specimens using fluoroscopy and computed tomography (CT), revealing multiple cases of benign and malignant neoplasms, including one instance of metastatic bone cancer in a hadrosaur vertebra suggestive of hemangiosarcoma rather than osteosarcoma.[143] This study established that neoplasms were present in Mesozoic archosaurs but lacked definitive histological confirmation for specific tumor types like osteosarcoma due to the limitations of early imaging techniques.The first confirmed case of osteosarcoma in a dinosaur was reported in 2020 from the fibula of a Centrosaurus apertus, a ceratopsian dinosaur from the Late CretaceousDinosaur Park Formation in Alberta, Canada, dating to approximately 76–77 million years ago.[144] Diagnosis involved multimodal approaches, including gross examination showing a large, irregular bone mass with malignant proliferation, CT imaging revealing aggressive periosteal reactions forming a Codman triangle and permeative bone destruction without healing signs, and thin-section histopathology demonstrating disorganized osteoid production by malignant osteoblasts, lytic lesions, and spiculed reactive bone—features mirroring human osteosarcoma. This specimen, housed at the Royal Ontario Museum, represents advanced-stage disease that likely impaired mobility in the adult individual.Subsequent discoveries have expanded the record of osteosarcoma-like lesions in ornithischian dinosaurs. In 2021, analysis of the holotype skeleton of Bonapartesaurus rionegrensis, a hadrosaurid from the Late Cretaceous Allen Formation in Argentina (about 70 million years old), identified a proliferative bone overgrowth on the second metatarsal interpreted as a neoplasm, possibly osteosarcoma, based on irregular periosteal new bone formation and absence of traumatic remodeling. The lesion's cauliflower-like morphology and location suggest it caused lameness, complicating the dinosaur's terrestrial locomotion alongside co-occurring tail fractures. Additional suspected cases include proliferative lesions in theropod fossils.[145]Diagnostic criteria for osteosarcoma in fossils rely on comparative paleopathology, emphasizing fossilized evidence of osteoid matrix production, aggressive periosteal reactions without remodeling, and permeative cortical destruction, which differentiate it from non-malignant conditions like trauma (characterized by callus formation and healing) or infection (evidenced by sequestra or cloacae). These features are assessed via CT for internal architecture and histology of thin sections for cellular analogs, though challenges arise from diagenetic alterations mimicking pathological changes.Such findings imply that osteosarcoma afflicted large-bodied dinosaurs, particularly ornithischians with rapid growth rates, potentially linked to evolutionary pressures favoring fast skeletal development similar to human pediatric cases.[144] Environmental factors, including dietary or hormonal influences on bonemetabolism, have been hypothesized but remain untested without molecular data. Limitations include the absence of preserved soft tissues, DNA, or proteins, precluding genetic confirmation, and reliance on rare, well-preserved specimens, which may underestimate prevalence. A 2024 comprehensive review of vertebrate paleoneoplasms confirmed several dinosaur specimens with osteosarcoma-compatible lesions across theropods and ornithischians, underscoring the tumor's ancient origins and contributing to evolutionary oncology by paralleling spontaneous bone cancers in mammals without modern lifestyle confounders.[146] In 2025, a novel osteogenic tumor was reported in the ulna of a Late Jurassic mamenchisaurid dinosaur from Thailand, diagnosed using CT scanning and thin-section analysis as an aggressive neoplasm with features suggestive of osteosarcoma, including permeative growth and disorganized bone production; this represents the oldest confirmed bone tumor in a sauropodomorph and further highlights the prevalence of such pathologies in Mesozoic archosaurs.[147]