Chapter 91 Radiotherapy of Nonmalignant Diseases


Keloids and Hypertrophic Scars



Yüklə 1,08 Mb.
səhifə5/8
tarix16.08.2018
ölçüsü1,08 Mb.
#63356
1   2   3   4   5   6   7   8

Keloids and Hypertrophic Scars

Background and Clinical Aspects

Keloids are an excessive tissue proliferation about scars after skin injury from surgery, heat, chemical burns, inflammation (e.g., acne), or even spontaneous proliferation. They differ from hypertrophic scars by their typical infiltrative growth pattern, causing local pain and inflammatory reactions, and sometimes long-term progression; hypertrophic scars show thickening without surrounding reaction and can flatten spontaneously. Keloids appear mostly in the upper body and in regions with high skin tension (e.g., sternum, earlobes). The cause is still unknown, although there is a genetic and race-specific predisposition that is already noted during adolescence. Keloids at the earlobe after piercing are typical. In some patients the resulting lesions are severely disfiguring and painful (Fig. 9). Recurrence is common after treatment



Nonradiotherapeutic Treatment

Silicone bandages, pressure dressings, and cryosurgery have all been used to treat for keloids with varying efficacy.232-234 Intralesional injections remain the first-line therapy for most keloids. Corticosteroids, 5FU, and verapamil have all been directly injected into keloid lesions with symptom improvement. Up to 70% of patients respond to intralesional corticosteroid injection with flattening of keloids, although the recurrence rate is high in some studies (up to 50 percent at five years).235

Surgical excision may be indicated if injection therapy alone does not result in improvement. In patients treated with excision alone, recurrence rates range from 45-100%,236 therefore excision is typically combined with peri-operative postoperative injections of either triamcinolone or interferon.235
Radiotherapeutic Options

Radiotherapy should be considered in cases of repeat recurrences postoperatively or where there is a high-risk of recurrence (e.g., marginal resection, large lesion, unfavorable location). Primary RT can be considered in instances where resection would result in functional impairment and in actively proliferating disorders within about 6 months after the triggering trauma. Because proliferating fibroblasts and mesenchymal and inflammatory cells are the target cells for RT, fully matured keloids have minimal response to RT alone. Prophylactic RT immediately following excision is most effective and reduces the risk of recurrence to 20% to 25% in most series.

RT is initiated 24 hours after surgery. The target volume is limited to the scar plus a 1-cm margin; lead shielding can be constructed to protect normal tissue. An analysis of multicenter data on the use of post-operative RT for earlobe keloids show that higher dose per fraction and use of deeper penetrating electrons is preferable to standard 2 Gy fractionation schemes or use of brachytherapy techniques that have rapid dose fall off.237 Radiation dose is typically 12 to 20 Gy, delivered in 3 or 4 fractions within 1 week.238 Single-fraction RT with 7.5 to 10 Gy is also effective.239 Clinical end points are long-term control, low relapse rate, and good cosmesis.
Diseases of Bone

Gorham-Stout Syndrome

Gorham-Stout Syndrome, also known as disappearing bone disease or essential osteolysis, is a rare bone disorder of unknown etiology. It is characterized by painless bone destruction due to progressive proliferation of small blood or lymph vessels. There may also be significant osteoclast activation. The symptoms are nonspecific, but include muscular weakness, limb tenderness, and pathologic fracture occurring minimal trauma. Involvement of the cervical spine or skull base could be fatal. Case reports indicate limited efficacy of systemic therapies such as zoledronic acid and interferon-alpha. Radiation therapy has also been used. Heyd, et al. completed a national patterns-of-care study and literature review that summarizes the scant data available for this rare disorder.244 The 38 articles listed therein provide evidence from treatment of 44 patients that conventionally fractionated external beam RT (total dose of 36 to 45 Gy) may prevent disease progression in 77% to 80% of cases.


Pigmented villonodularsynovitis

Pigmented villonodular synovitis or tenosynovial giant cell tumor is a rare proliferative disorder of synovial tissue. Symptoms include sudden onset, unexplained joint swelling and pain that frequentlyinvolves a single joint. The knee and foot are most commonly affected, but there are reports of shoulder, hand, and hip involvement.245 Decreased motion, joint stiffness, and increased pain occur as the disorder progresses. Surgical resection with either synovectomy or joint replacement is the treatment of choice.

Radiation therapy is indicated in cases of diffuse disease, bulky disease resulting in bone destruction, or in the rare instance of multiple recurrences after resection. Although intra-synovial injection of radioactive isotopes post-operatively has been used in the past for high-risk patients,248 most institutions use external beam radiation therapy. RT to a dose of 35 to 50 Gy has been effective. MRI is essential for delineating disease pre- and post-operatively. Final dose of RT should be tailored to amount of residual disease.251

Vertebral Hemangiomas

Hemangiomas are benign proliferations of blood vessels that can affect any tissue and are typically asymptomatic. About 50% of hemangiomas involving the vertebral body are associated with pain and therefore may require treatment. Treatment options include surgical resection or more conservative interventions such as vetebroplasty or intralesional injections.252 Radiation therapy either alone or post-operatively has been successful in reducing pain caused by vertebral hemangiomas.253 In this study, a total of 84 patients with 96 symptomatic lesions were irradiated for a symptomatic vertebral hemangioma. At a median 68 months follow-up, 90% of patients had either complete or partial pain relief. Radiation doses ≥34 Gy resulted in significantly improved pain relief. A total radiation dose of 36-40 Gy delivered in 2 Gy per fraction has been recommended.254


Heterotopic Ossification

Background and Clinical Aspects

Heterotopic ossification (HO) is a common complication of total hip arthroplasty, hip trauma, or acetabular fracture. HO occurs when the soft tissues around the hip become ossified. Following trauma, primitive mesenchymal cells in the surrounding soft tissues are transformed into osteoblastic tissue that then forms mature bone. The hip is the most common joint affected; HO typically occurs around the femoral neck and adjacent to the greater trochanter. The risk factors for development of HO are unknown, but the incidence is greater in men and occurs in more than 80% in patients who have a history of ipsilateral or contralateral HO. It is also more common in patients with a known history of osteoarthritis, ankylosing spondylitis, diffuse and Paget’s disease.255 Hip stiffness is the primary symptom and the diagnosis is made radiographically. Pain is typically not associated with HO.



Nonradiotherapeutic Treatment

The treatment for HO is surgical excision followed by some form of HO prophylaxis. Prophylaxis is only applied to patients at high risk for developing HO. A meta-analysis showed that NSAIDs are effective in reducing the risk of post-operative HO.256 Indomethacin is the most commonly used NSAID for HO prophylaxis. Indomethacin is a prostaglandin synthase inhibitor that also suppresses mesenchymal cells. The limited data available have not shown a clear benefit to the use of selective cyclooxygenase-2 inhibitors in HO prophylaxis. Bisphosphonates have been used for prophylaxis because they delay mineralization of osteoid and appear to have some efficacy in preventing HO if used at the appropriate time. In one study, the cost of bisophosphonate use was prohibitive for routine use when compared to indomethacin.259
Radiotherapeutic Options

External-beam radiation is an effective method for prevention of HO after total hip arthroplasty. Prophylactic radiation therapy for the prevention of HO has been used sine the 1970s. A single fraction of 700 or 800 cGy to the at-risk region (Figure 17) is recommended and should be delivered in the peri-operative period, either preoperatively (within 24 hours) or postoperatively (within 72 hours).260-262 When comparing radiation therapy and NSAIDs, there is no clear benefit for use of one modality over another. A prospective, randomized study demonstrated that radiation therapy and indomethacin are both effective in the prevention of postoperative HO.263 Although one meta-analysis of seven randomized studies concluded that radiotherapy is more effective than NSAIDs for HO prophylaxis,264 a more recent analysis of 9 studies involving 1295 patients found no statistically significant difference between the two.265 An economic analysis using the same 9 studies and the meta-analysis suggest that radiation therapy is not cost effective when compared to use of NSAIDs.266 This analysis has yet to be validated.
REFERENCES:

* = Selected Key References



1. Bahn RS. Graves' ophthalmopathy. N Engl J Med. Feb 25 2010;362(8):726-738.

2. Platta CS, Mackay C, Welsh JS. Pituitary adenoma: a radiotherapeutic perspective. Am J Clin Oncol. Aug 2010;33(4):408-419.

3. Shields CJ, Winter DC, Kirwan WO, Redmond HP. Desmoid tumours. Eur J Surg Oncol. Dec 2001;27(8):701-706.

4. Hessenbruch A. A brief history of x-rays. Endeavour. Dec 2002;26(4):137-141.

5. Mehta MP, Goetowski PG, Kinsella TJ. Radiation induced thyroid neoplasms 1920 to 1987: a vanishing problem? Int J Radiat Oncol Biol Phys. Jun 1989;16(6):1471-1475.

6. Inskip PD. Thyroid cancer after radiotherapy for childhood cancer. Med Pediatr Oncol. May 2001;36(5):568-573.

7. Glicksman AS. Malignant radiation of benign conditions. Ann Intern Med. Jul 1978;89(1):130-131.

8. Trott KR, Kamprad F. Radiobiological mechanisms of anti-inflammatory radiotherapy. Radiother Oncol. Jun 1999;51(3):197-203.

9. Rodel F, Keilholz L, Herrmann M, Sauer R, Hildebrandt G. Radiobiological mechanisms in inflammatory diseases of low-dose radiation therapy. Int J Radiat Biol. Jun 2007;83(6):357-366.

10. Hill RP, Rodemann HP, Hendry JH, Roberts SA, Anscher MS. Normal tissue radiobiology: from the laboratory to the clinic. Int J Radiat Oncol Biol Phys. Feb 1 2001;49(2):353-365.

11. Draper GJ, Sanders BM, Kingston JE. Second primary neoplasms in patients with retinoblastoma. Br J Cancer. May 1986;53(5):661-671.

12. Kleinerman RA, Tucker MA, Abramson DH, Seddon JM, Tarone RE, Fraumeni JF, Jr. Risk of soft tissue sarcomas by individual subtype in survivors of hereditary retinoblastoma. J Natl Cancer Inst. Jan 3 2007;99(1):24-31.

13. Leer JW, van Houtte P, Seegenschmiedt H. Radiotherapy of non-malignant disorders: where do we stand? Radiother Oncol. May 2007;83(2):175-177.

14. Trott KR, Kamprad F. Estimation of cancer risks from radiotherapy of benign diseases. Strahlenther Onkol. Aug 2006;182(8):431-436.

15. Jansen JT, Broerse JJ, Zoetelief J, Klein C, Seegenschmiedt HM. Estimation of the carcinogenic risk of radiotherapy of benign diseases from shoulder to heel. Radiother Oncol. Sep 2005;76(3):270-277.

16. Leer JW, van Houtte P, Davelaar J. Indications and treatment schedules for irradiation of benign diseases: a survey. Radiother Oncol. Sep 1998;48(3):249-257.

17. Seegenschmiedt MH, Katalinic A, Makoski H, Haase W, Gademann G, Hassenstein E. Radiation therapy for benign diseases: patterns of care study in Germany. Int J Radiat Oncol Biol Phys. Apr 1 2000;47(1):195-202.

18. Micke O, Seegenschmiedt MH. Consensus guidelines for radiation therapy of benign diseases: a multicenter approach in Germany. Int J Radiat Oncol Biol Phys. Feb 1 2002;52(2):496-513.

19. Mitsuhashi N, Furuta M, Sakurai H, et al. Outcome of radiation therapy for patients with Kasabach-Merritt syndrome. Int J Radiat Oncol Biol Phys. Sep 1 1997;39(2):467-473.

20. Louis DN OH, Wiestler OD, et al. WHO classification of tumours of the nervous system. Lyon IARC Press; 2007.

21. Morokoff AP, Zauberman J, Black PM. Surgery for convexity meningiomas. Neurosurgery. Sep 2008;63(3):427-433; discussion 433-424.

22. Nakamura M, Struck M, Roser F, Vorkapic P, Samii M. Olfactory groove meningiomas: clinical outcome and recurrence rates after tumor removal through the frontolateral and bifrontal approach. Neurosurgery. May 2007;60(5):844-852; discussion 844-852.

23. Simpson D. The recurrence of intracranial meningiomas after surgical treatment. J Neurol Neurosurg Psychiatry. Feb 1957;20(1):22-39.

24. Dowd CF, Halbach VV, Higashida RT. Meningiomas: the role of preoperative angiography and embolization. Neurosurg Focus. Jul 15 2003;15(1):E10.

25. Oka H, Kurata A, Kawano N, et al. Preoperative superselective embolization of skull-base meningiomas: indications and limitations. J Neurooncol. Oct 1998;40(1):67-71.

26. Yano S, Kuratsu J. Indications for surgery in patients with asymptomatic meningiomas based on an extensive experience. J Neurosurg. Oct 2006;105(4):538-543.

27. Wen PY, Quant E, Drappatz J, Beroukhim R, Norden AD. Medical therapies for meningiomas. J Neurooncol. Sep 2010;99(3):365-378.

28. Goldsmith BJ, Wara WM, Wilson CB, Larson DA. Postoperative irradiation for subtotally resected meningiomas. A retrospective analysis of 140 patients treated from 1967 to 1990. J Neurosurg. Feb 1994;80(2):195-201.

29. Mendenhall WM, Morris CG, Amdur RJ, Foote KD, Friedman WA. Radiotherapy alone or after subtotal resection for benign skull base meningiomas. Cancer. Oct 1 2003;98(7):1473-1482.

30. Minniti G, Amichetti M, Enrici RM. Radiotherapy and radiosurgery for benign skull base meningiomas. Radiat Oncol. 2009;4:42.

31. Nutting C, Brada M, Brazil L, et al. Radiotherapy in the treatment of benign meningioma of the skull base. J Neurosurg. May 1999;90(5):823-827.

32. Vendrely V, Maire JP, Darrouzet V, et al. [Fractionated radiotherapy of intracranial meningiomas: 15 years' experience at the Bordeaux University Hospital Center]. Cancer Radiother. Jul-Aug 1999;3(4):311-317.

33. Adler JR, Jr., Gibbs IC, Puataweepong P, Chang SD. Visual field preservation after multisession cyberknife radiosurgery for perioptic lesions. Neurosurgery. Feb 2008;62 Suppl 2:733-743.

34. Paulsen F, Doerr S, Wilhelm H, Becker G, Bamberg M, Classen J. Fractionated Stereotactic Radiotherapy in Patients with Optic Nerve Sheath Meningioma. Int J Radiat Oncol Biol Phys. Feb 5 2011.

35. Davidson L, Fishback D, Russin JJ, et al. Postoperative Gamma Knife surgery for benign meningiomas of the cranial base. Neurosurg Focus. 2007;23(4):E6.

36. Feigl GC, Samii M, Horstmann GA. Volumetric follow-up of meningiomas: a quantitative method to evaluate treatment outcome of gamma knife radiosurgery. Neurosurgery. Aug 2007;61(2):281-286; discussion 286-287.

37. Ganz JC, Reda WA, Abdelkarim K. Gamma Knife surgery of large meningiomas: early response to treatment. Acta Neurochir (Wien). Jan 2009;151(1):1-8.

38. Han JH, Kim DG, Chung HT, et al. Gamma knife radiosurgery for skull base meningiomas: long-term radiologic and clinical outcome. Int J Radiat Oncol Biol Phys. Dec 1 2008;72(5):1324-1332.

39. Hasegawa T, Kida Y, Yoshimoto M, Koike J, Iizuka H, Ishii D. Long-term outcomes of Gamma Knife surgery for cavernous sinus meningioma. J Neurosurg. Oct 2007;107(4):745-751.

40. Iwai Y, Yamanaka K, Ikeda H. Gamma Knife radiosurgery for skull base meningioma: long-term results of low-dose treatment. J Neurosurg. Nov 2008;109(5):804-810.

41. Kollova A, Liscak R, Novotny J, Jr., Vladyka V, Simonova G, Janouskova L. Gamma Knife surgery for benign meningioma. J Neurosurg. Aug 2007;107(2):325-336.

42. Kondziolka D, Mathieu D, Lunsford LD, et al. Radiosurgery as definitive management of intracranial meningiomas. Neurosurgery. Jan 2008;62(1):53-58; discussion 58-60.

43. Takanashi M, Fukuoka S, Hojyo A, Sasaki T, Nakagawara J, Nakamura H. Gamma knife radiosurgery for skull-base meningiomas. Prog Neurol Surg. 2009;22:96-111.

44. Zachenhofer I, Wolfsberger S, Aichholzer M, et al. Gamma-knife radiosurgery for cranial base meningiomas: experience of tumor control, clinical course, and morbidity in a follow-up of more than 8 years. Neurosurgery. Jan 2006;58(1):28-36; discussion 28-36.

45. DiBiase SJ, Kwok Y, Yovino S, et al. Factors predicting local tumor control after gamma knife stereotactic radiosurgery for benign intracranial meningiomas. Int J Radiat Oncol Biol Phys. Dec 1 2004;60(5):1515-1519.

46. Noel G, Bollet MA, Calugaru V, et al. Functional outcome of patients with benign meningioma treated by 3D conformal irradiation with a combination of photons and protons. Int J Radiat Oncol Biol Phys. Aug 1 2005;62(5):1412-1422.

47. Vernimmen FJ, Harris JK, Wilson JA, Melvill R, Smit BJ, Slabbert JP. Stereotactic proton beam therapy of skull base meningiomas. Int J Radiat Oncol Biol Phys. Jan 1 2001;49(1):99-105.

48. Weber DC, Lomax AJ, Rutz HP, et al. Spot-scanning proton radiation therapy for recurrent, residual or untreated intracranial meningiomas. Radiother Oncol. Jun 2004;71(3):251-258.

49. Wenkel E, Thornton AF, Finkelstein D, et al. Benign meningioma: partially resected, biopsied, and recurrent intracranial tumors treated with combined proton and photon radiotherapy. Int J Radiat Oncol Biol Phys. Dec 1 2000;48(5):1363-1370.

50. Laws ER, Sheehan JP, Sheehan JM, Jagnathan J, Jane JA, Jr., Oskouian R. Stereotactic radiosurgery for pituitary adenomas: a review of the literature. J Neurooncol. Aug-Sep 2004;69(1-3):257-272.

51. Brada M, Rajan B, Traish D, et al. The long-term efficacy of conservative surgery and radiotherapy in the control of pituitary adenomas. Clin Endocrinol (Oxf). Jun 1993;38(6):571-578.

52. Breen P, Flickinger JC, Kondziolka D, Martinez AJ. Radiotherapy for nonfunctional pituitary adenoma: analysis of long-term tumor control. J Neurosurg. Dec 1998;89(6):933-938.

53. Gittoes NJ, Bates AS, Tse W, et al. Radiotherapy for non-function pituitary tumours. Clin Endocrinol (Oxf). Mar 1998;48(3):331-337.

54. Littley MD, Shalet SM, Beardwell CG, Ahmed SR, Applegate G, Sutton ML. Hypopituitarism following external radiotherapy for pituitary tumours in adults. Q J Med. Feb 1989;70(262):145-160.

55. Landolt AM, Haller D, Lomax N, et al. Octreotide may act as a radioprotective agent in acromegaly. J Clin Endocrinol Metab. Mar 2000;85(3):1287-1289.

56. Sheehan JP, Kondziolka D, Flickinger J, Lunsford LD. Radiosurgery for residual or recurrent nonfunctioning pituitary adenoma. J Neurosurg. Dec 2002;97(5 Suppl):408-414.

57. Stripp DC, Maity A, Janss AJ, et al. Surgery with or without radiation therapy in the management of craniopharyngiomas in children and young adults. Int J Radiat Oncol Biol Phys. Mar 1 2004;58(3):714-720.

58. Luu QT, Loredo LN, Archambeau JO, Yonemoto LT, Slater JM, Slater JD. Fractionated proton radiation treatment for pediatric craniopharyngioma: preliminary report. Cancer J. Mar-Apr 2006;12(2):155-159.

59. Winkfield KM, Linsenmeier C, Yock TI, et al. Surveillance of craniopharyngioma cyst growth in children treated with proton radiotherapy. Int J Radiat Oncol Biol Phys. Mar 1 2009;73(3):716-721.

60. Lee M, Kalani MY, Cheshier S, Gibbs IC, Adler JR, Chang SD. Radiation therapy and CyberKnife radiosurgery in the management of craniopharyngiomas. Neurosurg Focus. 2008;24(5):E4.

61. Chiou SM, Lunsford LD, Niranjan A, Kondziolka D, Flickinger JC. Stereotactic radiosurgery of residual or recurrent craniopharyngioma, after surgery, with or without radiation therapy. Neuro Oncol. Jul 2001;3(3):159-166.

62. Kobayashi T, Kida Y, Mori Y, Hasegawa T. Long-term results of gamma knife surgery for the treatment of craniopharyngioma in 98 consecutive cases. J Neurosurg. Dec 2005;103(6 Suppl):482-488.

63. Selch MT, DeSalles AA, Wade M, et al. Initial clinical results of stereotactic radiotherapy for the treatment of craniopharyngiomas. Technol Cancer Res Treat. Feb 2002;1(1):51-59.

64. Ulfarsson E, Lindquist C, Roberts M, et al. Gamma knife radiosurgery for craniopharyngiomas: long-term results in the first Swedish patients. J Neurosurg. Dec 2002;97(5 Suppl):613-622.

65. Hasegawa T, Kondziolka D, Hadjipanayis CG, Lunsford LD. Management of cystic craniopharyngiomas with phosphorus-32 intracavitary irradiation. Neurosurgery. Apr 2004;54(4):813-820; discussion 820-812.

66. Voges J, Sturm V, Lehrke R, Treuer H, Gauss C, Berthold F. Cystic craniopharyngioma: long-term results after intracavitary irradiation with stereotactically applied colloidal beta-emitting radioactive sources. Neurosurgery. Feb 1997;40(2):263-269; discussion 269-270.

67. Gormley WB, Sekhar LN, Wright DC, Kamerer D, Schessel D. Acoustic neuromas: results of current surgical management. Neurosurgery. Jul 1997;41(1):50-58; discussion 58-60.

68. Samii M, Matthies C. Management of 1000 vestibular schwannomas (acoustic neuromas): the facial nerve--preservation and restitution of function. Neurosurgery. Apr 1997;40(4):684-694; discussion 694-685.

69. Kondziolka D, Lunsford LD, McLaughlin MR, Flickinger JC. Long-term outcomes after radiosurgery for acoustic neuromas. N Engl J Med. Nov 12 1998;339(20):1426-1433.

70. Flickinger JC, Kondziolka D, Niranjan A, Maitz A, Voynov G, Lunsford LD. Acoustic neuroma radiosurgery with marginal tumor doses of 12 to 13 Gy. Int J Radiat Oncol Biol Phys. Sep 1 2004;60(1):225-230.

71. Hansasuta A, Choi CY, Gibbs IC, et al. Multi-session Stereotactic Radiosurgery for Vestibular Schwannomas: Single Institution Experience with 383 Cases. Neurosurgery. May 6 2011.

72. Meijer OW, Vandertop WP, Baayen JC, Slotman BJ. Single-fraction vs. fractionated linac-based stereotactic radiosurgery for vestibular schwannoma: a single-institution study. Int J Radiat Oncol Biol Phys. Aug 1 2003;56(5):1390-1396.

Yüklə 1,08 Mb.

Dostları ilə paylaş:
1   2   3   4   5   6   7   8




Verilənlər bazası müəlliflik hüququ ilə müdafiə olunur ©www.genderi.org 2024
rəhbərliyinə müraciət

    Ana səhifə