ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Treatment for tenosynovial giant cell tumor and other benign neoplasms affecting soft tissue and bone

Treatment for tenosynovial giant cell tumor and other benign neoplasms affecting soft tissue and bone
Literature review current through: Jan 2024.
This topic last updated: Apr 10, 2023.

INTRODUCTION — The majority of benign soft tissue masses are nonneoplastic and of little clinical consequence. However, some locally aggressive fibromatoses (desmoid tumors), infiltrating lipomas, neurofibromas, and vertebral hemangiomas may cause significant morbidity and, in some cases, mortality.

Treatment for a variety of benign diseases affecting soft tissue and bone will be reviewed here. These include Gorham disease, desmoplastic fibroma of bone, symptomatic vertebral hemangioma, and tenosynovial giant cell tumor (pigmented villonodular synovitis). Treatment for other conditions affecting soft tissue, such as desmoid tumors, keloids, ganglia, and Dupuytren's contracture, is discussed separately. (See "Desmoid tumors: Epidemiology, molecular pathogenesis, clinical presentation, diagnosis, and local therapy" and "Keloids and hypertrophic scars" and "Ganglion cysts of the wrist and hand" and "Dupuytren's contracture", section on 'Other interventions'.)

GORHAM DISEASE — Gorham disease (also known as Gorham-Stout disease, disappearing bone disease, or essential osteolysis) is a rare disorder characterized by relatively painless bone destruction with massive osteolysis due to progression of lymphangiomatous tissue [1]. There may also be significant osteoclast activation. Why this occurs is not clear, but increased levels of interleukin 6 may contribute to the osteolysis [2].

The pattern is that of lytic destruction of cortical bone and extension across joints to adjacent bone. There is usually little or no soft tissue mass.

Treatment — Treatment consists of resection when this is feasible from the standpoints of the medical condition of the patient, the technical aspects of the procedure, and the likelihood of retained function. There is observational evidence that radiation therapy also may be effective [3-5]:

In one report, two patients with disease involving C1 and the occiput were managed with 45 to 50 Gy alone [4]. Both patients experienced cessation of progression and were otherwise well at follow-up exceeding 12 years. There was no remodeling of bone in either of these patients.

Others have demonstrated benefit from radiation therapy in treating chylothorax related to chest wall involvement [6] and a lesion in the mandible [7].

A patterns of care study from Germany with literature review concluded that radiation therapy (30 to 45 Gy) may prevent disease progression in 77 to 80 percent of cases [5].

The role of systemic therapy has not been systematically evaluated. There are reports of benefit from bisphosphonates alone [8] and with sunitinib plus low-dose metronomic paclitaxel in two patients, one with generalized lymphatic anomaly and one with lymphatic malformation in Gorham disease, both of whom had severe exacerbation during puberty [9]. The first child presented with florid pulmonary failure and pleural effusion; the other presented with severe pain due to bone destruction of the pelvis and inability to walk. Both patients experienced clinical and radiologic response without major toxicities.

DESMOPLASTIC FIBROMA OF BONE — Desmoplastic fibroma of bone, known as collagenous fibroma, is a benign tumor of bone that has a histologic appearance similar to that of a desmoid tumor. This is an uncommon tumor that represents fewer than 0.1 percent of bone tumors. These lesions appear in young adults, with approximately equal frequency in males and females. Affected patients typically present with a history of a painless, slowly growing mass, often of relatively long duration [10].

These lesions can also appear as soft tissue tumors; in these cases, the lesions are predominantly subcutaneous, but fascial involvement is common, and approximately one-quarter involve skeletal muscle [10]. When they arise in bone, the most frequently involved sites are the pelvic and other flat bones, the jawbones, and for the long bones, the diaphyseal regions. The tumors are often large, ranging in size from 1 to 20 cm. They are firm and pearl gray in color on gross examination. The lesional cells are bland stellate and spindle-shaped fibroblasts separated by a collagenous matrix; there is typically no mitotic activity [10]. A characteristic (2;11)(q31;q12) translocation has been reported in several affected patients [11]. The beta-catenin pathway does not seem to have the same essential role in the tumorigenesis of desmoplastic fibroma as it has in desmoid-type fibromatosis [12]. (See "Desmoid tumors: Epidemiology, molecular pathogenesis, clinical presentation, diagnosis, and local therapy".)

Desmoplastic fibroma of bone is managed with surgical resection, with extremely few exceptions. One patient with a 7.6 cm lesion in the iliac crest responded well to a course of fractionated radiation therapy (60 Gy) [13], as did a patient with a mandibular lesion who refused surgery [14].

Recurrent disease appears to be rare. In one study, none of the 39 patients developed a recurrence at a median follow-up of 11 years [10].

SYMPTOMATIC VERTEBRAL HEMANGIOMA — Hemangiomas are benign neoplasms consisting of blood vessels. Autopsy series suggest a prevalence of 10 to 12 percent for vertebral body hemangiomas [15], but most remain asymptomatic [16-18].

Approximately one-half of vertebral body hemangiomas are associated with painful symptoms, and in a few cases, they cause neurologic emergencies such as acute spinal cord compression [16]. The diagnosis is usually confirmed by computed tomography (CT) or magnetic resonance imaging (MRI) (image 1) [19].

Options for treatment include surgical procedures (such as decompressive laminectomy, vertebrectomy, or ligation of the segmental arteries) and conservative methods (such as embolization, vertebroplasty, and/or intralesional alcohol injection) [15]. Radiation therapy may be applied as a treatment for painful vertebral hemangiomas, both as a single modality and for postsurgical prevention of local relapse [20-22].

In one report, a total of 84 patients with 96 symptomatic lesions were irradiated for a symptomatic vertebral hemangioma (98 percent for pain; 29 percent had additional neurologic symptoms) [20]. Prior to undergoing radiation therapy, 31 patients had been treated with resection, embolization, or vertebroplasty. The median total dose was 34 Gy. At a median follow-up of 68 months, 62 percent had complete symptom relief, 29 percent had partial pain relief, and 10 percent had no pain relief. Total doses ≥34 Gy resulted in significantly greater symptom relief and a lower recurrence rate.

TENOSYNOVIAL GIANT CELL TUMOR — Tenosynovial giant cell tumor (TGCT), historically known as pigmented villonodular synovitis (PVNS), is a rare [23] but well-recognized proliferative lesion of synovial tissue. Due to the presence of a characteristic translocation TGCT is considered to be a clonal neoplastic lesion; it can recur locally and cause substantial morbidity, but metastases are rare.

Histopathology and presentation — TGCT is characterized by hypervascular proliferative synovium containing multinucleated giant cells, macrophages, and hemosiderin. The multinucleated cells express features of osteoclasts [24]. Progressive nodular disease near or in the joints limits function and may destroy adjacent bone.

TGCT occurs in two histopathologic forms:

Localized form, which involves a discrete section of the synovium

Diffuse form, which involves the entire synovium and accounts for the majority of cases

TGCT almost always involves a single joint; the knee and ankle synovial structures are most commonly affected (image 2), while involvement of the shoulder, elbow, wrist/hand (picture 1), and hip is less common [23,25-27]. TGCT can present with attacks of pain, swelling, arthritis, and spontaneous hemarthrosis. (See "Overview of hemarthrosis", section on 'Tenosynovial giant cell tumor/pigmented villonodular synovitis' and "Basic calcium phosphate (BCP) crystal arthritis, including Milwaukee shoulder syndrome", section on 'Differential diagnosis'.)

Initial imaging often consists of plain radiographs, which typically reveal well-circumscribed areas of bone erosion. Ultrasonography can also be used to identify hypervascular areas of tumor and optimally guide synovial biopsy [28].

However, magnetic resonance imaging (MRI) is key to establishing the correct diagnosis [29,30]. MRI shows a characteristic lack of signal on both T1 and T2 images, which has been attributed to the presence of large amounts of hemosiderin in the synovium (image 3). (See "Radiologic evaluation of knee tumors in adults", section on 'Tenosynovial giant cell tumor' and "Imaging evaluation of the painful hip in adults", section on 'Pigmented villonodular synovitis and synovial osteochondromatosis'.)

Treatment-naïve patients — In patients with treatment-naïve TGCT, the principal treatment modality is resection [31,32]. Although limited data support the efficacy of radiation therapy, we do not offer this approach due to the risks of long-term toxicity and the availability of alternative systemic therapies for unresected and/or relapsed disease.

Although marginal excision represents the optimal treatment for localized TGCT, diffuse TGCT is more difficult to eradicate and is optimally treated with total or near total synovectomy [33]. The approach is similar to that used for inflammatory arthritis. (See "Synovectomy for inflammatory arthritis of the knee".)

Patients with diffuse disease can have multiple recurrences and bulky disease, resulting in significant bone destruction. Although observational data suggest some local disease control with adjuvant or perioperative radiation therapy, no randomized trials have evaluated this approach [34-40], and it is not our standard practice.

Examples of relevant data include the following:

In one meta-analysis of 35 observational studies containing over 600 patients with TGCT of the knee, perioperative radiation therapy was associated with a reduced rate of recurrence (odds ratio 0.31, 95% CI 0.14-0.70), although the evidence was of low quality [33].

In the largest series of 50 patients, 49 of whom had microscopic or gross residual disease prior to radiation therapy, 47 maintained local control at an average follow-up of 94 months [40].

Similarly, in multiple observational cohort studies of patients with diffuse TGCT and in a national patterns of care study, the addition of radiation therapy to a dose of 35 to 50 Gy was associated with decreased local recurrence rates [34-40].

Despite these results, radiation therapy may cause long-term toxicities and subsequent morbidity, especially in young patients with TGCT, a nonlethal tumor. Examples of such toxicities include joint stiffness due to radiation fibrosis, skin changes, tissue necrosis, and radiation-associated secondary malignancies, including transformation into malignant sarcoma [27]. (See "Radiation-associated sarcomas" and "Overview of cancer survivorship care for primary care and oncology providers", section on 'Risk of subsequent primary cancer'.)

In addition, available data suggest that both systemic therapies and radiation therapy may be used effectively in the setting of relapse, making the incorporation of radiation therapy into the treatment strategy for treatment-naïve disease less appealing [33,40]. As such, it is our practice to reserve radiation therapy for patients with recurrent or relapsed disease who are nonsurgical candidates and who either fail or choose to avoid the use of systemic therapy due to potential toxicities. (See 'Recurrent and/or relapsed disease' below.)

Postoperative intra-articular administration of radioactive isotopes has been advocated in the past for patients thought to be at high risk for recurrence [37,41,42], but this approach has fallen out of favor.

Recurrent and/or relapsed disease — The local recurrence rate among patients with localized TGCT is low (approximately 10 percent in one large series), and it is higher with diffuse disease (between 19 and 44 percent) [23,43].

Although TGCT is a nonlethal disease with a low risk of systemic metastases, patients with locoregional recurrences may experience severe long-term morbidity and poor quality of life, including postoperative complications, compromised joint function, secondary arthritis, effusions, joint replacement, and possible amputation (image 2) [44].

Approach to therapy — Treatment options are individualized based on patient characteristics, symptoms, extent of progression, expected postoperative morbidity, and availability of systemic therapies. Patients with recurrent and/or relapsed disease may undergo observation or receive further surgical interventions, such as subtotal resection, joint replacement, or amputation. A decision regarding surgery versus systemic therapy should be made in a multidisciplinary setting. In patients where further surgery would result in significant morbidity or functional impairment, we suggest the colony stimulating factor 1 receptor (CSF1R) inhibitor pexidartinib. Radiation therapy and/or enrollment in clinical trials may be reserved for those who are not surgical candidates and fail approved systemic therapies or who prefer to avoid the potential toxicities of systemic treatment [28,40]. (See 'Pexidartinib (CSF1R inhibitor)' below.)

Pexidartinib (CSF1R inhibitor) — In TGCT, a translocation in CSF1-COL6A3, t(1;2)(p13;q35-37), leads to overexpression of colony stimulating factor 1 (CSF1), with subsequent recruitment of the colony stimulating factor 1 receptor (CSF1R)-expressing macrophages that constitute the tumor mass [45,46]. Thus, in patients with unresectable relapsed and/or recurrent TGCT, agents that inhibit CSF1R, such as the oral agent pexidartinib [44,47-52], represent a logical therapeutic approach. Pexidartinib is approved by the US Food and Drug Administration (FDA) for patients with TGCT associated with severe morbidity or functional limitations and not amenable to surgery [53].

However, pexidartinib carries a black box warning for potentially fatal hepatotoxicity [54]; therefore, clinicians who offer pexidartinib to patients with TGCT, a nonlethal disease, should also provide a risk-benefit discussion regarding this particular toxicity. For patients who opt against pexidartinib, radiation therapy is a reasonable alternative. (See "Chemotherapy hepatotoxicity and dose modification in patients with liver disease: Molecularly targeted agents", section on 'Pexidartinib'.)

In addition, the following prescribing guidelines are recommended in discussions regarding pexidartinib [53]:

Pexidartinib can only be prescribed through a Risk Evaluation and Mitigation Strategy (REMS) program (TURALIO REMS), a registration and monitoring program for the safe use of pexidartinib.

Pexidartinib is administered at 250 mg orally twice a day with a low-fat meal (no more than approximately 11 to 14 grams of total fat). Patients should avoid taking pexidartinib with higher-fat meals (55 to 65 grams of total fat), which result in higher drug concentrations and increased risk of hepatoxicity. Patients may be referred to a dietician, if necessary.

Given the risk of hepatoxicity, liver biochemical tests should be monitored prior to and during therapy with pexidartinib. Further details on the frequency of monitoring and other issues related to hepatoxicity with pexidartinib are discussed separately. (See "Chemotherapy hepatotoxicity and dose modification in patients with liver disease: Molecularly targeted agents", section on 'Pexidartinib'.)

In a randomized, placebo-controlled, double-blinded phase III trial (ENLIVEN) of 120 patients with unresectable TGCT, pexidartinib improved objective response rates at week 25 (39 versus 0 percent in the placebo group) [44]. At a median follow-up of six months, no responders had progressed. Pexidartinib also improved functional outcomes in the affected joint at a follow-up of 25 weeks, including increased mean range of motion (15.1 versus 6.2 percent), improved physical function, and decreased stiffness. In addition, although not statistically significant, a numerically greater proportion of patients had at least a ≥30 percent decrease in their pain score (on a scale from 1 to 10) without an increase in narcotics (31 versus 15 percent). Depigmentation of the hair was the most common adverse event (67 versus 3 percent with placebo). Grade ≥3 adverse events included hepatotoxicity, cholestasis, periorbital edema, and hypertension. Eight patients discontinued therapy, mostly for hepatotoxicity. (See "Ocular side effects of systemically administered chemotherapy", section on 'Periorbital edema'.)

For patients who do not tolerate pexidartinib or who choose to avoid it due to the risk of toxicity, radiation therapy is a reasonable alternative, with limited data suggesting low recurrence rates and good functional outcomes. As examples, in one meta-analysis including approximately 130 patients with recurrence after synovectomy, radiation therapy reduced recurrence by 70 percent, although the evidence was of low quality [33]. In another study evaluating radiation therapy in 50 patients, including two-thirds with relapsed disease treated with at least two previous surgeries, long-term local control and good joint function were achieved in 94 and 82 percent of patients, respectively [40]. However, radiation therapy may also cause specific long-term toxicities, such as joint stiffness due to radiation fibrosis, skin changes, tissue necrosis, and radiation-associated secondary malignancies [27].

Investigational agents

NilotinibNilotinib and other multitargeted tyrosine kinase inhibitors (such as sunitinib, sorafenib, and imatinib) inhibit CSF1R (which functions as a receptor tyrosine kinase) [55-57]. For example, a phase II study of 56 patients with progressive disease not amenable to conservative surgical treatment demonstrated disease stabilization in a large proportion of patients with nilotinib (12-week progression-free rate 93 percent) [49].

ImatinibImatinib has also demonstrated activity in observational data [55,57]. For example, at least one report documents a brief complete response with imatinib in a patient with relapsed TGCT [55].

Emactuzumab – The monoclonal antibody emactuzumab (RG7155), which inhibits CSF1R activation, remains an investigational agent in TGCT [50,58]. In a phase I trial of 68 patients with locally advanced TGCT of the soft tissues, emactuzumab demonstrated durable responses, with an objective response rate of 64 percent at two years, and was associated with improvements in quality of life [58].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Soft tissue sarcoma" and "Society guideline links: Bone sarcomas".)

SUMMARY AND RECOMMENDATIONS

Benign neoplasms affecting soft tissue and bone – The majority of nonmalignant proliferative soft tissue and bone lesions are of modest clinical consequence. However, some locally aggressive proliferative lesions, despite their nonmalignant nature, can cause significant morbidity and, in some cases, mortality. These include the following:

Desmoid tumors (aggressive fibromatosis) (see "Desmoid tumors: Epidemiology, molecular pathogenesis, clinical presentation, diagnosis, and local therapy" and "Desmoid tumors: Systemic therapy")

Desmoplastic fibroma of bone (see 'Desmoplastic fibroma of bone' above)

Vertebral hemangioma (see 'Symptomatic vertebral hemangioma' above)

Tenosynovial giant cell tumor (pigmented villonodular synovitis) (see 'Tenosynovial giant cell tumor' above)

Tenosynovial giant cell tumor – For patients with treatment-naïve tenosynovial giant cell tumor (TGCT) and those with recurrent/relapsed disease in whom surgical resection is feasible, we suggest surgery with either total or near total synovectomy rather than radiation or systemic therapy (Grade 2C). (See 'Treatment-naïve patients' above and 'Recurrent and/or relapsed disease' above.)

For patients in whom further surgery would result in significant morbidity or functional impairment, options include pexidartinib or radiation therapy. Pexidartinib carries a black box warning for potentially fatal hepatotoxicity; therefore, clinicians who offer pexidartinib to patients with TGCT, a nonlethal disease, should also provide a risk-benefit discussion regarding this particular toxicity. For those who prefer to avoid the potential toxicities associated with pexidartinib, we offer radiation therapy. (See 'Pexidartinib (CSF1R inhibitor)' above.)

For patients who fail systemic therapies such as pexidartinib, we offer radiation therapy or clinical trials. A decision between these options should be made in a multidisciplinary setting and should factor in patient characteristics, symptoms, and extent of progression. (See 'Investigational agents' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Thomas F DeLaney, MD, who contributed to earlier versions of this topic review.

  1. Manisali M, Ozaksoy D. Gorham disease: correlation of MR findings with histopathologic changes. Eur Radiol 1998; 8:1647.
  2. Devlin RD, Bone HG 3rd, Roodman GD. Interleukin-6: a potential mediator of the massive osteolysis in patients with Gorham-Stout disease. J Clin Endocrinol Metab 1996; 81:1893.
  3. Mawk JR, Obukhov SK, Nichols WD, et al. Successful conservative management of Gorham disease of the skull base and cervical spine. Childs Nerv Syst 1997; 13:622.
  4. Dunbar SF, Rosenberg A, Mankin H, et al. Gorham's massive osteolysis: the role of radiation therapy and a review of the literature. Int J Radiat Oncol Biol Phys 1993; 26:491.
  5. Heyd R, Micke O, Surholt C, et al. Radiation therapy for Gorham-Stout syndrome: results of a national patterns-of-care study and literature review. Int J Radiat Oncol Biol Phys 2011; 81:e179.
  6. Duffy BM, Manon R, Patel RR, Welsh JS. A case of Gorham's disease with chylothorax treated curatively with radiation therapy. Clin Med Res 2005; 3:83.
  7. Ebert CS Jr, Zdanski C, Ardeshirpour F, et al. Recurrent infantile myofibromatosis: a report of conservative management and discussion of treatment strategies. Ear Nose Throat J 2008; 87:E4.
  8. Hammer F, Kenn W, Wesselmann U, et al. Gorham-Stout disease--stabilization during bisphosphonate treatment. J Bone Miner Res 2005; 20:350.
  9. Rössler J, Saueressig U, Kayser G, et al. Personalized Therapy for Generalized Lymphatic Anomaly/Gorham-Stout Disease With a Combination of Sunitinib and Taxol. J Pediatr Hematol Oncol 2015; 37:e481.
  10. Miettinen M, Fetsch JF. Collagenous fibroma (desmoplastic fibroblastoma): a clinicopathologic analysis of 63 cases of a distinctive soft tissue lesion with stellate-shaped fibroblasts. Hum Pathol 1998; 29:676.
  11. Bernal K, Nelson M, Neff JR, et al. Translocation (2;11)(q31;q12) is recurrent in collagenous fibroma (desmoplastic fibroblastoma). Cancer Genet Cytogenet 2004; 149:161.
  12. Hauben EI, Jundt G, Cleton-Jansen AM, et al. Desmoplastic fibroma of bone: an immunohistochemical study including beta-catenin expression and mutational analysis for beta-catenin. Hum Pathol 2005; 36:1025.
  13. Sanfilippo NJ, Wang GJ, Larner JM. Desmoplastic fibroma: a role for radiotherapy? South Med J 1995; 88:1267.
  14. Nag HL, Kumar R, Bhan S, et al. Radiotherapy for desmoplastic fibroma of bone: a case report. J Orthop Surg (Hong Kong) 2003; 11:90.
  15. Acosta FL Jr, Dowd CF, Chin C, et al. Current treatment strategies and outcomes in the management of symptomatic vertebral hemangiomas. Neurosurgery 2006; 58:287.
  16. Pastushyn AI, Slin'ko EI, Mirzoyeva GM. Vertebral hemangiomas: diagnosis, management, natural history and clinicopathological correlates in 86 patients. Surg Neurol 1998; 50:535.
  17. Bartels RH, Grotenhuis JA, Van Der Spek JA. Symptomatic vertebral hemangiomas. J Neurosurg Sci 1991; 35:187.
  18. Brunot S, Berge J, Barreau X, et al. [Long term clinical follow up of vertebral hemangiomas treated by percutaneous vertebroplasty]. J Radiol 2005; 86:41.
  19. Persaud T. The polka-dot sign. Radiology 2008; 246:980.
  20. Heyd R, Seegenschmiedt MH, Rades D, et al. Radiotherapy for symptomatic vertebral hemangiomas: results of a multicenter study and literature review. Int J Radiat Oncol Biol Phys 2010; 77:217.
  21. Rades D, Bajrovic A, Alberti W, Rudat V. Is there a dose-effect relationship for the treatment of symptomatic vertebral hemangioma? Int J Radiat Oncol Biol Phys 2003; 55:178.
  22. Miszczyk L, Tukiendorf A. Radiotherapy of painful vertebral hemangiomas: the single center retrospective analysis of 137 cases. Int J Radiat Oncol Biol Phys 2012; 82:e173.
  23. Ehrenstein V, Andersen SL, Qazi I, et al. Tenosynovial Giant Cell Tumor: Incidence, Prevalence, Patient Characteristics, and Recurrence. A Registry-based Cohort Study in Denmark. J Rheumatol 2017; 44:1476.
  24. Darling JM, Goldring SR, Harada Y, et al. Multinucleated cells in pigmented villonodular synovitis and giant cell tumor of tendon sheath express features of osteoclasts. Am J Pathol 1997; 150:1383.
  25. Müller LP, Bitzer M, Degreif J, Rommens PM. Pigmented villonodular synovitis of the shoulder: review and case report. Knee Surg Sports Traumatol Arthrosc 1999; 7:249.
  26. Mankin H, Trahan C, Hornicek F. Pigmented villonodular synovitis of joints. J Surg Oncol 2011; 103:386.
  27. Stephan SR, Shallop B, Lackman R, et al. Pigmented Villonodular Synovitis: A Comprehensive Review and Proposed Treatment Algorithm. JBJS Rev 2016; 4.
  28. Gouin F, Noailles T. Localized and diffuse forms of tenosynovial giant cell tumor (formerly giant cell tumor of the tendon sheath and pigmented villonodular synovitis). Orthop Traumatol Surg Res 2017; 103:S91.
  29. Bhimani MA, Wenz JF, Frassica FJ. Pigmented villonodular synovitis: keys to early diagnosis. Clin Orthop Relat Res 2001; :197.
  30. Dürr HR, Stäbler A, Maier M, Refior HJ. Pigmented villonodular synovitis. Review of 20 cases. J Rheumatol 2001; 28:1620.
  31. Hamlin BR, Duffy GP, Trousdale RT, Morrey BF. Total knee arthroplasty in patients who have pigmented villonodular synovitis. J Bone Joint Surg Am 1998; 80:76.
  32. Ogilvie-Harris DJ, Weisleder L. Arthroscopic synovectomy of the knee: is it helpful? Arthroscopy 1995; 11:91.
  33. Mollon B, Lee A, Busse JW, et al. The effect of surgical synovectomy and radiotherapy on the rate of recurrence of pigmented villonodular synovitis of the knee: an individual patient meta-analysis. Bone Joint J 2015; 97-B:550.
  34. Horoschak M, Tran PT, Bachireddy P, et al. External beam radiation therapy enhances local control in pigmented villonodular synovitis. Int J Radiat Oncol Biol Phys 2009; 75:183.
  35. Blanco CE, Leon HO, Guthrie TB. Combined partial arthroscopic synovectomy and radiation therapy for diffuse pigmented villonodular synovitis of the knee. Arthroscopy 2001; 17:527.
  36. Bisbinas I, De Silva U, Grimer RJ. Pigmented villonodular synovitis of the foot and ankle: a 12-year experience from a tertiary orthopedic Oncology Unit. J Foot Ankle Surg 2004; 43:407.
  37. Chin KR, Barr SJ, Winalski C, et al. Treatment of advanced primary and recurrent diffuse pigmented villonodular synovitis of the knee. J Bone Joint Surg Am 2002; 84-A:2192.
  38. Berger B, Ganswindt U, Bamberg M, Hehr T. External beam radiotherapy as postoperative treatment of diffuse pigmented villonodular synovitis. Int J Radiat Oncol Biol Phys 2007; 67:1130.
  39. Heyd R, Micke O, Berger B, et al. Radiation therapy for treatment of pigmented villonodular synovitis: results of a national patterns of care study. Int J Radiat Oncol Biol Phys 2010; 78:199.
  40. Griffin AM, Ferguson PC, Catton CN, et al. Long-term outcome of the treatment of high-risk tenosynovial giant cell tumor/pigmented villonodular synovitis with radiotherapy and surgery. Cancer 2012; 118:4901.
  41. Shabat S, Kollender Y, Merimsky O, et al. The use of surgery and yttrium 90 in the management of extensive and diffuse pigmented villonodular synovitis of large joints. Rheumatology (Oxford) 2002; 41:1113.
  42. Franssen MJ, Boerbooms AM, Karthaus RP, et al. Treatment of pigmented villonodular synovitis of the knee with yttrium-90 silicate: prospective evaluations by arthroscopy, histology, and 99mTc pertechnetate uptake measurements. Ann Rheum Dis 1989; 48:1007.
  43. Mastboom MJL, Palmerini E, Verspoor FGM, et al. Surgical outcomes of patients with diffuse-type tenosynovial giant-cell tumours: an international, retrospective, cohort study. Lancet Oncol 2019; 20:877.
  44. Tap WD, Gelderblom H, Palmerini E, et al. Pexidartinib versus placebo for advanced tenosynovial giant cell tumour (ENLIVEN): a randomised phase 3 trial. Lancet 2019; 394:478.
  45. West RB, Rubin BP, Miller MA, et al. A landscape effect in tenosynovial giant-cell tumor from activation of CSF1 expression by a translocation in a minority of tumor cells. Proc Natl Acad Sci U S A 2006; 103:690.
  46. Möller E, Mandahl N, Mertens F, Panagopoulos I. Molecular identification of COL6A3-CSF1 fusion transcripts in tenosynovial giant cell tumors. Genes Chromosomes Cancer 2008; 47:21.
  47. Tap WD, Wainberg ZA, Anthony SP, et al. Structure-Guided Blockade of CSF1R Kinase in Tenosynovial Giant-Cell Tumor. N Engl J Med 2015; 373:428.
  48. Kroot EJ, Kraan MC, Smeets TJ, et al. Tumour necrosis factor alpha blockade in treatment resistant pigmented villonodular synovitis. Ann Rheum Dis 2005; 64:497.
  49. Gelderblom H, Cropet C, Chevreau C, et al. Nilotinib in locally advanced pigmented villonodular synovitis: a multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol 2018; 19:639.
  50. Cassier PA, Italiano A, Gomez-Roca CA, et al. CSF1R inhibition with emactuzumab in locally advanced diffuse-type tenosynovial giant cell tumours of the soft tissue: a dose-escalation and dose-expansion phase 1 study. Lancet Oncol 2015; 16:949.
  51. Gouin F. Nilotinib in locally advanced pigmented villonodular synovitis: challenges of a new targeted therapy. Lancet Oncol 2018; 19:584.
  52. Gelderblom H, Wagner AJ, Tap WD, et al. Long-term outcomes of pexidartinib in tenosynovial giant cell tumors. Cancer 2021; 127:884.
  53. US Food and Drug Administration (FDA) prescribing guidelines for pexidartinib. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/211810s009lbl.pdf (Accessed on October 19, 2022).
  54. Lewis JH, Gelderblom H, van de Sande M, et al. Pexidartinib Long-Term Hepatic Safety Profile in Patients with Tenosynovial Giant Cell Tumors. Oncologist 2021; 26:e863.
  55. Blay JY, El Sayadi H, Thiesse P, et al. Complete response to imatinib in relapsing pigmented villonodular synovitis/tenosynovial giant cell tumor (PVNS/TGCT). Ann Oncol 2008; 19:821.
  56. Gelderblom H, Perol D, Chevreau C, et al. An open-label international multicentric phase II study of nilotinib in progressive pigmented villonodular synovitis (PVNS) not amenable to conservative surgical treatment (abstract). J Clin Oncol 2013; 31(suppl; abstr 10516). http://meetinglibrary.asco.org/content/117842-132 (Accessed on June 11, 2013).
  57. Cassier PA, Gelderblom H, Stacchiotti S, et al. Efficacy of imatinib mesylate for the treatment of locally advanced and/or metastatic tenosynovial giant cell tumor/pigmented villonodular synovitis. Cancer 2012; 118:1649.
  58. Cassier PA, Italiano A, Gomez-Roca C, et al. Long-term clinical activity, safety and patient-reported quality of life for emactuzumab-treated patients with diffuse-type tenosynovial giant-cell tumour. Eur J Cancer 2020; 141:162.
Topic 7733 Version 45.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟