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

Clinical manifestations, prevention, and treatment of radiation-induced fibrosis

Clinical manifestations, prevention, and treatment of radiation-induced fibrosis
Author:
Elisabeth Weiss, MD
Section Editor:
Steven E Schild, MD
Deputy Editor:
Sadhna R Vora, MD
Literature review current through: Jan 2024.
This topic last updated: Jan 24, 2023.

INTRODUCTION — Radiation-induced fibrosis can develop as a late effect of radiation therapy (RT) in skin and subcutaneous tissue, the lungs, the gastrointestinal and genitourinary tracts, muscles, or other organs, depending upon the treatment site. Radiation-induced fibrosis may cause both cosmetic and functional impairment, which can lead to death or significant deterioration in quality of life.

The development of radiation-induced fibrosis is influenced by multiple factors, including radiation dose and volume, fractionation schedule, previous or concurrent treatments, genetic susceptibility, and comorbidities such as diabetes mellitus. Although radiation-induced fibrosis originally was assumed to be a slow, irreversible process, contemporary studies suggest that it is not necessarily a fixed process.

The prevention of radiation-induced fibrosis has focused on improvements in RT technique, which have resulted in higher doses to the tumor target and decreased doses to normal tissue, thus potentially preventing the development of radiation-induced fibrosis. Furthermore, established radiation-induced fibrosis may be treatable with novel therapeutic approaches, particularly the combination of pentoxifylline and vitamin E.

The pathogenesis, diagnosis, clinical manifestations, prevention, and treatment of radiation-induced fibrosis will be discussed here. Radiation-induced changes in the lungs and gastrointestinal tract are discussed separately. (See "Radiation-induced lung injury" and "Diagnosis and management of chronic radiation enteritis".)

PATHOGENESIS — Radiation-induced fibrosis is similar to inflammation, wound healing, and fibrosis of any origin. Typical histologic features include the presence of inflammatory infiltrates, particularly macrophages in the earlier stages of fibrosis, differentiation of fibroblasts into postmitotic fibrocytes, and changes in the vascular connective tissue with excessive production and deposition of extracellular matrix proteins and collagen [1,2].

The development of fibrosis is mediated by cytokine expression that probably begins immediately after radiation therapy (RT) and continues for months or years. Inflammatory mediators that have been implicated include tumor necrosis factor-alpha (TNF-alpha) and interleukins 6 and 1 (IL-6 and IL-1) [2-5]. Fibrogenic cytokines that modulate the proliferation and differentiation of fibroblasts and the synthesis of extracellular matrix proteins and matrix metalloproteinases include transforming growth factor beta (TGF-beta) and platelet-derived growth factor (PDGF). Other growth factors, such as the connective tissue growth factor (CTGF), which is secreted by fibroblasts and endothelial cells, also promote fibrosis [6,7].

Radiation-induced alterations of the vascular system, particularly damage to endothelial cells, appear to contribute significantly to late toxicity. Damage to the myocardium after RT for breast cancer has been associated with changes of the microvasculature, resulting in reduced perfusion and fibrosis [8]. Hyalinoid changes of the capillary wall and a rarefication of the capillary net are typically seen in fibrotic areas [9].

ETIOLOGIC FACTORS — A number of factors influence the response to radiation, thus increasing the incidence of complications.

RT parameters — The risk of radiation therapy (RT)-induced fibrosis is increased with higher radiation doses and larger treated volumes [10-13]. The radiation dose that causes fibrosis can vary substantially in different tissues, as is illustrated by the following examples:

Fibrosis in both connective and vascular tissues is generally associated with total radiation doses of 60 Gy or higher.

By contrast, the heart is relatively sensitive to radiation, and relatively low doses of radiation therapy (RT) can cause peri-, myo-, or endocardial fibrosis. Thus, incidental radiation to the heart should be minimized, and radiation doses to the whole heart should be kept small. Similarly, the lungs are relatively sensitive to radiation. (See "Cardiotoxicity of radiation therapy for breast cancer and other malignancies" and "Radiation-induced lung injury".)

The technique of RT administration can have a major effect on the dose of radiation received by normal tissues. Examples of situations where the risk of radiation-induced fibrosis is influenced by the RT technique include the following:

Skin induration is more frequent in women treated with overlapping tangential breast fields and internal mammary lymph node fields [14].

The risk of radiation-induced fibrosis is increased with high single doses of RT (eg, intraoperative radiation treatment, single-fraction external beam RT, brachytherapy) [15].

Fibrosis may be more common when different RT techniques are combined. As an example, the combination of external beam RT with interstitial and intracavitary brachytherapy for gynecological tumors can cause radiation-induced fibrosis of the ureter and vagina, as well as late toxicity to the large and small bowel [16,17].

The use of multiple fractions per day increases the risk of radiation-induced fibrosis. In a randomized trial in patients with lung cancer, the risk of dysphagia was higher two years after treatment with hyperfractionated RT compared with conventional fractionation [18].

Postoperative, as opposed to preoperative, administration of RT is associated with an increased risk of late radiation complications in sarcoma patients [10]. In men with prostate cancer, patients with a history of abdominal surgery or a transurethral resection of the prostate (TURP) had an increased risk for late RT complications [19].

Higher doses of RT not only increase the incidence of fibrosis, but also shorten the latent period until late sequelae are apparent [9].

During RT planning, organ-specific guidelines are used to limit the dose to normal tissue. With this information, the risk for radiation-related sequelae can be estimated, and volume-dependent dose limits can be generated.

Genetic factors — Genetic variation in the response to radiation may be associated with an increased incidence of radiation-induced fibrosis.

As an example, there is a higher risk of subcutaneous fibrosis in patients with breast cancer who have specific modifications of the ATM gene [20]. Patients with other diseases associated with deficient DNA repair may also be more likely to develop symptoms of radiation-induced fibrosis. Fortunately, dramatic increases in the risk of radiation fibrosis are limited to the rare occurrence of these known genetic disorders.

Connective tissue diseases — Patients with connective tissue diseases, such as scleroderma, rheumatoid arthritis, or systemic lupus erythematosus, may have an increased incidence of toxicity from RT, but the magnitude of this risk is not clear [21-23]. In a review of eight observational studies that included 404 patients with connective tissue diseases who were treated with RT, there was a statistically significant association with late RT-induced complications in normal tissues (fibrosis, osteonecrosis, bone fractures) [21]. While not usually an absolute contraindication to RT, the severity of connective tissue disease should be taken into account when formulating the treatment plan.

Multiple factors may contribute to the association of RT complications with connective tissue diseases. The development of radiation-induced fibrosis may be affected by the concurrent use of medications such as corticosteroids and cytotoxic drugs. In addition to increasing RT toxicity, these drugs can cause osteopenia and bone fractures, which may be attributed to RT. Sequelae of connective tissue disorders, such as dry eye and dry mouth symptoms in Sjögren's disease, may mimic radiation-induced fibrosis.

Concurrent systemic therapy — Combining RT with systemic cancer treatment has been assumed to cause a higher risk of radiation-induced toxicities [24], but clinical studies have given conflicting results. The following illustrate the range of results that have been reported:

In a randomized trial comparing sequential with concurrent chemotherapy in breast cancer patients receiving RT, the risk of late effects, including subcutaneous fibrosis, was significantly increased in those receiving concurrent chemotherapy [25].

A review of eight studies of women with cervical cancer who were treated either with RT plus chemotherapy or with RT alone found that there were no significant differences in the rates of late complications in seven of eight trials [26].

In women with breast cancer, the incidence of lung fibrosis was increased after combined RT and tamoxifen. In a trial of women receiving RT with or without tamoxifen following mastectomy, those receiving tamoxifen had a significant increase in the incidence of lung fibrosis (relative risk 2.0) [27]. Tamoxifen is thought to induce transforming growth factor beta (TGF-beta) secretion, which is a key factor in the signaling cascade leading to fibrosis. Tamoxifen may also increase the incidence of subcutaneous fibrosis when given concurrently with RT, but this has not been a consistent finding in all studies [28,29].

In men, hormonal therapy given concurrently with RT for prostate cancer can increase the incidence of acute and late gastrointestinal and genitourinary toxicity [19,30-32].

Targeted therapies are increasingly combined with RT for a wide variety of tumor entities. While often the combination was tolerated well, increased toxicities have repeatedly been reported [33] (eg, erlotinib [34,35], bevacizumab and erlotinib [36], bevacizumab and oxaliplatin [37], BRAF inhibitors [38]).

Immune-related pneumonitis from therapy with checkpoint inhibitors, such as pembrolizumab, was observed more frequently in lung cancer patients who had previous RT, particularly if treated with curative-intent RT [39,40].

Diabetes mellitus — Diabetes mellitus increases the risk of late RT complications in normal tissue. The increased risk has been attributed to changes in the micro- and macrovasculature, which result in decreased tissue perfusion and impaired normal tissue repair.

Several reports have described a higher risk for gastrointestinal and particularly genitourinary toxicity in diabetic patients [41,42]. Diabetes mellitus has also been identified as a risk factor for the development of ischemic heart disease following breast RT [43] and for radiation pneumonitis [44,45]. Patients with severe diabetes have even been reported to be excluded from stereotactic RT for lung cancer in one clinic, but this is not common practice [46].

CLINICAL MANIFESTATIONS — Radiation-induced fibrosis can cause a wide range of clinical manifestations, including cutaneous induration, lymphedema, restrictions in joint motion, strictures and stenoses in hollow organs, and ulcerations. An overview of common manifestations of radiation-induced fibrosis in various organ systems and the associated clinical symptoms is presented in the table (table 1). Other radiation-induced fibrosis-related symptoms exist but are less frequent.

Radiation-induced fibrosis typically presents 4 to 12 months after radiation therapy (RT) and progresses over several years. Although radiation-induced fibrosis can occur without acute symptoms during or immediately after RT, late toxicity often follows acute reactions in the same organ, either immediately or after a latency period.

Late toxicities following acute effects are termed consequential late effects and are directly dependent upon the degree and duration of acute toxicities. Consequential late effects are well described for bladder, bowel, lung, skin and mucosal membranes [47].

Skin and subcutaneous tissue — Radiation-induced fibrosis of the skin and subcutaneous tissue is seen most commonly in breast cancer patients in areas with overlapping treatment fields following breast-conserving surgery with postoperative RT or after mastectomy and RT. (See "Overview of long-term complications of therapy in breast cancer survivors and patterns of relapse", section on 'Chest wall and breast complications' and "Radiation therapy techniques for newly diagnosed, non-metastatic breast cancer".)

The risk for developing radiation-induced fibrosis after conventional RT for breast cancer is low, particularly with the use of modern skin-sparing megavoltage equipment. Signs and symptoms can include skin retraction and induration, pain, necrosis and ulceration, restricted arm and neck movement, lymphedema, and brachial or cervical plexopathy [48]. The risk for radiation-induced fibrosis and telangiectasia after brachytherapy alone seems to depend on the skin dose delivered [15,49,50]. Proton therapy also did not increase the risk for radiation fibrosis compared with accelerated partial breast irradiation with photons [51], although higher skin toxicity was observed. Adding a radiation boost to the tumor bed after whole breast RT improves local control, but also increases the rate of fibrosis in breast cancer patients [52]. (See "Overview of cancer pain syndromes".)

Radiation-induced fibrosis of the chest wall has also been reported following stereotactic body RT for lung lesions. (See "Radiation therapy techniques in cancer treatment", section on 'Stereotactic radiation therapy techniques'.)

Head and neck — Trismus is a frequent late effect that occurs in 8 to 35 percent of head and neck cancer patients following RT, which is caused by inflammation and fibrosis in the muscles of mastication [53]. Radiation-induced fibrosis results in a loss of the ability to fully open the mouth, which can progress over time. The risk of radiation-induced fibrosis in the head and neck region is increased in patients who have had surgery, and in those treated with RT for recurrent tumor. (See "Management of late complications of head and neck cancer and its treatment", section on 'Trismus'.)

In addition to trismus, RT can cause skin and subcutaneous fibrosis, resulting in induration and lymphedema. (See "Management of late complications of head and neck cancer and its treatment", section on 'Lymphedema and fibrosis'.)

Gastrointestinal tract — The radiation sensitivity of different parts of the gastrointestinal tract differs, with tolerance doses varying between 40 to 70 Gy. The manifestations of toxicity are dependent upon the area irradiated.

Radiation-induced fibrosis in the esophagus may lead to strictures, ulcerations, and fistula formation. The risk of complications may be increased when RT is combined with chemotherapy, with up to 20 percent of patients developing strictures after combined modality treatment [16]. (See "Overview of gastrointestinal toxicity of radiation therapy", section on 'Esophagitis'.)

Late small bowel toxicity is more common after pelvic RT, with its higher doses of RT, rather than after abdominal irradiation. The incidence of radiation-induced fibrosis is low after pelvic RT doses of 50 Gy, whereas a 5 percent incidence of small bowel toxicity has been reported with pelvic irradiation to doses >70 Gy for gynecologic tumors [54]. (See "Diagnosis and management of chronic radiation enteritis".)

Radiation-induced fibrosis of the rectum resulting in strictures occurs primarily in patients with cervix or prostate cancer when high curative doses of RT are used. (See "Radiation proctitis: Clinical manifestations, diagnosis, and management".)

Genitourinary tract — Radiation-induced changes can affect the bladder, ureters, urethra, or vagina depending upon the site and dose of RT. Examples include the following:

Fibrotic constriction of the bladder can occur after doses of 65 to 70 Gy to small treatment volumes and after lower doses when the entire bladder is irradiated. Another potential late complication of bladder irradiation is hemorrhagic cystitis.

Ureteral strictures are rare but occur in 1 to 3 percent of cases after gynecological treatment involving brachytherapy, which delivers high single doses to the distal ureters [17].

In men treated with 60 to 70 Gy for prostate cancer, the incidence of urethral strictures is 0 to 5 percent for those without a prior transurethral resection of the prostate (TURP) and 5 to 15 percent with an antecedent TURP [17]. Interstitial brachytherapy for prostate cancer results in similar urethral stricture rates [55].

External beam RT and brachytherapy for gynecologic tumors can cause radiation-induced fibrosis of the vulva and vagina, with symptomatic shortening and narrowing of the vagina, dryness, and dyspareunia in up to 70 percent of patients [56]. Symptoms may also be seen after RT for other pelvic tumors if the female reproductive tract is included in the RT treatment fields.

DIAGNOSIS AND ASSESSMENT — The diagnosis and assessment of severity of radiation-induced fibrosis depends upon the tissues involved.

Fibrosis of the skin and subcutaneous tissue is usually diagnosed by palpation and inspection. Radiation-induced fibrosis is limited to the volume treated with radiation therapy (RT). To quantify the extent of radiation-induced fibrosis, the surface area and depth of involvement should be measured. This evaluation can be repeated to assess the potential effect of any therapeutic intervention.

Endoscopy or imaging with contrast agents (eg, esophagograms) can be used to demonstrate the loss of elasticity of an organ wall as a result of radiation-induced fibrosis.

Ultrasound may quantify the extent of radiation-induced fibrosis in the neck [57,58].

In other anatomic regions, functional tests (eg, for salivary gland function or nerve function in plexus lesions associated with radiation-induced fibrosis) might be helpful, but are non-specific for the diagnosis of radiation-induced fibrosis.

The Radiation Therapy Oncology Group has developed organ-specific classification systems that can be used to describe the severity of RT side effects [59].

The differential diagnosis of radiation-induced fibrosis often includes recurrent tumor, and magnetic resonance imaging (MRI) may be useful in excluding this possibility [60,61]. Persistent parenchymal enhancement (late gadolinium enhancement) after MRI contrast application is associated with areas of local fibrosis. T1 mapping is being investigated to assess diffuse fibrosis [62].

Biopsies and other surgical interventions in areas with radiation fibrosis should be performed only when necessary to exclude tumor recurrence, eg, in situations with clinically unclear changes on repeated imaging or in the presence of ulcerations, since wound healing may be impaired in previously irradiated areas [9]. Furthermore, surgery may result in worsening of fibrosis.

PREVENTION

Radiation therapy techniques — The prevention of radiation-induced fibrosis depends upon minimizing the dose and volume of normal tissue irradiated.

With modern conformal radiation therapy (RT) techniques, high doses are delivered to the tumor while normal tissue should largely be spared [63,64]. Using intensity-modulated radiation therapy (IMRT), a long-term reduction in the incidence of radiation-induced late effects, such as breast induration [65] and telangiectasis [66] for breast cancer RT [67,68], and reduced xerostomia [69] for head and neck cancer were observed. In addition, low rates of lung fibrosis were observed following IMRT for lung cancer [70]. However, the effect of breast IMRT on quality of life has not yet been clearly demonstrated because of the important role of postsurgical cosmesis and breast volume. Using IMRT, a long-term reduction in the incidence of radiation-induced fibrosis should be expected particularly for breast [71] and has been shown for head and neck cancer patients [72,73].

Proton beam therapy is an increasingly available RT technique that potentially can spare neighboring critical structures better than RT using photons, eg, for patients with locally advanced lung cancer where toxicity of the heart and esophagus are critical [74]. Use of multiple fields and scanning techniques is expected to reduce the increased rates of skin toxicity seen with proton therapy use in breast cancer [51].

Supplemental techniques that are helpful to avoid complications of RT include the use of equipment such as belly boards that minimize intestinal irradiation and thereby avoid small bowel toxicity [75], treating patients with a full bladder to avoid intestinal sequelae [76], using shielded vaginal cylinders for gynecologic brachytherapy and rectal spacers for prostate cancer RT [77], and avoiding large single doses. Minimizing acute toxicity is thought to be important in reducing consequential late effects [47].

Pentoxifylline — Pentoxifylline is a xanthine derivative that inhibits platelet aggregation and enhances microvascular blood flow. It has been used alone and in combination with tocopherol, both to prevent and treat radiation-induced fibrosis. Pentoxifylline may also inhibit fibroblast proliferation and the production of extracellular matrix.

A randomized trial in which pentoxifylline alone was assessed for the prevention of radiation-induced fibrosis included 40 patients with either lung or breast cancer who were receiving RT that included normal lung in the treatment field [78]. Another study randomized 91 patients with thoracic RT to pentoxifylline and tocopherol during and for three months after RT versus no treatment and found decreased acute and long-term pulmonary toxicity [79]. A randomized trial testing pentoxifylline and vitamin E versus standard follow-up after breast cancer RT indicated a significant benefit for the prevention of radiation fibrosis [80]. (See "Radiation-induced lung injury", section on 'Prevention'.)

A randomized study in 78 postoperative head and neck cancer patients also identified a positive effect on late skin changes, fibrosis, and soft tissue necrosis with pentoxifylline alone [81].

Although statistically significant benefit associated with pentoxifylline treatment was observed and prophylactic treatment was recommended for high-risk patients, eg, those with poor pulmonary function, the activity of pentoxifylline to prevent pulmonary toxicity requires confirmation, and prophylactic use of pentoxifylline is currently not established as a routine management approach.

Amifostine — Amifostine may reduce the incidence and severity of acute side effects from RT at various treatment sites, although given the mixed results and its toxicity, it is not routinely used.

Multiple randomized clinical trials have evaluated amifostine for the prevention of xerostomia in patients receiving RT or chemoradiotherapy for head and neck cancer. These trials have given conflicting results, and the role of amifostine in this setting remains uncertain. (See "Management and prevention of complications during initial treatment of head and neck cancer", section on 'Amifostine'.)

Although an initial randomized trial showed benefit from amifostine in patients receiving lung irradiation, a subsequent larger trial did not confirm these results. (See "Radiation-induced lung injury", section on 'Prevention'.)

In one randomized trial, amifostine decreased the incidence of acute cystitis.

Amifostine requires intravenous or subcutaneous administration and is associated with significant side effects. Amifostine has not been assessed for the treatment of established radiation-induced fibrosis.

TREATMENT

Symptomatic treatment — The treatment of radiation-induced fibrosis depends upon the location and extent of the fibrosis, as well as the severity of symptoms. Therapeutic approaches include medical management of symptoms, conservative interventions such as dilatation and stent implantation for stenoses (eg esophagus, ureter), and surgical treatment of adhesions and strictures.

In patients thought to be at high risk for radiation-induced fibrosis, early initiation of active and passive physical therapy measures is helpful.

A special type of massage (Louis-Paul Guitay [LPG]) has been shown to reduce skin fibrosis after radiation therapy (RT) for breast cancer [82].

In women receiving pelvic RT, regular vaginal dilatation and topical estrogen cream may be useful to prevent symptomatic shortening and narrowing of the vagina [83,84].

For patients with early signs or symptoms of trismus, forced opening of the mouth is recommended. In a prospective study, a mouth-opening device was shown to reduce trismus compared with conventional approaches after 12 weeks of training and exercise [53,85].

Preliminary results on the use of botulinum toxin type A for trismus, cervical dystonia, and neuralgias associated with radiation-induced fibrosis were encouraging [86].

Lymph drainage for lymph edema and physical therapy to treat articular contractions are other conservative ways that may ameliorate manifestations of late radiation-induced fibrosis.

In a small study on patients with severe radiation-induced fibrosis after breast-conserving therapy for breast cancer, partial mastectomy and latissimus dorsi reconstruction have been helpful to reduce symptoms [87].

Adequate analgesic medication is important as well, since radiation-induced fibrosis can be painful.

Any surgical or mechanical intervention potentially can worsen the underlying fibrosis, thereby exacerbating symptoms. The risks of active interventions therefore need to be weighed against the possible benefits [9,16]. Although conservative approaches are favored, surgery may be required, particularly if a tumor recurrence cannot otherwise be excluded.

Pentoxifylline plus tocopherol — Pentoxifylline has been used alone and in combination with tocopherol, a scavenger of reactive oxygen, to treat radiation-induced fibrosis, as well as to prevent pulmonary toxicity. (See 'Pentoxifylline' above.)

The potential clinical utility of pentoxifylline, either alone or with tocopherol (vitamin E), to treat established radiation-induced fibrosis was initially suggested by case reports and observational series in a variety of settings, in which treatment resulted in healing or symptomatic improvement. These observations included:

Partial reversal of soft tissue fibrosis following RT [48,88-90]

Healing of radiation-induced soft tissue and bone necrosis [91-95]

Improvement in symptoms of trismus [96]

Reduction in pelvic fibrosis after childhood RT [97,98]

Improvement of radiation-induced lumbosacral polyradiculopathy [99]

Multiple small randomized trials have suggested that pentoxifylline and or vitamin E may have a role in minimizing radiation fibrosis [14,80,100,101]. The two largest of these trials illustrate the range of results seen with this approach:

In one trial, 83 women with breast cancer were randomly assigned to treatment with vitamin E plus pentoxifylline or vitamin E plus placebo for 12 months [100]. Increase in arm volume was significantly greater in patients receiving vitamin E plus placebo rather vitamin E plus pentoxifylline. Arm abduction, which was significantly impaired at baseline, improved significantly in both groups; the improvement was greater in those receiving pentoxifylline although the difference was not statistically significant.

In a second trial, 68 women with long-standing lymphedema of the arm following axillary surgery and RT were randomly assigned to receive pentoxifylline plus tocopherol or matched placebos for six months [101]. No effect was observed at 6 or 12 months in arm volume, or in secondary parameters including fibrosis of the breast, chest wall, and regional lymph node areas was seen.

More prolonged therapy may be an important factor in reversing the effects of radiation-induced fibrosis. In a detailed longitudinal series, regression of superficial radiation-induced fibrosis following irradiation for breast cancer was assessed in 44 women with 55 areas of superficial radiation-induced fibrosis during and after treatment with pentoxifylline (800 mg/day) plus tocopherol (1000 units/day) [1]. Seven patients were treated for 6 to 12 months and therapy was then discontinued, while 37 patients remained on treatment for 24 to 48 months.

Overall, the mean estimated maximal regression in surface area of radiation-induced fibrosis was 68 percent, and this regression required an average of 24 months of treatment. The time to maximum regression of radiation-induced fibrosis was shorter in patients with fibrosis of less than six years duration (16 versus 28 months for those with fibrosis of longer duration). Among the seven patients who were treated for 6 to 12 months, a rebound increase in fibrotic area was observed after cessation of treatment.

For osteoradionecrosis in the mandible, a combination of pentoxifylline and tocopherol with clodronate (PENTOCLO) has been reported as a successful approach [95]. The PENTOCLO treatment is preceded by anti-inflammatory, antibacterial, and antimycotic therapy for four to six weeks, followed by PENTOCLO therapy until healing. Healing was achieved in 16 of 27 patients after a median of 82 days. Healing took longer in patients after radiochemotherapy (169 days) compared with surgery and radiotherapy (49 days). These results were confirmed in a prospective cohort study [102].

In summary, these data suggest that the combination of pentoxifylline plus tocopherol can reverse superficial radiation-induced fibrosis in some patients [103]. The optimal dose and duration of therapy and the role of tocopherol are unknown. However, some data suggest that a prolonged course of treatment may be necessary to induce maximal regression of radiation-induced fibrosis and to maintain any benefit. These results require additional confirmation. Furthermore, it is not clear whether these results can be extrapolated to radiation-induced fibrosis in other tissues, or whether treatment with pentoxifylline plus tocopherol needs to be continued indefinitely.

Hyperbaric oxygen — Hyperbaric oxygen has been evaluated as a treatment for late toxicities, including radiation-induced fibrosis, in multiple studies investigating the benefit for patients treated in the head and neck, breast, and pelvic area in particular. Small prospective studies identified some effect in reducing lymphedema and peripheral nerve damage.

Hyperbaric oxygen has been evaluated as a treatment for radiation-induced fibrosis in at least three small studies:

Two observational studies, which together included a total of 31 women, found that hyperbaric oxygen decreased lymphedema following surgery plus RT for breast cancer [104,105]. The observed benefit persisted for at least one year.

A double-blind trial in 34 women treated for radiation-induced brachial plexopathy did not demonstrate any benefit from hyperbaric oxygen [106]. However, two of the women who were treated with hyperbaric oxygen did have substantial improvement in their lymphedema. (See "Overview of cancer pain syndromes".)

However, a meta-analysis of 14 studies (753 patients) that randomized hyperbaric oxygen use versus no therapy found that hyperbaric oxygen can prevent osteoradionecrosis and proctitis, but it had no effect on other radiation-induced late effects [107].

SUMMARY AND RECOMMENDATIONS — Radiation-induced fibrosis can develop as a late effect in multiple tissues, as a function of the dose and volume of radiation as well as the radiation sensitivity of a given tissue. Radiation-induced fibrosis may cause both cosmetic and functional impairment, which can lead to a significant deterioration in quality of life or even death.

The primary approach to prevention of radiation-induced fibrosis is through the use of appropriate radiation therapy doses and techniques that minimize the radiation exposure for normal tissue. (See 'Radiation therapy techniques' above.)

For patients with established radiation-induced fibrosis, treatment is primarily symptomatic and depends upon the organ system involved. (See 'Symptomatic treatment' above.)

For patients with established symptomatic subcutaneous fibrosis, we suggest a combination of pentoxifylline and tocopherol (Grade 2C). The optimal duration of therapy is unknown, but treatment may be required for two or more years to observe and maintain any benefit. (See 'Pentoxifylline plus tocopherol' above.)

  1. Delanian S, Porcher R, Rudant J, Lefaix JL. Kinetics of response to long-term treatment combining pentoxifylline and tocopherol in patients with superficial radiation-induced fibrosis. J Clin Oncol 2005; 23:8570.
  2. Haase O, Rodemann HP. Fibrosis and cytokine mechanisms: relevant in hadron therapy? Radiother Oncol 2004; 73 Suppl 2:S144.
  3. Bentzen SM. Preventing or reducing late side effects of radiation therapy: radiobiology meets molecular pathology. Nat Rev Cancer 2006; 6:702.
  4. Abdollahi A, Li M, Ping G, et al. Inhibition of platelet-derived growth factor signaling attenuates pulmonary fibrosis. J Exp Med 2005; 201:925.
  5. Martin M, Lefaix J, Delanian S. TGF-beta1 and radiation fibrosis: a master switch and a specific therapeutic target? Int J Radiat Oncol Biol Phys 2000; 47:277.
  6. Vozenin-Brotons MC, Milliat F, Sabourin JC, et al. Fibrogenic signals in patients with radiation enteritis are associated with increased connective tissue growth factor expression. Int J Radiat Oncol Biol Phys 2003; 56:561.
  7. Yarnold J, Brotons MC. Pathogenetic mechanisms in radiation fibrosis. Radiother Oncol 2010; 97:149.
  8. Stewart FA, Hoving S, Russell NS. Vascular damage as an underlying mechanism of cardiac and cerebral toxicity in irradiated cancer patients. Radiat Res 2010; 174:865.
  9. Herrmann T, Baumann M, Dörr W. Clinical Radiation Biology, Elsevier, Munich 2006.
  10. Davis AM, O'Sullivan B, Turcotte R, et al. Late radiation morbidity following randomization to preoperative versus postoperative radiotherapy in extremity soft tissue sarcoma. Radiother Oncol 2005; 75:48.
  11. Geara FB, Komaki R, Tucker SL, et al. Factors influencing the development of lung fibrosis after chemoradiation for small cell carcinoma of the lung: evidence for inherent interindividual variation. Int J Radiat Oncol Biol Phys 1998; 41:279.
  12. Graham MV, Purdy JA, Emami B, et al. Clinical dose-volume histogram analysis for pneumonitis after 3D treatment for non-small cell lung cancer (NSCLC). Int J Radiat Oncol Biol Phys 1999; 45:323.
  13. Collette S, Collette L, Budiharto T, et al. Predictors of the risk of fibrosis at 10 years after breast conserving therapy for early breast cancer: a study based on the EORTC Trial 22881-10882 'boost versus no boost'. Eur J Cancer 2008; 44:2587.
  14. Delanian S, Porcher R, Balla-Mekias S, Lefaix JL. Randomized, placebo-controlled trial of combined pentoxifylline and tocopherol for regression of superficial radiation-induced fibrosis. J Clin Oncol 2003; 21:2545.
  15. Cuttino LW, Heffernan J, Vera R, et al. Association between maximal skin dose and breast brachytherapy outcome: a proposal for more rigorous dosimetric constraints. Int J Radiat Oncol Biol Phys 2011; 81:e173.
  16. Coia LR, Myerson RJ, Tepper JE. Late effects of radiation therapy on the gastrointestinal tract. Int J Radiat Oncol Biol Phys 1995; 31:1213.
  17. Marks LB, Carroll PR, Dugan TC, Anscher MS. The response of the urinary bladder, urethra, and ureter to radiation and chemotherapy. Int J Radiat Oncol Biol Phys 1995; 31:1257.
  18. Saunders M, Dische S, Barrett A, et al. Continuous hyperfractionated accelerated radiotherapy (CHART) versus conventional radiotherapy in non-small-cell lung cancer: a randomised multicentre trial. CHART Steering Committee. Lancet 1997; 350:161.
  19. Peeters ST, Heemsbergen WD, van Putten WL, et al. Acute and late complications after radiotherapy for prostate cancer: results of a multicenter randomized trial comparing 68 Gy to 78 Gy. Int J Radiat Oncol Biol Phys 2005; 61:1019.
  20. Andreassen CN, Overgaard J, Alsner J, et al. ATM sequence variants and risk of radiation-induced subcutaneous fibrosis after postmastectomy radiotherapy. Int J Radiat Oncol Biol Phys 2006; 64:776.
  21. Hölscher T, Bentzen SM, Baumann M. Influence of connective tissue diseases on the expression of radiation side effects: a systematic review. Radiother Oncol 2006; 78:123.
  22. Gold DG, Miller RC, Petersen IA, Osborn TG. Radiotherapy for malignancy in patients with scleroderma: The Mayo Clinic experience. Int J Radiat Oncol Biol Phys 2007; 67:559.
  23. Lin A, Abu-Isa E, Griffith KA, Ben-Josef E. Toxicity of radiotherapy in patients with collagen vascular disease. Cancer 2008; 113:648.
  24. Girinsky T, Cosset JM. [Pulmonary and cardiac late effects of ionizing radiations alone or combined with chemotherapy]. Cancer Radiother 1997; 1:735.
  25. Toledano A, Garaud P, Serin D, et al. Concurrent administration of adjuvant chemotherapy and radiotherapy after breast-conserving surgery enhances late toxicities: long-term results of the ARCOSEIN multicenter randomized study. Int J Radiat Oncol Biol Phys 2006; 65:324.
  26. Kirwan JM, Symonds P, Green JA, et al. A systematic review of acute and late toxicity of concomitant chemoradiation for cervical cancer. Radiother Oncol 2003; 68:217.
  27. Bentzen SM, Skoczylas JZ, Overgaard M, Overgaard J. Radiotherapy-related lung fibrosis enhanced by tamoxifen. J Natl Cancer Inst 1996; 88:918.
  28. Azria D, Gourgou S, Sozzi WJ, et al. Concomitant use of tamoxifen with radiotherapy enhances subcutaneous breast fibrosis in hypersensitive patients. Br J Cancer 2004; 91:1251.
  29. Taylor ME, Perez CA, Halverson KJ, et al. Factors influencing cosmetic results after conservation therapy for breast cancer. Int J Radiat Oncol Biol Phys 1995; 31:753.
  30. Sanguineti G, Agostinelli S, Foppiano F, et al. Adjuvant androgen deprivation impacts late rectal toxicity after conformal radiotherapy of prostate carcinoma. Br J Cancer 2002; 86:1843.
  31. Schultheiss TE, Hanks GE, Hunt MA, Lee WR. Incidence of and factors related to late complications in conformal and conventional radiation treatment of cancer of the prostate. Int J Radiat Oncol Biol Phys 1995; 32:643.
  32. Peeters ST, Hoogeman MS, Heemsbergen WD, et al. Volume and hormonal effects for acute side effects of rectum and bladder during conformal radiotherapy for prostate cancer. Int J Radiat Oncol Biol Phys 2005; 63:1142.
  33. Niyazi M, Maihoefer C, Krause M, et al. Radiotherapy and "new" drugs-new side effects? Radiat Oncol 2011; 6:177.
  34. Um SJ, Lee SK, Yang DK, et al. Fatal interstitial lung disease after erlotinib administration in a patient with radiation fibrosis. Clin Respir J 2009; 3:181.
  35. Arakawa H, Johkoh T, Sakai F, et al. Exacerbation of radiation fibrosis with erlotinib: another pattern of radiation recall phenomenon. Jpn J Radiol 2011; 29:587.
  36. Socinski MA, Stinchcombe TE, Moore DT, et al. Incorporating bevacizumab and erlotinib in the combined-modality treatment of stage III non-small-cell lung cancer: results of a phase I/II trial. J Clin Oncol 2012; 30:3953.
  37. Landry JC, Feng Y, Cohen SJ, et al. Phase 2 study of preoperative radiation with concurrent capecitabine, oxaliplatin, and bevacizumab followed by surgery and postoperative 5-fluorouracil, leucovorin, oxaliplatin (FOLFOX), and bevacizumab in patients with locally advanced rectal cancer: ECOG 3204. Cancer 2013; 119:1521.
  38. Satzger I, Degen A, Asper H, et al. Serious skin toxicity with the combination of BRAF inhibitors and radiotherapy. J Clin Oncol 2013; 31:e220.
  39. Shaverdian N, Lisberg AE, Bornazyan K, et al. Previous radiotherapy and the clinical activity and toxicity of pembrolizumab in the treatment of non-small-cell lung cancer: a secondary analysis of the KEYNOTE-001 phase 1 trial. Lancet Oncol 2017; 18:895.
  40. Voong KR, Hazell SZ, Fu W, et al. Relationship Between Prior Radiotherapy and Checkpoint-Inhibitor Pneumonitis in Patients With Advanced Non-Small-Cell Lung Cancer. Clin Lung Cancer 2019; 20:e470.
  41. Herold DM, Hanlon AL, Hanks GE. Diabetes mellitus: a predictor for late radiation morbidity. Int J Radiat Oncol Biol Phys 1999; 43:475.
  42. Maruyama Y, Van Nagell JR Jr, Utley J, et al. Radiation and small bowel complications in cervical carcinoma therapy. Radiology 1974; 112:699.
  43. Darby SC, Ewertz M, McGale P, et al. Risk of ischemic heart disease in women after radiotherapy for breast cancer. N Engl J Med 2013; 368:987.
  44. Zhou H, Cao K, Cao P, Jiang W. [Impact of diabetes mellitus on clinicopathological factors and relation with radiation pneumonitis in 332 patients with lung cancer]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2013; 38:138.
  45. Kalman NS, Hugo GD, Mahon RN, et al. Diabetes mellitus and radiation induced lung injury after thoracic stereotactic body radiotherapy. Radiother Oncol 2018; 129:270.
  46. Yamashita H, Takahashi W, Haga A, Nakagawa K. Radiation pneumonitis after stereotactic radiation therapy for lung cancer. World J Radiol 2014; 6:708.
  47. Dörr W, Hendry JH. Consequential late effects in normal tissues. Radiother Oncol 2001; 61:223.
  48. Delanian S, Balla-Mekias S, Lefaix JL. Striking regression of chronic radiotherapy damage in a clinical trial of combined pentoxifylline and tocopherol. J Clin Oncol 1999; 17:3283.
  49. Strnad V, Hildebrandt G, Pötter R, et al. Accelerated partial breast irradiation: 5-year results of the German-Austrian multicenter phase II trial using interstitial multicatheter brachytherapy alone after breast-conserving surgery. Int J Radiat Oncol Biol Phys 2011; 80:17.
  50. Rabinovitch R, Winter K, Kuske R, et al. RTOG 95-17, a Phase II trial to evaluate brachytherapy as the sole method of radiation therapy for Stage I and II breast carcinoma--year-5 toxicity and cosmesis. Brachytherapy 2014; 13:17.
  51. Galland-Girodet S, Pashtan I, MacDonald SM, et al. Long-term cosmetic outcomes and toxicities of proton beam therapy compared with photon-based 3-dimensional conformal accelerated partial-breast irradiation: a phase 1 trial. Int J Radiat Oncol Biol Phys 2014; 90:493.
  52. Bartelink H, Maingon P, Poortmans P, et al. Whole-breast irradiation with or without a boost for patients treated with breast-conserving surgery for early breast cancer: 20-year follow-up of a randomised phase 3 trial. Lancet Oncol 2015; 16:47.
  53. Sciubba JJ, Goldenberg D. Oral complications of radiotherapy. Lancet Oncol 2006; 7:175.
  54. Perez CA, Fox S, Lockett MA, et al. Impact of dose in outcome of irradiation alone in carcinoma of the uterine cervix: analysis of two different methods. Int J Radiat Oncol Biol Phys 1991; 21:885.
  55. Díez P, Mullassery V, Dankulchai P, et al. Dosimetric analysis of urethral strictures following HDR (192)Ir brachytherapy as monotherapy for intermediate- and high-risk prostate cancer. Radiother Oncol 2014; 113:410.
  56. Pötter R, Knocke TH, Fellner C, et al. Definitive radiotherapy based on HDR brachytherapy with iridium 192 in uterine cervix carcinoma: report on the Vienna University Hospital findings (1993-1997) compared to the preceding period in the context of ICRU 38 recommendations. Cancer Radiother 2000; 4:159.
  57. Leung SF, Zheng Y, Choi CY, et al. Quantitative measurement of post-irradiation neck fibrosis based on the young modulus: description of a new method and clinical results. Cancer 2002; 95:656.
  58. Yang X, Yoshida E, Cassidy RJ, et al. Quantitative Ultrasonic Nakagami Imaging of Neck Fibrosis After Head and Neck Radiation Therapy. Int J Radiat Oncol Biol Phys 2015; 92:407.
  59. Radiation Therapy Oncology Group Common Toxicity Criteria http://www.rtog.org/ResearchAssociates/AdverseEventReporting/CooperativeGroupCommonToxicityCriteria.aspx (Accessed on March 14, 2011).
  60. Hoeller U, Bonacker M, Bajrovic A, et al. Radiation-induced plexopathy and fibrosis. Is magnetic resonance imaging the adequate diagnostic tool? Strahlenther Onkol 2004; 180:650.
  61. Semiz Oysu A, Ayanoglu E, Kodalli N, et al. Dynamic contrast-enhanced MRI in the differentiation of posttreatment fibrosis from recurrent carcinoma of the head and neck. Clin Imaging 2005; 29:307.
  62. Padhani AR, Yarnold JR, Regan J, Husband JE. Magnetic resonance imaging of induration in the irradiated breast. Radiother Oncol 2002; 64:157.
  63. Eisbruch A, Ship JA, Dawson LA, et al. Salivary gland sparing and improved target irradiation by conformal and intensity modulated irradiation of head and neck cancer. World J Surg 2003; 27:832.
  64. Mundt AJ, Lujan AE, Rotmensch J, et al. Intensity-modulated whole pelvic radiotherapy in women with gynecologic malignancies. Int J Radiat Oncol Biol Phys 2002; 52:1330.
  65. Donovan E, Bleakley N, Denholm E, et al. Randomised trial of standard 2D radiotherapy (RT) versus intensity modulated radiotherapy (IMRT) in patients prescribed breast radiotherapy. Radiother Oncol 2007; 82:254.
  66. Barnett GC, Wilkinson JS, Moody AM, et al. Randomized controlled trial of forward-planned intensity modulated radiotherapy for early breast cancer: interim results at 2 years. Int J Radiat Oncol Biol Phys 2012; 82:715.
  67. Keller LM, Sopka DM, Li T, et al. Five-year results of whole breast intensity modulated radiation therapy for the treatment of early stage breast cancer: the Fox Chase Cancer Center experience. Int J Radiat Oncol Biol Phys 2012; 84:881.
  68. Mukesh MB, Qian W, Wilkinson JS, et al. Patient reported outcome measures (PROMs) following forward planned field-in field IMRT: results from the Cambridge Breast IMRT trial. Radiother Oncol 2014; 111:270.
  69. Gupta T, Agarwal J, Jain S, et al. Three-dimensional conformal radiotherapy (3D-CRT) versus intensity modulated radiation therapy (IMRT) in squamous cell carcinoma of the head and neck: a randomized controlled trial. Radiother Oncol 2012; 104:343.
  70. Jiang ZQ, Yang K, Komaki R, et al. Long-term clinical outcome of intensity-modulated radiotherapy for inoperable non-small cell lung cancer: the MD Anderson experience. Int J Radiat Oncol Biol Phys 2012; 83:332.
  71. McCormick B, Hunt M. Intensity-modulated radiation therapy for breast: is it for everyone? Semin Radiat Oncol 2011; 21:51.
  72. Zheng Y, Han F, Xiao W, et al. Analysis of late toxicity in nasopharyngeal carcinoma patients treated with intensity modulated radiation therapy. Radiat Oncol 2015; 10:17.
  73. Lambrecht M, Nevens D, Nuyts S. Intensity-modulated radiotherapy vs. parotid-sparing 3D conformal radiotherapy. Effect on outcome and toxicity in locally advanced head and neck cancer. Strahlenther Onkol 2013; 189:223.
  74. Zhang X, Li Y, Pan X, et al. Intensity-modulated proton therapy reduces the dose to normal tissue compared with intensity-modulated radiation therapy or passive scattering proton therapy and enables individualized radical radiotherapy for extensive stage IIIB non-small-cell lung cancer: a virtual clinical study. Int J Radiat Oncol Biol Phys 2010; 77:357.
  75. Martin J, Fitzpatrick K, Horan G, et al. Treatment with a belly-board device significantly reduces the volume of small bowel irradiated and results in low acute toxicity in adjuvant radiotherapy for gynecologic cancer: results of a prospective study. Radiother Oncol 2005; 74:267.
  76. Kim TH, Chie EK, Kim DY, et al. Comparison of the belly board device method and the distended bladder method for reducing irradiated small bowel volumes in preoperative radiotherapy of rectal cancer patients. Int J Radiat Oncol Biol Phys 2005; 62:769.
  77. Chao M, Ow D, Ho H, et al. Improving rectal dosimetry for patients with intermediate and high-risk prostate cancer undergoing combined high-dose-rate brachytherapy and external beam radiotherapy with hydrogel space. J Contemp Brachytherapy 2019; 11:8.
  78. Ozturk B, Egehan I, Atavci S, Kitapci M. Pentoxifylline in prevention of radiation-induced lung toxicity in patients with breast and lung cancer: a double-blind randomized trial. Int J Radiat Oncol Biol Phys 2004; 58:213.
  79. Misirlioglu CH, Demirkasimoglu T, Kucukplakci B, et al. Pentoxifylline and alpha-tocopherol in prevention of radiation-induced lung toxicity in patients with lung cancer. Med Oncol 2007; 24:308.
  80. Jacobson G, Bhatia S, Smith BJ, et al. Randomized trial of pentoxifylline and vitamin E vs standard follow-up after breast irradiation to prevent breast fibrosis, evaluated by tissue compliance meter. Int J Radiat Oncol Biol Phys 2013; 85:604.
  81. Aygenc E, Celikkanat S, Kaymakci M, et al. Prophylactic effect of pentoxifylline on radiotherapy complications: a clinical study. Otolaryngol Head Neck Surg 2004; 130:351.
  82. Bourgeois JF, Gourgou S, Kramar A, et al. A randomized, prospective study using the LPG technique in treating radiation-induced skin fibrosis: clinical and profilometric analysis. Skin Res Technol 2008; 14:71.
  83. PITKIN RM, BRADBURY JT. THE EFFECT OF TOPICAL ESTROGEN ON IRRADIATED VAGINAL EPITHELIUM. Am J Obstet Gynecol 1965; 92:175.
  84. Stahl JM, Qian JM, Tien CJ, et al. Extended duration of dilator use beyond 1 year may reduce vaginal stenosis after intravaginal high-dose-rate brachytherapy. Support Care Cancer 2019; 27:1425.
  85. Li YH, Chang WC, Chiang TE, et al. Mouth-opening device as a treatment modality in trismus patients with head and neck cancer and oral submucous fibrosis: a prospective study. Clin Oral Investig 2019; 23:469.
  86. Stubblefield MD, Levine A, Custodio CM, Fitzpatrick T. The role of botulinum toxin type A in the radiation fibrosis syndrome: a preliminary report. Arch Phys Med Rehabil 2008; 89:417.
  87. van Geel AN, Lans TE, Haen R, et al. Partial mastectomy and m. latissimus dorsi reconstruction for radiation-induced fibrosis after breast-conserving cancer therapy. World J Surg 2011; 35:568.
  88. Haddad P, Kalaghchi B, Amouzegar-Hashemi F. Pentoxifylline and vitamin E combination for superficial radiation-induced fibrosis: a phase II clinical trial. Radiother Oncol 2005; 77:324.
  89. Okunieff P, Augustine E, Hicks JE, et al. Pentoxifylline in the treatment of radiation-induced fibrosis. J Clin Oncol 2004; 22:2207.
  90. Futran ND, Trotti A, Gwede C. Pentoxifylline in the treatment of radiation-related soft tissue injury: preliminary observations. Laryngoscope 1997; 107:391.
  91. Dion MW, Hussey DH, Doornbos JF, et al. Preliminary results of a pilot study of pentoxifylline in the treatment of late radiation soft tissue necrosis. Int J Radiat Oncol Biol Phys 1990; 19:401.
  92. Delanian S, Depondt J, Lefaix JL. Major healing of refractory mandible osteoradionecrosis after treatment combining pentoxifylline and tocopherol: a phase II trial. Head Neck 2005; 27:114.
  93. Delanian S, Lefaix JL. Complete healing of severe osteoradionecrosis with treatment combining pentoxifylline, tocopherol and clodronate. Br J Radiol 2002; 75:467.
  94. Delanian S, Chatel C, Porcher R, et al. Complete restoration of refractory mandibular osteoradionecrosis by prolonged treatment with a pentoxifylline-tocopherol-clodronate combination (PENTOCLO): a phase II trial. Int J Radiat Oncol Biol Phys 2011; 80:832.
  95. Robard L, Louis MY, Blanchard D, et al. Medical treatment of osteoradionecrosis of the mandible by PENTOCLO: preliminary results. Eur Ann Otorhinolaryngol Head Neck Dis 2014; 131:333.
  96. Chua DT, Lo C, Yuen J, Foo YC. A pilot study of pentoxifylline in the treatment of radiation-induced trismus. Am J Clin Oncol 2001; 24:366.
  97. Letur-Könirsch H, Guis F, Delanian S. Uterine restoration by radiation sequelae regression with combined pentoxifylline-tocopherol: a phase II study. Fertil Steril 2002; 77:1219.
  98. Gothard L, Cornes P, Brooker S, et al. Phase II study of vitamin E and pentoxifylline in patients with late side effects of pelvic radiotherapy. Radiother Oncol 2005; 75:334.
  99. Delanian S, Lefaix JL, Maisonobe T, et al. Significant clinical improvement in radiation-induced lumbosacral polyradiculopathy by a treatment combining pentoxifylline, tocopherol, and clodronate (Pentoclo). J Neurol Sci 2008; 275:164.
  100. Magnusson M, Höglund P, Johansson K, et al. Pentoxifylline and vitamin E treatment for prevention of radiation-induced side-effects in women with breast cancer: a phase two, double-blind, placebo-controlled randomised clinical trial (Ptx-5). Eur J Cancer 2009; 45:2488.
  101. Gothard L, Cornes P, Earl J, et al. Double-blind placebo-controlled randomised trial of vitamin E and pentoxifylline in patients with chronic arm lymphoedema and fibrosis after surgery and radiotherapy for breast cancer. Radiother Oncol 2004; 73:133.
  102. Dissard A, P Dang N, Barthelemy I, et al. Efficacy of pentoxifylline-tocopherol-clodronate in mandibular osteoradionecrosis. Laryngoscope 2020; 130:E559.
  103. Anscher MS. The irreversibility of radiation-induced fibrosis: fact or folklore? J Clin Oncol 2005; 23:8551.
  104. Gothard L, Stanton A, MacLaren J, et al. Non-randomised phase II trial of hyperbaric oxygen therapy in patients with chronic arm lymphoedema and tissue fibrosis after radiotherapy for early breast cancer. Radiother Oncol 2004; 70:217.
  105. Teas J, Cunningham JE, Cone L, et al. Can hyperbaric oxygen therapy reduce breast cancer treatment-related lymphedema? A pilot study. J Womens Health (Larchmt) 2004; 13:1008.
  106. Pritchard J, Anand P, Broome J, et al. Double-blind randomized phase II study of hyperbaric oxygen in patients with radiation-induced brachial plexopathy. Radiother Oncol 2001; 58:279.
  107. Bennett MH, Feldmeier J, Hampson NB, et al. Hyperbaric oxygen therapy for late radiation tissue injury. Cochrane Database Syst Rev 2016; 4:CD005005.
Topic 7055 Version 26.0

References

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