Accelerated Partial Breast Irradiation: An Opportunity for Therapeutic De-escalation

Adjuvant radiation therapy (RT) after breast conserving surgery has been consistently shown to reduce local recurrence and to improve breast cancer survival after breast conserving surgery.1 Traditionally, adjuvant RT consisted of standard fractionation whole breast irradiation (WBI), which required daily treatment for 5 to 7 weeks. As such, this led to noncompliance, increased costs of care, as well as patients electing to pursue mastectomy to avoid such long durations of therapy.2 In addition, the burdens of such an approach led to many early deintensification trials in low-risk patients evaluating the omission of RT in lieu of endocrine therapy alone.3,4

The last 2 decades has brought a plethora of new radiotherapy approaches for patients which are forcing clinicians to rethink the assumptions made based on standard fractionation WBI. With respect to WBI, mature outcomes have supported the transition to moderately hypofractionated WBI and more recently, ultrashort WBI, though long-term outcomes with 5 fraction WBI remain limited.5–8 Concurrently, multiple randomized trials have evaluated the utilization of partial breast irradiation (PBI), allowing for a reduction in treatment volumes and therefore the potential for reduced toxicities and treatment duration.9 The purpose of this review is to evaluate outcomes with PBI (not including intraoperative RT) while evaluating the role of therapeutic de-escalation for early-stage breast cancers. Only the PBI techniques for which the definitive pathology is available at the time of the RT delivery have been considered to tailor the treatment to the individual, according to the principle of high-precision medicine.

DISCUSSION Outcomes

Over the past decade, multiple randomized trials have been published comparing WBI with various modern PBI techniques with long-term follow-up. Outcomes from these studies are summarized in Table 1 and consistently demonstrate low rates of local recurrence with both PBI and WBI at 5 to 20 years of follow-up; in addition, these studies have identified either no statistically significant difference or <1% in local recurrence between approaches.10–17 For example, Polgar et al10 recently published 20-year outcomes from their randomized trial comparing interstitial/electron PBI to WBI; no difference in local control was noted (9.6% PBI vs. 7.9% WBI, P=0.59). Similar results have been seen in the NSABP B-39 trial, which did not meet equivalence at 10 years with a 0.7% difference in local control (4.6% vs. 3.9%), as well as the Randomized Trial of Accelerated partial breast irradiation (RAPID) trial that demonstrated no difference in local recurrence at 8 years (3.0% vs. 2.8%).11,17 Importantly, the randomized trial data have shown that local control is comparable with PBI irrespective of technique (brachytherapy and external beam RT).

TABLE 1 - Randomized Trials Evaluating Partial Breast Irradiation No. Patients Partial Breast Technique(s) Follow-up (y) Inclusion Criteria Local Recurrence Toxicity/Cosmetic Outcomes NSABP B-3911 4216 Interstitial, applicator, 3D-CRT 10.2 18 y or older, pTis, pT1-2 (≤3 cm), N0-1, negative margins 3.9% WBI vs. 4.6% PBI Grade 3: 7% WBI vs. 10% PBI RAPID17 2135 3D-CRT 8.6 40 y or older, pTis, pT1-2 (≤3 cm), N0-mi, negative margins; no ILC 2.8% WBI vs. 3.0% PBI Late toxicity grade 2+: 13% WBI vs. 32% PBI Adverse cosmesis 18% worse with PBI at 7 y GEC-ESTRO12 1184 Interstitial 6.6 40 y or older, pTis, pT1-2 (≤3 cm), N0-mi, negative margins (2 mm, 5 mm ILC); no LVSI 0.9% WBI vs. 1.4% PBI Late grade 2-3 toxicity: 5.7% PBI vs. 3.2% PBI Florence16 520 IMRT 10.7 40 y or older, pT1-2 (≤2.5 cm), N0-1, negative margins (5 mm); no EIC, multifocal 2.5% WBI vs. 3.7% PBI PBI reduced acute, chronic toxicity, improved cosmetic outcomes IMPORT-LOW14 2018 IMRT 6.0 50 y or older, pT1-2 (≤3 cm), N0-1, negative margins (2 mm) 1.1% WBI vs. 0.5% PBI vs. 0.2% SIB PBI less change in breast appearance Barcelona15 102 3D-CRT 10.3 60 y or older, pT1-2 (≤3 cm), N0, negative margins (3 mm), grade 2 or less; no ILC, no EIC 4% both arms Similar excellent/good cosmesis, No difference late skin toxicity NIO10 258 Interstitial brachytherapy/electron 17 40 y or older, pT1, N0-1mi, negative margins, No ILC/DCIS, No EIC, grade 1-2 7.9% WBI vs. 9.6% PBI Improved cosmetic outcomes with HDR PBI

3D-CRT indicates 3-dimensional conformal radiotherapy; DCIS, ductal carcinoma in situ; EIC, extensive intraductal component; GEC-ESTRO, Groupe Européen de Curiethérapie- European SocieTy for Radiotherapy and Oncology; HDR, high dose rate; ILC, invasive lobular carcinoma; IMPORT-LOW, Intensity modulated partioal organ radiotherapy trial; IMRT, intensity-modulated radiotherapy; LVSI, lymphovascular invasion; PBI, partial breast irradiation; RAPID, Randomized Trial of Accelerated partial breast irradiation; SIB, simultaneous integrated boost; WBI, whole breast irradiation.

Beyond local control, PBI has demonstrated the ability to improve toxicity and cosmetic outcomes. For example, Polgar et al10,18 found that with long-term follow-up, PBI was associated with improved rates of excellent/good cosmesis, whereas the Groupe Européen de Curiethérapie- European SocieTy for Radiotherapy and Oncology (GEC-ESTRO) trial demonstrated reduced breast pain with PBI, with no difference in quality of life. More recently, using advanced techniques including intensity-modulated radiation therapy (IMRT), the Florence trial demonstrated reduced rates of acute and chronic toxicity, as well as improved cosmetic outcomes. Similar outcomes were seen in the IMPORT-LOW trial, which used intensity modulated PBI, with reduced changes in breast appearance, and breast firmness. Quality of life data from Intensity modulated partioal organ radiotherapy trial demonstrated fewer adverse events with PBI as well, which is consistent with quality of life data from the Florence trial.19,20 The data have also demonstrated that when using less advanced techniques to deliver PBI (eg, 3-dimensional conformal RT) that outcomes may be worse with PBI as compared with WBI though this was not seen in NSABP B39;21 data from RAPID and RTOG 0319 have demonstrated increased toxicity and deteriorated cosmetic outcomes though accurate dosimetric evaluation and once a day fractionation may minimize late toxicity and provide satisfactory cosmesis.17,22 The use of more advanced techniques, such as IMRT, can increase the confidence of radiation oncologists in prescribing PBI, especially when ultrashort fractionations are applied.

Utilization

With comparable clinical outcomes and reduced toxicity rates, one would expect that PBI would represent the most common strategy utilized in women with early-stage breast cancer, particularly given current guidelines that support its utilization in women 45 to 50 years or older with early-stage node negative breast cancers or ductal carcinoma in situ.23,24 This is consistent with data that have demonstrated up to 75% of patients with early-stage breast cancer would be eligible for PBI.25 However, recent data have demonstrated that only 15% of patients who may be eligible for accelerated PBI receive the treatment with additional data demonstrating no increase in PBI utilization since 2008 despite the growing data supporting its utilization.26,27 These findings support that there remains an opportunity for clinicians to greater embrace PBI as an approach for women with early-stage breast cancer after breast-conserving surgery. As PBI enables the delivery of RT in a short, safe, and effective manner, the controversial question of RT-related risks outweighing the benefits will fade, alongside the decisional conflict of both clinicians and patients.28,29

As patient selection is at the core of risk-adapted RT options, consistent with American Society for Radiation Oncology and GEC-ESTRO criteria, assessment of HER2 and KI-67 might contribute to better assesment of optimal candidates;30 for example, the APT trial demonstrated that use of HER2-targeted therapies for HER2 over-expressed cancers led to local recurrence rates approximating luminal A breast cancers, allowing for risk adoption to PBI in these patients.31 In addition, the LUMINA trial (NCT01791829) is currently testing the role of these immunohistochemical biomarkers to identify Luminal A breast cancers, avoiding the cost of gene expression profiling for all patients, whereas deintensification based on multigene assays such as Prosigna and Oncotype Dx to define the risk of recurrence score is also being investigated in the EXPERT trial (NCT02889874) and IDEA Study (NCT02400190).

Rethinking Deintensification

For decades, when evaluating options to deintensify therapy for patients with low-risk early-stage breast cancer, the focus has been removing RT.3,4 Meta-analyses have demonstrated that RT omission while increasing the risk of local recurrence, did not impact survival.32,33 Given the duration of the historical RT regimens and potential radiation-induced toxicities, this seemed appropriate. However, with modern RT approaches including short-course PBI and significant reductions in toxicities, reevaluation of deintensification should occur; this is particularly relevant, given growing data on long-term toxicities of endocrine therapy, as well as noncompliance.34,35 Importantly, previous data have shown that in low-risk patients, endocrine therapy does not impact the rates of distant metastases as compared with RT alone;36,37 in addition, RT provides improved local control as compared with endocrine therapy alone with modest increases in contralateral breast cancers.36,38 Population data have shown no difference in outcomes with RT alone as compared with endocrine therapy alone in low-risk breast cancer patients with modeling studies demonstrating comparable costs and quality of life outcomes.38–40 Because of the non-negligible rate of endocrine therapy discontinuation over time, a propensity score matching analysis using the National Cancer Data Base found that patients undergoing RT alone had better overall survival than those receiving endocrine therapy alone.41 Murphy and colleagues confirmed the association of endocrine therapy nonadherence with distant metastases and disease-free survival, stressing the importance of considering RT alone for elderly women.42 In a survey of elderly women with biologically favorable tumors, 71% of whom received both RT and endocrine therapy, respondents noted they would rather receive RT than endocrine therapy, which was perceived as having the most negative effect on their quality of life.43

PBI Planning

PBI planning will be dependent on the technique utilized; however, the use of smaller targets than WBI allows for sparing of organs at risk including the heart, lung, skin, chest wall, and uninvolved breast tissue. In addition, the use of PBI may be associated with reduced risk of secondary cancers.44,45 For interstitial, and applicator brachytherapy techniques, after placement of catheters or applicators, patients should undergo computed tomography–based planning. Volume expansions range from 1.5 to 2 cm beyond the cavity and optimization can be utilized to meet/exceed standardized constraints from randomized and prospective trials, as well as guidelines to ensure target coverage while limiting dose to critical structures including the skin and chest wall.46

External beam PBI represents an opportunity to use noninvasive techniques to improve the therapeutic ratio of breast radiation; more advanced RT modalities such as IMRT may allow for the optimal balance between target volume coverage and organ at risk sparing, paving the way for lower rates of toxicity and dose-escalation regimens. As such, more recent trials have incorporated use of IMRT to allow for improved homogeneity, as well as conformality, reducing hot spots and dose to uninvolved breast tissue, which may be the reason that these trials demonstrated reduced toxicities as compared with WBI while older PBI techniques did not. Moving beyond IMRT, newer techniques have been reported. Data from the Cleveland Clinic incorporated deep inspiratory breath hold in conjunction with daily cone-beam computed tomography; incorporating advanced planning and treatment delivery allowed for reductions in target margins from 2 to 1 cm.47 The transition to conformal, highly accurate delivery of external beam PBI that delivers high doses in as few as 5 fractions requires complex planning and delivery which can be considered consistent with stereotactic body RT (SBRT), which the American Association of Physicists in Medicine defines as, “the delivery of large doses in a few fractions, which results in a high biological effective dose (BED). In order to minimize the normal tissue toxicity, conformation of high doses to the target and rapid fall-off doses away from the target is critical. The practice of SBRT therefore requires a high level of confidence in the accuracy of the entire treatment delivery process.48” For example, the regimen used in the Florence trial was 30 Gy in 5 fractions, which is comparable to 50 Gy in 25 fractions with a 5 fraction boost additionally.16 These doses/fraction coupled with reduced margins in order to provide a highly conformal low side-effect treatment is consistent with SBRT use in the management of lung and brain tumors.

Future Directions

At this time, the role of PBI continues to evolve. With respect to deintensification, the EUROPA trial is comparing RT alone to endocrine therapy alone.49 Likewise, the GEC-ESTRO Breast Cancer Working Group has conceived a phase III randomized trial using multicatheter interstitial brachytherapy, EPOPE, which is designed to test 30 to 32 Gy in 7 to 8 fractions versus 16 Gy in a single fraction, with the primary endpoint of disease-free survival.50 In addition, trials are evaluating shorter courses of PBI. For example, initial outcomes from the TRIUMPH-T trial demonstrated the safety and feasibility of a 3 fraction PBI approach with further outcomes expected in the years to come; in addition, advances in treatment technology have allowed for the evaluation of a single-fraction postoperative PBI approach.51,52 Studies are ongoing evaluating the use of SBRT to deliver PBI with outcomes demonstrating the safety and efficacy of this approach while proceeding with dose escalation; the safety of dose escalation has also led to increased use of SBRT-like approaches preoperatively which are being evaluated in prospective trials at this time.53,54

CONCLUSIONS

PBI has demonstrated comparable clinical outcomes as compared with whole breast irradiation, with new techniques demonstrating reduced toxicity profiles. This transition allows for the reevaluation of treatment deintensification approaches in the management of early-stage breast cancer. To maximize therapeutic ratios with PBI, complex treatment planning incorporating stereotactic body RT approaches should be considered.

REFERENCES 1. Darby S, McGale P, Correa C, et al. Effect of radiotherapy after breast-conserving surgery on 10-year recurrence and 15-year breast cancer death: meta-analysis of individual patient data for 10,801 women in 17 randomised trials. Lancet. 2011;378:1707–1716. 2. Ma AM, Barone J, Wallis AE, et al. Noncompliance with adjuvant radiation, chemotherapy, or hormonal therapy in breast cancer patients. Am J Surg. 2008;196:500–504. 3. Hughes KS, Schnaper LA, Bellon JR, et al. Lumpectomy plus tamoxifen with or without irradiation in women age 70 years or older with early breast cancer: Long-term follow-up of CALGB 9343. J Clin Oncol. 2013;31:2382–2387. 4. Kunkler IH, Williams LJ, Jack WJL, et al. Breast-conserving surgery with or without irradiation in women aged 65 years or older with early breast cancer (PRIME II): a randomised controlled trial. Lancet Oncol. 2013;16:266–273. 5. Whelan TJ, Pignol JP, Levine MN, et al. Long-term results of hypofractionated radiation therapy for breast cancer. N Engl J Med. 2010;362:513–520. 6. Haviland JS, Owen JR, Dewar JA, et al. The UK Standardisation of Breast Radiotherapy (START) trials of radiotherapy hypofractionation for treatment of earl breast cancer: 10-year follow-up results of two randomized controlled trials. Lancet Oncol. 2013;14:1086–1094. 7. Brunt AM, Haviland JS, Sydenham M, et al. Ten-year results of FAST: a randomized controlled trial of 5-fraction whole-breast radiotherapy for early breast cancer. J Clin Oncol. 2020;38:3261–3272. 8. Brunt AM, Haviland JS, Wheatley DA, et al. Hypofractionated breast radiotherapy for 1 week versus 3 weeks (FAST-Forward): 5-year efficacy and late normal tissue effects results from a multicenter, non-inferiority, randomised, phase 3 trial. Lancet. 2020;395:1613–1626. 9. Shah C, Arthur D, Vicini F. Efficacy, improved quality of life, and cost-effectiveness of partial breast irradiation: a triple-threat approach. JAMA Oncol. 2020;6:1859–1860. 10. Polgar C, Major T, Takacsi-Nagy Z, et al. Breast-conserving surgery followed by partial or whole breast irradiation: twenty-year results of a phase 3 clinical study. Int J Radiat Oncol Biol Phys. 2021;109:998–1006. 11. Vicini FA, Cecchini RS, White JR, et al. Long-term primary results of accelerated partial breast irradiation after breast-conserving surgery for early-stage breast cancer: a randomised, phase 3, equivalence trial. Lancet. 2019;394:2155–2164. 12. Strnad V, Ott OJ, Hildebrandt G, et al. 5-year results of accelerated partial breast irradiation using sole interstitial multicatheter brachytherapy versus whole-breast irradiation with boost after breast-conserving surgery for low-risk invasive and in-situ carcinoma of the female breast: a randomised, phase 3, non-inferiority trial. Lancet. 2016;387:229–238. 13. Offersen B, Alsner J, Nielsen HM, et al. Partial bs whole-breast irradiation for breast cancer patients in the randomized DBCG PBI trial. ESTRO. 2021. 14. Coles CE, Griffin CL, Kirby AM, et al. Partial-breast radiotherapy after breast conservation surgery for patients with early breast cancer (UK IMPORT LOW): 5-year results from a multicenter, randomized, controlled, phase 3, non-inferiority trial. Lancet. 2017;390:1048–1060. 15. Li X, Sanz J, Foro P, et al. Long-term results of a randomized partial irradiation trial compared to whole breast irradiation in the early stage and low-risk breast cancer patients after conservative surgery. Clin Transl Oncol. 2021;23:2127–2132. 16. Meattini I, Marrazzo L, Saieva C, et al. Accelerated partial-breast irradiation compared with whole-breast irradiation for early breast cancer: long-term results of the randomized Phase III APBI-IMRT-Florence trial. J Clin Oncol. 2020;38:4175–4183. 17. Whelan TJ, Julian JA, Berrang TS, et al. External beam accelerated partial breast irradiation versus whole breast irradiation after breast conserving surgery in women with ductal carcinoma in situ and node-negative breast cancer (RAPID): a randomised controlled trial. Lancet. 2019;394:2165–2172. 18. Schafter R, Strnad V, Polgar C, et al. Quality-of—life results for accelerated partial breast irradiation with interstitial brachytherapy vs. whole-breast irradiation in early stage breast cancer after breast-conserving surgery (GEC-ESTRO): 5-year results of a randomised, phase 3 trial. Lancet Oncol. 2018;19:834–844. 19. Bhattacharya IS, Haviand JS, Kirby AM, et al. Patient-reported outcomes over 5 years after whole- or partial-breasts radiotherapy: longitudinal analysis of the IMPORT-LOW (CRUK/06/003) Phase III randomized controlled trial. J Clin Oncol. 2019;37:305–317. 20. Meattini I, Saieva C, Miccinesi G, et al. Accelerated partial breast irradiation using intensity modulated radiotherapy versus whole breast irradiation: health-related quality of life final analysis from the Florence phase 3 trial. Eur J Cancer. 2017;76:17–26. 21. Julian TB, Constantino JP, Vicini FA, et al. Early toxicity results with 3D conformal external beam (CEBT) from the NSABP B-39/RTOG 0413 accelerated partial breast irradiation (APBI) trial. J Clin Oncol. 2011;29:S1011. 22. Rabinovitch R, Moughan J, Vicini F, et al. Long-term update of NRG Oncology RTOG 0319: a Phase 1 and 2 trial to evaluate 3-dimensional conformal radiation therapy confined to the region of the lumpectomy cavity for Stage I and II breast carcinoma. Int J Radiat Oncol Biol Phys. 2016;96:1054–1059. 23. Correa C, Hariss EE, Leonardi MC, et al. Accelerated partial breast irradiation: executive summary for the update of an ASTRO evidence-based consensus statement. Pract Radiat Oncol. 2017;l7:73–79. 24. Shah C, Vicini F, Shaitelman SF, et al. The American Brachytherapy Society consensus statement for accelerated partial-breast irradiation. Brachytherapy. 2018;17:154–170. 25. Manyam BV, Tendulkar R, Cherian S, et al. Evaluating candidacy for hypofractionated radiation therapy, accelerated partial breast irradiation, and endocrine therapy after breast conserving surgery: a Surveillance Epidemiology and End Results (SEER) analysis. Am J Clin Oncol. 2018;41:526–531. 26. Obi E, Parker SM, Asha W, et al. Trends in the delivery of accelerated partial breast irradiation therapy. ASTRO. 2022. 27. Ladbury C, Liu J, Radany E, et al. An examination of nationwide trends in accelerated partial breast irradiation—the replacement of breast brachytherapy with intraoperative radiotherapy and external beam radiation. Radiother Oncol. 2022;166:79–87. 28. Raphael DB, Russell NS, Immink JM, et al. Risk communication in a patient decision aid for radiotherapy in breast cancer: how to deal with uncertainty? Breast. 2020;51:105–113. 29. Nichol AM, Chan EK, Lucas S, et al. The use of hormone therapy alone versus hormone therapy and radiation therapy for breast cancer in elderly women: a population-based study. Int J Radiat Oncol Biol Phys. 2017;98:829–839. 30. Kirwan CC, Coles CE, Bliss J. PRIMETIME Protocol Working Group PRIMETIME Protocol Working Group. It’s PRIMETIME. Postoperative avoidance of radiotherapy: biomarker selection of women at very low risk of local recurrence. Clin Oncol (R Coll Radiol). 2016;28:594–596. 31. Bellon JR, Guo H, Barry WT, et al. Local-regional recurrence in women with small node-negative, HER2-positive breast cancer: results from a prospective multi-institutional study (the APT trial). Breast Cancer Res Treat. 2019;176:303–310. 32. Matuschek C, Bölke E, Haussmann J, et al. The benefit of adjuvant radiotherapy after breast conserving surgery in older patients with low risk breast cancer- a meta-analysis of randomized trials. Radiat Oncol. 2017;12:60. 33. Chesney TR, Yin JX, Rajaee N, et al. Tamoxifen with radiotherapy compared with Tamoxifen alone in elderly women with early-stage breast cancer treated with breast conserving surgery: a systematic review and meta-analysis. Radiother Oncol. 2017;123:1–9. 34. Howell A, Cuzick J, Baum M, et al. Results of the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial after completion of 5 years’ adjuvant treatment for breast cancer. Lancet. 2005;365:60–62. 35. Chirgwin JH, Giobbie-Hurder A, Coates AS, et al. Treatment adherence and its impact on disease-free survival in the breast international group 1-98 trial of tamoxifen and letrozole, alone and in sequence. J Clin Oncol. 2016;34:2452–2459. 36. Fisher B, Bryant J, Dignam JJ, et al. Tamoxifen, radiation therapy, or both for prevention of ipsilateral breast tumor recurrence after lumpectomy in women with invasive breast cancers of one centimeter or less (NSABP-B21). J Clin Oncol. 2002;20:4141–4149. 37. Blamey RW, Bates T, Chetty U, et al. Radiotherapy or tamoxifen after conserving surgery for breast cancers of excellent prognosis: British Association of Surgical Oncology (BASO) II trial. Eur J Cancer. 2013;49:2294–2302. 38. Ward MC, Vicini F, Chadha M, et al. Radiation therapy without hormone therapy for women age 70 or above with low-risk early breast cancer; a microsimulation. Int J Radiat Oncol Biol Phys. 2019;105:296–306. 39. Biszek SM, Yin H, Bedrosian I, et al. Lumpectomy plus hormone or radiation therapy alone for women aged 70 years or older with hormone receptor-positive early stage breast cancer in the modern era: an analysis of the National Cancer Database. Int J Radiat Oncol Biol Phys. 2019;105:795–802. 40. Ward MC, Vicini F, Al-Hilli Z, et al. Cost-effectiveness analysis of endocrine therapy alone versus partial-breast irradiation alone versus combined treatment for low-risk hormone-positive early-stage breast cancer in women aged 70 years or older. Breast Cancer Res Treat. 2020;182:355–365. 41. Jhawar SR, Alpert N, Taioli E, et al. Adjuvant radiation therapy alone is associated with improved overall survival compared to hormonal therapy alone in older women with estrogen receptor positive early stage breast cancer. Cancer Med. 2020;9:8345–8354. 42. Murphy CT, Li T, Wang LS, et al. Comparison of adjuvant radiation therapy alone versus radiation therapy and endocrine therapy in elderly women with early-stage, hormone receptor-positive breast cancer treated with breast-conserving surgery. Clin Breast Cancer. 2015;15:381–389. 43. Savard MF, Alzahrani MJ, Saunders D, et al. Experiences and perceptions of older adults with lower-risk hormone receptor-positive breast cancer about adjuvant radiotherapy and endocrine therapy: a patient survey. Curr Oncol. 2021;28:5215–5226. 44. Pignol JP, Hoekstra N, Wilke D, et al. Estimation of annual secondary lung cancer deaths using various adjuvant breast radiotherapy techniques for early-stage cancer. Front Oncol. 2021;11:713328. 45. Hoekstra N, Fleury E, Lara TRM, et al. Long-term risk of secondary cancer for various whole and partial breast irradiation techniques. Radiother Oncol. 2018;128:428–433. 46. Shah C, Vicini F, Beriwal S, et al. American Brachytherapy Society radiation oncology alternative payment model task force: quality measures and metrics for brachytherapy. Brachytherapy. 2022;21:63–74. 47. Shah C, Obi E, Tom MC, et al. Initial outcomes with image-guided partial breast irradiation delivered with intensity-modulated radiation therapy. Breast J. 2020;26:227–230. 48. Benedict SH, Yenice KM, Followill D, et al. Report No. 101- Stereotactic body radiation therapy: the report of AAPM Task Group 101. Med Phys. 2010;37:4078–4101. 49. Exclusive endocrine therapy or partial breast irradiation for women aged > 70 years early stage breast cancer (EUROPA). Available at: https://clinicaltrials.gov/ct2/show/NCT04134598. Accessed September 2, 2022. 50. Hannoun-Levi JM, Chamorey E, Boulahssass R, et al. Breast Cancer Working Group of the GEC-ESTRO. Endocrine therapy with accelerated partial breast irradiation or exclusive ultra-accelerated partial breast irradiation for women aged ≥ 60 years with early-stage breast cancer (EPOPE): the rationale for a GEC-ESTRO randomized phase III-controlled trial. Clin Transl Radiat Oncol. 2021;29:1–8. 51. Khan AJ, Chen PY, Yashar C, et al. Three-fraction accelerated partial breast irradiation (APBI) delivered with brachytherapy applicators is feasible and safe: first results from the TRIUMPH-T trial. Int J Radiat Oncol Biol Phys. 2019;104:67–74. 52. Trifiletti DM, Showalter TN, Libby B, et al. Intraoperative breast radiation therapy with image guidance: findings from CT images obtained in a prospective trial of intraoperative high-dose-rate brachytherapy with CT on rails. Brachytherapy. 2015;14:919–924. 53. Rahimi A, Simmons A, Kim DN, et al. Preliminary results of a multi-institutional Phase I dose escalation trial using single-fraction stereotactic partial breast irradiation for early stage breast cancer. Int J Radiat Oncol Biol Phys. 2022;112:663–670. 54. Bosma SCJ, Leij F, Vreeswijk S, et al. Five-year results of the preoperative accelerated partial breast irradiation (PABI) trial. Int J Radiat Oncol Biol Phys. 2020;106:958–967.

留言 (0)

沒有登入
gif