Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Critical Reviews in Oncology / Hematology 167 (2021) 103500

Contents lists available at ScienceDirect

Critical Reviews in Oncology / Hematology


journal homepage: www.elsevier.com/locate/critrevonc

Thoracic re-irradiation with 3D-conformal or more advanced techniques: A


systematic review of treatment safety by the Re-irradiation Study Group of
the Italian Association of Radiation and Oncology AIRO
Marta Maddalo a, 1, Elisa D’Angelo b, *, 1, Francesco Fiorica c, Angela Argenone d,
Melissa Scricciolo e, Salvatore Cozzi f, Alessia Nardangeli g, Francesco Dionisi h,
Gianluca Costantino a, Stefano Vagge i, Antonio Pontoriero j, Vittorio Donato k, 2,
Mariangela Massaccesi l, on behalf of the Re-irradiation Study Group of the Italian Association of
Radiation Oncology
a
Department of Radiation Oncology, ASST Spedali Civili of Brescia, P.le Spedali Civili 1, 25123, Brescia, Italy
b
Department of Radiotherapy, University Hospital of Modena, L. del Pozzo 71, 41121, Modena, Italy
c
Department of Radiation Oncology and Nuclear Medicine, State Hospital Mater Salutis AULSS 9, 37045, Legnago (VR), Italy
d
Radiotherapy Unit, AORN San PIO, Via dell’Angelo, 82100, Benevento, Italy
e
Radiation Therapy Unit, Ospedale dell’Angelo, Via Paccagnella 11, 30174, Venezia, Italy
f
Radiation Therapy Unit, Azienda USL-IRCCS di Reggio Emilia, 42122, Reggio Emilia, Italy
g
Fondazione Policlinico Universitario "A. Gemelli" IRCCS, UOC di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed
Ematologia, L. Gemelli 1, 00168, Roma, Italy
h
Department of Research and Advanced Technology, Radiation Oncology Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Roma, Italy
i
Department of Radiation Oncology, Azienda Ospedaliera Universitaria San Martino di Genova-IST, Istituto Nazionale Ricerca sul Cancro, Genoa, Italy
j
Dept. of Radiation Oncology, University of Messina, 98125, Messina, Italy
k
Radiation Oncology Division, Oncology and Speciality Medicine Department, San Camillo-Forlanini Hospital, 00152, Roma, Italy
l
Radiation Oncology Department, Gemelli-ART, Università Cattolica S. Cuore, L. Gemelli 1, 00168, Roma, Italy

A R T I C L E I N F O A B S T R A C T

Keywords: Re-irradiation (re-RT) is a treatment modality that has been actively investigated in recurrent lung cancer or in
Re-irradiation lung metastases appeared in previously irradiated areas. A literature search, according PRISMA recommenda­
Recurrent lung cancer tions and a meta-analysis technique were performed with the aims to identify possible factors related to the
Lung metastases
toxicity incidence and severity of ≥ G3 acute toxicity. 1243 patients and 36 studies, met inclusion criteria. Our
Stereotactic body radiotherapy
3D conformal radiotherapy
results, showed that there was no difference in ≥ G3 acute (10,5%) toxicity rate with respect to different ra­
Carbon ion radiotherapy diation techniques, cumulative dose and re-irradiation total dose and fractionation. Factors eventually related to
Proton therapy severe toxicity were described. The frequent lack of a sufficient description of the treatment’s intent, the het­
Toxicity erogeneity in technique and radiotherapy regimen, makes balancing risk and benefit of re-RT based on published
data even more difficult.

1. Introduction systemic therapies has led to a substantial change in the natural history
of lung cancer. Nowadays a substantial number of patients can be
The development of increasingly effective and selective local and treated with more than a purely palliative approach, resulting into an

* Corresponding author at: Radiation Oncology Unit, University Hospital of Modena, Via del pozzo, 71, 41124, Modena, Italy.
E-mail addresses: marta.maddalo@aol.com (M. Maddalo), dangelo.elisa@policlinico.mo.it (E. D’Angelo), francesco.fiorica@aulss9.veneto.it (F. Fiorica), angela.
argenone@ao-rummo.it (A. Argenone), melissa.scricciolo@aulss3.veneto.it (M. Scricciolo), salvatore.cozzi@ausl.re.it (S. Cozzi), alessia.nardangeli@guest.
policlinicogemelli.it (A. Nardangeli), francescodionisi2@gmail.com (F. Dionisi), stefano.vagge@hsanmartino.it (S. Vagge), antonio.pontoriero@unime.it
(A. Pontoriero), vdonato@scamilloforlanini.rm.it (V. Donato), mariangela.massaccesi@policlinicogemelli.it (M. Massaccesi).
1
Equally contribute.
2
President of AIRO (Italian Association of Radiotherapy and Clinical Oncology), Italy.

https://doi.org/10.1016/j.critrevonc.2021.103500
Received 24 March 2021; Received in revised form 20 August 2021; Accepted 10 October 2021
Available online 21 October 2021
1040-8428/© 2021 Elsevier B.V. All rights reserved.
M. Maddalo et al. Critical Reviews in Oncology / Hematology 167 (2021) 103500

increasing number of long-term survivors. These patients can develop remains an issue when considering thoracic re-irradiation.
during the follow-up metachronous oligometastatic progression, local A wide heterogeneity of indications, techniques and treatment reg­
recurrent cancer or a second primary tumor and can be treated a second imens makes it difficult to easily draw definitive safety considerations
time either with a curative purpose or with palliative intent. from currently available clinical data. The aim of this review, performed
Radiation therapy represents a mainstay of local lung cancer treat­ within the framework of the AIRO Re-irradiation Study Group, is to
ment, both in the curative and the palliative settings, with or without systematically analyze the most recent literature in order to obtain a
combined systemic therapy. Approximately 20 %–50 % of patients with clearer picture of re-irradiation-induced toxicity focusing on identifi­
Non Small Cell Lung Cancer (NSCLC) and 36–52 % of patients with cation of the incidence and severity of radiation pneumonitis/pulmo­
Small Cell Lung Cancer (SCLC) develop a loco-regional failure after nary fibrosis (RP/PF), esophagitis, chest wall pain/rib fracture,
primary radiotherapy (O’Rourke et al., 2010; Auperin et al., 2006; radiation dermatitis and any severe/fatal adverse events G4-G5 (AE).
Turrisi et al., 1999). Survival and local control data will also be reported, to clarify
Radiation oncologists frequently face recurrences, second primary whether thoracic re-irradiation provides sufficient tumor control with
tumors or metastasis developed within a previously irradiated thoracic acceptable toxicity.
area (Jeremic et al., 2001).
Local treatments for patients already irradiated for chest malig­ 2. Methods
nancies include radical surgery, radiofrequency ablation (RFA), and
radiation therapy. Re-irradiation has become an effective option, both 2.1. Selection of studies
for palliative and radical treatment intent in patients not suitable for
surgery. Usually thoracic re-irradiation is defined as at least two courses Using recommendations of the Preferred Reporting Items for Sys­
of thoracic radiotherapy with an overlap of at least the 50 % isodose tematic Reviews and Meta-Analyses (PRISMA) (Moher et al., 2009) a
levels between the two courses. Recent advances in radiation techniques literature search on MEDLINE, EMBASE, OVID, and Cochrane databases,
and technology, such as image-guided radiotherapy (IGRT), stereotactic published from January 1995 to October 2019, was performed (Fig. 1).
body radiotherapy (SBRT) and particle therapy (in particular proton Key words used were (‘reirradiation’/exp OR‘re-irradiation’ OR ‘reir­
beam therapy - PBT), have the potential to widen the therapeutic win­ radiation’ AND ‘thoracic’) and (re irradiation/exp OR re irradiation OR
dow and thus facilitate thoracic re-irradiation. Nevertheless, the toler­ re irradiation AND lung).
ance of repeated exposure of previously irradiated organs at risk (OARs)

Fig. 1. Flow chart of studies screened, excluded, and identified for analysis according PRISMA.

2
M. Maddalo et al. Critical Reviews in Oncology / Hematology 167 (2021) 103500

2.2. Selection criteria and data extraction published between 2002 and 2019 in 10 different countries. Only 2
studies had a prospective design while the remaining 34 were retro­
Retrospective and prospective studies with at least 10 patients were spective (Chao et al., 2017; Wu et al., 2003). The analyzed population of
included in the analysis. English language, full-text articles and presence each study varied greatly, ranging from 10 (Chao et al., 2017; Wegner
of detailed toxicity data were the other inclusion criteria. References et al., 2018) to 102 patients (McAvoy et al., 2014) with a total number of
listed in the screened articles were also evaluated and cross-referenced 1283 patients who had been treated with re-irradiation in the thoracic
to ensure completeness. Abstracts, letters, proceedings from scientific region. Treatment intent (i.e. palliation versus cure) was generally not
meetings, editorials, reviews without original data and duplicate studies well defined. Median follow-up after re-irradiation was 15 months
were excluded during the initial database screening. Thoracic re- (range 6.5–47.3 months), with six (Chao et al., 2017; McAvoy et al.,
irradiation was reported for recurring primary, lymph node recur­ 2014; Ebara et al., 2007; Ceylan et al., 2017; Ren et al., 2018; Schlampp
rence, or both. et al., 2019) of thirty-six studies reporting a median follow up of <10
Five authors (MM, AA, MS, AN and ED) performed the selection of months.
studies independently, then the decision for full-text articles eligible for
quantitative analysis was performed by three reviewers (FF, MM, ED) 3.1. Re-irradiation site, dose and time interval from first radiotherapy
and any disagreement was resolved by discussing the issues that led to
disagreement. In our selection, the degree of volume overlap between the previous
and re-irradiation plans was often not well detailed. When available, the
2.3. Review of the trials reported locations of re-irradiated lesions indicated that the re-
irradiation volumes were likely to have been fully encompassed by at
Studies were first reviewed for pertinence of patients and treatments least the 50 % isodose of the previous radiotherapy plan and most often,
characteristics. Methodological quality of studies was assessed with a wholly or partially, within the high dose region.
checklist for quality appraisal of case series studies produced by the Previous radiotherapy was delivered with a median dose of 60 Gy,
Institute of Health Economics (IHE) after modification to improve ranging from 28.8 to 80 Gy using a conventional treatment (2D or 3D
applicability (www.ihe.ca/research-programs/rmd/cssqac/cssqac-abo radiotherapy, with a fraction dose ≤ 3 Gy/die) in 15 studies (Wu et al.,
ut). The following items were evaluated for each study: a clearly 2003; Ebara et al., 2007; Ren et al., 2018; Schlampp et al., 2019; Oka­
stated aim, prospective data collection, multi-center study, consecutive moto et al., 2002; Tada et al., 2005; Kelly et al., 2010; Liu et al., 2012;
patients, described characteristics of patients, clearly stated eligibility Reyngold et al., 2013; Kruser et al., 2014; Trovo et al., 2014; Owen et al.,
criteria, described intervention, reported losses to follow-up and adverse 2014; Hong et al., 2019; Sumodhee et al., 2019) and a combination of
events, conclusions of the study supported by the results. Studies conventional treatment and stereotactic radiotherapy in 8 studies
reporting data on re-irradiation with different intent (palliative versus (Ceylan et al., 2017; Trakul et al., 2012; McAvoy et al., 2013; Meijneke
curative) and techniques (conventional fractionation, SBRT, proton or et al., 2013; Griffioen et al., 2014; Kilburn et al., 2014; Patel et al., 2015;
carbon ion radiotherapy) were equally accepted and included. To Binkley et al., 2016). In the remaining studies, stereotactic radiotherapy
improve the comparability of the different re-irradiation regimens and alone was used in five studies (Hearn et al., 2014; Peulen et al., 2011;
to assess the relationship between radiation dose and toxicity, when the Yoshitake et al., 2013; Caivano et al., 2018; Ogawa et al., 2018),
nominal treatment dose was not reported in the study, equivalent dose carbon-ion or proton therapy in two studies (Karube et al., 2017a;
in 2 Gy fractions (EQD2) was calculated according to D × (D/n + α/β)/(2 Hayashi et al., 2018), brachytherapy in two (Wegner et al., 2018; Gill
+ α/β), derived from the linear-quadratic model. The α/β-value used for et al., 2015) and in two other studies (McAvoy et al., 2013; Ohguri et al.,
the target volume was 10, while it was 3 for organs at risk. We calculated 2012) a combination of conventional and proton therapy was used while
cumulative equivalent dose assuming no repair between irradiation se­ in 3 studies (Chao et al., 2017; Tada et al., 2005; Ohguri et al., 2012) the
ries (Abusaris et al., 2012). To determine the pooled G≥3 toxicity rate, technique of previous radiotherapy was not described. The mean time
overall survival at 1 and 2 years, locoregional progression free survival elapsed since previous irradiation ranged from 7.9 to 51 months, the
at 1 and 2 years and symptom relief, an established meta-analysis median interval time was 18 months.
technique over single arm studies was performed. For this purpose, we A stereotactic radiotherapy technique was used to re-irradiate pa­
calculated the estimated population proportion of toxicity, 1 and 2 year tients in fourteen studies (Wegner et al., 2018; Ceylan et al., 2017; Kelly
overall survival, 1 and 2 year locoregional control and symptom relief et al., 2010; Liu et al., 2012; Reyngold et al., 2013; Trovo et al., 2014;
with 95 % CI for each separate study (Julious and Campbell, 1998). Owen et al., 2014; Trakul et al., 2012; Patel et al., 2015; Hearn et al.,
Pooled effect size aided the general evaluation of re-irradiation risk and 2014; Peulen et al., 2011; Caivano et al., 2018; Ogawa et al., 2018; Gill
effect. Heterogeneity across studies was examined by the Cochran Q et al., 2015), while in ten studies (Wu et al., 2003; Ebara et al., 2007;
chi-square test and the I 2 statistic. Studies with an I 2 statistic of 25–50 Schlampp et al., 2019; Okamoto et al., 2002; Tada et al., 2005; Kruser
%, 50–75 %, and >75 % were considered to have low, moderate, and et al., 2014; Griffioen et al., 2014; Yoshitake et al., 2013; Ohguri et al.,
high heterogeneity, respectively (Higgins et al., 2003). We used ran­ 2012; Cetingoz et al., 2009) a conventional treatment (2D or 3D
dom-effects models because there was great subjectivity given the lack radiotherapy with a fraction dose ≤ 3 Gy/die) was used. In four studies
of related control groups in the non-comparative studies, and a tendency (Chao et al., 2017; McAvoy et al., 2013; Hayashi et al., 2018; Karube
towards high heterogeneity. et al., 2017b) radiation was delivered using proton or carbon ion ther­
apy, while in eight studies (McAvoy et al., 2014; Ren et al., 2018; Hong
3. Results et al., 2019; Sumodhee et al., 2019; Meijneke et al., 2013; Kilburn et al.,
2014; Binkley et al., 2016; Sumita et al., 2016) various re-irradiation
The literature search yielded 293 citations. After duplicates removal, techniques were used (conventional, stereotactic, proton therapy).
272 articles were evaluated, of these 232 were excluded with reasons Re-irradiation prescription doses were variable across different studies,
(Fig. 1): treatment was not radiotherapy (n = 61) or re-irradiation (23), but also between patients in the same study (see Table 1). Chemotherapy
data available only for technique (n = 12) or radiobiology (n = 16), was given concurrently with re-irradiation in 187 patients (14.6 %). In
other re-irradiation site (n = 76), treatment in pediatric population (n = 27 patients (2.1 %) re-irradiation was delivered after surgery and in 6
4), other exclusion criteria (n = 36). Reviews without original data (n = patients (0.5 %) re-irradiation was given concomitantly with
3), 1 tox reported out of review’s criteria (n = 1), were excluded after a hyperthermia.
second screening. Thirty-six studies met the inclusion criteria. The main
features of the included trials are shown in Table 1. These studies were

3
Table 1

M. Maddalo et al.
Characteristics of the included studies in the meta-analysis.
First Author Study type: Accrual Median FU Previous RT Previous total dose Time elapsed Re-RT Re-RT total dose Gy CHT Relevant toxicity (N◦ of pts)
Journal Retrospective (R) period (months) technique Gy (months) technique median (range) (%)
Year Prospective (P) median (range) between RT
N◦ Patients/N◦ and re-RT
Treatments* median (range)

Okamoto, Int. J. R 1979– 2000 15 2D 60 (30− 80) 23 (5–87) 2D 3D 50 (10− 70) 15 7 G3 radiation pneumonitis
Radiation 34/34
Oncology Biol.
Phys. 2002
Wu, Int. J. Radiation P 1999− 2001 15 3D 66 (30− 78) 13 (6− 42) 3D 51 (46− 60) 0 2 G3 pulmonary fibrosis
Oncology Biol. 23/23
Phys. 2003
Tada, Int. J. Clin. R 1992− 2002 unknown unknown (50− 69.6) 16 (5–60) unknown 50 1 1 G3 radiation pneumonitis
Oncol. 2005 19/19
Ebara, Anticancer R 1990− 2004 6,5 2D (50− 70) 13 (6− 47) 2D (30− 60) 36 3 G3 pulmonary toxicity
Research 2007 44/44
Cetingoz, Journal of R 1992− 2006 135 2D (28,8− 67,2) 8 (4− 44) 2D (5− 30) 3 1 G3 acute esophagitis
BUON 2009 38/38
Kelly, Int. J. R 2004− 2008 15 3D IMRT 61.5 (30–79.2) 22 (0–92) SBRT 50 0 7 G3 pneumonitis 3 G3 esophagitis 2 G3 chest
Radiation 36/36 wall ulcers 2 G3 cough
Oncology Biol.
Phys. 2010
Peulen, R 1994− 2004 12 SBRT various (see original 14 (5–54) SBRT various (see original 0 3 G3 Cough 4 G3 Dyspnea 1 G3 Pneumonitis 1 G3
Radiotherapy and 29/32 article) article) Stenosis of airway 3 G5 Bleeding 1 G3 Pleural
Oncology 2011 effusion G3 Dermatitis G3 Pain 2 G4 Other
(fistula, VCS fibrosis)
Ohguri,Lung Cancer R 1992− 2009 11 unknown 70 (30− 85) 8 (1− 28) unknown 50 (29− 70) 46 1 neuropathy brachial plexus
2012 33/33
4

Trakul, Journal of R 2004− 2010 15 3D IMRT BED 87.5 16 (5–80) SBRT various (see original 0 9 Pneumonitis > G2
Thoracic 15/17 SBRT (60–112.5) article)
Oncology 2012
Liu, Int. J. Radiation R 2004− 2010 16 CFRT 63 (30− 79.2) 21 (0− 106) SBRT 50 0 14 G3 radiation pneumonitis 1 G5 radiation
Oncology Biol. 72/72 pneumonitis
Phys. 2012
Reyngold, Radiation R 2004− 2011 126 2D 3D IMRT 61 (30–80) 37 (1− 180) SBRT various (see original 0 G3 pulmonary toxicity 2 G3 chest wall pain 1 G4
Oncology 2013 39/39 article) skin toxicity
Yoshitake, R 2004− 2011 126 SBRT 48 12 (6− 36) unknown (60− 70) 24 All G1-G2
Anticancer 17/17

Critical Reviews in Oncology / Hematology 167 (2021) 103500


Research 2013
McAvoy, R 2006− 2011 11 unknown 63 (40–74) 36 (2–376) Proton RBE 66 (16.4− 75) 24 3 G3 esophageal toxicity 7 G3 pulmonary toxicity
Radiotherapy and 33/33 1 G3 cardiac toxicity 1 G4 esophagitis (fistula) 2
Oncology 2013 G4 pulmonary toxicity
Meijneke, R 2005− 2012 12 SBRT CFRT various (see original 17 (2–33) SBRT CFRT various (see original 0 All G1-G2
Radiotherapy and 20/20 article) article)
Oncology 2013
McAvoy, Int. J. R 2006− 2013 6,5 2D 3D IMRT EQD2 70 (33− 276) 17 (0− 376) Proton EQD2 60.5 33 3 early discontinuation
Radiation 102/102 SBRT (25.2− 155) 7 > G3 esophageal toxicity 10 > G3 pulmonary
Oncology Biol. Proton toxicity 1 G3 cardiac toxicity
Phys. 2014
Griffioen, Lung R 2004− 2013 193 unknown 59.8 (24–70) 51 (5− 189) IMRT 60 (39–66) 8 1 G3 esophagitis 1 G3 dermatitis 3 G5 bleeding
Cancer 2014 24/24
Kruser, Am. J. Clin. R 2004− 2010 265 3D IMRT NSCLC 57 19 2D 3D IMRT NSCLC 30 (12–60) 0 3 G3 acute pulmonary toxicity 1 G4 late
Oncol. 2014 48/48 (30–80.5) SCLC 45 SCLC 375 (25− 45) pulmonary toxicity
(12–54)
Trovò, Int. J. R unkown 18 3D IMRT (50− 60) 18 (1− 60) SBRT 30 0 4 G3 radiation pneumonitis 1 G5 radiation
Radiation 17/17 pneumonitis 1 G5 hemoptysis
(continued on next page)
Table 1 (continued )

M. Maddalo et al.
First Author Study type: Accrual Median FU Previous RT Previous total dose Time elapsed Re-RT Re-RT total dose Gy CHT Relevant toxicity (N◦ of pts)
Journal Retrospective (R) period (months) technique Gy (months) technique median (range) (%)
Year Prospective (P) median (range) between RT
N◦ Patients/N◦ and re-RT
Treatments* median (range)

Oncology Biol.
Phys. 2014
Hearn, Int. J. R 2004− 2012 138 SBRT various (see original 15 (10− 26) SBRT (50− 60) 0 All G1-G2
Radiation 10/10 article)
Oncology Biol.
Phys. 2014
Kilburn, R 2001− 2012 17 3D SBRT CFRT 66 (45–80.5) 18 (6− 61) 3D IMRT various (see original 0 1 G3 dyspnea (oxygen) 1 G5 aorta-esophageal
Radiotherapy and 33/33 SBRT 50 (22.5–60) SBRT article) fistula
Oncology 2014
Patel, J. Radiat. R 2008- 2011 unknown CFRT SBRT 612 8 (3− 26) SBRT 30 (15–50) 0 All G1-G2
Oncol. 2015 26/29
Binkley, Int. J. R 2008- 2014 17 CFRT HFRT SBRT 40 (25− 54) 16 (1− 71) SBRT HFRT SBRT 40 (25− 54) 26 3 G>2 acute esophagitis 2 G>2 acute chest wall
Radiation 38/38 SBRT HFRT (49,5− 51 Gy) CFRT HFRT 60(60− 64,5) tox 2 G > 2 late rib fracture 2 recurrent laryngeal
Oncology Biol. CFRT 66 (45− 71.6 hypoFRT CFRT 66 (60− 72) neuropathy 1 late brachial plexopathy 1 late
Phys. 2015 Gy) hypoFRT 60 (50− 60) Horner’s syndrome
Gill, Frontiers in R unkown 252 post-op BRT unkown 46 (11− 114) SBRT various (see original 0 1 G3 esophageal stricture
Oncology 2015 13/13 article)
Owen, Frontiers in R 2006- 2012 212 CFRT EQD2 60 (42.3− 70) 18 (2− 113) SBRT various (see original 17 All G1-G2
Oncology 2015 18/27 article)
Sumita, Radiation R 2007- 2014 221 CFRT NSCLC: 60 SCLC: 27 (11− 92) CFRT NSCLC: 60 SCLC: 50 5 1 acute G3 pneumonitis
Oncology 2016 21/21 Proton (1) 43.1 Proton
Ceylan, Oncol. Res. R 2005- 2015 9 2D 3D IMRT 54 (40–57) 14 (4− 56) SBRT various (see original 57 All G1-G2
Treat. 2017 28/28 VMAT SBRT article)
Chao, Journal of P, multi- 2010- 2015 7,8 unknown unknown 19 (4− 151) Proton 66.6 (30–74) 68 3 G4 neutropenia 1 G4 pericardial effusion 1 G5
5

Thoracic institutional bronchial hemorrhage 1 G5 neutropenic sepsis 1


Oncology 2017 57/57 G5 Anorexia 1 G5 Pneumonitis 1 5 Hypoxic
respiratory failure/pleural effusion 1 G5
Tracheoesophageal fistula
Karube, R 2007− 2014 29 carbon-ion various (see original 20 (8− 99) carbon-ion 66 0 All G1-G2
Radiotherapy and 29/29 article)
Oncology 2017
Caivano, Radiation R 2011− 2016 13 SBRT BED 97.2 Gy 18 (6− 66) SBRT BED 100.6 0 1 G3 acute dyspnea 1 G3 late dyspnea 1 G3 late
Oncology 2018 22/27 (40− 120) (48− 151.2) chest pain 2 G3 pulmonary fibrosis
Ogawa Radiation R 2004− 2017 26 SBRT (48− 52) 18 (4− 80) SBRT (48− 60) All G1-G2
Oncology 2018 31/31

Critical Reviews in Oncology / Hematology 167 (2021) 103500


Wegner, Lung R unkown 465 BRT 120 41 (2− 85) SBRT 50 0 All G1-G2
Cancer 10/10
Management
2018
Hayashi, Cancer R 2006− 2016 18 carbon-ion RBE (28− 68,4) 17 (6− 139) carbon-ion (48− 72) 0 All G1-G2
Science 95/95 stage I (68− 76)
stage IIA-IIIB
Ren, Radiation R 2010− 2017 9 3D IMRT 56 (30− 120) 16 (2− 96) 3D SBRT 3D (40− 60) SBRT 28 18 G3 Pneumonitis 1 G4 Pneumonitis
Oncology 67/67 (40− 50)
Hong, Cancer res. R 2005 174 3D IMRT 60 (45− 66) 15 (4− 56) IMRT 50 (35− 65) 10 All G1-G2
treat. 31/31 Helical Helical
SBRT
Schlampp, Strahler R 2010− 2015 8,2 3D 60 14 (3− 103) IMRT 385 (20− 60) 0 2 G3 and 1 G5 Pneumonitis 1 tracheal fistula
Onkology 62/62
Sumodhee, BMC R 2007− 2015 473 3D 66 (44− 70) 23 (6− 102) SBRT (50− 60) 22 All G1-G2
Cancer 46/46

*Number of patients treated or number of lesion treated.


3D: three dimensional conformal radiotherapy; IMRT: intensity modulated radiation therapy; SBRT: Sterotactic body radiotherapy; HFRT: hyperfractionated radiotherapy; CFRT: conformal radiotherapy; hypoFRT:
hypofractionated radiotherapy.
M. Maddalo et al. Critical Reviews in Oncology / Hematology 167 (2021) 103500

Fig. 4. Pooled objective 2 y overall survival in re-irradiated patients for


included studies.
Fig. 2. Pooled rate of ≥G3 toxicity in re-irradiated patients for included
Abbreviations: Q: cochran’s Q-statistic, I2: I2-statistic (both are used to evaluate
studies.
heterogeneity across studies), ES: Effect Size.
Abbreviations: Q: cochran’s Q-statistic, I2: I2-statistic (both are used to evaluate
heterogeneity across studies), ES: Effect Size.

Fig. 5. Pooled objective 1 y local control in re-irradiated patients for included


studies.
Abbreviations: Q: cochran’s Q-statistic, I2: I2-statistic (both are used to evaluate
heterogeneity across studies), ES: Effect Size.

Fig. 3. Pooled objective 1 y overall survival in re-irradiated patients for maximum point doses or as cumulative doses from the previous irradi­
included studies. ation and the re-irradiation. Data on acute toxicity were analyzed in all
Abbreviations: Q: cochran’s Q-statistic, I2: I2-statistic (both are used to evaluate studies: ≥ G3 acute toxicity was observed in 135 patients (10.5 %).
heterogeneity across studies), ES: Effect Size. There was no difference in ≥ G3 acute toxicity rate with respect to
different radiation techniques, cumulative dose and re-irradiation total
3.2. Toxicity and Radiation tolerance of organ at risk dose and fractionation. Fourteen patients (1.09 %) died because of likely
treatment related acute toxicities such as neutropenia, fatal hemor­
All studies reported the organs at risk (OARs) constraints either as rhages and pneumonia. Globally, the pooled effect size for toxicity ≥ G3

6
M. Maddalo et al. Critical Reviews in Oncology / Hematology 167 (2021) 103500

2007; Okamoto et al., 2002; Tada et al., 2005; Kruser et al., 2014;
Ohguri et al., 2012; Cetingoz et al., 2009). The pooled analysis showed
that 79.5 % of patients who experienced symptoms obtained relief (95 %
CI: 72–87.8 %) (Fig. 7).

4. Discussion

The advent of more efficient local interventions and increasingly


multimodal therapies has led to an improvement in lung cancer treat­
ment outcome. Therefore, an increasing number of patients may need
re-treatment for recurrent or metastatic disease and re-irradiation is
among the possible approaches. Thoracic re-irradiation is one of the
most significant challenges for modern radiotherapy, as the control of
symptoms, the possibility for cure and the risk of side effects has to be
carefully balanced. The major issue in this context is the difficulty to
predict the probability for severe toxicity for a given patient due to the
lack of knowledge of normal tissue complication probabilities (NTCP)
and data on organ at risk (OARs) tolerance in the heterogeneous cohort
of patients that undergo re-irradiation. Currently, dosimetric constraints
are only available for patients that underwent SBRT, as first treatment
approach (Benedict et al., 2010; Grimm et al., 2011; Milano et al., 2008).
Fig. 6. Pooled objective 2 y local control in re-irradiated patients for included This review attempts to extract useful information regarding severe
studies. toxicity after thoracic re- irradiation from available clinical data.
Abbreviations: Q: cochran’s Q-statistic, I2: I2-statistic (both are used to evaluate First, some issues should be considered when thoracic toxicity is
heterogeneity across studies), ES: Effect Size.
reported. The majority of the studies included in the analysis scored
toxicity according to Common Terminology Criteria for Adverse Event
(CTCAE), but there were 5 series in which the scale used was either the
RTOG/EORTC (Wu et al., 2003; Ebara et al., 2007; Okamoto et al., 2002;
Cetingoz et al., 2009) or the WHO (Tada et al., 2005) and 2 studies in
which the scale was not specified (Wegner et al., 2018; Trakul et al.,
2012). The use of different toxicity scores has an impact on the
comparability of toxicity data and limits the accuracy of results’
comparison.
As a second issue, thoracic re-irradiation has so far mainly performed
with palliative intent, which results in a median follow up of only 15
Fig. 7. Pooled objective symptoms relief in re-irradiated patients for included
studies. months (range 6.5–47.3 months) and as a consequence in a paucity late
Abbreviations: Q: cochran’s Q-statistic, I2: I2-statistic (both are used to evaluate toxicity data. This limits the extrapolation of existing data to patient
heterogeneity across studies), ES: Effect Size. cohorts where re-irradiation is performed in a fully or quasi-curative
intent.
was 9.1 % (95 %CI: 6.3–13.1 %) with high heterogeneity (I2 = 87.063, P Only few studies in this review explicitly reported symptom relief,
< 0.001) (Fig. 2). No difference in heterogeneity was evident analyzing but an albeit ill defined “benefit” was seen globally in 795% of cases. In a
studies using the different modalities of re-irradiation. meta-analysis (Drodge et al., 2014) published in 2014 collecting data of
older studies with a smaller proportion of patients treated with
radical-intent re-irradiation the authors reported a similar overall
3.3. Outcomes average symptom improvement of 69.2 %. Thus, even if the description
of symptoms varied across different studies the benefit of re-irradiation
Thirty-three studies reported data on 1-yr OS and the pooled 1-yr OS in the palliative setting seems well documented.
rate was 60.9 % (95 %CI: 54.8–67.6 %) with high heterogeneity (I 2 = Over the recent past, a shift towards SBRT higher doses per fraction
85.5 %, P = 0.001) (Fig. 3). No difference in heterogeneity was observed accentuated the uncertainties on late toxicities prediction. In a recent
analyzing studies using the different modalities of re-irradiation. Two- meta-analysis (Viani et al., 2020) on effectiveness and safety of SBRT
year OS was reported by 33 studies. The pooled effect size for 2-yr OS re-irradiation the authors found a correlation between the cumulative
was 43.9 % (95 %CI: 38.7–49.9 %) with moderate heterogeneity (I 2 = EQD2 dose and the development of any grade 3 toxicity. The risk of
74.7 %, P = 0.001) (Fig. 4). Analyzing only studies using stereotactic toxicity was significantly higher (3% vs. 15 %, P = 0.03) when the EQD2
radiotherapy and proton/carbon ion, the pooled result was respectively was stratified into EQD2 ≤ 145 Gy or EQD2 > 145 Gy.
50.8 % (95 % CI: 42–61.6 %) with a moderate heterogeneity (I2 = 65.2 In a further attempt to provide the appropriate “flavor” of the results
%, P = 0.001) and 50.4 % (95 % CI: 38.8–65.5 %) with a moderate reported as quantitative global data based on the meta-analysis in the
heterogeneity (I2 = 60.6 %, P = 0.055). No difference in heterogeneity results section, we here discuss these data adding more specific infor­
was observed analyzing studies using the other different modalities of mation and conclusions on three peculiar topics: severe acute toxicity,
re-irradiation. particle therapy and the combination of re-irradiation with
Locoregional progression free survival at 1 and 2 year was reported chemotherapy.
by 27 and 26 studies, respectively. The pooled 1-yr locoregional pro­
gression free survival rate was 65.1 % (95 % CI: 59.7–71.1 %) with high 4.1. Severe acute toxicity
heterogeneity (I 2 = 83.7 %, P = 0.001) (Fig. 5). Instead, the pooled 2-yr
locoregional progression free survival rate was 47.2 % (95 % CI: A review published in 2011 by Jeremic (Jeremic et al., 2001) on
40.4–55.1 %) with high heterogeneity (I 2 = 83.7 %, P = 0.001) (Fig. 6). chest re-irradiation reported low rates of severe acute toxicity, mainly
Only six studies evaluated improvements in symptoms (Ebara et al., esophagitis (4–6 %) and pneumonitis (5–21 %), but the authors specified

7
M. Maddalo et al. Critical Reviews in Oncology / Hematology 167 (2021) 103500

that many of the studies included that covered a pre-1990 therapy were with treatment for out-of-field relapses (28 % of patients). The authors
particularly limited by the absence of formal toxicity scoring systems. hypothesized a protective role of inflammation induced by first course of
In the present review, data about the safety of re-irradiation with radiotherapy to explain the high rate of pulmonary toxicity out of field.
conventional techniques were collected from different studies (see However, looking more closely at the available data, the cumulative
Table 1). However, many of these works collected data from re- G2-G3 rate of radiation pneumonitis was similar for patients treated for
irradiation addressed both with symptomatic and “radical” intent. in-field (5 of the 11 patients, 45 %) and out-of-field relapses (13 of the 25
Cetingoz et al. (2009) reported the outcome of 38 patients. Of the 26 patients, 52 %). In the same paper, the Authors reported that chest wall
patients who showed toxicity, only 1 had G3 esophagitis (3%). Ac­ pain was associated with in-field recurrence.
cording to Ebara et al. (2007) in 44 patients, 3 G3 pulmonary toxicity The combined approach of radiotherapy and Hyperthermia (Ohguri
were observed (7%), and 14 % of patients experienced a pulmonary et al., 2012) or brachytherapy, as part of initial course of treatment, does
toxicity of any grade. Tada et al. (2005) in a series of 19 patients not seem to further enhance toxicity (Gill et al., 2015).
mentioned only 1 G3 pneumonitis in the oldest patient of the series (84 Schröder et al. (Schroder et al., 2020) in a retrospective analysis (not
year’s old), and a 20 % of cumulative G2-G3 lung toxicity. A study from included in the present systematic review due to the recent publication)
Wu et coll. (Wu et al., 2003), showed similar rates (< 10 %) of severe attempted to estimate the tolerance of thoracic organs-at-risk (OAR) to
lung toxicity with 2 G3 pulmonary fibrosis. Only one study by Okamoto re-irradiation by calculating accumulated dose distributions using
et al. (2002) reported a higher rate of G3 pneumonitis in 7/34 (21 %). In deformable image registration and dose conversion to EQD2. In their
our pooled analysis acute toxicity ≥ G3 was observed in 135 patients series of 40 patients, even if most OARs received cumulative EQD2 doses
(10.5 %). There was no difference in ≥ G3 acute toxicity rate with between 70 and 100 Gy and maximum doses to individual OARs above
respect to different radiation techniques. 100 Gy, only one patient suffered from a high-grade complication.
In 2015 De Bari et al. (2015) performed a review on the feasibility of
SBRT after high dose radiotherapy for lung cancer. They reported a ≥ G3 4.2. Particle therapy
pulmonary toxicity of 3–28 %. This wide range of toxicity reflects the
heterogeneity of studies due to the intent of treatment, schedules, and Proton beam therapy (PBT) has emerged as a potentially promising
patients’ characteristics. lung re-treatment modality, given its reduced integral dose and thus
In our analysis, severe toxicity such as pneumonitis/pulmonary ideally improved normal tissue sparing. Nevertheless, few studies have
fibrosis (RP/PF), esophagitis, chest wall pain/rib fracture, radiation analyzed the role of PBT in thorax re-irradiation.
dermatitis and severe/fatal G4-G5 adverse events (AE) were reported by Chao et al. (2017) in a prospective multi-institutional trial reported
different studies. acute and late > G3 toxicity in a high proportion (42 %) of patients. The
Different predictive factors for severe toxicity were identified. One of paper also reported a consistent rate of G4-G5 toxicity (four cases of
the most important factors seems to be the location of the treatment site. grade 4 toxicity and 6 cases of grade 5 toxicity were observed, see
A Swedish retrospective analysis (Peulen et al., 2011) of 32 patients that Table 1). This higher rate of severe toxicity in comparison with all the
underwent high-dose SBRT, reported that 5 patients died of massive other series of the present selection could be explained by the pro­
hemorrhage, but the authors didn’t found a correlation with the spective nature of the study and the consequent more accurate adverse
maximum dose to vessel wall or the dose to the entire circumference of event report. The authors observed a correlation of the proximity of
large mediastinal vessels. In the same report 8 patients experienced G3 re-irradiated lesions to the bronchial tree, the mean doses to heart and
lung toxicity and the authors concluded that a larger clinical target esophagus obtained from the composite dose to these organ and con­
volumes (CTV) and central tumour localization were associated with an current chemotherapy (p = 0.003) with higher toxicity and were
increased rate of severe toxicity. The correlation between the central therefore advocating a cautious selection of patients in relation to the
position of the tumor and re-irradiation toxicity has also been docu­ predictive factors identified. Notably, authors had selected patients
mented by Schlapp and Sumodhee (Schlampp et al., 2019; Sumodhee based on treatment volume, those with high volume >250 cm3 were
et al., 2019). Sumodhee et al. reported that hilar tumors and tumor size considered untreatable.
> 33 mm correlated with G5 hemoptysis and alveolitis. According to McAvoy et al. (2014, 2013) registered a G3 or higher esophageal and
Binkley et al. (2016), re-irradiation of non-peripheral lesions was asso­ pulmonary toxicity in 9% and 21 % pts respectively. Toxicity was related
ciated with increased toxicity, but no patient experienced a severity > to the high cumulative doses to these organs (significant Dmax (p =
G3. 0.001) and larger esophageal V60 (p < 0.001)), but not with concomi­
In 2012, Liu et al. (2012) published the results on re-irradiation with tant chemotherapy. Peripheral lesions had significantly lower cardiac
SBRT in 72 pts initially treated with conventional fractionation (median toxicity rates (p = 0.02) than central ones.
dose of 63 Gy). Fourteen and one patients experienced G3 and G5 ra­ Overall, the crude rates of toxicity reported for PBT are similar to
diation pneumonitis respectively. The dosimetric analysis of composite what has been reported for conventional radiation therapy modalities
plans showed that V10 and mean lung dose (MLD) of the previous plan (Wu et al., 2003; Okamoto et al., 2002; Cetingoz et al., 2009), keeping in
and the V10-V40 and MLD were related to lung toxicity. Also Hong et al. mind, that the latter was usually performed with a more palliative
(2019) reported a statistically significant difference in the cumulative intent, thus with lower doses and hypothetical smaller irradiated
MLD for patients with and without acute pulmonary symptoms (p = volumes.
0.004). Only two studies on re-irradiation with Carbon Ion Radiotherapy
Interestingly, pulmonary severe toxicity could also be related to the (CIRT) had been included in our analysis, both showing the feasibility of
dose received by other OARs. In a retrospective study on 17 pts re- the treatment with only few cases of > G3 toxicity (Hayashi et al., 2018;
treated with relatively low doses (30 Gy in 5–6 fractions), Trovò et al. Karube et al., 2017b).
(Trovo et al., 2014) identified that heart maximum dose, D5 mean of 10 Hayashi et coll. (Hayashi et al., 2018) reported the data of 95 pa­
Gy and D10 mean of 3 Gy were correlated with higher risk of > G3 tients treated for primary or metastatic lung tumors: one patient
pneumonitis and hemoptysis. A retrospective analysis in definitive developed Pneumonitis G4, 1 reported chest pain G3, and 1 developed a
treatment of lung malignancies, established the role of heart dosimetric bronchopleural fistula G5. This patient had received long-term Bev­
parameters in predicting the post-radiotherapy pneumonitis, although acizumab before and after CIRT and the fistula was detected in the re­
the mechanism of this effect remains unclear (Huang et al., 2011). gion exposed to >70.0 Gy of relative biological effectiveness (RBE).
In contrast with what was reported thus far, a group from the MD Based on their experience, the authors suggested the criteria for selec­
Anderson Cancer Center published the results on 36 patients (Kelly tion of appropriate patients for CIRT: solitary recurrent, metastatic or
et al., 2010) and showed that the G3 lung toxicity was only associated secondary tumors in the lung or mediastinum, not suitable for surgery,

8
M. Maddalo et al. Critical Reviews in Oncology / Hematology 167 (2021) 103500

no previously treatment with Bevacizumab, with at least 24 months The high rate of symptoms relief could still justify the use of re-
between primary treatments and re-irradiation, CTV volume for new irradiation with lower doses in a palliative scenario. However, dose
irradiation <80.0 mL. schedules, both for palliative and curative intent are yet to be
Even if our analysis does not support the use of one technique over investigated.
the others in terms of incidence of adverse events, a comparison between Importantly the conclusions regarding the comparison of the
CIRT and photon or proton therapy is challenging because differences in different treatment modalities, given that different NTCP model might
physical dose deposition but more importantly because of significant have to be applied. To offer the best therapeutic option to patients
uncertainties in the precise estimation of the α/β ratio of high-linear referred to re-irradiation in loco-regional recurrence or metastatic dis­
energy transfer (LET) radiation (CIRT), and, thus, RBE. ease, great attention should be paid to comorbidities and age to more
precisely predict the benefit of thoracic re-irradiation over systemic
4.3. Chemotherapy therapy or best supportive care.
Besides, unknown factors such as genetic susceptibility could also
The impact of concomitant chemotherapy on the development of affect OAR tolerance to radiotherapy (Tang et al., 2020; Xu et al., 2019).
serious toxicity in the re-irradiation setting is not clear as the results are To achieve reliable results from retrospective series an accurate se­
heterogeneous. In our analysis, concurrent chemotherapy was admin­ lection of patients and a proper indication about intent, dose and tech­
istered in 11/36 studies (Chao et al., 2017; McAvoy et al., 2014; Ebara nique are warranted. Prospective clinical trials evaluating molecular
et al., 2007; Ceylan et al., 2017; Okamoto et al., 2002; Tada et al., 2005; predictors to assess the real advantages of re-irradiation in terms of
McAvoy et al., 2013; Binkley et al., 2016; Ohguri et al., 2012; Cetingoz disease control and toxicity with different techniques and in different
et al., 2009; Sumita et al., 2016). populations, above all in oligometastic disease, are needed to provide
Only in the study of Chao et al. (2017), there seem to be a correlation more accurate selection criteria for thoracic re-irradiation. Defining a
with an increase in severe toxicity with concomitant chemotherapy, standard shared nomenclature to be used in re-irradiation prospective
while, in the reports by McAvoy et al. (2014, 2013) and Binkley et al. and retrospective data collection would be welcomed.
(2016), this was not confirmed.
Interestingly, Yang et al. (2020) in a recent paper not included in the Funding
present selection reported a significant association between subsequent
chemotherapy following thoracic re-irradiation (p = 0.009) with lethal This research did not receive any specific grant from funding
lung events, and a trend in favor of taxane-based regimens versus those agencies in the public, commercial, or not-for-profit sectors.
not receiving taxane-based regimens.
In the era of a preferentially integrated and multidisciplinary treat­ Declaration of Competing Interest
ment approach, even though the goal of re-irradiation is still mostly
palliation, the combined approach could provide durable local control The authors certify that they have NO affiliations with or involve­
and potentially improve progression-free and overall survival in an ment in any organization or entity with any financial interest (such as
increasing number of patients. honoraria; educational grants; participation in speakers’ bureaus;
Recently, in clinical practice, despite a lack of unequivocal preclin­ membership, employment, consultancies, stock ownership, or other
ical and clinical experimental data, patients with recurrent or metastatic equity interest; and expert testimony or patent- licensing arrangements),
NSCLC and SCLC have frequently been referred to targeted therapy and or non-financial interest (such as personal or professional relationships,
immunotherapy in association with re-irradiation in order to gain affiliations, knowledge or beliefs) in the subject matter or materials
greater efficacy and increase survival (Evans et al., 2017; Arcangeli discussed in this manuscript.
et al., 2019a; Arcangeli et al., 2019b). Although existing data on efficacy
are encouraging, long-term result, particularly acute and late toxicity, Acknowledgments
are not yet enough, and combined treatments should be performed with
caution. The Authors thank the Scientific Committee and Board of the AIRO
for the critical revision and final approval of the paper.
5. Conclusion
References
This is a comprehensive and systematic review on thoracic re-
irradiation. A meta-analysis technique has been used to address sur­ Abusaris, H., Hoogeman, M., Nuyttens, J.J., 2012. Re-irradiation: outcome, cumulative
vival and toxicity outcomes, but its clinical and scientific validity is dose and toxicity in patients retreated with stereotactic radiotherapy in the
abdominal or pelvic region. Technol. Cancer Res. Treat. 11, 591–597.
limited by the heterogeneity of the included studies. Different treatment Arcangeli, S., Jereczek-Fossa, B.A., Alongi, F., Aristei, C., Becherini, C., Belgioia, L., et al.,
paradigms (palliative versus curative intent, in field recurrence versus 2019a. Combination of novel systemic agents and radiotherapy for solid tumors -
edge of field recurrence, local relapse versus separate primary versus part i: an airo (italian association of radiotherapy and clinical oncology) overview
focused on treatment efficacy. Crit. Rev. Oncol. Hematol. 134, 87–103.
oligometastatic lung metastases) makes it even harder to draw reliable Arcangeli, S., Jereczek-Fossa, B.A., Alongi, F., Aristei, C., Becherini, C., Belgioia, L., et al.,
conclusions. 2019b. Combination of novel systemic agents and radiotherapy for solid tumors -
The inconsistency of the data is also due to the retrospective nature part ii: an airo (italian association of radiotherapy and clinical oncology) overview
focused on treatment toxicity. Crit. Rev. Oncol. Hematol. 134, 104–119.
of the majority of the studies analyzed - many of these including a small Auperin, A., Le Pechoux, C., Pignon, J.P., Koning, C., Jeremic, B., Clamon, G., et al.,
number of patients - the possible role of chemotherapy and the hetero­ 2006. Concomitant radio-chemotherapy based on platin compounds in patients with
geneity of radiation therapy modalities, volumes treated, and locally advanced non-small cell lung cancer (nsclc): a meta-analysis of individual
data from 1764 patients. Ann. Oncol. 17, 473–483.
scheduling. Benedict, S.H., Yenice, K.M., Followill, D., Galvin, J.M., Hinson, W., Kavanagh, B., et al.,
This analysis shows that the main problem of the existing data is the 2010. Stereotactic body radiation therapy: the report of aapm task group 101. Med.
frequent lack of a sufficient description of the treatment’s intent, which Phys. 37, 4078–4101.
Binkley, M.S., Hiniker, S.M., Chaudhuri, A., Maxim, P.G., Diehn, M., Loo Jr., B.W., et al.,
makes balancing risk and benefit of re-irradiation based on published
2016. Dosimetric factors and toxicity in highly conformal thoracic reirradiation. Int.
data even more difficult. J. Radiat. Oncol. Biol. Phys. 94, 808–815.
Thus the 10–25 % rate of acute severe pulmonary toxicity could be Caivano, D., Valeriani, M., De Matteis, S., Bonome, P., Russo, I., De Sanctis, V., et al.,
reasonable in the curative setting of re-irradiation both with photon RT 2018. Re-irradiation in lung disease by sbrt: a retrospective, single institutional
study. Radiat. Oncol. 13, 87.
techniques and highly conformal techniques such as IMRT, SBRT, PBT, Cetingoz, R., Arican-Alicikus, Z., Nur-Demiral, A., Durmak-Isman, B., Bakis-Altas, B.,
or CIRT. Kinay, M., 2009. Is re-irradiation effective in symptomatic local recurrence of non

9
M. Maddalo et al. Critical Reviews in Oncology / Hematology 167 (2021) 103500

small cell lung cancer patients? A single institution experience and review of the O’Rourke, N., Roque, I.F.M., Farre Bernado, N., Macbeth, F., 2010. Concurrent
literature. J. B.U.O.N. 14, 33–40. chemoradiotherapy in non-small cell lung cancer. Cochrane Database Syst. Rev.,
Ceylan, C., Hamaci, A., Ayata, H., Berberoglu, K., Kilic, A., Guden, M., et al., 2017. Re- CD002140
irradiation of locoregional nsclc recurrence using robotic stereotactic body Ogawa, Y., Shibamoto, Y., Hashizume, C., Kondo, T., Iwata, H., Tomita, N., et al., 2018.
radiotherapy. Oncol. Res. Treat. 40, 207–214. Repeat stereotactic body radiotherapy (sbrt) for local recurrence of non-small cell
Chao, H.H., Berman, A.T., Simone 2nd, C.B., Ciunci, C., Gabriel, P., Lin, H., et al., 2017. lung cancer and lung metastasis after first sbrt. Radiat. Oncol. 13, 136.
Multi-institutional prospective study of reirradiation with proton beam radiotherapy Ohguri, T., Imada, H., Yahara, K., Moon, S.D., Yamaguchi, S., Yatera, K., et al., 2012. Re-
for locoregionally recurrent non-small cell lung cancer. J. Thorac. Oncol. 12, irradiation plus regional hyperthermia for recurrent non-small cell lung cancer: a
281–292. potential modality for inducing long-term survival in selected patients. Lung Cancer
De Bari, B., Filippi, A.R., Mazzola, R., Bonomo, P., Trovo, M., Livi, L., et al., 2015. 77, 140–145.
Available evidence on re-irradiation with stereotactic ablative radiotherapy Okamoto, Y., Murakami, M., Yoden, E., Sasaki, R., Okuno, Y., Nakajima, T., et al., 2002.
following high-dose previous thoracic radiotherapy for lung malignancies. Cancer Reirradiation for locally recurrent lung cancer previously treated with radiation
Treat. Rev. 41, 511–518. therapy. Int. J. Radiat. Oncol. Biol. Phys. 52, 390–396.
Drodge, C.S., Ghosh, S., Fairchild, A., 2014. Thoracic reirradiation for lung cancer: a Owen, D., Olivier, K.R., Song, L., Mayo, C.S., Miller, R.C., Nelson, K., et al., 2014. Safety
literature review and practical guide. Ann. Palliat. Med. 3, 75–91. and tolerability of sbrt after high-dose external beam radiation to the lung. Front.
Ebara, T., Tanio, N., Etoh, T., Shichi, I., Honda, A., Nakajima, N., 2007. Palliative re- Oncol. 4, 376.
irradiation for in-field recurrence after definitive radiotherapy in patients with Patel, N.R., Lanciano, R., Sura, K., Yang, J., Lamond, J., Feng, J., et al., 2015. Stereotactic
primary lung cancer. Anticancer Res. 27, 531–534. body radiotherapy for re-irradiation of lung cancer recurrence with lower biological
Evans, T., Ciunci, C., Hertan, L., Gomez, D., 2017. Special topics in immunotherapy and effective doses. J. Radiat. Oncol. 4, 65–70.
radiation therapy: reirradiation and palliation. Transl. Lung Cancer Res. 6, 119–130. Peulen, H., Karlsson, K., Lindberg, K., Tullgren, O., Baumann, P., Lax, I., et al., 2011.
Gill, B.S., Clump, D.A., Burton, S.A., Christie, N.A., Schuchert, M.J., Heron, D.E., 2015. Toxicity after reirradiation of pulmonary tumours with stereotactic body
Salvage stereotactic body radiotherapy for locally recurrent non-small cell lung radiotherapy. Radiother. Oncol. 101, 260–266.
cancer after sublobar resection and i(125) vicryl mesh brachytherapy. Front. Oncol. Ren, C., Ji, T., Liu, T., Dang, J., Li, G., 2018. The risk and predictors for severe radiation
5, 109. pneumonitis in lung cancer patients treated with thoracic reirradiation. Radiat.
Griffioen, G.H., Dahele, M., de Haan, P.F., van de Ven, P.M., Slotman, B.J., Senan, S., Oncol. 13, 69.
2014. High-dose, conventionally fractionated thoracic reirradiation for lung tumors. Reyngold, M., Wu, A.J., McLane, A., Zhang, Z., Hsu, M., Stein, N.F., et al., 2013. Toxicity
Lung Cancer 83, 356–362. and outcomes of thoracic re-irradiation using stereotactic body radiation therapy
Grimm, J., LaCouture, T., Croce, R., Yeo, I., Zhu, Y., Xue, J., 2011. Dose tolerance limits (sbrt). Radiat. Oncol. 8, 99.
and dose volume histogram evaluation for stereotactic body radiotherapy. J. Appl. Schlampp, I., Rieber, J., Adeberg, S., Bozorgmehr, F., Heussel, C.P., Steins, M., et al.,
Clin. Med. Phys. 12, 3368. 2019. Re-irradiation in locally recurrent lung cancer patients. Strahlentherapie und
Hayashi, K., Yamamoto, N., Karube, M., Nakajima, M., Tsuji, H., Ogawa, K., et al., 2018. Onkologie : Organ der Deutschen Rontgengesellschaft … [et al] 195, 725–733.
Feasibility of carbon-ion radiotherapy for re-irradiation of locoregionally recurrent, Schroder, C., Stiefel, I., Tanadini-Lang, S., Pytko, I., Vu, E., Guckenberger, M., et al.,
metastatic, or secondary lung tumors. Cancer Sci. 109, 1562–1569. 2020. Re-irradiation in the thorax - an analysis of efficacy and safety based on
Hearn, J.W., Videtic, G.M., Djemil, T., Stephans, K.L., 2014. Salvage stereotactic body accumulated eqd2 doses. Radiother. Oncol. 152, 56–62.
radiation therapy (sbrt) for local failure after primary lung sbrt. Int. J. Radiat. Oncol. Sumita, K., Harada, H., Asakura, H., Ogawa, H., Onoe, T., Murayama, S., et al., 2016. Re-
Biol. Phys. 90, 402–406. irradiation for locoregionally recurrent tumors of the thorax: a single-institution,
Higgins, J.P., Thompson, S.G., Deeks, J.J., Altman, D.G., 2003. Measuring inconsistency retrospective study. Radiat. Oncol. 11, 104.
in meta-analyses. BMJ 327, 557–560. Sumodhee, S., Bondiau, P.Y., Poudenx, M., Cohen, C., Naghavi, A.O., Padovani, B., et al.,
Hong, J.H., Kim, Y.S., Lee, S.W., Lee, S.J., Kang, J.H., Hong, S.H., et al., 2019. High-dose 2019. Long term efficacy and toxicity after stereotactic ablative reirradiation in
thoracic re-irradiation of lung cancer using highly conformal radiotherapy is locally relapsed stage iii non-small cell lung cancer. BMC Cancer 19, 305.
effective with acceptable toxicity. Cancer Res. Treat. 51, 1156–1166. Tada, T., Fukuda, H., Matsui, K., Hirashima, T., Hosono, M., Takada, Y., et al., 2005.
Huang, E.X., Hope, A.J., Lindsay, P.E., Trovo, M., El Naqa, I., Deasy, J.O., et al., 2011. Non-small-cell lung cancer: reirradiation for loco-regional relapse previously treated
Heart irradiation as a risk factor for radiation pneumonitis. Acta Oncol. 50, 51–60. with radiation therapy. Int. J. Clin. Oncol. 10, 247–250.
Jeremic, B., Shibamoto, Y., Acimovic, L., Nikolic, N., Dagovic, A., Aleksandrovic, J., Tang, Y., Yang, L., Qin, W., Yi, M.X., Liu, B., Yuan, X., 2020. Impact of genetic variant of
et al., 2001. Second cancers occurring in patients with early stage non-small-cell hipk2 on the risk of severe radiation pneumonitis in lung cancer patients treated
lung cancer treated with chest radiation therapy alone. J. Clin. Oncol. 19, with radiation therapy. Radiat. Oncol. 15, 9.
1056–1063. Trakul, N., Harris, J.P., Le, Q.T., Hara, W.Y., Maxim, P.G., Loo Jr., B.W., et al., 2012.
Julious, S.A., Campbell, M.J., 1998. Sample size calculations for paired or matched Stereotactic ablative radiotherapy for reirradiation of locally recurrent lung tumors.
ordinal data. Stat. Med. 17, 1635–1642. J. Thorac. Oncol. 7, 1462–1465.
Karube, M., Yamamoto, N., Shioyama, Y., Saito, J., Matsunobu, A., Okimoto, T., et al., Trovo, M., Minatel, E., Durofil, E., Polesel, J., Avanzo, M., Baresic, T., et al., 2014.
2017a. Carbon-ion radiotherapy for patients with advanced stage non-small-cell lung Stereotactic body radiation therapy for re-irradiation of persistent or recurrent non-
cancer at multicenters. J. Radiat. Res. 58, 761–764. small cell lung cancer. Int. J. Radiat. Oncol. Biol. Phys. 88, 1114–1119.
Karube, M., Yamamoto, N., Tsuji, H., Kanematsu, N., Nakajima, M., Yamashita, H., et al., Turrisi 3rd, A.T., Kim, K., Blum, R., Sause, W.T., Livingston, R.B., Komaki, R., et al.,
2017b. Carbon-ion re-irradiation for recurrences after initial treatment of stage i 1999. Twice-daily compared with once-daily thoracic radiotherapy in limited small-
non-small cell lung cancer with carbon-ion radiotherapy. Radiother. Oncol. 125, cell lung cancer treated concurrently with cisplatin and etoposide. N. Engl. J. Med.
31–35. 340, 265–271.
Kelly, P., Balter, P.A., Rebueno, N., Sharp, H.J., Liao, Z., Komaki, R., et al., 2010. Viani, G.A., Arruda, C.V., De Fendi, L.I., 2020. Effectiveness and safety of reirradiation
Stereotactic body radiation therapy for patients with lung cancer previously treated with stereotactic ablative radiotherapy of lung cancer after a first course of thoracic
with thoracic radiation. Int. J. Radiat. Oncol. Biol. Phys. 78, 1387–1393. radiation: a meta-analysis. Am. J. Clin. Oncol. 43, 575–581.
Kilburn, J.M., Kuremsky, J.G., Blackstock, A.W., Munley, M.T., Kearns, W.T., Hinson, W. Wegner, R.E., Ahmed, N., Hasan, S., Schumacher, L.Y., Colonias, A., 2018. Lung
H., et al., 2014. Thoracic re-irradiation using stereotactic body radiotherapy (sbrt) stereotactic body radiotherapy after past ablative therapy: a single institution case
techniques as first or second course of treatment. Radiother. Oncol. 110, 505–510. series. Lung Cancer Manag. 7, LMT05.
Kruser, T.J., McCabe, B.P., Mehta, M.P., Khuntia, D., Campbell, T.C., Geye, H.M., et al., Wu, K.L., Jiang, G.L., Qian, H., Wang, L.J., Yang, H.J., Fu, X.L., et al., 2003. Three-
2014. Reirradiation for locoregionally recurrent lung cancer: outcomes in small cell dimensional conformal radiotherapy for locoregionally recurrent lung carcinoma
and non-small cell lung carcinoma. Am. J. Clin. Oncol. 37, 70–76. after external beam irradiation: a prospective phase i-ii clinical trial. Int. J. Radiat.
Liu, H., Zhang, X., Vinogradskiy, Y.Y., Swisher, S.G., Komaki, R., Chang, J.Y., 2012. Oncol. Biol. Phys. 57, 1345–1350.
Predicting radiation pneumonitis after stereotactic ablative radiation therapy in Xu, L., Jiang, J., Li, Y., Zhang, L., Li, Z., Xian, J., et al., 2019. Genetic variants of sp-
patients previously treated with conventional thoracic radiation therapy. Int. J. d confer susceptibility to radiation pneumonitis in lung cancer patients undergoing
Radiat. Oncol. Biol. Phys. 84, 1017–1023. thoracic radiation therapy. Cancer Med. 8, 2599–2611.
McAvoy, S.A., Ciura, K.T., Rineer, J.M., Allen, P.K., Liao, Z., Chang, J.Y., et al., 2013. Yang, W.C., Hsu, F.M., Chen, Y.H., Shih, J.Y., Yu, C.J., Lin, Z.Z., et al., 2020. Clinical
Feasibility of proton beam therapy for reirradiation of locoregionally recurrent non- outcomes and toxicity predictors of thoracic re-irradiation for locoregionally
small cell lung cancer. Radiother. Oncol. 109, 38–44. recurrent lung cancer. Clin. Transl. Radiat. Oncol. 22, 76–82.
McAvoy, S., Ciura, K., Wei, C., Rineer, J., Liao, Z., Chang, J.Y., et al., 2014. Definitive Yoshitake, T., Shioyama, Y., Nakamura, K., Sasaki, T., Ohga, S., Shinoto, M., et al., 2013.
reirradiation for locoregionally recurrent non-small cell lung cancer with proton Definitive fractionated re-irradiation for local recurrence following stereotactic body
beam therapy or intensity modulated radiation therapy: predictors of high-grade radiotherapy for primary lung cancer. Anticancer Res. 33, 5649–5653.
toxicity and survival outcomes. Int. J. Radiat. Oncol. Biol. Phys. 90, 819–827.
Meijneke, T.R., Petit, S.F., Wentzler, D., Hoogeman, M., Nuyttens, J.J., 2013.
Marta Maddalo is a full time radiation oncologist at the university hospital ASST Spedali
Reirradiation and stereotactic radiotherapy for tumors in the lung: dose summation
Civili di Brescia (Italy). She specialized “cum laude” in May 2015 with a dissertation
and toxicity. Radiother. Oncol. 107, 423–427.
entitled “A Patient-Reported Outcome Measure to screen for Symptom in the Head and
Milano, M.T., Constine, L.S., Okunieff, P., 2008. Normal tissue toxicity after small field
Neck Cancer Population: Translation and Pilot Study on Testing Feasibility and Utility of
hypofractionated stereotactic body radiation. Radiat. Oncol. 3, 36.
the Vanderbilt Head and Neck Symptom Survey (VHNSS)”. The thesis followed a period
Moher, D., Liberati, A., Tetzlaff, J., Altman, D.G., Group P, 2009. Preferred reporting
abroad as a visiting doctor at the Medical Oncology Department of the Vanderbilt Ingram
items for systematic reviews and meta-analyses: the prisma statement. J. Clin.
Cancer Center in Nashville, Tennessee. She gained particular interest in treating cancer of
Epidemiol. 62, 1006–1012.
the head and neck, lung, breast and anal canal, and collaborated as investigator on na­
tional prospective protocols and retrospective data series collections.

10
M. Maddalo et al. Critical Reviews in Oncology / Hematology 167 (2021) 103500

Elisa D’Angelo is a full-time radiation oncologist at University Hospital of Modena. She’s irradiation. Active researcher in the field of radiation oncology, author of several peer
involved in about ten national and international clinical controlled trials as principal reviewed papers. Phd candidate. Reviewer for numerous journals in the field of radiation
investigator or sub-investigator, and author of several peer-reviewed papers. She is a oncology.
member and counsellor of Italian and European most important oncological societies, and
reviewer for journals in the oncological field. Major area of interest: lung cancer, head and
Gianluca Costantino, August 2012, Master’s Degree in Medicine and Surgery at the
neck cancer, radiogenomics, immune-oncology.
University of Turin 2016–2020, Residency training in Radiation Oncology at the Univer­
sity of Brescia; graduated magna cum laude. January 2021, MD Radiation Oncologist at
Francesco Fiorica, MD, PhD; Head of Department of Radiation Oncology and Nuclear Radiation Oncology Department ASST Valcamonica. May 2021 to present MD Radiation
Medicine, AULSS 9 Scaligera, Verona, Italy. Obtained his MD degree in University of Oncologist Currently at Humanitas-Gavazzeni Bergamo.
Palermo (honor’s degree), his radiation oncology certification at University of Modena and
his PhD in Geriatric Oncology in University of Catania. Dr Fiorica‘s interests include
Stefano Vagge, MD, PhD is a full-time radiation oncologist at IRCCS Ospedale Policlinico
development of biomarkers from tissue and from diagnostic images and translational
San Martino, Genoa. He obtained his MD and the RO board certification at the University
research.
of Genoa. He reached a Ph.D. in translational and clinical onco-hematology at the
Department of Internal Medicine (DIMI) at the University of Genoa. His main field of
Angela Argenone, is a radiation oncologist with more than 10 years of experience. She has research and clinical practice are thoracic cancers, onco-hematology, lower G.I. and skin
collaborated within multiple Italian medical trials and active member and counsellor of cancers.
important scientific societies. She is authors of numerous scientific papers.
Antonio Pontoriero is Senior Researcher (Assistant Professor) Department of BIOMORF,
Melissa Scricciolo, is a Radiation Oncologist and at present, working at Radiotherapy Section of Radiological Science, Operative Unit of Radiation Oncology Messina University
Department of Hospital “Ospedale dell’Angelo” of Venice. She deal with head and neck School of Medicine, Messina, Italy. July 2007 - present Co-Director, Messina Cyberknife
cancer, lymphoma and lung cancer, and is member of Head and Neck Young Investigator Program. Authors of many peer-reviewed publications. Main field of research are Re-
group of EORTC, of coordination group AIRO Re-irradiation 2020–2021 and of coordi­ Irradiation, Prostate Cancer Gynecological Cancer, Stereotactic Body Radiotherapy,
nation group AIRO Triveneto 2020–2021. Cyberknife.

Salvatore Cozzi, is a radiation oncologist graduated in 2013. Actually working at Radi­ Vittorio Donato, President of Italian Association of Radiotherapy and Clinical Oncology.
ation Therapy Unit, USL-IRCCS of Reggio Emilia. Major area of interest are lung cancer, Researcher from 1991 to 2006 at University of Rome “La Sapienza”. From 2006 has
breast cancer and brachytherapy. Author and co-Author of 11 publications. become Head of Radiotherapy Department at “San Camillo Forlanini” Hospital of Rome
and from 2017 Chief of Oncology and Specialized Medicine Department.
Alessia Nardangeli is a Radiation Oncologist at the department of Radiation Oncology at
Fondazione Policlinico Universitario “A. Gemelli” IRCSS, Università Cattolica del Sacro Mariangela Massaccesi is a Radiation Oncologist at the Department of Radiation
Cuore in Rome, where she attended medical studies and completed residency in Radiation Oncology at the Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Università
Oncology in 2016. Her research interest include re-irradiation and gynecologic cancer. Cattolica del Sacro Cuore in Rome, where she attended medical studies, and completed
residency in Radiation Oncology in 2007. Dr. Massaccesi is the author and co-author of
more than 60 peer reviewed paper. Her research interest include reirradiation, lung can­
Francesco Dionisi, radiation oncologist with > 10 year of experience in the field. Major
cer, molecular imaging and new techniques for image guidance and radiation delivery.
areas of expertise: proton therapy, moving targets, head and neck cancer, liver cancer, re-

11

You might also like