Skip to main content

Prospective phase II trial of preoperative hypofractionated proton therapy for extremity and truncal soft tissue sarcoma: the PRONTO study rationale and design

Abstract

Background

Oncologic surgical resection is the standard of care for extremity and truncal soft tissue sarcoma (STS), often accompanied by the addition of pre- or postoperative radiation therapy (RT). Preoperative RT may decrease the risk of joint stiffness and fibrosis at the cost of higher rates of wound complications. Hypofractionated, preoperative RT has been shown to provide acceptable outcomes in prospective trials. Proton beam therapy (PBT) provides the means to decrease dose to surrounding organs at risk, such as the skin, bone, soft tissues, and adjacent joint(s), and has not yet been studied in patients with extremity and truncal sarcoma.

Methods

Our study titled “PROspective phase II trial of preoperative hypofractionated protoN therapy for extremity and Truncal soft tissue sarcOma (PRONTO)” is a non-randomized, prospective phase II trial evaluating the safety and efficacy of preoperative, hypofractionated PBT for patients with STS of the extremity and trunk planned for surgical resection. Adult patients with Eastern Cooperative Group Performance Status ≤ 2 with resectable extremity and truncal STS will be included, with the aim to accrue 40 patients. Treatment will consist of 30 Gy radiobiological equivalent of PBT in 5 fractions delivered every other day, followed by surgical resection 2–12 weeks later. The primary outcome is rate of major wound complications as defined according to the National Cancer Institute of Canada Sarcoma2 (NCIC-SR2) Multicenter Trial. Secondary objectives include rate of late grade ≥ 2 toxicity, local recurrence-free survival and distant metastasis-free survival at 1- and 2-years, functional outcomes, quality of life, and pathologic response.

Discussion

PRONTO represents the first trial evaluating the use of hypofractionated PBT for STS. We aim to prove the safety and efficacy of this approach and to compare our results to historical outcomes established by previous trials. Given the low number of proton centers and limited availability, the short course of PBT may provide the opportunity to treat patients who would otherwise be limited when treating with daily RT over several weeks. We hope that this trial will lead to increased referral patterns, offer benefits towards patient convenience and clinic workflow efficiency, and provide evidence supporting the use of PBT in this setting.

Trial registration: NCT05917301 (registered 23/6/2023).

Background

Soft tissue sarcoma (STS) represents a rare and heterogeneous group of malignancies of mesenchymal origin which develops in bone and connective tissue. The American Cancer Society estimates 17,370 new cases and 7280 deaths due to cancers of the soft tissue, bone, and joints in the United States in 2023 [1]. Standard of care treatment approach for STS of the extremity and trunk typically involves limb sparing surgery and radiation therapy (RT), which has been shown to provide comparable outcomes to upfront amputation, with local control rates of 80–100% [2,3,4]. Moreover, multiple studies have demonstrated improved outcomes for patients with STS who are managed at high volume centers with appropriate expertise given the rarity of these tumors [5, 6].

Radiation may be delivered pre- or postoperatively using external beam radiation (EBRT), intraoperatively via a linear accelerator or brachytherapy, or using a combination of EBRT and brachytherapy. Preoperative RT may decrease the risk of joint stiffness and fibrosis after surgery due to the lower dose and smaller volumes delivered in comparison to postoperative RT while providing equivalent rates of local control [7,8,9]. Correspondingly, preoperative RT is now the preferred approach over postoperative RT according to the 2021 American Society for Therapeutic Radiation Oncology (ASTRO) Executive Summary [10]. This is particularly true for patients who are at higher risk for local recurrence with surgery alone, such as clinical scenarios in which there is concern for inability to achieve widely negative margins. These patients are more likely to see the benefits of decreased risk of local recurrence with radiation, with preoperative radiation associated with lower rates of irreversible late toxicities than postoperative radiation.

Wound complications continue to be a common toxicity experienced by patients who undergo preoperative RT. While modern techniques, smaller margins, and skin sparing volumes have decreased the rates of wound complications, 20–40% of patients still experience this adverse outcome, with 10–20% of patients requiring reoperation [11, 12]. Late toxicity is also a common complication which affects quality of life, occurring in 10–35% of patients treated with preoperative RT and surgical resection.

Hypofractionated preoperative RT has been shown to provide acceptable outcomes in retrospective studies and prospective trials [13,14,15,16,17]. Hypofractionation provides several potential advantages over conventional fractionation. These include patient convenience, shorter interval from diagnosis and treatment initiation to definitive surgical intervention, and radiobiological improvement in therapeutic ratio due to the low α/β of STS suggesting a benefit to higher doses per fraction [18, 19]. It also has the potential to increase patient referral for RT, particularly for patients who must travel long distances for daily treatment, and was shown to increase the number of patients treated with preoperative RT by three-fold at The University of California, Los Angeles (UCLA) after initiation of their trial, with a concomitant increase in the catchment area [13]. Previous prospective trials studying the use of both conventionally fractionated and hypofractionated preoperative RT in STS are summarized in Table 1.

Table 1 Prospective trials reporting outcomes after preoperative photon irradiation for extremity and truncal soft tissue sarcoma

The traditional form of RT is with photons, which can be delivered via 3D conformal or intensity-modulated radiotherapy (IMRT) techniques. These techniques typically use multiple beams with differing angles of delivery in order to treat the target to curative doses while attempting to minimize dose to nearby organs at risk. Unfortunately, these attempts are limited by the inherent characteristics of photons, which deposit a meaningful percentage of dose beyond the target, as shown in Fig. 1.

Fig. 1
figure 1

Representative color wash images in axial (left), coronal (middle), and sagittal (right) planes comparing hypofractionated preoperative proton (top row) versus photon (bottom row) dose deposition for a lower extremity sarcoma, with the dose increasing as the color ranges from blue to green to red. As shown, the target receives similar coverage in the two plans, while the normal tissue outside of the target receives lower doses in the proton plan

In contrast, protons have a finite range determined by the energy with which they are accelerated, allowing the delivery of high doses of radiation with little to no dose deposited beyond the target [25]. Thus, proton beam therapy (PBT) has the potential to decrease the rates of acute and late toxicities associated with photon irradiation by limiting dose to surrounding organs at risk such as the bone, adjacent joint(s), and uninvolved fascial compartments.

One limitation of PBT is the higher entrance dose in comparison with photon irradiation. Correspondingly, this would have the potential to increase the risk of radiation dermatitis and wound complications if not accounted for. However, consideration of the proton beam orientation with the aim of minimizing exposure of tissues which will be manipulated during surgical intervention, such as incision sites and adjacent flaps, may mitigate this risk.

To explore this potential risk, our group conducted a dosimetric analysis, comparing target coverage and dose to surrounding organs at risk when treating extremity STS with identical hypofractionated regimens of photon and proton radiation as those which will be used in this trial [26]. Doses to skin were statistically similar between the two modalities for most of the dosimetric endpoints outside of mean and maximum doses, which were slightly lower with PBT than photon RT. Thus, PBT also has the potential to provide similar or potentially lower rates of wound complications and toxicity than those associated with preoperative photon irradiation.

We therefore hypothesize that employing hypofractionated PBT will further reduce RT-associated toxicity, thus serving as a highly effective method for minimizing the risk of local recurrence without incurring additional toxicity or delaying surgery for technically resectable disease.

PBT has been utilized in the treatment of STS for several decades, mostly in the setting of re-irradiation and dose escalation, however it has not yet been studied in the setting of hypofractionation [27,28,29,30,31,32,33]. We present a trial where patients with STS undergo preoperative, hypofractionated PBT followed by surgery. We will establish toxicity rates associated with hypofractionated PBT and explore the effects of preoperative, hypofractionated PBT on surgical complications and local recurrence-free survival (LRFS).

Methods/design

This study titled “PROspective phase II trial of preoperative hypofractionated protoN therapy for extremity and Truncal soft tissue sarcOma (PRONTO)” is a prospective, single arm, phase II clinical trial designed to assess the safety and efficacy of preoperative, hypofractionated PBT for patients with extremity and truncal STS planned for surgical resection. We plan to accrue 40 patients who meet all inclusion and no exclusion criteria, as outlined in Table 2. The trial was activated April 2024, and we anticipate accrual to begin May 2024. With an anticipated accrual of 24–36 months for all 40 patients, we aim to complete accrual by late 2026 or early 2027.

Table 2 Inclusion and exclusion criteria

Trial organization

This trial was designed by the Departments of Radiation Oncology and Molecular Radiation Sciences, Oncology, Orthopaedic Surgery, and Physical Medicine and Rehabilitation of Johns Hopkins University School of Medicine. It is carried out by the Johns Hopkins Proton Therapy Center together with the Department of Radiation Oncology and Molecular Radiation Sciences at Johns Hopkins University School of Medicine. It is an investigator-initiated trial.

Investigators

Patients will be recruited by the Departments of Radiation Oncology and Molecular Radiation Sciences and Orthopaedic Surgery of Johns Hopkins University School of Medicine. All investigators cooperating in this trial are experienced oncologists from the fields of radiation oncology and orthopaedic surgery.

Ethical and legal considerations

The study protocol was approved by the Clinical Research Review Committee and the Institutional Review Boards of Johns Hopkins University School of Medicine (IRB00335181). The trial is carried out by adhering to local legal and regulatory requirements. All patients will sign informed consent before enrollment on trial after the nature, scope, and potential consequences of participation are explained by a physician.

Study objectives and endpoints

The primary outcome is the rate of major wound complications occurring within 90 days after surgery, as defined according to the National Cancer Institute of Canada Sarcoma2 (NCIC-SR2) Multicenter Trial [7, 8]. This includes “secondary operation under general or regional anaesthesia for wound repair (debridement, operative drainage, and secondary wound closure including rotationplasty, free flaps, or skin grafts), or wound management without secondary operation…[including] an invasive procedure without general or regional anaesthesia (mainly aspiration of seroma), readmission for wound care such as intravenous antibiotics, or persistent deep packing for 120 days or longer.” Secondary objectives include safety and tolerability (acute grade ≥ 3 adverse events based on National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0 [CTCAE v5.0]), 1- and 2-year LRFS and distant metastasis-free survival (DMFS) rates, incidence of CTCAE v5.0 late grade ≥ 2 radiation toxicity (fibrosis, lymphedema, or joint stiffness), functional outcomes using the Musculoskeletal Tumor Rating Scale (MSTS) and Toronto Extremity Salvage Score (TESS), quality of life assessed via Functional Assessment of Cancer Therapy-General (FACT-G) forms, and pathologic response (complete response, positive margins, and percentage necrosis in comparison with pre-treatment biopsy when available).

Pretreatment evaluation

Table 3 outlines time flow for all work up, enrollment, interventions, and assessments.

Table 3 Time flow for all work up, enrollment, interventions, and assessments

Initial work up includes clinical evaluation, staging computed tomography (CT) and/or magnetic resonance imaging (MRI) of the primary site, chest CT, histologic confirmation of STS, and determination of eligibility and resectability by clinical assessment and laboratory studies.

Treatment assignment and schedule

All eligible patients who provide informed consent are registered and follow work-up and treatment as outlined in Fig. 2.

Fig. 2
figure 2

Flow chart outlining trial schema, study procedures, follow-up, and planned analysis. Abbreviations: STS, soft tissue sarcoma; yrs, years; ECOG PS, Eastern Cooperative Oncology Group Performance Status; GyE, Gy radiobiologic equivalent; PBT, proton beam therapy; CTV, clinical target volume; OTV, on treatment visit; CTCAE, common terminology criteria for determining adverse events version 5.0; q3-6mo x2yrs, every 3–6 months for 2 years; CT, computed tomography; MRI, magnetic resonance imaging; SOC, standard of care

Treatment details

Enrolled patients undergo CT simulation. Due to the variety of sites in which STS may occur and the mobility of extremities, careful consideration of immobilization is required prior to simulation. Immobilization must be reproducible to a high degree. Examples include the use of large vac loc or body fix bags for lower extremity immobilization with the addition of aquaplast mold to immobilize the foot and/or knee.

After completing simulation, patients begin PBT within 1–3 weeks from their simulation, treating with 5 fractions given every other weekday, to a total dose of 30 Gy radiobiological equivalent for a total of 10–12 calendar days from treatment start to completion. Clinical target volumes include gross tumor with a 3 cm margin longitudinally and 1.5 cm margin radially excluding natural barriers of spread, in addition to any surrounding edema seen on T2 weighted MRI, cropped 0.3 cm from the skin. In place of the planning target volumes (PTVs) typically used when treating with photon radiation, robust optimization of PBT is used to account for setup and range uncertainties. Target and organ at risk planning goals are listed in Table 4A, B.

Table 4 (A) Target dose and coverage parameters, (B) Normal tissue constraints

Patients undergo daily kilovoltage x-ray and cone beam CT (CBCT) before daily PBT to assist with treatment set-up. Quality assurance (QA) verification CT +/− MRI are performed if feasible within the first 2 fractions and repeated during the treatment course as needed.

Oncologic surgical resection occurs within 2–12 weeks of completing PBT. Patients will be followed in the postoperative setting according to standard of care surveillance for STS, as outlined in Fig. 2.

Outcomes measured and follow-up

Medical and demographic details, performance status (Eastern Cooperative Group Performance Status scale), laboratory studies, and imaging are captured at baseline. CTCAE v5.0 toxicity, functional status using MSTS and TESS forms, and quality of life assessed by FACT-G forms are captured at baseline, during PBT, after PBT but prior to surgery, within 3 months after surgery, and every 3–6 months thereafter. Pathologic response is evaluated using rates of pathologic complete response, positive margins, and percentage necrosis in comparison with pre-treatment biopsy. Postoperative wound complications are assessed after surgery at 2 weeks, 4–6 weeks, and every 3–6 months thereafter. LRFS is assessed clinically and by postoperative MRI +/− CT of the primary site at 3 and 6 months after surgery, CT and/or MRI every 3–6 months for the first 2 years, and every 6–12 months thereafter. DMFS is assessed via chest CT or X-ray at 3 months after surgery, every 3–6 months for the first 2 years, and every 6–12 months thereafter. Patients will be followed for a minimum of 2 years on study.

Statistical considerations

The primary outcome, rate of major wound complications, will be estimated as the number of major wound complications occurring within 90 days after surgery during the study period divided by the number of evaluable patients who have completed PBT and surgery. A sample size of 36 evaluable patients will allow us to obtain a two-sided 90% exact confidence interval with a width less than 0.3 if the incidence is between 30 and 60%, based on those reported in multiple prospective trials in patients receiving preoperative, hypofractionated photon-based EBRT followed by surgical resection for STS [13, 24].

Guarding against the potential that 10% of patients may be not evaluable, we plan to accrue 40 patients in total in order to provide 36 evaluable patients. Patients will be considered evaluable as long as they have received 1 fraction of hypofractionated PBT and completed surgery.

Safety of hypofractionated PBT in isolation will be analyzed by calculating the incidence of CTCAE v5.0 grade ≥ 3 adverse events occurring between receipt of the first fraction of PBT and the day of surgery. Specific adverse events will be reported as frequencies. Patient or cancer characteristics associated with acute PBT-related toxicity will be explored with logistic regression models provided a sufficient number of events. Tolerability will be defined and reported as the frequency of patients who stop treatment with PBT due to an adverse event.

LRFS and DMFS will be reported at 1- and 2-years post-enrollment based upon estimates produced using Kaplan–Meier methods. Patient and tumor characteristics associated with LRFS and DMFS will be explored using Cox proportional hazards models. A sensitivity analysis will be performed including patients who completed all 5 fractions of their hypofractionated PBT if this does not apply to the entire population.

Early stopping rules

Based on existing literature, we assume the rate of major wound complication within 90 days after surgery when treated with preoperative photon RT is about 25–30%. Therefore, to minimize risks, safety will be monitored by a Bayesian stopping rule for the rate of major wound complications greater than 60%. Table 5 summarizes the continuous stopping rule for the 36 evaluable patients, evaluated in cohorts of 3 patients, starting from 6th evaluable patient.

Table 5 Stopping rule for safety

At any time if the stopping criterion is met, accrual to the trial will be temporarily suspended, and the principal investigators and study team will review the toxicity data to recommend modification or termination of the trial.

Discussion

While previous trials have provided evidence supporting hypofractionated photon RT and ongoing trials are assessing conventionally fractionated PBT, PRONTO represents the first trial evaluating the use of hypofractionated PBT for STS to our knowledge. We aim to prove the safety and efficacy of this approach and to compare our results to historical outcomes established by previous trials.

Given the low number of proton centers and limited availability, the short course of PBT outlined in this protocol is worthy of acknowledgement, as it may provide the opportunity to treat patients who would otherwise be limited when treating with daily RT over several weeks, such as those who must travel from long distances for treatment. We hope that this trial will lead to increased referral patterns, offer benefits towards patient convenience and clinic workflow efficiency, and provide evidence supporting the use of PBT in this setting.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

STS:

Soft tissue sarcoma

RT:

Radiation therapy

PBT:

Proton beam therapy

PRONTO:

PROspective phase II trial of preoperative hypofractionated protoN therapy for extremity and Truncal soft tissue sarcOma

EBRT:

External beam radiation

ASTRO:

American Society for Therapeutic Radiation Oncology

N:

Patient number

BED:

Biologically effective dose

R0 resection:

Surgery with negative margins

Fx:

Fractions

3DCRT:

3D conformal radiation therapy

Wk:

Week(s)

Yr(s):

Year(s)

G4:

Grade 4

IMRT:

Intensity-modulated radiation therapy

Mo:

Month(s)

G3:

Grade 3

G2:

Grade 2

D:

Day(s)

G5:

Grade 5

Q2d:

Every other day

SBRT:

Stereotactic body radiation therapy

R1 resection:

Surgery with microscopically positive margins

LRFS:

Local recurrence-free survival

DMFS:

Distant metastasis-free survival

CTCAE v5.0:

National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0

MSTS:

Musculoskeletal Tumor Rating Scale

TESS:

Toronto Extremity Salvage Score

FACT-G:

Functional Assessment of Cancer Therapy-General

CT:

Computed tomography

MRI:

Magnetic resonance imaging

ECOG PS:

Eastern Cooperative Oncology Group Performance Status

GyE:

Gy radiobiologic equivalent

CTV:

Clinical target volume

OTV:

On treatment visit

SOC:

Standard of care

GTV:

Gross tumor volume

Vx < y:

Volume of organ at risk receiving x dose < y percentage or volume

CBCT:

Cone beam CT

QA:

Quality assurance

References

  1. American Cancer Society. Cancer Facts and Figures, 2023 Atlanta: American Cancer Society; 2023. https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2023/2023-cancer-facts-and-figures.pdf.

  2. Rosenberg SA, Tepper J, Glatstein E, Costa J, Baker A, Brennan M, et al. The treatment of soft-tissue sarcomas of the extremities: prospective randomized evaluations of (1) limb-sparing surgery plus radiation therapy compared with amputation and (2) the role of adjuvant chemotherapy. Ann Surg. 1982;196(3):305–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Yang JC, Chang AE, Baker AR, Sindelar WF, Danforth DN, Topalian SL, et al. Randomized prospective study of the benefit of adjuvant radiation therapy in the treatment of soft tissue sarcomas of the extremity. J Clin Oncol. 1998;16(1):197–203.

    Article  CAS  PubMed  Google Scholar 

  4. McGee L, Indelicato DJ, Dagan R, Morris CG, Knapik JA, Reith JD, et al. Long-term results following postoperative radiotherapy for soft tissue sarcomas of the extremity. Int J Radiat Oncol Biol Phys. 2012;84(4):1003–9.

    Article  PubMed  Google Scholar 

  5. Gutierrez JC, Perez EA, Moffat FL, Livingstone AS, Franceschi D, Koniaris LG. Should soft tissue sarcomas be treated at high-volume centers? An analysis of 4205 patients. Ann Surg. 2007;245(6):952–8.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Derbel O, Heudel PE, Cropet C, Meeus P, Vaz G, Biron P, et al. Survival impact of centralization and clinical guidelines for soft tissue sarcoma (A prospective and exhaustive population-based cohort). PLoS ONE. 2017;12(2):e0158406.

    Article  PubMed  PubMed Central  Google Scholar 

  7. O’Sullivan B, Davis AM, Turcotte R, Bell R, Catton C, Chabot P, et al. Preoperative versus postoperative radiotherapy in soft-tissue sarcoma of the limbs: a randomised trial. Lancet. 2002;359(9325):2235–41.

    Article  PubMed  Google Scholar 

  8. Davis AM, O’Sullivan B, Turcotte R, Bell R, Catton C, Chabot P, et al. Late radiation morbidity following randomization to preoperative versus postoperative radiotherapy in extremity soft tissue sarcoma. Radiother Oncol. 2005;75(1):48–53.

    Article  PubMed  Google Scholar 

  9. Al-Absi E, Farrokhyar F, Sharma R, Whelan K, Corbett T, Patel M, et al. A systematic review and meta-analysis of oncologic outcomes of pre- versus postoperative radiation in localized resectable soft-tissue sarcoma. Ann Surg Oncol. 2010;17(5):1367–74.

    Article  PubMed  Google Scholar 

  10. Salerno KE, Alektiar KM, Baldini EH, Bedi M, Bishop AJ, Bradfield L, et al. Radiation therapy for treatment of soft tissue sarcoma in adults: executive summary of an ASTRO Clinical Practice Guideline. Pract Radiat Oncol. 2021;11(5):339–51.

    Article  PubMed  Google Scholar 

  11. Wang D, Zhang Q, Eisenberg BL, Kane JM, Li XA, Lucas D, et al. Significant reduction of late toxicities in patients with extremity sarcoma treated with image-guided radiation therapy to a reduced target volume: results of radiation therapy oncology group RTOG-0630 Trial. J Clin Oncol. 2015;33(20):2231–8.

    Article  PubMed  PubMed Central  Google Scholar 

  12. O’Sullivan B, Griffin AM, Dickie CI, Sharpe MB, Chung PW, Catton CN, et al. Phase 2 study of preoperative image-guided intensity-modulated radiation therapy to reduce wound and combined modality morbidities in lower extremity soft tissue sarcoma. Cancer. 2013;119(10):1878–84.

    Article  PubMed  Google Scholar 

  13. Kalbasi A, Kamrava M, Chu FI, Telesca D, Van Dams R, Yang Y, et al. A phase II trial of 5-day neoadjuvant radiotherapy for patients with high-risk primary soft tissue sarcoma. Clin Cancer Res. 2020;26(8):1829–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Pennington JD, Eilber FC, Eilber FR, Singh AS, Reed JP, Chmielowski B, et al. Long-term outcomes with ifosfamide-based hypofractionated preoperative chemoradiotherapy for extremity soft tissue sarcomas. Am J Clin Oncol. 2018;41(12):1154–61.

    Article  PubMed  Google Scholar 

  15. Koseła-Paterczyk H, Szacht M, Morysiński T, Ługowska I, Dziewirski W, Falkowski S, et al. Preoperative hypofractionated radiotherapy in the treatment of localized soft tissue sarcomas. Eur J Surg Oncol. 2014;40(12):1641–7.

    Article  PubMed  Google Scholar 

  16. Kubicek GJ, LaCouture T, Kaden M, Kim TW, Lerman N, Khrizman P, et al. Preoperative radiosurgery for soft tissue sarcoma. Am J Clin Oncol. 2018;41(1):86–9.

    Article  PubMed  Google Scholar 

  17. Parsai S, Lawrenz J, Kilpatrick S, Rubin B, Hymes C, Gray M, et al. Early outcomes of preoperative 5-fraction radiation therapy for soft tissue sarcoma followed by immediate surgical resection. Adv Radiat Oncol. 2020;5(6):1274–9.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Thames HD, Suit HD. Tumor radioresponsiveness versus fractionation sensitivity. Int J Radiat Oncol Biol Phys. 1986;12(4):687–91.

    Article  CAS  PubMed  Google Scholar 

  19. van Leeuwen CM, Oei AL, Crezee J, Bel A, Franken NAP, Stalpers LJA, et al. The alfa and beta of tumours: a review of parameters of the linear-quadratic model, derived from clinical radiotherapy studies. Radiat Oncol. 2018;13(1):96.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Kraybill WG, Harris J, Spiro IJ, Ettinger DS, DeLaney TF, Blum RH, et al. Phase II study of neoadjuvant chemotherapy and radiation therapy in the management of high-risk, high-grade, soft tissue sarcomas of the extremities and body wall: Radiation Therapy Oncology Group Trial 9514. J Clin Oncol. 2006;24(4):619–25.

    Article  CAS  PubMed  Google Scholar 

  21. Lansu J, Bovée JVMG, Braam P, van Boven H, Flucke U, Bonenkamp JJ, et al. Dose reduction of preoperative radiotherapy in myxoid liposarcoma: a nonrandomized controlled trial. JAMA Oncol. 2021;7(1):e205865.

    Article  PubMed  Google Scholar 

  22. Meyer JM, Perlewitz KS, Hayden JB, Doung YC, Hung AY, Vetto JT, et al. Phase I trial of preoperative chemoradiation plus sorafenib for high-risk extremity soft tissue sarcomas with dynamic contrast-enhanced MRI correlates. Clin Cancer Res. 2013;19(24):6902–11.

    Article  CAS  PubMed  Google Scholar 

  23. Gobo Silva ML, Lopes de Mello CA, Aguiar Junior S, D'Almeida Costa F, Stevanato Filho PR, Santoro Bezerra T, et al. Neoadjuvant hypofractionated radiotherapy and chemotherapy for extremity soft tissue sarcomas: Safety, feasibility, and early oncologic outcomes of a phase 2 trial. Radiother Oncol. 2021;159:161–7.

  24. Bedi M, Singh R, Charlson J, Kelly T, Johnstone C, Wooldridge A, et al. Could five be the new twenty-five? Long-term oncologic outcomes from a phase ii, prospective, 5-fraction pre-operative radiation therapy trial in patients with localized soft tissue sarcoma. Adv Radiat Oncol. 2022;7:100850.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Wilson RR. Radiological use of fast protons. Radiology. 1946;47(5):487–91.

    Article  CAS  PubMed  Google Scholar 

  26. Thomas R, Chen H, Gogineni E, Halthore A, Floreza B, Esho-Voltaire T, et al. Comparing ultra-hypofractionated proton versus photon therapy in extremity soft tissue sarcoma. Int J Part Ther. 2023;9(3):30–9.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Frisch S, Timmermann B. The evolving role of proton beam therapy for sarcomas. Clin Oncol (R Coll Radiol). 2017;29(8):500–6.

    Article  CAS  PubMed  Google Scholar 

  28. Kozak KR, Adams J, Krejcarek SJ, Tarbell NJ, Yock TI. A dosimetric comparison of proton and intensity-modulated photon radiotherapy for pediatric parameningeal rhabdomyosarcomas. Int J Radiat Oncol Biol Phys. 2009;74(1):179–86.

    Article  PubMed  Google Scholar 

  29. Verma V, Rwigema JM, Malyapa RS, Regine WF, Simone CB. Systematic assessment of clinical outcomes and toxicities of proton radiotherapy for reirradiation. Radiother Oncol. 2017;125(1):21–30.

    Article  PubMed  Google Scholar 

  30. Haas RL, Miah AB, LePechoux C, DeLaney TF, Baldini EH, Alektiar K, et al. Preoperative radiotherapy for extremity soft tissue sarcoma; past, present and future perspectives on dose fractionation regimens and combined modality strategies. Radiother Oncol. 2016;119(1):14–21.

    Article  PubMed  Google Scholar 

  31. Gaito S, Abravan A, Richardson J, Lowe M, Indelicato DJ, Burnet N, et al. Skin toxicity profile of photon radiotherapy versus proton beam therapy in paediatric and young adult patients with sarcomas. Clin Oncol (R Coll Radiol). 2021;3:507–16.

    Article  Google Scholar 

  32. DeLaney TF, Haas RL. Innovative radiotherapy of sarcoma: proton beam radiation. Eur J Cancer. 2016;62:112–23.

    Article  PubMed  Google Scholar 

  33. DeLaney TF, Chen YL, Baldini EH, Wang D, Adams J, Hickey SB, et al. Phase 1 trial of preoperative image guided intensity modulated proton radiation therapy with simultaneously integrated boost to the high risk margin for retroperitoneal sarcomas. Adv Radiat Oncol. 2017;2(1):85–93.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This trial is supported through the Robert L. Sloan Fund for Cancer Research, awarded from the Albert L. Tucker and Elizabeth T. Tucker Foundation and Sibley Memorial Hospital Foundation.

Author information

Authors and Affiliations

Authors

Contributions

All authors: conceptualization, methodology, validation, investigation, resources, writing—review and editing, and visualization. EG: writing—original draft preparation and funding acquisition. CH: formal analysis. CD: supervision, project administration, and funding acquisition. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Emile Gogineni.

Ethics declarations

Ethics approval and consent to participate

The study was conducted in accordance with the Declaration of Helsinki and approved by the Clinical Research Review Committee and the Institutional Review Boards of Johns Hopkins University School of Medicine: #00335181. All participants enrolled in the trial will be required to provide written informed consent.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gogineni, E., Chen, H., Hu, C. et al. Prospective phase II trial of preoperative hypofractionated proton therapy for extremity and truncal soft tissue sarcoma: the PRONTO study rationale and design. Radiat Oncol 19, 56 (2024). https://doi.org/10.1186/s13014-024-02447-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13014-024-02447-0

Keywords