ArticlePDF AvailableLiterature Review

Neoadjuvant Therapy for Neuroendocrine Neoplasms: Recent Progresses and Future Approaches

Frontiers
Frontiers in Endocrinology
Authors:

Abstract and Figures

Neuroendocrine neoplasms (NENs) are a heterogeneous group of tumors, their treatment being challenging and requiring a multidisciplinary approach. Though the only curative treatment is surgery, up to 50% of patients are diagnosed with metastatic disease. In the last years, neoadjuvant chemo(radio)therapy has become part of the standard of care in the treatment of different cancer types. However, evidence of its efficacy and safety in NEN patients has not yet been confirmed in the literature. The aim of the present review is to perform an extensive review of the scientific evidence for neoadjuvant therapy in patients with gastroenteropancreatic and thoracic NENs.
Content may be subject to copyright.
Neoadjuvant Therapy for
Neuroendocrine Neoplasms: Recent
Progresses and Future Approaches
Andrea Lania
1,2
*, Francesco Ferraù
3,4
, Manila Rubino
5
, Roberta Modica
6
,
Annamaria Colao
6
and Antongiulio Faggiano
7,8
on behalf of NIKE group
1
Endocrinology, Diabetology and Andrology Unit, Humanitas Clinical and Research CenterIRCCS, Rozzano, Italy,
2
Department of Biomedical Sciences, Humanitas University, Rozzano, Italy,
3
Department of Human Pathology of Adulthood
and Childhood G. Barresi, University of Messina, Messina, Italy,
4
Endocrine Unit, University Hospital G. Martino, Messina,
Italy,
5
Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO,
IRCCS, Milan, Italy,
6
Endocrinology, Department of Clinical Medicine and Surgery, Federico IIUniversity of Napoli, Napoli,
Italy,
7
Endocrinology, Department of Experimental Medicine, Sapienza, University of Rome, Rome, Italy,
8
Department of
Experimental Medicine, Division of Medical Physiopathology, Sapienza University of Rome, Rome, Italy
Neuroendocrine neoplasms (NENs) are a heterogeneous group of tumors, their treatment
being challenging and requiring a multidisciplinary approach. Though the only curative
treatment is surgery, up to 50% of patients are diagnosed with metastatic disease. In the
last years, neoadjuvant chemo(radio)therapy has become part of the standard of care in
the treatment of different cancer types. However, evidence of its efcacy and safety in NEN
patients has not yet been conrmed in the literature. The aim of the present review is to
perform an extensive review of the scientic evidence for neoadjuvant therapy in patients
with gastroenteropancreatic and thoracic NENs.
Keywords: neuroendocrine tumors, somatostatin (analogs and derivatives), peptide receptor radionuclide therapy,
everolimus, capecitabine, temozolomide, chemotherapy
INTRODUCTION
Although neuroendocrine neoplasms (NENs) are considered rare malignancies, their incidence has
rapidly increased in the last decades. Since several patients are diagnosed with metastatic disease,
curative surgery is usually not an option (1), palliative surgical intervention possibly being effective
in controlling clinical symptoms and improving patients quality of life (24). Neoadjuvant therapy,
with the aim of reducing tumor size and disease burden, can potentially change the clinical scenario
making it suitable for curative surgery as already demonstrated in other cancer types (58). While it
is conceivable that neoadjuvant chemo-and radiotherapy might be effective in NENs (9), reliable
evidence is still lacking in this eld and study results are difcult to compare due the heterogeneity
of both neoadjuvant therapies used, and series studied. Moreover, although the available therapeutic
options (i.e., somatostatin analogs, everolimus, chemotherapy, sunitinib, and peptide receptor
radionuclide therapy PRRT) are currently not included in any therapeutic algorithm with specic
neoadjuvant purpose, most of them have been used with this intent, even successfully (9).
Neoadjuvant chemotherapy, radiotherapy, and PRRT have been shown to provide variable
results in terms of tumor down-sizing (10). The aim of the present minireview is to perform an
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6514381
Edited by:
Antimo Migliaccio,
University of Campania Luigi Vanvitelli,
Italy
Reviewed by:
Daniela Pasquali,
University of Campania Luigi Vanvitelli,
Italy
Dario Giuffrida,
Mediterranean Institute of Oncology
(IOM), Italy
*Correspondence:
Andrea Lania
andrea.lania@humanitas.it
Specialty section:
This article was submitted to
Cancer Endocrinology,
a section of the journal
Frontiers in Endocrinology
Received: 09 January 2021
Accepted: 27 April 2021
Published: 26 July 2021
Citation:
Lania A, Ferraù F, Rubino M,
Modica R, Colao A and Faggiano A
(2021) Neoadjuvant Therapy for
Neuroendocrine Neoplasms: Recent
Progresses and Future Approaches.
Front. Endocrinol. 12:651438.
doi: 10.3389/fendo.2021.651438
MINI REVIEW
published: 26 July 2021
doi: 10.3389/fendo.2021.651438
extensive review of the scientic evidence for neoadjuvant
therapy in patients with pancreatic, gastrointestinal, and
thoracic NENs.
PANCREATIC NEUROENDOCRINE
NEOPLASMS
Pancreatic neuroendocrine neoplasms (pNENs) account for 1 to
2% of all pancreatic tumors and most of them are sporadic and
nonfunctioning. Their incidence has been increasing and
survival rates, although improved, remain poor compared with
other primary sites, with an overall survival of 3.6 years (1).
Often pNENs present with advanced disease at diagnosis and the
treatment of metastatic unresectable pNENs remains debated
and the role of neoadjuvant therapies is still uncertain. However,
among gastrointestinal NENs, data on the possible role of
neoadjuvant therapy are mainly related to pNENs even if the
results seem to be somewhat contradictory and difcult to
interpret due to the extreme heterogeneity of the data and the
incompleteness of the information provided (Table 1). In a large
series of high-grade gastrointestinal neuroendocrine carcinomas,
where pancreas was the most common primary site (361
patients), neoadjuvant or adjuvant therapy resulted in better
overall survival (OS) in patients with early-stage disease
compared with those treated with resection alone. Details of
neoadjuvant therapies were not available, but single and multi-
agent chemotherapy besides radiotherapy were included. The
positive effect of neoadjuvant or adjuvant therapy on OS seems
to suggest the importance of these treatments to lower the
incidence of both micrometastases and possibly to enhance
tumor resection thus lowering the risk of local and systemic
recurrences (26).
Patients who received neoadjuvant or adjuvant therapy had
better over-all survival, suggesting that high incidence of
micrometasta-sis contributes to the poor surgical outcomes. In
TABLE 1 | Neoadjuvant therapies in gastroenteropancreatic NENs.
Year Article type Neoadjuvant treatment Outcome Reference
Pancreas
2019 research article no details available no changes in OS (11)
2008 research article PRRT Partial response and R0 resection (12)
2009 case report PRRT Partial response and R0 resection (13)
2010 case report PRRT Partial response and R0 resection (14)
2011 research article PRRT Stabilization or partial response (15)
2012 case report PRRT Partial response and R0 resection (16)
2015 research article PRRT Better PFS and lower morbidity (17)
2015 research article PRRT Improved PFS (17)
2017 research article PRRT Improved PFS (18)
2018 case report PRRT Partial response and R0 resection (19)
2015 research article chemo Improved survival (20)
2016 research article chemo Improved OS (21)
2017 research article chemo no changes in tumor size (22)
2018 research article chemo Improved OS and RFS (23)
2011 case report chemo, radio R0 resection (24)
2017 research article chemo, radio Stabilization or partial response and R0 resection (25)
2018 research article chemo, radio Low recurrence risk and improved OS (26)
2012 research article chemo, radio No effects (27)
2012 research article PRRT, chemo Radiological response (28)
2020 research article PRRT, chemo Stabilization or partial response (29)
2020 research article PRRT, chemo Improved PFS and OS (30)
Esophagous
2018 case report chemo complete response, complete resection (31)
1989 case report/review chemo almost complete response, complete resection (32)
1995 case report chemo almost complete response, complete resection (33)
1999 case report chemo almost complete response, complete resection (34)
2002 case report chemo partial response, reduced tumor burden (35)
2003 case report chemo partial response, reduced tumor burden (3)
Rectum
2017 research article chemo, radio almost complete response, complete resection (36)
2018 case report chemo reduction of primary lesion/grading (4)
Miscellanea
2009 research article (midgut tumor) PRRT partial response, partial resection (15)
2012 research article (midgut tumor) PRRT partial response, complete resection (37)
2012 research article (midgut tumor) SSA-PRRT partial response, partial resection (38)
2012 research article (duodenal tumor) PRRT partial response (29)
2015 case report (small bowel tumor) PRRT no response (39)
Chemo, chemotherapy; Radio, radiotherapy; PRRT, peptide receptor radionuclide therapy; SSA, somatostatin analogs; OS, overall survival; R0, microscopically margin-negative
resection; PFS, progression-free survival; RFS, relapse free survival.
Lania et al. Neoadjuvant Therapy for NENs
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6514382
addition, the relatively high proportion of margin-positive re-
section raises the question of whether there is a role of
downstaging with neoadjuvant therapy, aimed at enhancing
resection and lowering risk of systemic recurrence.
Xie et al. recently described the largest group of pNENs in
whom neoadjuvant therapies were used. The study included
4,892 patients who underwent curative-intent surgical
resection. Authors showed that neoadjuvant therapy was
mainly prescribed in patients <65 years, with grade 3 pNENs
localized in the head of the pancreas and associated to the
presence of metastasis. In this setting, Authors did not nd
any signicant improvement in OS even in patients with grade 1
and 2 pNENs thus suggesting that neoadjuvant therapy should
be used with caution given the lack of conclusive data (11). It is
worth noting that, the main limitation of the Study was the lack
of any detail regarding the type of neoadjuvant therapies used.
PRRT with radiolabeled somatostatin analogues, over the
years, has evolved as an important therapeutic option for the
treatment of inoperable or metastasized, well/moderately
differentiated, NETs, particularly of the GEP (Table 1)(9).
Conversely, neoadjuvant PRRT based on either 177Lu-
octreotate or 90Y-DOTATATE have been shown to provide
variable results as tumor downsizing possibly due to the
heterogeneous inclusion criteria, the variable length of follow-
up and the different response criteria used (10). Kwekkeboom
et al. described a series of 310 patients with pNENs treated with
177Lu-octreotate PRRT. A partial response was observed in four
of them, all undergoing a subsequent R0 resection (12). Similar
results were conrmed by other case reports and small series (13,
14,16,19). Van Vliet et al. described the results of 177Lu-
octreotate as neoadjuvant therapy in 29 patients with borderline,
unresectable or oligometastatic nonfunctioning pNENs (14).
After PRRT, successful surgery was performed in nine patients
and all resection specimens showed brosis/sclerosis or necrosis
thus conrming the effects of 177Lu-octreotate on tumor tissues.
As a result, the median PFS was 69 months for patients with
successful surgery and 49 months for the other patients (14). On
the same line, other case reports and case series conrmed a
benecial effect of neoadjuvant PRRT with 90Y-DOTATATE on
tumor and/or metastases downsizing in patients with pNENs,
thus leading to a successful surgical intervention (15,17). Recently,
Partelli et al. reported a series of 23 resectable or potentially
resectable G1-G2 pNENs who underwent neoadjuvant PRRT
(177Lu-octreotate in three and 90Y-DOTATATE in 20 patients)
comparedto 23 patients who underwentupfront surgicaloperation.
PRRT did result in a reduction of primary tumor size and a
reduction in the number of positive lymph nodes compared with
controls. Interestingly, though both the rates of disease-specic
survival and median PFS from time of diagnosis were similar
between groups, in the subgroup of patients who underwent an
R0 resection, a trend toward a prolonged PFS was observed in the
PRRT group (18).
The administration of chemotherapy prior to surgical
resection is increasingly used for patients with adenocarcinomas
of the pancreas with the aim to improve surgical results. Similarly,
different regimens of chemotherapies have been described as
neoadjuvant treatment in pNENs, all these studies being biased
by their retrospective design and by the heterogeneity of
chemotherapy regimens used (Table 1). Dumont et al.
evaluated the effect of different neoadjuvant chemotherapy
regimens (i.e., 5-uorouracil, streptozotocin, doxorubicin,
cisplatin, etoposide, and oxaliplatine) on segmental portal
hypertension (SHP), the feasibility of surgery and the
prognostic inuence of a complete surgery in 42 patients with
G1/G2 locally advanced pNETs associated to SHP. A complete
resection was achieved in 13 out of 28 cases underwent surgery
and a not statistically signicant trend towards improved 5-year
survival was observed in patients with R0 resections compared to
R1/R2 resections and no resection at all (20). A retrospective
analysis of 59 patients with a histologic diagnosis of pancreatic
neuroendocrine carcinomas (pNECs) described ve patients who
underwent neoadjuvant treatment with etoposide and cisplatin
before surgery. Four of them had a curative resection and one
patient with stage IV disease remained with residual small liver
metastases (21). A further retrospective observational study
analyzed the efcacy of neoadjuvant 5-uorouracil (5-FU),
doxorubicin, and streptozocin (FAS) chemotherapy in 29
patients with non-metastatic locally advanced well-differentiated
pNENs. In this series, neoadjuvant FAS did not induce a clinically
signicant change inthe size of the primary tumor in upto 90% of
treated patients thus suggesting that localized disease does not
benet from this preoperative treatment in terms of tumor
downstaging (22). Preoperative FAS treatment has been further
evaluated in a retrospective series of 27 patients with pancreatic
neuroendocrine liver metastases (NELM) who underwent liver
resection. Despite being associated with higher rates of
synchronous disease, lymph node metastases, and larger tumor
size, patients who underwent preoperative FAS had similar
overall survival OS and RFS as patients who did not. Similarly,
in patients who presented with synchronous liver metastases the
median OS and RFS were signicantly greater among patients
who received preoperative FAS. Authors concluded that
preoperative FAS could be considered for patients with
advanced synchronous pancreatic NELM (23).
Few data are available on the effects of association of
chemotherapy and radiotherapy as neoadjuvant therapy in
pNENs, all these data being mainly obtained from case reports
(Table 1). A poorly differentiated pNEC metastatic to the breast
and lung was successfully managed with neoadjuvant
chemotherapy (5-FU treatment followed by carboplatin and
etoposide) and radiotherapy, followed by radical surgical
resection (24). Among 33 patients with pNENs undergoing
surgical resection with curative intent, 16 underwent surgery
alone, while 17 underwent adjuvant or neoadjuvant external
beam radiotherapy in addition to surgery. Fluoropyrimidine-
based chemotherapy was delivered concurrently in 14 patients
receiving radiotherapy. Local control in patients receiving
combined modality therapy was like those who had surgery
alone. Although the Authors conclude that the role of
neoadjuvant radiotherapy remains unclear, it has been
hypothesized that patients who underwent radiotherapy had
more aggressive or extensive disease than the surgery alone
Lania et al. Neoadjuvant Therapy for NENs
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6514383
group thus explaining the lack of any signicant effect of
combined neoadjuvant treatments (27).
Capecitabine combined with temozolomide (CAPTEM) has
been frequently used in the treatment of pNENs. In particular,
Strosberg et al. demonstrated that CAPTEM regimen was
extremely effective for treatment of metastatic pNENs,
resulting in an objective response rate of 70% and median PFS
of 18 months (40). These data have been strengthened by a recent
metanalysis conrming that capecitabine combined with
temozolomide is effective for treating patients with advanced
NENs, disease control rate being 72.8% (41). Neoadjuvant
CAPTEM regimen with or without radiation has been
successfully applied in six pNENs with borderline resectable
disease. All patients had radiological evidence of tumor regression
after neoadjuvant treatment (two partial responses and four
stabilization) and all of them could undergo successful resection
of the primary tumor with negative margins in 4/6 patients (25).
The efcacy of CAPTEM regimen in the neoadjuvant setting was
further conrmed by Ostwal et al. who studied 30 patients with
locally advanced pNENs or pancreatic neuroendocrine hepatic
metastases receiving neoadjuvant CAPTEM. Partial response was
observed in 13 of them, while a stable disease was found in 16
patientsthus suggesting that neoadjuvant CAPTEMmight improve
the radicality of the surgical procedure (29).
The association of PRRT and chemotherapy has also been used
as neoadjuvant therapy in pNENs, taking advantage from the
radiosensitising effects of 5-FU. In this respect, the combination
of PRRT with 177Lu-octreotate and 5FU chemotherapy was found
to be effective in ve nonfunctioning pancreatic and one duodenum
NEN with inoperable disease, resulting in radiological response in
all pNENs. Only one patient underwent surgery successfully after
treatment and remained12 months postoperatively aliveand free of
disease (28). Finally, combined PRRT and chemotherapy
sandwiching two cycles of CAPTEM between two cycles of
PRRT, has been proposed in neoadjuvant setting. This regimen
resulted in favorable response rates with effective control of
symptoms and longer PFS and OS in NEN patients with
aggressive, both FDG- and SSTR-avid, metastatic progressive
disease (30).
GASTROINTESTINAL NEUROENDOCRINE
NEOPLASMS
As for pNENs, neoadjuvant treatments have been proposed for other
gastrointestinal NENS, data being mainly based on few case reports
(Table 1). Neoadjuvant chemotherapy can be effective in patients
with esophageal neuroendocrine carcinoma (ENEC), which are rare
but aggressive neoplasms. In 2018, Yamamoto et al. reported the case
of a patient with an ENEC who received neoadjuvant chemotherapy
using etoposide and cisplatin. One course of chemotherapy led to
tumor downstaging at endoscopy and to the absence of FDG
accumulation at PET-CT examination. Seven weeks after
chemotherapy, a thoracoscopic esophagectomy was performed and
the histopathological examination of the resected specimen revealed
no residual cancer cells, demonstrating a complete response with
neoadjuvant treatment (31). Other few cases of ENECs treated with
neoadjuvant chemotherapy have been reported (3235). In three
patients, cisplatin or combinationchemotherapycausedanalmost
complete regression of the neoplasm with evidence of only
microscopic foci of tumor in the resected esophageal specimen
(3234), while in other three cases treated with carboplatin/
etoposide or combination chemotherapy a signicant reduction in
tumor burden was observed (35).
Neoadjuvant chemotherapy or chemoradiotherapy has been
anecdotally reported to be effective also in rectal NENs (Table 1).
In a study reporting on the management of patients with high grade
rectum or anal canal neuroendocrine carcinomas, two cases were
treated with preoperative pelvic chemoradiation. One of them
received radiotherapy followed by consolidative cisplatin/5-FU, low
anterior resection, and postoperative cisplatin/etoposide, while the
second patient received induction oxaliplatin/irinotecan, followed by
radiotherapy, trans anal excision, and additional oxaliplatin/
irinotecan. Both patients had only microscopic foci of residual
carcinoma at surgery (36). In another case report, a 50-year-old
woman diagnosed with a liver mass and a G3 rectal NEN was treated
with two cycles of neoadjuvant chemotherapy with etoposide and
nedaplatin, this treatment being effective in rectal tumor but not liver
metastasis shrinkage. Subsequently, the patient was switched to
irinotecan plus nedaplatin, associated to octreotide LAR 30 mg/
month because of neuroendocrine symptoms and MRI abdomen
scan showed no signicant changes in lesions size. Therefore, surgery
was suggested and histopathological examination showed that the
tumor downgraded from G3 to G2 thus suggesting that neoadjuvant
chemotherapy may be effective in reducing primary lesion size and
possibly grading, offering favourable conditions for less demolitive
and more effective surgery.
The possible role of neoadjuvant PRRT and PRRT +
chemotherapy combination was evaluated in small series of
advanced gastrointestinal NENs (Table 1). Sowa-Staszczak et al.
reported on neoadjuvant 90YDOTA-TATE treatment of ve
patients with foregut tumors, including three with pancreatic,
and one with midgut NEN. According to RECIST criteria, disease
stabilization was observed in four and partial responses in two
patients, one with pancreatic and the second with the midgut
NEN. In this latter case, tumor size decreased from 11 to 7.2 cm
one month after PRRT. Five months later, a further reduction in
tumor size was observed, enabling qualication for a laparotomy,
which was performed 11 months after PRRT. However, only
partial removal of the tumor was possible due to inltration of
the large vessels (15). In another study on 89 patients with
disseminated and inoperable gastrointestinal NENs, Authors
described one patient with a midgut tumor who was successfully
treated with PRRT in a neoadjuvant setting, thus enabling an
effective surgical intervention (37). The case of a 43-year-old man
complaining of abdominal pain, vomiting, weight loss and ushes,
who underwent CT examination that revealed upper and middle
abdomen tumor was reported by Sowa-Staszczak et al. (38).
Histopathological examination of tumor specimen obtained
during exploratory laparotomy showed a well-differentiated NET
according to the 2000 WHO classication. The patient received
ve cycles of chemotherapy (streptozocin and 5-FU) without any
Lania et al. Neoadjuvant Therapy for NENs
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6514384
response and then he underwent PRRT with 90Y-DOTA-TATE.
The subsequent CT scan revealed a reduction in tumor size and the
patient wastherefore candidate toa second laparotomy for a partial
excisionof the tumor. Then he was treated with long-acting SSAand
two additional courses of 90Y-DOTA-TATE that induced a further
reduction of tumor size, potentially enabling a further laparotomy
for curativesurgery (38). Barber et al. reported their experience with
PRRT as neoadjuvant treatment in ve patients with NENs, one of
them being diagnosed with a locoregional recurrence of a duodenal
tumor. The patient was treated with one cycle of 177 Lu-
DOTATATE, with a partial scintigraphy and biochemical
response (28). On the other hand, Frilling et al. reported the case
of a patientwith a small-bowelwell differentiatedNEN metastasised
to the root of the mesentery, who underwent four cycles of
neoadjuvant PRRT with 177Lu-DOTATATE. A following 68Ga-
DOTATATE PET/CT demonstrated high tracer uptake in the
mesenteric and aortocaval tumor foci with signicantly higher
SUV than pre-treatment imaging with no change in size of either
the mesenteric or the aortocaval lesions. The patient then
underwent a modied liver free multivisceral transplantation (39).
Overall, the few reported experiences would suggest that
neoadjuvant chemotreatment can be a successful management
strategy in esophageal NEN, while too little data are available about
chemo/radio-treatment of other non-pancreatic gastrointestinal
NENs in a neoadjuvant setting. However, PRRT seems represent
an option in selected cases in this context.
THORACIC NEUROENDOCRINE
NEOPLASMS
Lung neuroendocrineneoplasms represent approximately 2030%
of all NENs. Based on clinical, histological and molecular data, lung
NENs are classied in two main categories well differentiated
neuroendocrine tumors (carcinoids) and poorly differentiated
neuroendocrine carcinomas (NECs). Furthermore, lung
carcinoids (LC) are classied in typical (low grade) and atypical
carcinoids (intermediate grade) and lung NECs in large-cells and
small-cells carcinomas (LCNECand SCLC, respectively). Only low-
quality evidence guides the therapeutic management of LC, and
everolimus is the only approved drugs. However currently used
systemic therapeutic options include somatostatin analogues,
alkylating- and oxaliplatin-based chemotherapies and PRRT. Few
data are available on the efcacy of neoadjuvant treatment in
thoracic NENs and all these data come from small series and case
reports (Table 2).
Srirajaskanthan el al. reported two patients with lung NENs
that received a preoperative chemotherapy with 5-FU, cisplatin
and streptozotocin, that induced a good response with
consequently a curative resection, both patients being disease
free at 36 months after surgery (42). A multicentric study by
Daddi et al. reported six of 247 patients with atypical carcinoids
treated with neoadjuvant chemotherapy as an initial diagnosis of
SCLC was performed on ne needle aspiration biopsy. Though
no data on the results of neoadjuvant treatment were clearly
shown, Authors found an association between adjuvant and
neoadjuvant treatments and a worse prognosis. These data do
not support the efcacy of neoadjuvant therapies in terms of
complete regression of the metastatic disease. However, these
treatments might be effective in alleviating clinical signs and
symptoms (43). The same Authors reported ve patients with
poorly differentiated NECs who underwent to induction therapy
and surgery without disease recurrence at 5 years, but no
information was available on the chemotherapy regimens
used (44).
More data are available on the role of neoadjuvant therapies in
LCNEC. In the multicenter retrospective study by Veronesi et al.,
15% of 144 patients who underwent surgical resection for LCNEC,
received neoadjuvant chemotherapy (i.e., platin/etoposide,
gemcitabine, vinorelbine and taxol). In this study no association
was found between neoadjuvant chemotherapy and survival except
for stage I patients in whom induction or post-operative
chemotherapy tended to be associated to a longer OS (OS rate at
3 years 100% vs 58%) (45). Sarkaria et al. retrospectively analyzed
100 patients with LCNEC operated at Memorial Sloan-Kettering
Cancer Center. Twenty-four patients received neo-adjuvant
platinum-based chemotherapy and 68% showed a partial
response and 31% were characterized by a stable disease. The
correlation analysis did not show any association between OS and
neoadjuvant or adjuvant chemotherapy. The authors also
TABLE 2 | Neoadjuvant therapies in lung NENs.
Year Article type Lung NENs Neoadjuvant treatment Outcome Reference
2009 case report undened chemo complete response and resection (42)
2014 research article atypical carcinoids chemo no response, worse prognosis (43)
2004 research article NEC chemo complete resection (44)
2006 research article LCNEC chemo longer OS (stage I) (45)
2011 research article LCNEC chemo longer OS (stage IB-IIA) (46)
2010 research article LCNEC chemo higher 5-year survival rate (47)
2019 research article LCNEC, SCLC chemo higher 5-year survival rate (48)
2018 case report LCNEC chemo downsizing of the tumor, complete resection (49)
2019 case report LCNEC chemo downsizing of the tumor, complete resection (50)
2010 research article LCNEC chemo worse 5-year OS (51)
2015 research article Thymus NENs chemo or radio no effects (52)
2008 case report Thymus NENs chemo downsizing of the tumor, complete resection (53)
NEC, neuroendocrine carcinoma; LCNEC, large cell neuroendocrine carcinoma; SCLC, small cell neuroendocrine carcinoma; Chemo, chemotherapy; radio, radiotherapy;
OS, overall survival.
Lania et al. Neoadjuvant Therapy for NENs
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6514385
performed a subgroup analysis in patients with completely resected
advancedstage (IBIIIA) diseaseand, in these patients, neoadjuvant
andadjuvantchemotherapyresultedinanimprovedOS(2vs7.4
years) and 5 years OS rate (37% vs 51%) (46). Saji et al.
retrospectively conrmed a positive effect of perioperative
chemotherapy on survival in 45 patients with LCNEC. In this
study, seven patients received a neoadjuvant chemotherapy
(cisplatin and paclitaxel in four and cisplatin/topotecan in three,
respectively) thus leading to a statistically signicant higher 5-year
survival rate (87.5% vs 58%) (47). Similar results were obtained by
Ogawa et al. retrospectively evaluating a series LCNEC and SCLC
who underwent complete resection. Seventy patients (31 with
LCNEC and 32 with SCLC) received perioperative platinum-
based chemotherapy and a signicant improvement of the 5-year
OS rates was observed (74.5% vs. 34.7%). Multivariate analysis
revealed that perioperative chemotherapy, sublobar resection, and
lymph node metastasis were independently associated with survival
(48). The efcacy of perioperative chemotherapy in patients with
LCNEC has been further conrmed by some case reports (49,50).
In particular, Mauclet et al. reported a case of a 41-years old women
with a large LCNEC with mediastinal involvement. After an
ineffective rst line chemotherapy with cisplatin etoposide,
patient underwent to palliative radiotherapy and second line
therapy with Nivolumab that led to a downsizing of the tumor.
Patient underwent surgery with the complete removal of the tumor
and histology showed an absence of viable tumor cells, while
necrosis and brosis were observed (50). Finally, the retrospective
analysis of 63 patients with LCNEC showed that neoadjuvant
platin-etoposide based chemotherapy was associated with a trend
towards a worse 5-year OS rate despite a partial response in 12 cases
was observed. Authors suggested that the negative association
between neoadjuvant chemotherapy and survival could be due to
the fact that only patients with stage III tumors received induction
chemotherapy (51).
NENs of thethymus are very rare tumors, accounting for0.4% of
all carcinoid tumors. Based on WHO 2015, also thymic NENs are
classied in two main histopathological and clinical categories: well
differentiated tumors, typical and atypical carcinoids and poorly
differentiated tumors, small cell and large cell carcinoma. These
tumors could be associated to ectopic hormonal secretion, in
particular adrenocorticotropic hormone secretion or to multiple
endocrine neoplasia type 1. The prognosis of patients with thymic
NENs is poor because of the high incidence of local recurrenceand
distant metastasis and 5-year OS vary from 3070%. As for other
lung NENs, few data are available on neoadjuvat therapy in thymic
NENs (Table 2). Few data are available on neoadjuvant therapy in
thymic NENs. Filosso et al. reported 25 patients with primary
thymic NENs treated by induction therapy (19 by chemotherapy
and six by radiotherapy), these treatments having no impact on
survival (52). Dham et al. described a clinical case of a 40-year-old
man with unresectable typicalcarcinoid of the thymus. Threeweeks
of treatment with sunitinib 50 mg per day for 4 weeks with 2 weeks
off and octreotide LAR 30 mg very 4 weeks, induced a tumor
shrinkagethat led to curativesurgery of the mediastinal mass andno
evidence of disease recurrence was evident 12 months after
surgery (53).
CONCLUSIONS
Data on neoadjuvant treatment of NEN patients are scanty,
mainly based on inhomogeneous and often incomparable
retrospective studies of limited numbers of patients and
prospective studies are necessary to clarify the role of
neoadjuvant therapy in this clinical setting. Available literature
on pancreatic NEN patients suggests PRRT to be variably
successful as a neoadjuvant approach, as well as chemotherapy
to be more promising in patients with advanced synchronous
pancreatic NELM, whilein the same settingcombined chemo/
radio/PRRT-therapies would not be supported by sufcient
evidence. Neoadjuvant PRRT, chemo or chemoradio-therapies
have been anectodotally reported to be effective in non-pancreatic
gastrointestinal NENs. Among thoracic NENs, neoadjuvant
chemotherapy has been inconsistently reported to be benecial
in LCNC patients, also in relationship to disease stage, while little
evidence would suggest neoadjuvant treatment to be negligible in
thymic tumors. Therefore, it is advisable to use a neoadjuvant
approach with caution, as the effects on quality of life and long-
term results in terms of prolonged survival remain yet to be
conrmed and the choice of this therapeutic approach should be
discussed for each single patient in a multidisciplinary setting.
AUTHOR CONTRIBUTIONS
AL conceived and authored the nal draft of the manuscript. FF,
MR, and RM provided further content, added key references, and
authored sections of the manuscript. AF and AC revised the
manuscript. All authors contributed to the article and approved
the submitted version.
REFERENCES
1. Dasari A, Shen C, Halperin D, Zhao B, Zhou S, Xu Y, et al. Trends in the
Incidence, Prevalence, and Survival Outcomes in Patients With
Neuroendocrine Tumors in the United States. JAMA Oncol (2017) 3:1335
42. doi: 10.1001/jamaoncol.2017.0589
2. Perysinakis I, Aggeli C, Kaltsas G, Zografos GN. Neoadjuvant Therapy for
Advanced Pancreatic Neuroendocrine Tumors: An Emerging Treatment
Modality? Hormones (Athens) (2016) 15(1):1522. doi: 10.14310/horm.
2002.1636
3. Howe JR, Cardona K, Fraker DL, Kebebew E, Untch BR, Wang YZ, et al. The
Surgical Management of Small Bowel Neuroendocrine Tumors: Consensus
Guidelines of the North American Neuroendocrine Tumor Society. Pancreas
(2017) 46(6):71531. doi: 10.1097/MPA.0000000000000846
4. Tsoli M, Spei ME, Wallin G, Kaltsas G, Daskalakis K. Association of a
Palliative Surgical Approach to Stage IV Pancreatic Neuroendocrine
Neoplasms With Survival: A Systematic Review and Meta-Analysis. Cancers
(Basel) (2020) 12(8):2246. doi: 10.3390/cancers12082246
5. Oba A, Ho F, Bao QR, Al-Musawi MH, Schulick RD, Del Chiaro M.
Neoadjuvant Treatment in Pancreatic Cancer. Front Oncol (2020) 10:245.
doi: 10.3389/fonc.2020.00245
6. Lee JS, Yost SE, Yuan Y. Neoadjuvant Treatment for Triple Negative Breast
Cancer: Recent Progresses and Challenges. Cancers (Basel) (2020) 12(6):1404.
doi: 10.3390/cancers12061404
Lania et al. Neoadjuvant Therapy for NENs
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6514386
7. Leng XF, Daiko H, Han YT, Mao YS. Optimal Preoperative Neoadjuvant
Therapy for Resectable Locally Advanced Esophageal Squamous Cell
Carcinoma. Ann N Y Acad Sci (2020) 1482(1):21324. doi: 10.1111/
nyas.14508
8. Müller PC, Frey MC, Ruzza CM, Nickel F, Jost C, Gwerder C, et al.
Neoadjuvant Chemotherapy in Pancreatic Cancer: An Appraisal of the
Current High-Level Evidence. Pharmacology (2020) 23:111. doi: 10.1159/
000510343
9. Cives M, Strosberg J. Treatment Strategies for Metastatic Neuroendocrine
Tumors of the Gastrointestinal Tract. Curr Treat Options Oncol (2017) 18
(3):14. doi: 10.1007/s11864-017-0461-5
10. Hirmas N, Jadaan R, Al-Ibraheem A. Peptide Receptor Radionuclide Therapy
and the Treatment of Gastroentero-Pancreatic Neuroendocrine Tumors:
Current Findings and Future Perspectives. Nucl Med Mol Imaging (2018)
52(3):1909. doi: 10.1007/s13139-018-0517-x
11. Xie H, Liu J, Yadav S, Keutgen XM, Hobday TJ, Strosberg JR, et al. The Role of
Perioperative Systemic Therapy in Localized Pancreatic Neuroendocrine
Neoplasms. Neuroendocrinology (2020) 110(3-4):23445. doi: 10.1159/
000501126
12. Kwekkeboom DJ, de Herder WW, Kam BL, van Eijck CH, van Essen M, Kooij
PP, et al. Treatment With the Radiolabeled Somatostatin Analog [177 Lu-
DOTA 0,Tyr3]octreotate: Toxicity, Efcacy, and Survival. J Clin Oncol (2008)
26(13):212430. doi: 10.1200/JCO.2007.15.2553
13. Kaemmerer D, Prasad V, Daffner W, Hörsch D, Klöppel G, Hommann M,
et al. Neoadjuvant Peptide Receptor Radionuclide Therapy for an Inoperable
Neuroendocrine Pancreatic Tumor. World J Gastroenterol (2009) 15
(46):586770. doi: 10.3748/wjg.15.5867
14. Stoeltzing O, Loss M, Huber E, Gross V, Eilles C, Mueller-Brand J, et al. Staged
Surgery With Neoadjuvant 90Y-DOTATOC Therapy for Down-Sizing
Synchronous Bilobular Hepatic Metastases From a Neuroendocrine
Pancreatic Tumor. Langenbecks Arch Surg (2010) 395(2):18592.
doi: 10.1007/s00423-009-0520-x
15. Sowa-Staszczak A, Pach D, Chrzan R, Tromiuk M, Stefanska A, Tomaszuk
M, et al. Peptide Receptor Radionuclide Therapy as a Potential Tool for
Neoadjuvant Therapy in Patients With Inoperable Neuroendocrine Tumours
(Nets). Eur J Nucl Med Mol Imaging (2011) 38(9):166974. doi: 10.1007/
s00259-011-1835-8
16. Ezziddin S, Lauschke H, Schaefers M, Meyer C, van Essen M, Biersack HJ,
et al. Neoadjuvant Downsizing by Internal Radiation: A Case for Preoperative
Peptide Receptor Radionuclide Therapy in Patients With Pancreatic
Neuroendocrine Tumors. Clin Nucl Med (2012) 37(1):1024. doi: 10.1097/
RLU.0b013e318238f111
17. van Vliet EI, van Eijck CH, de Krijger RR, Nieveen van Dijkum EJ, Teunissen
JJ, Kam BL, et al. Neoadjuvant Treatment of Nonfunctioning Pancreatic
Neuroendocrine Tumors With [177Lu-DOTA0,Tyr3]Octreotate. J Nucl Med
(2015) 56(11):164753. doi: 10.2967/jnumed.115.158899
18. Partelli S, Bertani E, Bartolomei M, Perali C, Muffatti F, Grana CM, et al.
Peptide Receptor Radionuclide Therapy as Neoadjuvant Therapy for
Resectable or Potentially Resectable Pancreatic Neuroendocrine Neoplasms.
Surgery (2018) 163(4):7617. doi: 10.1016/j.surg.2017.11.007
19. da Silva TN, van Velthuysen MLF, van Eijck CHJ, Teunissen JJ, Hoand J, de
Herder WW. Successful Neoadjuvant Peptide Receptor Radionuclide Therapy
for an Inoperable Pancreatic Neuroendocrine Tumour. Endocrinol Diabetes
Metab Case Rep (2018) 2018:180015. doi: 10.1530/EDM-18-0015
20. Dumont F, Goudard Y, Caramella C, GoereD, Baudin E, Elias D. Therapeutic
Strategies for Advanced Pancreatic Neuroendocrine Tumors With Segmental
Portal Hypertension. World J Surg (2015) 39(8):197480. doi: 10.1007/
s00268-015-3030-8
21. Crippa S, Partelli S, Belori G, Palucci M, Muffatti F, Adamenko O, et al.
Management of Neuroendocrine Carcinomas of the Pancreas (WHO G3): A
Tailored Approach Between Proliferation and Morphology. World J
Gastroenterol (2016) 722(45):994453. doi: 10.3748/wjg.v22.i45.9944
22. Prakash L, Bhosale P, Cloyd J, Kim M, Parker N, Yao J, et al. Role of
Fluorouracil, Doxorubicin, and Streptozocin Therapy in the Preoperative
Treatment of Localized Pancreatic Neuroendocrine Tumors. J Gastrointest
Surg (2017) 21(1):15563. doi: 10.1007/s11605-016-3270-4
23. Cloyd JM, Omichi K, Mizuno T, Kawaguchi Y, Tzeng CD, Conrad C, et al.
Preoperative Fluorouracil, Doxorubicin, and Streptozocin for the Treatment
of Pancreatic Neuroendocrine Liver Metastases. Ann Surg Oncol (2018) 25
(6):170915. doi: 10.1245/s10434-018-6468-8
24. Satahoo-Dawes S, Palmer J, Manning EW III, Levi J. Breast and Lung
Metastasis From Pancreatic Neuroendocrine Carcinoma. World J Radiol
(2011) 3(1):327. doi: 10.4329/wjr.v3.i1.32
25. Ambe CM, Nguyen P, Centeno BA, Choi J, Strosberg J, Kvols L, et al.
Multimodality Management of Borderline ResectablePancreatic
Neuroendocrine Tumors: Report of a Single-Institution Experience. Cancer
Control (2017) 24(5):1073274817729076. doi: 10.1177/1073274817729076
26. Alese OB, Jiang R, Shaib W, Wu C, Akce M, Behera M, et al. High-Grade
Gastrointestinal Neuroendocrine Carcinoma Management and Outcomes: A
National Cancer Database Study. Oncologist (2019) 24(7):91120.
doi: 10.1634/theoncologist.2018-0382
27. Zagar TM, White RR, Willett CG, Tyler DS, Papavassiliou P, Papalezova KT,
et al. Resected Pancreatic Neuroendocrine Tumors: Patterns of Failure and
Disease-Related Outcomes With or Without Radiotherapy. Int J Radiat Oncol
Biol Phys (2012) 83(4):112631. doi: 10.1016/j.ijrobp.2011.09.041
28. Barber TW, Hofman MS, Thomson BN, Hicks RJ. The Potential for Induction
Peptide Receptor Chemoradionuclide Therapy to Render Inoperable
Pancreatic and Duodenal Neuroendocrine Tumours Resectable. Eur J Surg
Oncol (2012), 38(1):6471. doi: 10.1016/j.ejso.2011.08.129
29. Ostwal V, Basu S, Bhargava P, Shah M, Parghane RV, Srinivas S, et al.
Capecitabine-Temozolomide (CAPTEM) in Advanced Grade 2 and Grade 3
Neuroendocrine Neoplasms (Nens) - Benets of Chemotherapy in NENs
With Signicant 18FDG Uptake. Neuroendocrinology (2020). doi: 10.1159/
000511987
30. Parghane RV, Ostwal V, Ramaswamy A, Bhandare M, Chaudhari V, Talole S,
et al. Long-Term Outcome of SandwichChemo-PRRT: A Novel Treatment
Strategy for Metastatic Neuroendocrine Tumors With Both FDG- and SSTR-
avid Aggressive Disease. Eur J Nucl Med Mol Imaging (2021) 48(3):91323.
doi: 10.1007/s00259-020-05004-5
31. Yamamoto M, Ozawa S, Koyanagi K, Oguma J, Kazuno A, Ninomiya Y, et al.
Effectiveness of Neoadjuvant Chemotherapy With Etoposide and Cisplatin
Followed by Surgery for Esophageal Neuroendocrine Carcinoma: A Case
Report. J Thorac Dis (2018) 10(6):E4505. doi: 10.21037/jtd.2018.05.170
32. Walker SJ, Steel A, Cullen MH, Matthews HR. Treatment of Oesophageal
Small Cell Carcinoma by Combined Chemotherapy and Surgical Resection:
Report of Two Cases and Review of Published Cases. Thorax (1989) 44:7512.
doi: 10.1136/thx.44.9.751
33. Muto I, Nishimaki T, Aizawa K, Suzuki T, Tanaka O, Hatakeyama K. Primary
Small Cell Carcinoma of the Esophagus: Report of a Case. Surg Today (1995)
25(9):8303. doi: 10.1007/BF00311462
34. Nimura Y, Koide N, Nishio A, Watanabe H, Yazawa K, Adachi W, et al.
Effective Treatment With Chemotherapy and Surgical Resection for Small
Cell Carcinoma of the Esophagus: Report of a Case. Hepatogastroenterology
(1999) 46(27):177881.
35. Makino H, Tajiri T, Onda M, Sasajima K, Miyashita M, Nomura T, et al.
Effectiveness of Preoperative Chemotherapy Using Carboplatin (CBDCA) and
Surgery Against an Esophageal Small Cell Carcinoma. Dis Esophagus (2002)
15(3):23741. doi: 10.1046/j.1442-2050.2002.00244.x
36. Voong KR, Rashid A, Crane CH, Minsky BD, Krishnan S, Yao JC, et al.
Chemoradiation for High-grade Neuroendocrine Carcinoma of the Rectum
and Anal Canal. Am J Clin Oncol (2017) 40(6):55560. doi: 10.1097/
COC.0000000000000211
37. Pach D, Sowa-Staszczak A, Kunikowska J, Krolicki L, Tromiuk M, Stefanska
A, et al. Repeated Cycles of Peptide Receptor Radionuclide Therapy (PRRT)
Results and Side-Effects of the Radioisotope 90Y-DOTA TATE, 177lu-Dota
TATE or 90Y/177Lu-DOTA TATE Therapy in Patients With Disseminated
NET. Radiother Oncol (2012) 102(1):4550. doi: 10.1016/j.radonc.2011.08.006
38. Sowa-Staszczak A, Pach D, Stefanska A, Szybinski P, Kulig J, Tomaszewska R,
et al. Case Report of a Patient With Initially Inoperable Well-Differentiated
Midgut Neuroendocrine Tumor (WDNT)PRRT and Long-Acting
Somatostatin Analogs as the Neoadjuvant Therapy. Nucl Med Rev Cent East
Eur (2012) 15(2):1379.
39. Frilling A, Giele H, Vrakas G, Reddy S, Macedo R, Al-Nahhas A, et al.
Modied Liver-Free Multivisceral Transplantation for a Metastatic Small
Bowel Neuroendocrine Tumor: A Case Report. Transplant Proc (2015) 47
(3):85862. doi: 10.1016/j.transproceed.2015.01.007
Lania et al. Neoadjuvant Therapy for NENs
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6514387
40. Strosberg JR, Fine RL, Choi J, Nasir A, Coppola D, Chen DT, et al. First-Line
Chemotherapy With Capecitabine and Temozolomide in Patients With
Metastatic Pancreatic Endocrine Carcinomas. Cancer (2011) 117(2):26875.
doi: 10.1002/cncr.25425
41. Lu Y, Zhao Z, Wang J, Lv W, Lu L, Fu W, et al. Safety and Efcacy of
Combining Capecitabine and Temozolomide (CAPTEM) to Treat Advanced
Neuroendocrine Neoplasms: A Meta-Analysis. Med (Baltimore) (2018) 97
(41):e12784. doi: 10.1097/MD.0000000000012784
42. Srirajaskanthan R, Toumpanakis C, Karpathakis A, Marelli L, Quigley AM,
Dusmet M, et al. Surgical Management and Palliative Treatment in Bronchial
Neuroendocrine Tumours: A Clinical Study of 45 Patients. Lung Cancer
(2009) 65(1):6873. doi: 10.1016/j.lungcan.2008.10.025
43. Daddi N, Schiavon M, Filosso PL, Cardillo G, Ambrogi MC, De Palma A, et al.
Prognostic Factors in a Multicentre Study of 247 Atypical Pulmonary
Carcinoids. Eur J Cardiothorac Surg (2014) 45(4):67786. doi: 10.1093/ejcts/
ezt470
44. Daddi N, Ferolla P, Urbani M, Semeraro A, Avenia N, Ribacchi R, et al.
Surgical Treatment of Neuroendocrine Tumors of the Lung. Eur J
Cardiothorac Surg (2004) 26(4):8137. doi: 10.1016/j.ejcts.2004.05.052
45. Veronesi G, Morandi U, Alloisio M, Terzi A, Cardillo G, Filosso P, et al. Large
Cell Neuroendocrine Carcinoma of the Lung: A Retrospective Analysis of 144
Surgical Cases. Lung Cancer (2006) 53(1):1115. doi: 10.1016/
j.lungcan.2006.03.007
46. Sarkaria IS, Iyoda A, Roh MS, Sica G, Kuk D, Sima CS, et al. Neoadjuvant and
Adjuvant Chemotherapy in Resected Pulmonary Large Cell Neuroendocrine
Carcinomas: A Single Institution Experience. Ann Thorac Surg (2011) 92
(4):11806. doi: 10.1016/j.athoracsur.2011.05.027 discussion 1186-7.
47. Saji H, Tsuboi M, Matsubayashi J, Miyajima K, Shimada Y, Imai K, et al.
Clinical Response of Large Cell Neuroendocrine Carcinoma of the Lung to
Perioperative Adjuvant Chemotherapy. Anticancer Drugs (2010) 21(1):8993.
doi: 10.1097/CAD.0b013e328330fd79
48. Ogawa H, Tanaka Y, Kitamura Y, Shimizu N, Doi T, Hokka D, et al. Efcacy of
Perioperative Chemotherapy for Pulmonary High-Grade Neuroendocrine
Carcinomas: A Propensity Score Matching Analysis. J Thorac Dis (2019) 11
(4):114554. doi: 10.21037/jtd.2019.04.56
49. Tang H, Wang H, Xi S, He C, Chang Y, Wang Q, et al. Perioperative
Chemotherapy With Pemetrexed and Cisplatin for Pulmonary Large-Cell
Neuroendocrine Carcinoma: A Case Report and Literature Review. Onco
Targets Ther (2018) 11:255763. doi: 10.2147/OTT.S160565
50. Mauclet C, Duplaquet F, Pirard L, Rondelet B, Dupont M, Pop-Stanciu C,
et al. Complete Tumor Response of a Locally Advanced Lung Large-Cell
Neuroendocrine Carcinoma After Palliative Thoracic Radiotherapy and
Immunotherapy With Nivolumab. Lung Cancer (2019) 128:536.
doi: 10.1016/j.lungcan.2018.12.006
51. Fournel L, Falcoz PE, Alifano M, Charpentier MC, Boudaya MS, Magdeleinat
P, et al. Surgical Management of Pulmonary Large Cell Neuroendocrine
Carcinomas: A 10-Year Experience. Eur J Cardiothorac Surg (2013) 43
(1):1114. doi: 10.1093/ejcts/ezs174
52. Filosso PL, Yao X, Ahmad U, Zhan Y, Huang J, Rufni E, et al. Outcome of
Primary Neuroendocrine Tumors of the Thymus: A Joint Analysis of the
International Thymic Malignancy Interest Group and the European Society of
Thoracic Surgeons Databases. J Thorac Cardiovasc Surg (2015) 149(1):1039.
doi: 10.1016/j.jtcvs.2014.08.061
53. Dham A, Truskinovsky AM, Dudek AZ. Thymic Carcinoid Responds to
Neoadjuvant Therapy With Sunitinib and Octreotide: A Case Report. J Thorac
Oncol (2008) 3(1):947. doi: 10.1097/JTO.0b013e31815eb7a2
Conict of Interest: The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be construed as a
potential conict of interest.
Publishers Note: All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their afliated organizations, or those of
the publisher, the editors and the reviewers. Any product that may be evaluated in
this article, or claim that may be made by its manufacturer, is not guaranteed or
endorsed by the publisher.
Copyright © 2021 Lania, Ferraù, Rubino, Modica, Colao and Faggiano. This is an
open-access article distributed under the terms of the Creative Commons Attribution
License (CC BY). The use, distribution or reproduction in other forums is permitted,
provided the original author(s) and the copyright owner(s) are credited and that the
original publication in this journal is cited, in accordance with accepted academic
practice. No use, distribution or reproduction is permitted which does not comply with
these terms.
Lania et al. Neoadjuvant Therapy for NENs
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6514388
... The use of PRRT has been also evaluated in a neoadjuvant setting, but data are scanty, especially in pulmonary NETs. PRRT seems to be useful as neoadjuvant in some pNETs, but long-term data are not available [114]. ...
... Neoadjuvant therapies, including both radiation and chemotherapy alone or in combination, have been utilized in several studies to downstage tumors, with the aim of improving R0 surgical margin rates and controlling disease progression when surgery is not feasible [63,64] [64][65][66][67]. CAPTEM is currently the most widely used regimen, demonstrating efficacy in both progression-free and overall survival in a recent systematic review and meta-analysis [68]. ...
Article
Full-text available
Simple Summary Pancreatic neuroendocrine tumors (PNETs) represent a small portion of pancreatic neoplasms and can manifest as either functional or non-functional. Managing these tumors presents a challenge due to their rarity and heterogenous tumor biology. This manuscript offers a comprehensive review of PNET types, classification, and management strategies, with a focus on the evolving landscape of pancreatic surgery. It aims to assist clinicians in facilitating the management of these increasingly common tumors and optimizing patient outcomes. Abstract The incidence of pancreatic neuroendocrine tumors (PNETs) is on the rise primarily due to the increasing use of cross-sectional imaging. Most of these incidentally detected lesions are non-functional PNETs with a small proportion of lesions being hormone-secreting, functional neoplasms. With recent advances in surgical approaches and systemic therapies, the management of PNETs have undergone a paradigm shift towards a more individualized approach. In this manuscript, we review the histologic classification and diagnostic approaches to both functional and non-functional PNETs. Additionally, we detail multidisciplinary approaches and surgical considerations tailored to the tumor’s biology, location, and functionality based on recent evidence. We also discuss the complexities of metastatic disease, exploring liver-directed therapies and the evolving landscape of minimally invasive surgical techniques.
... Improved understanding of pathological mechanisms in NENs remains a crucial issue, considering the steadily increasing incidence and prevalence of these neoplasms. Furthermore, the identification of the best treatment at the right time for any individual patient is a compelling issue in NENs and a deep knowledge of pathogenetic mechanisms will be essential to improve treatment strategies, as well as to identify new therapeutic targets [4,[161][162][163]. ...
... We could have missed interesting studies by excluding those that included patients with mucinous, signet-ring cell, or neuroendocrine tumors. However, these tumors may respond differently to neoadjuvant therapy than standard adenocarcinomas because of distinct tumor biology [85][86][87][88], which would have led to more heterogeneous results. Moreover, by excluding radiomics-based and image-based deep learning models, we might have missed potential studies that combine these models with predictors from other categories. ...
Article
Full-text available
Simple Summary Organ preservation strategies can be offered to patients with rectal cancer that show a strong response to preoperative treatment in order to avoid major surgery. Prediction models may help identify these patients before the start of preoperative treatment, when treatment can still be adapted. We systematically reviewed validated pretreatment prediction models for response to preoperative treatment in patients with rectal cancer. Sixteen studies were included in this review. All studies were considered to have a high risk of bias and external validation was missing. Nevertheless, some studies show promising results, which could serve as a foundation for future research. Our systematic review provides a comprehensive overview of the current state of the literature regarding pretreatment prediction models for response to preoperative treatment in patients with rectal cancer. Abstract Pretreatment response prediction is crucial to select those patients with rectal cancer who will benefit from organ preservation strategies following (intensified) neoadjuvant therapy and to avoid unnecessary toxicity in those who will not. The combination of individual predictors in multivariable prediction models might improve predictive accuracy. The aim of this systematic review was to summarize and critically appraise validated pretreatment prediction models (other than radiomics-based models or image-based deep learning models) for response to neoadjuvant therapy in patients with rectal cancer and provide evidence-based recommendations for future research. MEDLINE via Ovid, Embase.com, and Scopus were searched for eligible studies published up to November 2022. A total of 5006 studies were screened and 16 were included for data extraction and risk of bias assessment using Prediction model Risk Of Bias Assessment Tool (PROBAST). All selected models were unique and grouped into five predictor categories: clinical, combined, genetics, metabolites, and pathology. Studies generally included patients with intermediate or advanced tumor stages who were treated with neoadjuvant chemoradiotherapy. Evaluated outcomes were pathological complete response and pathological tumor response. All studies were considered to have a high risk of bias and none of the models were externally validated in an independent study. Discriminative performances, estimated with the area under the curve (AUC), ranged per predictor category from 0.60 to 0.70 (clinical), 0.78 to 0.81 (combined), 0.66 to 0.91 (genetics), 0.54 to 0.80 (metabolites), and 0.71 to 0.91 (pathology). Model calibration outcomes were reported in five studies. Two collagen feature-based models showed the best predictive performance (AUCs 0.83–0.91 and good calibration). In conclusion, some pretreatment models for response prediction in rectal cancer show encouraging predictive potential but, given the high risk of bias in these studies, their value should be evaluated in future, well-designed studies.
Chapter
Neuroendocrine neoplasms (NEN) represent a rare and heterogeneous group of malignancies, whose incidence is increasing trough the last decades, both because of better detection strategies and of a growing awareness. The aim of biochemical, radiological, and nuclear medicine improved techniques is to early identify and characterize these tumors, in order to offer new and tailored treatments. An updated overview of diagnosis, classification, and treatments in patients with NEN is provided, with the aim to summarize recent evidence in the field of NEN. Interesting novelties in pathological examination with new immunohistochemical panels, radiological and functional imaging techniques, as well as the latest published guidelines in NEN are discussed, with focus on therapeutic strategies according to primary. This concise but thorough examination of the current advancements in NEN aims to represent a useful tool for clinicians and researchers working in this field. NEN classification and characterization have changed trough years, aiming to obtain a precise tumor profile and the best therapeutic strategy. NEN diagnosis and treatment should be discussed by expert multidisciplinary group to improve management and survival rates.
Article
Full-text available
Background The finding of pancreatic cystic lesions (PCL) on incidental imaging is becoming increasingly common. International studies report a prevalence of 2.2–44.7% depending on the population, imaging modality and indication for imaging, and the prevalence increases with age. Patients with PCL are at risk of developing pancreatic cancer, a disease with a poor prognosis. This publication summarizes recommendations for the diagnosis and management of PCL and post-operative pancreatic exocrine insufficiency (PEI) from a group of local specialists. Methods Clinical evidence was consolidated from narrative reviews and consensus statements formulated during two online meetings in March 2022. The expert panel included gastroenterologists, hepatobiliary surgeons, oncologists, radiologists, and endocrinologists. Results Patients with PCL require careful investigation and follow-up due to the risk of malignant transformation of these lesions. They should undergo clinical investigation and pancreas-specific imaging to classify lesions and understand the risk profile of the patient. Where indicated, patients should undergo pancreatectomy to excise PCL. Following pancreatectomy, patients are at risk of PEI, leading to gastrointestinal dysfunction and malnutrition. Therefore, such patients should be monitored for symptoms of PEI, and promptly treated with pancreatic enzyme replacement therapy (PERT). Patients with poor response to PERT may require increases in dose, addition of a proton pump inhibitor, and/or further investigation, including tests for pancreatic function. Patients are also at risk of new-onset diabetes mellitus after pancreatectomy; they should be screened and treated with insulin if indicated. Conclusions These statements are an accurate summary of our approach to the diagnosis and management of patients with PCL and will be of assistance to clinicians treating these patients in a similar clinical landscape.
Article
Objectives To test the performance of the National Comprehensive Cancer Network (NCCN) CT resectability criteria for predicting the surgical margin status of PNET and to identify factors associated with margin-positive resection Methods 80 patients with preoperative CT and upfront surgery were retrospectively enrolled. Two radiologists assessed the CT resectability (resectable [R], borderline resectable [BR], unresectable [UR]) of the PNET according to NCCN criteria. Logistic regression was used to identify factors associated with resection margin status. κ statistics were used to evaluate inter reader agreements. Kaplan–Meier method with log-rank test was used to estimate and compare recurrence-free survival (RFS). Results 45 patients (56.2%) received R0 resection and 35 (43.8%) received R1 or R2 resection. R0 resection rates were 63.6–64.2%, 20.0–33.3%, and 0% for R, BR, and UR diseases, respectively (all p ≤ 0.002), with a good inter reader agreement (κ, 0.74). Tumor size (< 2 cm, 2–4 cm, and >4 cm; odds ratio (OR), 9.042–18.110; all p ≤ 0.007) and NCCN BR/UR diseases (OR, 5.918; p = 0.032) were predictors for R1 or R2 resection. The R0 resection rate was 91.7% for R disease <2 cm and decreased for larger R disease. R0 resection and smaller tumor size in R disease improved RFS. Conclusions NCCN resectability criteria can stratify patients with PNET into distinct groups of R0 resectability. Adding tumor size to R disease substantially improves the prediction of R0 resection, especially for PNETs < 2 cm. Advances in knowledge Tumor size and radiologic resectability independently predicted margin status of PNETs.
Article
This ENETS guidance paper, developed by a multidisciplinary working group, provides an update on the previous colorectal guidance paper in a different format. Guided by key clinical questions practical advice on the diagnosis and management of neuroendocrine tumours (NET) of the caecum, colon, and rectum is provided. Although covered in one guidance paper colorectal NET comprises a heterogeneous group of neoplasms. The most common rectal NET are often small G1 tumours that can be treated by adequate endoscopic resection techniques. Evidence from prospective clinical trials on the treatment of metastatic colorectal NET is limited and discussion of patients in experienced multidisciplinary tumour boards strongly recommended. Neuroendocrine carcinomas (NEC) and mixed neuroendocrine non-neuroendocrine neoplasms (MiNEN) are discussed in a separate guidance paper.
Article
Full-text available
Esophageal squamous cell carcinoma (ESCC) is one of the most common malignancies worldwide, especially in East Asia. ESCC accounts for more than 90% of esophageal cancer. Currently, neoadjuvant therapy in combination with surgical resection is the mainstay of treatment. However, the overall survival rate of patients with locally advanced ESCC is not satisfactory even when treated following the standard treatment guidelines. With neoadjuvant chemoradiotherapy, chemotherapy, or emerging immunotherapy, continuous exploration of efficacy in relation to ESCC is expected to improve overall survival further. Here, we review and summarize current evidence for efficacy of preoperative therapy for locally advanced ESCC.
Article
Full-text available
Objective: The primary aims of this study were to evaluate the long-term outcome of a "sandwich chemo-PRRT (SCPRRT)" regimen with regard to therapeutic response rate, progression-free survival (PFS), and overall survival (OS) rates in metastatic neuroendocrine tumors (NETs) with both somatostatin receptor (SSTR)- and fluorodeoxyglucose (FDG)-avid aggressive disease. Additionally, health-related quality of life (HRQoL) scales, clinical toxicity, and association of PFS and disease control rate (DCR) with various variables were also evaluated. Materials and methods: A total of 38 patients of the aforementioned cohort, who received SCPRRT (at least 2 cycles of each PRRT and chemotherapy) at our institute between January 2012 and December 2018, were included and analyzed in this retrospective study. Between two cycles of 177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT), two cycles of oral capecitabine and temozolomide (CAPTEM) were sandwiched. Therapeutic responses following SCPRRT were assessed by using pre-defined criteria. PFS and OS after first SCPRRT were determined. Eastern Cooperative Oncology Group (ECOG) and Karnofsky score were used for evaluation of HRQoL before and after SCPPRT in all 38 patients. Any adverse events were graded according to the Common Terminology Criteria for Adverse Events version 5.0 (CTCAE v5.0) of the National Cancer Institute. Associations of PFS and DCR with various variables were evaluated. Results: Response (complete response and partial response) to SCPRRT was seen in 28 patients (73%), 15 patients (39%), and 16 patients (42%) on symptomatic, biochemical, and molecular imaging response evaluation criteria respectively. A total of 17 patients (45%) had anatomical imaging response with DCR of 84% based upon the RECIST 1.1 criteria. Pre-therapy mean ECOG and KPS was 2.0 and 68, which changed to 1.0 and 75 respectively following SCPRRT. Long-term follow-up data was available and ranged from 12 to 65 months after the first SCPRRT. Median PFS and OS were not reached at a median follow-up of 36 months. An estimated PFS rate of 72.5% and OS rate of 80.4% was found at 36 months. Longer PFS was dependent upon high SSTR uptake and number of CAPTEM cycle (≥ 7 cycles), absence of skeletal metastasis, and no previous external beam radiotherapy (EBRT) exposure with significant P value. Higher DCR was dependent upon absence of skeletal metastasis with significant P value. SCPRRT was tolerated well with none developing grade 4 hematotoxicity and nephrotoxicity of any grade. Anemia (grade 3), thrombocytopenia (grade 3), and leukopenia (grade 3) were noticed in 1 patient (2.5%), 2 patients (5%), and 1 patient (2.5%) respectively in this study. Conclusion: Thus, favorable response rates with effective control of symptoms and longer PFS and OS without high-grade or life-threatening toxicities were important observations in the present study following SCPRRT in NET patients with aggressive, both FDG- and SSTR-avid, metastatic progressive disease. The study results indicate the potential role of "sandwich chemo-PRRT" in future therapeutic algorithms of aggressive, both SSTR- and FDG-positive subset of neuroendocrine tumors.
Article
Full-text available
The role of primary tumor resection in patients with pancreatic neuroendocrine neoplasms (PanNENs) and unresectable distant metastases remains controversial. We aimed to evaluate the effect of palliative primary tumor resection (PPTR) on overall survival (OS) in this setting. We searched the MEDLINE, Embase, Cochrane Library, Web of Science and SCOPUS databases up to January 2020 and used the Newcastle–Ottawa scale (NOS) criteria to assess quality/risk of bias. A total of 5661 articles were screened. In 10 studies, 5551 unique patients with stage IV PanNEN and unresectable metastases were included. The five-year OS for PanNEN patients undergoing PPTR in stage IV was 56.6% vs. 23.9% in the non-surgically treated patients (random effects relative risk (RR): 1.70; 95% CI: 1.53–1.89). Adjusted analysis of pooled hazard ratios (HR) confirmed longer OS in PanNEN patients undergoing PPTR (random effects HR: 2.67; 95% CI: 2.24–3.18). Cumulative OS analysis confirmed an attenuated survival benefit over time. The complication rate of PPTR was as high as 27%. In conclusion, PPTR may exert a survival benefit in stage IV PanNEN. However, the included studies were subject to selection bias, and special consideration should be given to PPTR anchored to a multimodal treatment strategy. Further longitudinal studies are warranted, with long-term follow-up addressing the survival outcomes associated with surgery in stage IV disease.
Article
Full-text available
Triple negative breast cancer (TNBC) is an aggressive breast cancer with historically poor outcomes, primarily due to the lack of effective targeted therapies. The tumor molecular heterogeneity of TNBC has been well recognized, yet molecular subtype driven therapy remains lacking. While neoadjuvant anthracycline and taxane-based chemotherapy remains the standard of care for early stage TNBC, the optimal chemotherapy regimen is debatable. The addition of carboplatin to anthracycline, cyclophosphamide, and taxane (ACT) regimen is associated with improved complete pathologic response (pCR). Immune checkpoint inhibitor (ICI) combinations significantly increase pCR in TNBC. Increased tumor infiltrating lymphocyte (TILs) or the presence of DNA repair deficiency (DRD) mutation is associated with increased pCR. Other targets, such as poly-ADP-ribosyl polymerase inhibitors (PARPi) and Phosphatidylinositol-3-kinase/Protein Kinase B/mammalian target of rapamycin (PI3K-AKT-mTOR) pathway inhibitors, are being evaluated in the neoadjuvant setting. This review examines recent progress in neoadjuvant therapy of TNBC, including platinum, ICI, PARPi, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) pathway targeted therapies, and novel tumor microenvironment (TME) targeted therapy, in addition to biomarkers for the prediction of pCR.
Article
Full-text available
Thanks to the development of modern chemotherapeutic regimens, survival after surgery for pancreatic ductal adenocarcinoma (PDAC) has improved and pancreatologists worldwide agree that the treatment of PDAC demands a multidisciplinary approach. Neoadjuvant treatment (NAT) plays a major role in the treatment of PDAC since only about 20% of patients are considered resectable at the time of diagnosis. Moreover, increasing data demonstrating the benefits of NAT for borderline resectable/locally advanced PDAC are driving a shift from up-front surgery to NAT in the multidisciplinary treatment of even resectable PDAC. Our understanding of the role of NAT in PDAC has evolved from tumor shrinkage to controlling potential micrometastases and selecting patients who may benefit from radical resection. The present review gives an overview on the current literature of NAT concepts for BR/LA PDAC and resectable PDAC.
Article
Introduction: Capecitabine-temozolomide (CAPTEM) chemotherapy, alone or with concurrent peptide receptor radionuclide therapy (PRRT), has activity in advanced WHO grade 2 and grade 3 neuroendocrine neoplasms (NENs). The objective of this study was to evaluate the activity of the CAPTEM in patients with grade 2 and grade 3 NENs and identify prognostic factors. Materials and methods: A retrospective analysis of patients with metastatic grade 2 and grade 3 NENs, who were having baseline significant dual uptake on 68Ga-DOTATATE/18F-fluorodeoxyglucose (FDG)-PET-CT scan and treated with CAPTEM chemotherapy between January 2014 and December 2019 at Tata Memorial Hospital, was conducted. The clinical variables and survival data were collected. Progression-free survival (PFS) was estimated using the Kaplan-Meier method. Results: A total of 68 patients received the CAPTEM regimen, of whom 29 patients (43%) received CAPTEM alone and 39 patients (57%) received concurrent PRRT. The primary sites were pancreas in 32 (47%) and small intestine in 12 (18%) patients. Mean Ki-67 index was 12.6% (range: 3-50). Forty-five patients (65%) were treatment naïve. There were no significant differences in baseline clinical variables between patients treated with CAPTEM alone or with CAPTEM-PRRT. Both regimens were well tolerated. With a median follow-up of 22.1 months, the median PFS for the entire cohort was 27.5 months. There was no statistical difference in the median PFS between patients receiving CAPTEM alone or CAPTEM-PRRT (33.7 vs. 22 months; p = 0.199). A Ki-67 index of >5% predicted for inferior PFS on multivariate analysis (24 versus 73.8 months; p = 0.04; hazard ratio -3.77; 95% confidence interval: 1.07-13.26). Conclusion: CAPTEM, alone or concurrent with PRRT, has a significant activity in grade 2 and grade 3 NENs with dual SSTR and 18FDG expression. A Ki-67 index >5% predicts strongly for inferior outcomes and should be further explored as a prognostic cutoff in grade 2 NENs. Early initiation of CAPTEM should be considered in this group of tumors with significant baseline 18FDG expression.
Article
At the time of diagnosis, only about 20% of patients with pancreatic ductal adenocarcinoma (PDAC) have resectable disease. PDAC treatment necessitates a multidisciplinary approach, and adjuvant chemotherapy after upfront resection is an established means of preventing recurrence. Neoadjuvant chemotherapy (NAT), originally introduced to downstage tumor size, is nowadays more frequently used for selection of patients with favorable tumor biology and to control potential micrometastases. While NAT is routinely applied in locally advanced (LA) PDAC, there is increasing evidence demonstrating benefits of NAT in borderline resectable (BR) PDAC. The concept of NAT has recently been tested in resectable PDAC, but to date NAT has been restricted to clinical trials, as the data are limited and no clear benefits have yet been shown in this patient group. This review summarizes the current evidence for NAT in resectable, BR, and LA PDAC, with a focus on high-level evidence and randomized controlled trials.
Article
Background: The role of perioperative systemic therapy (PST) in the management of localized pancreatic neuroendocrine neoplasms (PanNEN) is unclear. Objectives: We aimed to evaluate the benefit of PST compared to surgery alone (SA) in patients with localized PanNEN. Method: We selected patients with stages I-III PanNEN who underwent curative-intent surgical resection in National Cancer Database from 2006 to 2014. Patients who had both PST and surgical resection were matched with patients who received SA by propensity score at 1-to-1 ratio with nearest neighbor method. Results: Four thousand eight hundred and ninety-two patients were included in this study with median age of 60 years. Factors associated with significant more use of PST compared to SA included age <65 years, community medical facilities, grade 3 tumor, tumor in the pancreatic head, T34 tumor, and N1 tumor. Three hundred and one PST patients were matched with 301 SA patients. In the matched cohort, the PST group had significantly shorter overall survival (OS) compared to the SA group (median overall both not reached, p = 0.037). This finding was confirmed by multivariable Cox proportional hazards regression in the original cohort (hazard ratio [HR] 1.45, 95% CI 1.11-1.89, p = 0.006). Subgroup analyses showed that adjuvant therapy was not associated with improved OS in grades 1-2 PanNEN (HR 2.03, 95% CI 1.31-3.16, p = 0.002). Conclusions: PST stratified by grade and neoadjuvant or adjuvant therapy compared to SA was not associated with improved OS in patients with localized PanNEN. PST for localized PanNEN should be used with caution until prospective data are available.
Article
Background: Large-cell neuroendocrine carcinoma (LCNEC) and small cell lung cancer (SCLC) are categorized as high-grade neuroendocrine carcinoma (HGNEC). We analyzed the efficacy of perioperative chemotherapy for HGNEC and the prognostic factors. Methods: We retrospectively reviewed the medical records of patients who underwent tumor resection and were diagnosed with HGNEC between January 2001 and December 2014. The overall survival (OS) was estimated by the Kaplan-Meier method. Propensity score matching was performed to compare the OS between the treatment groups. Multivariate analyses using a Cox proportional hazards model were performed to search for prognostic factors for HGNEC. Results: We analyzed 146 HGNEC patients (LCNEC n=92, SCLC n=54) without synchronous multiple cancers, who underwent complete resection. Seventy patients (LCNEC n=31, SCLC n=32) received perioperative chemotherapy and all of them received a platinum-based anticancer drug. Perioperative chemotherapy significantly improved the 5-year OS rates of HGNEC patients (all stages: 74.5% vs. 34.7%, P<0.01, stage I: 88.5% vs. 40.0%, P<0.01). The efficacy of perioperative chemotherapy was similar between LCNEC and SCLC patients [LCNEC all stages: hazard ratio (HR) 0.27, P<0.01, LCNEC stage I: HR 0.27, P=0.01; SCLC all stages: HR 0.38, P=0.02, SCLC stage I: HR 0.34, P=0.06]. The survival benefit of perioperative chemotherapy for HGNEC patients was confirmed by propensity score matching analysis (HR 0.31, P<0.01). The multivariate analysis revealed that perioperative chemotherapy (HR 0.29, P<0.01), sublobar resection (HR 2.11, P=0.04), and lymph node metastasis (HR 3.34, P<0.01) were independently associated with survival. Conclusions: Surgical resection combined with perioperative chemotherapy was considered to be effective even for stage I HGNEC patients. Sublobar resection might increase the risk of death in HGNEC patients.
Article
Lung large-cell neuroendocrine carcinoma (L-LCNEC) is a rare subset of lung carcinoma associated with poor overall survival. Due to its rarity, little has been established about its optimal treatment in the advanced stage. We report the case of a 41-year-old woman diagnosed with an unresectable locally advanced L-LCNEC who presented an impressive tumor response to immunotherapy with nivolumab after non-curative thoracic radiotherapy. Salvage surgery was then performed, and pathologic analysis of the resected piece revealed the absence of residual viable tumor cells. Based on this case report, we discuss the literature regarding the efficacy of inhibitors of programmed death-1 protein (PD-1) in L-LCNEC and their use in association with radiotherapy and in the neoadjuvant setting.