Update on thymic epithelial tumors: a narrative review
Review Article

Update on thymic epithelial tumors: a narrative review

Luis Cabezón-Gutiérrez1,2 ORCID logo, Vilma Pacheco-Barcia1, Fátima Carrasco-Valero3, Magda Palka-Kotlowska1, Sara Custodio-Cabello1, Parham Khosravi-Shahi4

1Medical Oncology, Torrejón University Hospital, Madrid, Spain; 2Faculty of Medicine, Francisco de Vitoria University, Madrid, Spain; 3Internal Medicine, Torrejón University Hospital, Madrid, Spain; 4Medical Oncology, Gregorio Marañón University Hospital, Madrid, Spain

Contributions: (I) Conception and design: L Cabezón-Gutiérrez, V Pacheco-Barcia; (II) Administrative support: L Cabezón-Gutiérrez, V Pacheco-Barcia, M Palka-Kotlowska; (III) Provision of study materials or patients: L Cabezón-Gutiérrez, F Carrasco-Valero; (IV) Collection and assembly of data: L Cabezón-Gutiérrez, V Pacheco-Barcia, F Carrasco-Valero; (V) Data analysis and interpretation: L Cabezón-Gutiérrez, V Pacheco-Barcia, S Custodio-Cabello; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Luis Cabezón-Gutiérrez, MD, PhD. Medical Oncology, Torrejón University Hospital, Calle Mateo Inurria Nº1, Madrid, Spain; Faculty of Medicine, Francisco de Vitoria University, Ctra. Pozuelo-Majadahonda KM 1.800, 28223 Pozuelo de Alarcón, Madrid, Spain. Email: lcabezon@torrejonsalud.com.

Background and Objective: Thymoma, thymic carcinoma and thymic neuroendocrine tumors originate from the epithelial cells of the thymus and account for the thymic epithelial tumors (TETs). Although TETs are uncommon, they are the most frequent tumor type in the anterior mediastinum. Multidisciplinary approach is essential for their correct management. The aim of the present review is to summarize the update management for TETs.

Methods: For this review, we searched in Excerpta Medica database (EMBASE) and MEDLINE until 6 September 2023. The terms used in the search included thymoma, thymic carcinoma, thymic epithelial tumors, management, immunotherapy, multiple tyrosine kinases inhibitors.

Key Content and Findings: The therapeutic approach is based on histology and tumor stage and may involve surgery with or without neoadjuvant or adjuvant treatment. In the metastatic setting, platinum-based chemotherapy is the standard of care and patients who do not respond to first-line treatment have limited treatment options mainly because of the poor efficacy shown in subsequent lines of therapy.

Conclusions: Future research should focus on identifying predictive biomarkers for patients with TETs, and should implement multicenter collaborations and appropriate clinical trials tailored for rare tumor types. Immune check point inhibitors, mammalian target of rapamycin (mTOR) and antiangiogenic multikinase inhibitors have also been studied in this clinical setting.

Keywords: Thymoma; thymic carcinoma; thymic epithelial tumors (TETs); thymic neuroendocrine tumors and management


Received: 11 October 2023; Accepted: 14 January 2024; Published online: 26 April 2024.

doi: 10.21037/med-23-47


Introduction

Thymic epithelial tumors (TETs) include thymomas, thymic carcinomas, and neuroendocrine tumors of the thymus (NETTs). Although their incidence is low, they are the most common tumors of the anterior mediastinum (1).

The most common subgroup of TET is thymoma, which represents almost 50%, followed by thymic carcinoma (14–22%) and NETTs (2–5%) (1).

Epidemiologically, even though the distribution by age is quite similar, there is a slightly higher incidence in patients over 50 years of age (2), with the mean age at diagnosis being 50–60 years. The incidence by gender is similar, prevailing in men. The frequency of metastasis at diagnosis is higher in thymic carcinoma and NETTs than in thymomas (2), due to their more aggressive behavior with a greater tendency to disseminate systemically (1,3). Risk of developing a secondary malignancy is increased in this population, especially patients with thymomas. This is possibly due to treatments for their primary malignancy which includes radiotherapy (2).

In terms of survival, thymoma has an overall 5-year survival of approximately 78% (1). Thymic carcinomas and NETTs, as they are more aggressive entities with worse prognosis, the 5-year survival is 30% and 23%, respectively (1-4). No differences in prognosis have been observed between men and women in TETs (2). It has been observed that prognosis may be affected by histology, stage and the presence or absence of paraneoplastic syndromes (1).

This review aims to summarize the existing literature regarding the management for TETs. We present this article in accordance with the Narrative Review reporting checklist (available at https://med.amegroups.com/article/view/10.21037/med-23-47/rc).


Methods

For this review, we searched EMBASE and MEDLINE until 6 September 2023. The search strategy is described in Table 1. The terms used in the search included thymoma, thymic carcinoma, thymic epithelial tumors, management, immunotherapy, multiple tyrosine kinases inhibitors. One of the main methodological limitations that we found when conducting the literature search and in the preparation of this manuscript is the lack of randomized clinical trials in this type of rare tumors. Many of the articles included are older, reviews or retrospective case series.

Table 1

The search strategy summary

Items Specification
Date of search 6th September 2023
Databases and other sources searched MEDLINE, EMBASE
Search terms used Keywords: thymoma, thymic carcinoma, thymic neuroendocrine tumors, thymic epithelial tumors (TET), TET management, TET immunotherapy, TET multiple tyrosine kinases inhibitors
Timeframe January 1, 1950 to September 6, 2023
Inclusion and exclusion criteria Inclusion: (I) English and Spanish language; (II) case reports, case series, retrospective cohort series, prospective studies; (III) focusing on subtopics of histology and diagnosis
Exclusion: extra-thoracic tumors
Selection process L.C.G., V.P.B. and F.C.V. selected literature, all authors chose those for inclusion

Clinical and diagnostic

For the diagnostic management, we must consider the clinical presentation and the findings in the diagnostic tests, being of special interest the radiological and histopathological findings (1).

Concerning the clinical presentation, about 33% of patients with thymic tumors are asymptomatic at the time of diagnosis. In those patients who are symptomatic, 40% present with symptoms related to intrathoracic mass compression (chest pain, cough, hoarseness, superior vena cava syndrome or dyspnea), 30% present with neurological symptoms and 30% present with systemic symptoms (weight loss, night sweats or fever), which make them difficult to differentiate from lymphoma (2).

In thymic carcinomas, the usual clinical presentation is as described above, with no more frequent associations with other entities (5). However, in NETTs, in addition to the symptoms described, 50% are functionally active and can be associated with endocrinopathies, with up to 40% presenting associated Cushing’s syndrome, or less frequently, multiple endocrine neoplasia (MEN) I in 19 to 25% (3). The association of MEN type IIA with NETT is an unusual presentation, known in a few cases, and considered a variant of Sipple’s syndrome (described as incomplete Sipple’s syndrome) (6,7).

The most frequent thymoma’s association is myasthenia gravis (1), present in up to 82% (8), in contrast to non-thymoma TETs, where the association with myasthenia gravis is exceedingly rare (9).

Preoperative diagnosis of these thymic masses can be complex, but currently, imaging tests are available to assist in this process (10).

Despite the fact that chest radiography is used for the initial study to confirm the presence of a thymic mass (1), the most useful and frequently used diagnostic test is contrast enhanced computed tomography (CT). CT scans provide information on clinical tumor local stage with an evaluation of the organs and structures adjacent to it (1). It also provides information on the presence of pleural parietal deposits (also called “droplet metastases”), as well as on the density characteristics of the thymic neoplasm. The identification of areas whose density is different from thymoma, such as hemorrhage, calcification or necrosis, provides relevant information for staging-for instance, the presence of calcifications suggesting B1, B2, and B3 types of thymoma (2).

Thymomas frequently appear as well-defined rounded masses located anterior to the great vessels and heart. In contrast, thymic carcinoma is characterized by irregular margins and associated lymph nodes (2). In the case of NETTs, a lobulated thymic mass with heterogeneous enhancement and central areas of decreased attenuation secondary to areas of necrosis or hemorrhage are observed (3).

Magnetic resonance imaging (MRI) is usually reserved for cases where iodinated contrasts cannot be administered or to examine for the presence of cystic lesions or areas of local invasion (1,2). Fluorodeoxyglucose (FDG) positron emission tomography (PET)/CT may be considered for thymic carcinoma, given the high metabolism of this tumor, for the detection of occult metastases (1) or to characterize lesions suspicious for recurrence (5).

NETTs exhibit an overexpression of somatostatin receptors (SSTRs) on their cell membrane. Imaging techniques targeting SSTRs, such as 68Ga-DOTATOC/DOTATATE-PET, are employed to identify hormonally active tumors of this nature and devise suitable therapeutic strategies. This holds significance as the verification of receptor affinity through diagnostic imaging serves as a crucial determinant of the potential for peptide receptor radionuclide therapy (PRRT). PRRT, involving the use of receptor agonists or antagonists within the context of a theranostic approach, has gained widespread acceptance as an effective treatment modality for neuroendocrine neoplasms (NENs) since its introduction (11). While 68Ga, a positron emitter radionuclide, is exclusively utilized for diagnostic imaging, 90Yttrium-DOTA octreotide and 177-Lutetium DOTA octreotide are the most commonly employed regimens for PRRT (12).


Pathology

At the histological level, moderate atypicality with little associated mitosis and immature T-cell lymphocytes can be observed in thymoma. Vascular invasion and necrosis are usually absent. In contrast to thymomas, the histology of thymic carcinomas is characterized by marked atypicality, frequent mitosis, mature T- and B-cells and vascular invasion and necrosis. Immunohistochemically, thymoma is c-KIT (CD117) negative whereas in thymic carcinoma, in 60–80% of cases, epithelial cells are c-KIT positive, with frequent CD5-associated expression (13).

NETTs differ from the two previously described entities by the presence of elongated tumor cells, pleomorphic nuclei and arrangement in small, rosette-like acinar structures. Immunohistochemically, they are characterized by positivity for markers such as cytokeratin, Leu-7, synaptophysin and cytoplasmic chromogranin stain. Among the latter markers, TTF-1 positivity is noteworthy (3).

It is necessary to compare the histopathological differences among these tumor types, as they contribute to the postoperative diagnosis. This is imperative in scenarios where clinical presentation and imaging findings are merely suggestive (2).

Table 2 describes the histological classification for thymoma and Table 3, the histological classification for thymic carcinoma (1,14). NETTs are histologically classified into three categories: low grade (well differentiated), intermediate grade (moderately undifferentiated) and high grade (poorly differentiated) with well-differentiated (WD) carcinomas being the most frequent (3).

Table 2

Histological classification for thymoma according to the World Health Organization (14)

Histological subtypes of thymoma Obligatory criteria Optional criteria
Subtype A Occurrence of bland, spindle shaped epithelial cells Polygonal epithelial cells CD20+
Paucity or absence of immature T cells Epithelial cells
Atypical subtype A variant Criteria of type A with comedo-type tumor necrosis
Elevated mitotic count, nuclear crowding
Subtype AB Occurrence of bland, spindle shaped epithelial cells
Profusion of immature T cells
Subtype B
   Subtype B1 Thymus-like architecture and cytology Hassall’s corpuscles
Profusion of immature T cells with areas of medullary differentiation Perivascular spaces
Paucity of polygonal or dendritic epithelia cells without clustering
   Subtype B2 Elevated numbers of single or clustered polygonal or dendritic epithelial cells intermingled Criteria of type B1
Profusion of immature T cells Medullary islands
   Subtype B3 Sheets of polygonal slightly to moderately atypical epithelial cells Hassall’s corpuscles
Absent or rare intercellular bridges Perivascular spaces
Paucity or absence of intermingled T cells
Micronodular thymoma (MNT) with lymphoid stroma Nodules of bland spindle or oval epithelial cells surrounded by an epithelial cell-free lymphoid stroma Lymphoid follicles
Monoclonal B cells and/or plasma cells
Metaplastic thymoma Biphasic tumor formed of solid areas of epithelial cells in a background of bland-looking spindle cells Pleomorphism of epithelial cells
Absence of immature T cells Actin, keratin, or EMA-positive spindle cells
Other subtypes
   Microscopic thymoma Occurrence of bland, spindle shaped epithelial cells
   Sclerosing thymoma Paucity or absence of immature T cells
   Lipofibroadenoma

EMA, epithelial membrane antigen.

Table 3

Histological classification for thymic carcinoma according to the World Health Organization (14)

Histological subtypes of thymic carcinoma:
   Adenocarcinoma
    Adenocarcinoma, NOS
    Low grade papillary adenocarcinoma
    Thymic carcinoma with adenoid cystic carcinoma-like features
    Adenocarcinoma, enteric-type
   Squamous carcinoma
    Squamous cell carcinoma, NOS
    Basaloid carcinoma
    Lymphoepithelial carcinoma
   Adenosquamous carcinoma
    Salivary gland-like carcinoma
      Mucoepidermoid carcinoma
      Clear cell carcinoma
      Sarcomatoid carcinoma
      Carcinosarcoma
   Carcinoma undifferentiated, NOS
   Thymic carcinoma, NOS
   NUT carcinomas

NOS, not otherwise specified; NUT, nuclear protein in testis.

Despite the fact that there are relatively few studies on tumor mutational burden (TMB) in TETs, it is necessary to elucidate its role in this type of tumors. TMB stands as an indirect indicator of the ability and extent of tumors to produce new antigens, which is correlated to the suitability for immunotherapy (15).

Some of the most mutated genes in TETs are GTF2I, HRAS, TTN and TP53. GTF2I has been described as the predominant mutation in TETs, particularly in the case of the comparatively indolent type A and AB thymomas. However, its incidence is notably infrequent in the more aggressive types B and C. Patients with GTF2I mutations exhibit a more favorable prognosis, potentially attributable to their prevalence in relatively less aggressive subtypes (16). Comparing to thymomas, the incidence of GTF2I is decreased in thymic carcinomas (16).


Differential diagnosis and staging

The differential diagnosis of these tumors should be made primarily with: (I) lymphomas (both Hodgkin’s and non-Hodgkin’s), the most common, (II) extragonadal germ cell tumors and (III) metastatic carcinomas that may involve the mediastinum (1). In order to provide a differential diagnosis, the patient’s age, sex, clinical features and CT images should be considered (1,2).

One of the main differential diagnoses for TETs is lymphoma, however patients presenting with lymphoma tend to be younger compared to those with TET. They usually have constitutional symptoms, different from parathymic syndromes, such as night sweats, fever, weight loss and malaise. In contrast to TET, the physical examination of a patient with lymphoma may reveal lymphadenopathy.

There are several staging systems for this group of tumors; however, the most commonly used are the TNM 8th edition and the Masaoka-Koga staging system. Tables 4–6 describe these systems, respectively (1,2).

Table 4

TNM 8th edition staging system for TETs (1)

Primary tumor (T)
   TX: primary tumor cannot be assessed
   T0: no evidence of primary tumor
   T1: tumor encapsulated or extending into the mediastinal fat. It can involve the mediastinal pleura
      • T1a: tumor with no mediastinal pleura involvement
      • T1b: tumor with direct invasion of mediastinal pleura
   T2: tumor with direct invasion of the pericardium (either partial or full thickness)
   T3: tumor with direct invasion into any of the following: lung, brachiocephalic vein, superior vena cava, phrenic nerve, chest wall, or extrapericardial pulmonary artery or veins
   T4: tumor with invasion into any of the following: aorta (ascending, arch, or descending), arch vessel, intrapericardial pulmonary artery, myocardium, trachea, esophagus
Regional lymph nodes (N)
   NX: regional lymph nodes cannot be assessed
   N0: no regional Iymph node metastasis
   N1: metastasis in anterior (perithymic) lymph nodes
   N2: metastasis in deep intrathoracic or cervical lymph nodes
Distant metastasis (M)
   M0: no pleural, pericardial or distant metastasis
   M1: pleural, pericardial, or distant metastasis
      • M1a: separate pleural or pericardial nodule(s)
      • M1b: pulmonary intraparenchymal nodule or distant organ metastasis

TETs, thymic epithelial tumors.

Table 5

AJCC prognostic groups (1)

Stage T N M
Stage I T1a, b N0 M0
Stage II T2 N0 M0
Stage IIIA T3 N0 M0
Stage IIIB T4 N0 M0
Stage IVA Any T N0-N1 M1a
Any T N2 M0–M1a
Stage IVB Any T Any N M1b

AJCC, American Joint Committee on Cancer.

Table 6

Modified Masaoka clinical staging system for thymoma (1)

Masaoka stage Diagnostic criteria
Stage I Macroscopically and microscopically completely encapsulated
Stage II (A) Microscopic transcapsular invasion
(B) Macroscopic invasion into surrounding fatty tissue or grossly adherent to but not through mediastinal pleura or pericardium
Stage III Macroscopic invasion into neighboring organs (for example: lung, great vessels or pericardium)
(A) Without invasion of great vessels
(B) With invasion of great vessels
Stage IV (A) Pleural or pericardial dissemination
(B) Lymphogenous or hematogenous metastasis

In the most recent 2021 classification of thymic tumors by the World Health Organization (WHO), NETTs are categorized into three groups (17): low-grade typical carcinoids (TC), intermediate-grade atypical carcinoids (ACs), and two high-grade malignancies—specifically, large cell neuroendocrine carcinomas (LCNEC) and small cell carcinomas (SCC).


Localized disease

Surgical approach of thymoma and thymic carcinoma

Surgery is the main treatment strategy of patients diagnosed with thymoma and thymic carcinoma (18) and a complete surgical resection (R0) is a prognostic factor for recurrence and survival in these patients (19,20). Survival may differ according to the resection margins: complete R0 resection has an excellent prognosis, microscopic R1 has shown a 64% 10-year survival compared with macroscopic R2 that has shown a 36% 10-year survival (21-26). The best surgical approach is debatable and both an open surgery or a minimally invasive surgery could be performed on a case-by-case basis. For an open surgery, a median sternotomy can allow an extensive evaluation of mediastinal structures and surgical manipulation as well (4). If structures of the posterior mediastinum or the pulmonary hilar are infiltrated, a horizontal incision would be a better option (4).

Minimally invasive surgery could be considered for patients with early clinical stages (I-II) if a complete resection is feasible, taking into account that long-term data of the benefits of minimally invasive surgery compared to open surgery are lacking (27,28). Depending on the surgical approach, minimally invasive surgery could be divided in different categories: (I) unilateral transthoracic video-assisted thoracic surgery (VATS) thymectomy; (II) conventional subxiphoid VATS thymectomy; (III) transcervical VATS thymectomy; (IV) subxiphoid VATS thymectomy with double elevation of sternum. Open surgery has been compared with VATS thymectomy and robotic VATS (R-VATS) thymectomy and has been shown to have acceptable oncological outcomes and less perioperative complications (29,30). However, it should be taking into consideration that resecability is the first evaluation that should be performed in patients with localized disease and it is mainly based on the expertise of the surgeon (4). A thymectomy which includes the resection of the thymic tumor, residual thymus and perithymic fat is recommended (31). Furthermore, the resection of pleura, pericardium, phrenic nerve, lung and major vessels that are close to the thymus may be required as well.

Lymphadenectomy in thymic tumors is controversial and there the data on the prognostic significance is lacking. Systematic lymphadenectomy is recommended in stage II or higher, WHO histology B2/3, C, tumors >6 cm and NETTs (32). N1 could be resected with the total thymectomy but, N2 with the station R 2/4 and L 5/6 depend usually on the suspicious intraoperative findings (4). After curative therapy, if there is persistent or recurrent disease, salve surgery could be performed but the oncological outcomes are not well defined (33).


Thymoma

Resectable disease

Thymomas are classified as type A, type AB, type B1/B2/B3 in the WHO fifth edition which includes gene mutations and gene fusions (17). The Masaoka-Koga staging has been associated with survival and is based on the extension of the tumor (34). The prognostic relevance of molecular changes in thymomas has been recently highlighted as a distinctive feature and may allow future targeted treatments (35).

The symptoms and underlying autoimmune diseases that are diagnosed in patients with thymoma can have an impact in the workup required for diagnosis and in the treatment strategy. Myasthenia gravis can be present in up to 50% of patients with thymoma (36) and require an evaluation and treatment by a neurologist before a surgery can be performed because these patients have an increased surgical risk and could require a specific treatment prior to the intervention (37-39). Patients with a strong suspicion of having a resectable thymoma do not require a surgical biopsy nor a transpleural approached biopsy because there could be tumor seeding when the tumor capsule is ruptured (40).

Thymomas can invade local structures like pleura and lung but it is unlikely to spread to extrathoracic sites or lymph nodes (41,42). For patients with resected tumors the most important prognostic factor is the complete resection rate that depend on the adhesion to other structures (43): stage I and II have 10-year OS of 90% and 70%, respectively (40,44). Patients with tumors invading structures that can be resected or those patients with encapsulated tumors should be evaluated for surgery as the standard approach of resectable thymomas (45,46). A total resection of the thymus and a lymph node dissection is the most common surgical approach in patients with early stage without myasthenia gravis (47).

After surgery, clinicians should evaluate the benefit of postoperative radiotherapy (PORT) depending on the stage of thymoma and resection margins, these recommendations are summarized in Tables 7,8. Thymomas don’t usually metastasize to regional lymph nodes, therefore, extensive elective nodal radiation is not a recommendation (41-48). On the contrary, postoperative adjuvant chemotherapy has not demonstrated a benefit in the adjuvant setting (59,60).

Table 7

Postoperative radiotherapy in the adjuvant setting of thymomas, depending on margin status

Thymoma Resection margins Radiotherapy
Resected thymoma Clear/close margins 45–50 Gy
Microscopically positive resection margins 54 Gy (48,49)
Gross residual disease 60–70 Gy (1.8–2 Gy/fraction per day) (50,51)
Resected thymoma with capsular invasion R0 Can be considered
Incompletely resected thymomas Recommended (52-58)

Table 8

Postoperative radiotherapy in the adjuvant setting of thymomas, depending on tumor stage

Thymoma Resection margins Radiotherapy
Stage I
   No capsular invasion R0 Not recommended (53,57,58)
   Invasion of mediastinal fat or pleura R0 Can be considered
   Microscopic or grossly positive surgical margins R1/R2 Can be considered
Stage II May not benefit but can be considered
   Stage III thymoma Macroscopic invasion into neighboring organs Recommended

The surveillance of patients with resected thymomas should be done with a chest CT scan every 6 months during the first 2 years followed by an annual chest CT scan for a total of 10 years (61).

Potentially resectable disease

Patients with locally advanced thymomas where a complete resection is not feasible, may benefit from induction chemotherapy, surgery and PORT (62-69). Table 9 summarizes studies with a multidisciplinary approach of unresectable malignant thymomas. The preferred chemotherapy regimen for thymoma as first-line combination is the CAP regimen: cisplatin, doxorubicin, cyclophosphamide administered every 3 weeks with response rates of approximately 44% (55,70-72). However, a recent cohort study did not report differences between upfront surgery alone versus induction chemotherapy followed by surgery (76.7% vs. 77.4%, respectively, P=0.596) (63). For patients with oligometastastic disease that are diagnosed with solitary metastasis or ipsilateral pleural metastases two therapeutic approaches can be considered: (I) upfront surgery alone; (II) induction chemotherapy followed by surgery for patients with resectable disease (36,37).

Table 9

Studies with a multidisciplinary approach of unresectable malignant thymomas

Author Type of study Year Country/region N Treatment strategy Response rates and survival outcomes
Kanzaki et al. (65) Retrospective 2019 Japan 29 Preoperative CT or chemoradiotherapy + surgery 37% PR
5-year OS: 100%
10-year OS: 87%
Park et al. (63) Retrospective 2019 Korea 110 Induction CT + surgery Response rates not reported
5-year OS: 77.4% vs. 76.7% for surgery alone
Ruffini et al. (64) Retrospective 2019 Europe and United States 484 Induction CT + surgery + PORT Overall response rate: 10.8%
Note: thymic carcinoma and neuroendocrine thymic tumors included
Hassan et al. (69) Prospective 2009 Saudi Arabia 9 Induction CT (×3 cycles) + surgery + PORT + consolidation CT (×3 cycles) 77% major responses: 11% CR
4-year OS: 77%
Wright et al. (67) Retrospective 2008 United States 10 Induction CT (×2 cycles) + concurrent radiotherapy followed by surgery + postoperative CT if high risk 60% stable disease, 40% PR
5-year OS 69%
Kim et al. (68) Phase II 2004 United States 22 Induction CT (×3 cycles) + surgery + PORT + consolidation CT (×3 cycles) 77% major responses: 14% CR
5-year OS: 95%
7-year OS 79%

CT, chemotherapy; PR, partial response; OS, overall survival; PORT, postoperative radiotherapy; CR, complete response.

Neoadjuvant chemotherapy may be useful for achieving a complete R0 resection. Previous studies have reported response rates that range from 77–100% and an average R0 resection rate of 72%. One of the main controversies is that the data to recommend a multimodality approach with neoadjuvant therapy is based on small studies that could not be representative (73).


Thymic carcinoma

Resectable disease

Thymic carcinomas are infrequent tumors that harbor a worse prognosis than thymomas and can metastasize to lymph nodes and other organs (74-76). The main differences between thymomas and thymic carcinomas are based on histologic grounds because thymic carcinomas show malignant features as well as different genetic and immunohistochemical features (17,75). The standard of care of patients with resectable tumors at diagnosis is surgery. If a patient is resectable and undergoes resection the 5-year OS is 50–75% and survival rates vary according to stage: (I) stages 1 and 2: 91%; (II) stages 3 and 4: 31% (77).

As previously reported for thymomas, for most patients with thymic carcinomas the mainstay of surgery is a total resection of the thymus and a lymph node dissection (35). Tumor stage and the invasion of other structures can alter the possibility of performing a complete resection (47). In order to achieve an R0 resection, surgeons with specialized training may need to perform surgery over the pericardium and the adjacent lung parenchyma with the main goal of achieving negative margins that can impact long-term survival (47,78).

Thymic carcinomas have a higher risk of recurrence and adjuvant PORT is recommended in order to achieve local control (77). Therefore, after surgery, clinicians should evaluate the benefit of PORT depending on the stage of thymic carcinoma and the resection margins obtained: recommendations on PORT are summarized in Table 10. The benefit of PORT in thymoma and thymic neoplasms has been observed in retrospective data and is summarized in Table 11.

Table 10

Postoperative radiation in the adjuvant setting of thymic carcinomas

Thymic carcinoma Resection margins Radiotherapy
Resected thymic carcinoma Clear/close margins 45–50 Gy
Microscopically positive resection margins 54 Gy
Gross residual disease 60–70 Gy (1.8–2 Gy/fraction per day)
Resected thymic carcinoma with capsular invasion R0 Can be considered
Stage I R0 Not recommended

Table 11

Retrospective data on PORT for thymic neoplasms

Study Year Country N Stage (Masaoka) Thymic neoplasm Survival outcomes
Jackson et al. (79) 2017 United States 4,000 Any stage Thymoma ↑OS (HR 0.72, 95% CI: 0.59–0.87), not significant, for stage IIB o III or positive margins
No benefit of PORT in stage I or IIA
Thymic carcinoma ↑OS (HR 0.79, 95% CI: 0.64–0.97), not significant
Boothe et al. (80) 2016 United States 1,156 II and III Thymic malignancies ↑5-year OS after PORT (83% vs. 79%, P=0.03)
Rimner et al. (81) 2016 Global 1,263 II or III Thymoma ↑5-year OS (95% vs. 90%)
↑10-year OS (86% vs. 79%)
Lim et al. (82) 2015 United States 529 IIB, III or IV Thymoma ↑OS rate (76% vs. 66%)
↑RFS at 7-year (91% vs. 81%)
Benefit limited to stage III or IV
Omasa et al. (83) 2015 Japan 1,265 II or III Thymoma and thymic carcinoma ↑RFS in thymic carcinoma
No benefit in OS
No benefit of PORT for thymoma
Forquer et al. (56) 2010 United States 901 I–III Thymoma and thymic carcinoma PORT had no benefit in surgically resected stage I
↑5-year OS by adding PORT (76% vs. 66% for surgery alone, P=0.01) for stage II–III
Utsumi et al. (58) 2009 Japan 324 I–IV Thymoma 10-year OS in stage I and II with surgery alone: 100%
No benefit of PORT in stage I and II

↑, increase. PORT, postoperative radiotherapy; OS, overall survival; HR, hazard ratio; CI, confidence interval; RFS, relapse-free survival.

Thymic carcinomas with positive margins or residual disease may benefit of PORT supplemented with adjuvant chemotherapy with carboplatin and paclitaxel (84). Adjuvant chemoradiotherapy could be an option for patients with thymic carcinoma and macroscopic residual disease after surgery (84).

Potentially resectable disease

Thymic carcinomas that invade phrenic nerve(s), innominate vein or heart/great vessels are usually not suitable for upfront surgery because it is difficult to achieve an R0 resection, thus, a multimodal approach incorporating induction chemotherapy and postoperative RT is recommended (85). Prior to the start of induction chemotherapy, a diagnostic biopsy is recommended (84). An extensive evaluation on the risk of iatrogenic phrenic nerve injury should be performed prior to surgery because it can impair respiratory function.

Multimodality therapy approach based on previous studies of unresectable malignant thymomas, summarized in Table 10:

  • Induction chemotherapy based on combination regimens, with resecability rates that range from 36–69% (57,68,86) followed by complete surgery and adjuvant radiotherapy/chemotherapy has been shown to prolong free survival (55). The recommendation of chemotherapy regimen is the same as unresectable disease: cyclophosphamide/doxorubicin and cisplatin repeated every 3 weeks.
  • Reevaluate with imaging techniques if surgery is feasible. Patients who require a pleurectomy or extrapleural pneumonectomy because of the extent of disease should be discussed since the evidence of prolonged disease survival after performing an aggressive surgical approach is controversial (84).
  • If an R0 resection is not possible it should be discussed if a maximum debulking followed by adjuvant RT (PORT) can be performed (84). Patients with residual disease may benefit from adjuvant chemotherapy and PORT.

Recurrent disease

Patients who have a localized recurrent disease require an assessment of a radical approach of surgery and the consideration of PORT or chemotherapy (87-89). If an R0 resection is not feasible, the resection of resectable disease and radiotherapy for the non-resectable disease can be discussed (90). If the patient has metastastic widespread disease then the treatment approach should be palliative (84).

NETTs

NETTs are usually diagnosed in a more advanced stage compared to thymic carcinomas and are larger in size (91,92). In functional lesions, locally advanced invasive tumors or fast-growing mediastinal lesions a histological confirmation is recommended prior to the surgical approach (93). The resection should include invaded mediastinal structures to achieve an R0 resection. In advanced tumors where there is an invasion of great vessels, pleural deposits or lung invasion, a posterolateral thoracotomy combined with sternotomy could be performed (93).

NETTs harbor an aggressive behaviour and have a poor prognosis even when an R0 resection has been achieved. If a recurrence occurs, an extensive surgical approach should be considered at the multidisciplinary meeting and an adjuvant radiotherapy has been shown to be effective in this subgroup of patients (94-96).


Unresectable/advanced disease

Thymoma and thymic carcinoma

Unresectable disease is that which presents with extensive pleural and/or pericardial metastases, unreconstructable great vessel, heart, or tracheal involvement or otherwise technically unresectable disease, including those with distant metastases.

Treatments are individualized according to the symptoms, extent of disease, and performance status. A multidisciplinary team should evaluate on a case-by-case basis the best therapeutic approach for patients with TETs. Debulking surgery may also provide benefit to select patients with initially unresectable disease, so continued involvement of a multidisciplinary team, including a thoracic surgeon, is important.

Patients with locally advanced, unresectable disease (TNM stage IIIB or Masaoka-Koga stage IVB), thymoma, or thymic carcinoma should be treated with concurrent chemoradiotherapy (cisplatin and etoposide) when feasible. Extrapolating from treatment paradigms for locally advanced lung cancer, radiotherapy doses of 60 Gy are appropriate (5). In this setting, chemoradiotherapy can offer long-term survival benefit and control the symptoms of the disease (97).

In select patients with initially unresectable disease, it is appropriate to evaluate for debulking surgery, as this approach may improve survival outcomes (97).


First line

Chemotherapy is the primary palliative treatment modality for patients with more widespread disease (1). Up to six cycles of platinum-anthracycline based regimens as CAP (cyclophosphamide, doxorubicin, cisplatin), cisplatin and etoposide and carboplatin and paclitaxel are the chemotherapy regimens that have shown efficacy in this setting (1). Six first-line chemotherapy regimens are recommended, with the carboplatin-paclitaxel combination being the preferred regimen (84). In advanced thymoma, a pooled analysis of 10 prospective and 5 retrospective studies indicated that anthracycline-based and platinum chemotherapy was superior to platinum without anthracycline in overall response rate (ORR 69.4% vs. 37.8%) and cisplatin-based chemotherapy was superior to carboplatin-based chemotherapy (ORR 53.6% vs. 32.8%) (72).

Although several regimens are acceptable, cyclophosphamide, doxorubicin, and cisplatin (CAP) and cisplatin and etoposide (PE) have been used successfully for thymomas or thymic carcinomas. Data suggest that the CAP and ADOC regimens could be effective for thymic carcinomas, but they are more toxic than carboplatin/paclitaxel (98,99). The combination of carboplatin and paclitaxel is also used extensively, especially in patients with thymic carcinoma, while the CAP regimen is preferred in patients with thymoma (84). Table 12 summarizes the different chemotherapy regimens (100-104).

Table 12

Chemotherapy regimens in unresectable/advanced TETs

Name Study Patient population Dose Efficacy
PE Giaccone et al. (100) 16 patients with advanced thymoma Cisplatin (60 mg/m2 IV day 1) and etoposide (120 mg/m2 IV days 1 to 3), repeated every three weeks ORR: 56%
CR: 31%
PFS: 2.2 years
OS: 4.3 years
CAP Loehrer et al. (101) 29 patients with metastatic or progressive thymoma Cyclophosphamide (500 mg/m2 IV day 1), doxorubicin (50 mg/m2 IV day 1), and cisplatin (50 mg/m2 IV day 1), repeated every three weeks ORR: 50%
CR: 10%
OS: 38 months
CAP with prednisone Kim et al. (68) 22 patients with locally advanced unresectable thymoma Cyclophosphamide (500 mg/m2 IV day 1), doxorubicin (20 mg/m2/day as a continuous infusion, days 1 to 3), cisplatin (30 mg/m2 IV day 1 to 3) and prednisone (100 mg/day on days 1 to 5), repeated every three weeks ORR: 77%
CR: 14%.
ADOC Fornasiero et al. (102) 37 patients with locally advanced invasive thymoma Cisplatin (50 mg/m2 IV day 1), doxorubicin (40 mg/m2 IV day 1), vincristine (0.6 mg/m2 IV day 3), and cyclophosphamide (700 mg/m2 IV day 4), repeated every three weeks ORR: 92%
CR: 43%
OS: 15 months
CP Lemma et al. (103) 44 patients with advanced previously untreated thymoma (21) and thymic carcinoma (23) Carboplatin (area under the curve 6) and paclitaxel (225 mg/m2 IV) every three weeks Thymoma:
      ORR: 43%
      CR: 14%
      OS: NR
Thymic carcinoma:
      ORR: 22%
      CR: 0%
      OS: 20 months
VIP Loehrer et al. (104) 34 patients with advanced previously untreated thymoma and thymic carcinoma Etoposide (75 mg/m2 IV days 1 to 4), ifosfamide (1.2 g/m2 IV on days 1 to 4), and cisplatin (20 mg/m2 IV days 1 to 4), repeated every three weeks Only 28 patients were evaluable
ORR: 32%
CR: 0%
OS: 32 months

TETs, thymic epithelial tumors; IV, intravenous; ORR, overall response rate; CR, complete response; PFS, progression free survival; OS, overall survival; NR, not reached.


Subsequent therapy

There are no further recognized standard lines of treatment for patients with TETs who progress on initial chemotherapy. Despite them, many patients are candidates to receive a second line. None of the agents studied in this context has been assessed in randomized phase 3 trials.

Pemetrexed, everolimus, octreotide [long-acting release (LAR)] with or without prednisone, paclitaxel, 5-fluorouracil (5-FU), gemcitabine with or without capecitabine, sunitinib, ifosfamide and etoposide are second-line chemotherapy options for thymomas (105-116).

Pemetrexed, 5-FU, sunitinib, everolimus, paclitaxel, lenvatinib, gemcitabine with or without capecitabine, ifosfamide and pembrolizumab are second-line chemotherapy options for thymic carcinomas (99,105,109,117-122). Table 13 summarizes the different chemotherapy regimens.

Table 13

Systemic treatments in pretreated advanced thymic epithelial tumors

Study Patient population Phase Dose Efficacy
Palmieri et al. (106) N=30: 22 thymoma, 8 thymic carcinoma II Capecitabine (650 mg/m2 twice daily on days 1–14) and gemcitabine IV (1,000 mg/m2 on days 1 and 8 every 3 weeks) ORR: 40%
PFS: 11 months
OS: NR
Bluthgen et al. (107) N=20: 5 thymoma, 15 thymic carcinoma Retrospective study Oral etoposide 25 mg three times daily for 3 weeks, followed by 1 week off (4-week cycle) Thymoma:
      ORR: 20%
      SD: 80%
      PFS: 21 months
      OS: 99 months
Thymic carcinoma:
      ORR: 13%
      SD: 33%
      PFS: 4 months
      OS: 13 months
Zucali et al. (108) N=51: 32 thymoma, 19 thymic carcinoma II Everolimus 10 mg/day continuous Thymoma:
      ORR: 9%
      SD: 85%
      PFS: 16.6 months
      OS: NR
Thymic carcinoma:
      ORR: 16%
      SD: 58%
      PFS: 5.6 months
      OS: 14.7 months
Thomas et al. (109) N=41: 16 thymoma,25 thymic carcinoma II Sunitinib 50 mg orally once a day, in 6-week cycles (i.e., 4 weeks of treatment followed by 2 weeks without treatment) Thymoma:
      ORR: 6%
      SD: 75%
      PFS: 8.5 months
      OS: 15.5 months
Thymic carcinoma:
      ORR: 26%
      SD: 65%
      PFS: 7.2 months
      OS: NR
Antonarelli et al. (110) N=20: 8 thymoma, 12 thymic carcinoma Retrospective study Sunitinib 37.5 mg/day continuous daily dosing
Gbolahan et al. (111) N=27: 16 thymoma, 11 thymic carcinoma II Pemetrexed, 500 mg/m2 IV every 3 weeks Thymoma:
      ORR: 27%
      PFS: 12.1 months
      OS: 46.4 months
Thymic carcinoma:
      ORR: 9%
      PFS: 2.9 months
      OS: 9.8 months
Loehrer et al. (114) N=38: 32 thymoma, 5 thymic carcinoma, 1 thymic carcinoid II Octreotide in a dose of 0.5 mg subcutaneously 3 times a day, for a maximum of 1 year. Patients with stable disease at the end of two cycles, receive prednisone at a dose of 0.6 mg/kg per day ORR: 30%
SD: 37%
Octreotide:
      PFS: 2 months
Octreotide plus prednisone:
      PFS: 9.2 months
Thymoma:
      PFS: 8.8 months
      OS: NR
Thymic carcinoma:
      PFS: 4.5 months
      OS: 23.4 months
Highley et al. (116) N=15: 15 thymoma [only 7 patients received prior treatment (one chemotherapy)] Retrospective study Ifosfamide 1.5 g/m2 on days 1 to 5 ORR: 46%
CR: 38%
Estimated survival rate 5 years 57%
Conforti et al. (117) N=18: 5 thymoma, 12 thymic carcinoma, 1 mixed histology Multicentric, prospective study Ifosfamide (1 g/m2/day) and sodium-2-mercaptoethanesulfonate (1 g/m2/day), as continuous infusion, via a portable pumps for 14 consecutive days. Treatment was administered every 4 weeks ORR: 28%
SD: 39%
PFS: 5.4 months
Sato et al. (120) N=42: 42 thymic carcinoma II Lenvatinib 24 mg orally once daily in 4-week cycles ORR: 38%
SD: 57%
PFS: 9.3 months
OS: NR
Giaccone et al. (121) N=40: 40 thymic carcinoma II Pembrolizumab 200 mg every 3 weeks for up to 2 years ORR: 23%
SD: 53%
PFS: 4.2 months
OS: 24.9 months
Cho et al. (122) N=33: 7 thymoma, 26 thymic carcinoma II Pembrolizumab 200 mg every 3 weeks Thymoma:
      ORR: 29%
      SD: 71%
      PFS: 6.1 months
      Duration of response: NR
Thymic carcinoma:
      ORR: 19%
      SD: 54%
      PFS: 6.1 months
      Duration of response: 9.7 months

IV, intravenous; ORR, overall response rate; PFS, progression free survival; OS, overall survival; NR, not reached; SD, stable disease; CR, complete response.

Although immunotherapy studies have shown efficacy in patients with advanced thymoma, we do not offer immunotherapy, as high rates of immune-related adverse events (irAEs) have been reported in these patients (121,122). In clinical trials, pembrolizumab demonstrated durable responses in patients with thymic carcinoma, which may be more pronounced those whose tumors express programmed death-ligand 1 (PD-L1) (121-123). These patients should be carefully monitored for possible severe irAEs, including myocarditis, myasthenia gravis, and hepatitis. There are no randomized trials directly comparing immunotherapy with other subsequent-line regimens, such as chemotherapy.

The elevated incidence of irAEs in TETs patients that receive immune checkpoint inhibitors warrant additional biomarker studies to identify patients who can benefit the most from immunotherapy and could present less irAEs (122). In this context, immunologic biomarkers for the early identification and prediction identification of irAEs are currently being investigated (124,125). Biomarkers like immune gene expression, IL-17 or peripheral eosinophil counts have been associated with the development of irAEs in solid tumors (124).

Sunitinib or lenvatinib are multiple tyrosine kinases inhibitors, including vascular endothelial growth factor (VEGF) and c-KIT, are an appropriate option in patients with thymic carcinomas refractory to initial chemotherapy, based on data from phase II trials and retrospective studies (109,110,120).

There is no clear role for nivolumab or avelumab in patients with relapsed thymic carcinoma, as clinical trials evaluating these agents showed limited activity and significant toxicity (126,127). Similarly, everolimus is not routinely used due to severe toxicity (pneumonitis), despite initial studies that suggest some efficacy in relapsed thymoma and thymic carcinoma (128).

Later-line options for treatment-refractory thymomas and thymic carcinomas include etoposide, ifosfamide, pemetrexed, octreotide, fluorouracil, S-1, gemcitabine plus capecitabine and paclitaxel.

Arunachalam et al. performed a meta-analysis focused on the efficacy and safety of subsequent treatments for advanced thymic carcinoma after failure of first-line platinum-based chemotherapy (123). From the nineteen trials identified in the systemic literature review, three trials with one or two TC patients were removed to reduce publication bias. The pooled ORRs in patients receiving S-1 (46 patients), sunitinib (46 patients), or pembrolizumab (66 patients) were 28%, 24%, and 21%, respectively. Pembrolizumab obtained an extended duration of response with a pooled median OS of 23.8 months [95% confidence interval (CI): 12, not reached]. Patients who had received lenvatinib, sunitinib, capecitabine + gemcitabine, S-1, everolimus or pembrolizumab reported a median PFS of at least five months. S-1 or pembrolizumab trials reported a median OS of at least 20 months; this endpoint was not reached in trials evaluating lenvatinib, regorafenib, or sunitinib. Therefore, the study found limited treatment options upon relapse, and there is a need for further investigations into novel therapeutics and well-powered clinical trials to better inform on optimal treatments.

NETTs

Approximately 80% to 90% of WD thoracic NETTs express SSTRs on their cell surface, that bind with high affinity somatostatin analogs (SSAs) lanreotide autogel and octreotide LAR (2). SSAs should probably be chosen first line for patients with relatively low-volume, relatively asymptomatic, SSTR-positive disease (129-131).

There are other several systemic treatment options: everolimus, temozolomide-based chemotherapy, and peptide receptor radioligand therapy using a radiolabeled SSA such as lutetium Lu-177 dotatate (177Lu-dotatate).

Beyond SSAs, there are no data for selecting or sequencing these treatments except that 177Lu-dotatate is limited to SSTR-expressing tumors. Even in those tumors, there is no real basis for choosing 177Lu-dotatate over everolimus, or viceversa, as the second-line treatment. Most of the data on the effectiveness of these drugs is extrapolated from thoracic, gastroenteropancreatic or intestinal neuroendocrine tumors, with only data available from retrospective studies of patients with NETTs. Table 14 summarizes the different treatment options in G1/G2 advanced/metastatic NETTs.

Table 14

summarizes the different treatment options in G1/G2 advanced/metastatic NETTs

Name Study Patient population Dose Efficacy
Octreotide LAR Rinke et al. (130) 85 gastroenteropancreatic neuroendocrine tumors patients Octreotide LAR 30 mg intramuscularly in monthly intervals until tumor progression or death vs. placebo SD: 66.7% vs. 37.2%; P=0.0079
PFS: 4.3 and 6 months, HR =0.34; (95% CI: 0.20 to 0.59; P=0.000072)
OS: HR =0.81 (95% CI: 0.30 to 2.18)
Extended-release aqueous-gel formulation of lanreotide Caplin et al. (131) 204 patients with advanced, well-differentiated or moderately differentiated, nonfunctioning, somatostatin receptor-positive neuroendocrine tumors of grade 1 or 2 and documented disease-progression status Extended-release aqueous-gel formulation of lanreotide at a dose of 120 mg or placebo once every 28 days for 96 weeks SD: NR
PFS: HR =0.47; 95% CI: 0.30 to 0.73
OS: no differences
Everolimus Yao et al. (132) 302 patients with advanced, progressive, well-differentiated, non-functional neuroendocrine tumors of lung or gastrointestinal origin Randomly assigned in a 2:1 ratio to receive everolimus 10 mg per day orally or identical placebo, both with supportive care SD: 81% in the everolimus arm vs. 64% in the placebo arm
PFS: 11.0 vs. 3.9 months in the placebo group. HR =048 (95% CI: 0.35–0.67, P<0.00001)
OS: HR 0.64 (95% CI 0.40–1.05), one-sided P=0.037
Everolimus Lang et al. (133) 4 patients with progressing NETTs (two well-differentiated atypical carcinoids and two atypical carcinoids with large cell characteristics) Everolimus 10 mg/day until progression disease SD interval in all patients and mean PFS of 20.8 months
PFS interval was longer in well differentiated tumors (24 and 42 months, respectively) compared with large cell differentiation (7 and 10 months)
OS: NR
Temozolomide Ekeblad et al. (134) 36 patients with advanced and pretreated neuroendocrine tumor (1 gastric, 7 thymic and 13 bronchial carcinoids, 12 pancreatic endocrine tumors, 1 paraganglioma, 1 neuroendocrine foregut, and 1 neuroendocrine cecal cancer) Temozolomide 200 mg/m2 for 5 days every 4 weeks SD: 53% and 14% of ORR (in 7 NETTs, SD in 71% and 0% ORR)
PFS: 7 months (95% CI: 3–10)
OS: NR
Temozolomide Crona et al. (135) 28 patients with NETTs, of which 8 received temozolomide NR temozolomide dose SD: 75% and ORR 12.5%
PFS: median PFS of 20.5 months
OS: NR
Capecitabine plus temozolomide Saranga-Perry et al. (136) 3 patients with progressive NETTs Patient 1: capecitabine (700 mg/m2 b.i.d. days 1–14 every 28 days) and temozolomide (170 mg/m2 days 10–14) every 28 days SD 67% and ORR 33%
Patient 2: capecitabine (600 mg/m2 b.i.d. days 1–14 every 28 days) and temozolomide (190 mg/m2 days 10–14 every 28 days)
Patient 3: capecitabine (750 mg/m2 b.i.d. days 1–14 every 28 days) and temozolomide (180 mg/m2 days 10–14 every 28 days)
Radiolabeled somatostatin analog 177Lu-dotatate Strosberg et al. (137) 229 patients with advanced midgut NETs, high level of expression of somatostatin receptors 177Lu-Dotatate at a dose of 7.4 GBq every 8 weeks (four intravenous infusions, plus best supportive care including octreotide LAR administered intramuscularly at a dose of 30 mg) or octreotide LAR alone administered intramuscularly at a dose of 60 mg every 4 weeks ORR: 18% vs. 3%; P<0.001
PFS: not reached in the 177Lu-Dotatate group and was 8.4 months (95% CI: 5.8 to 9.1) in the control group (HR 0.21; 95% CI: 0.13 to 0.33; P<0.001)
OS: HR 0.40; P=0.004
Radiolabeled somatostatin analog 177Lu-dotatate van Essen et al. (138) Nine patients with bronchial, five with gastric and two with thymic carcinoids were treated. All patients had metastasised disease 177Lu-Dotatate at a dose of 7.4 GBq, injected in 30 min. The interval between treatments was 6–10 weeks. Patients were treated up to an intended cumulative dose of 22.2–29.6 GBq SD of two patients with NETTs was 50%

NETTs, neuroendocrine tumors of the thymus; LAR, long acting release; SD, stable disease; PFS, progression free survival; HR, hazard ratio; CI, confidence interval; OS, overall survival; ORR, overall response rate; NR, not reported.

Patients with intermediate to poorly-differentiated tumors respond to platinum-based chemotherapy regimens (135,139). In particular, treatment of poorly-differentiated NETTs with platinum-based regimens, such as carboplatin and etoposide, as per treatment guidelines for poorly-differentiated NETs at other sites.

New combinations of SSAs and other investigational drugs are therefore warranted, with the aim to improve clinical outcomes, while maintaining a good tolerability profile.


New therapeutics options

Immunotherapy administered alone or in combination with other agents is currently under study in several trials including patients with advanced B3 thymoma and thymic carcinoma which relapsed after at least one line of platinum-based chemotherapy. One of the main lines of research is the combination of antiangiogenic agents with chemotherapy or immunotherapy. Table 15 summarizes the main ongoing clinical trials in patients with advanced TETs.

Table 15

Ongoing clinical trials in patients with TETs (140)

Study ClinicalTrials.gov identifier Phase Patient population Drug Primary end point
Pembrolizumab in treating participants with unresectable T or TC NCT03295227 I Unresectable T or TC Pembrolizumab Safety
Combination of pembrolizumab and lenvatinib in pre-treated TC patients (PECATI) NCT04710628 II Advanced B3 T and TC relapsed after at least one line of P-ChT Pembrolizumab, lenvatinib PFS
Pembrolizumab and sunitinib malate in treating participants with refractory metastatic or unresectable TC NCT03463460 II Advanced TC relapsed after at least one line of P-ChT Pembrolizumab, sunitinib ORR
A Phase II, neo-adjuvant pembrolizumab, docetaxel, cisplatin therapy followed by surgery and pembrolizumab consolidation therapy in locally advanced thymic epithelial tumor (TET) NCT03858582 II Locally advanced TET Pembrolizumab, docetaxel, cisplatin Major pathologic response rate
Chemotherapy combined with pembrolizumab in treating patients with T and TC NCT04554524 IV First line in locally advanced or metastatic invasive T and TC that cannot be removed by surgery Carbo-paclitaxel/ nab-paclitaxel combined with pembrolizumab ORR
A pilot study to investigate the safety and clinical activity of avelumab in T and TC after progression on platinum-based chemotherapy NCT03076554 II Advanced T and TC relapsed after at least one line of P-ChT Avelumab Safety ORR
Nivolumab in patients with type B3 T and TC (NIVOTHYM) NCT03134118 II Advanced B3 T and TC relapsed after at least one line of P-ChT Nivolumab PFS
Trial of sunitinib in patients with type B3 T or TC in second and further lines (STYLE) NCT03449173 II Advanced B3 T and TC relapsed after at least one line of P-ChT Sunitinib ORR
Carboplatin and paclitaxel with or without ramucirumab in treating patients with locally advanced, recurrent or metastatic TC NCT03694002 II Advanced TC with no anti-cancer therapy for locally advanced or metastatic disease Carboplatin, paclitaxel, ramucirumab PFS
Ramucirumab and carbo-paclitaxel for untreated thymic carcinoma/B3 thymoma with carcinoma (RELEVENT) NCT03921671 II Chemotherapy-naïve patients with thymic carcinoma or B3 thymoma with areas of carcinoma Carboplatin, paclitaxel, ramucirumab ORR
A study of KC1036 in patients with advanced TC NCT05683886 II Advanced recurrent, unresectable and/or metastatic T KC1036 ORR
A study of KN046 in patients with TC who failed ICIs NCT04925947 II Advanced TC relapsed after P-ChT and at least one line of ICIs KN046 ORR
KN046 in subjects with TC NCT04469725 II Advanced TC relapsed after at least one line of P-ChT KN046 ORR
Bintrafusp alfa (M7824) in subjects with T and TC NCT04417660 II Advanced T and TC relapsed after at least one line of P-ChT Bintrafusp alfa (M7824) ORR
PT-112 in subjects with T and TC NCT05104736 II Advanced T and TC relapsed after at least one line of P-ChT PT-112 ORR
Atezolizumab in previously-treated patients with advanced TC NCT04321330 II Advanced TC who failed prior systemic therapy Atezolizumab ORR
ChT plus cetuximab followed by surgical resection in patients with locally advanced or recurrent T or TC NCT01025089 II Clinical Masaoka stage II–IVa T and TC Cetuximab, cisplatin, doxorubicin, and cyclophosphamide Major pathologic response rate
Nivolumab in combination with vorolanib in patients with refractory thoracic tumors NCT03583086 I/II Non-small cell lung cancer naïve to ICIs non-small cell lung cancer who have progressed on ICIs small cell lung cancer (who have progressed on platinum-based chemotherapy), and TC Oral vorolanib plus infusional nivolumab Adverse events ORR

TETs, thymic epithelial tumors; T, thymoma; TC, thymic carcinoma; P-ChT, platinum-based chemotherapy; ICIs, immune checkpoint inhibitors; PFS, progression-free survival; ORR, overall response rate.


Conclusions

TETs are rare and heterogeneous tumors that arise in the anterior mediastinum. Thymomas may be an incidental diagnosis discovered at chest imaging, and patients may present with symptoms due to the presence of a mass in the thorax or to a paraneoplastic phenomenon such as myasthenia gravis. The management of TETS requires a multidisciplinary approach (pathologists, medical oncologists, radiation oncologists and thoracic surgeons). Complete surgical resection is the initial treatment approach for all patients when preoperative evaluation suggests that a complete resection will be feasible and there are no medical contraindications to surgery. For patients with resected disease, the approach to postoperative radiation therapy is based on stage. In case of potentially resectable disease the recommendation is initial treatment with neoadjuvant chemotherapy and local treatment depending on the response.

In unresectable disease, RT alone, chemotherapy, or the combination is appropriate for patients in whom surgery is not technically feasible or is contraindicated, and may be of curative potential. Platinum-based chemotherapy is the treatment of choice in case of metastatic disease. However, patients with metastatic TETs have limited treatment options beyond platinum-based chemotherapy, due to the poor effectiveness showed by several other agents administered in subsequent lines of therapy. New therapies have been explored in this clinical setting such as the antiangiogenic multikinase inhibitors, mammalian target of rapamycin (mTOR) inhibitor, ICIs and their combinations.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://med.amegroups.com/article/view/10.21037/med-23-47/rc

Peer Review File: Available at https://med.amegroups.com/article/view/10.21037/med-23-47/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://med.amegroups.com/article/view/10.21037/med-23-47/coif). L.C.G. reports he received payment for presentations of Roche, Astra Zeneca, Brystol Myers Squibb, Merck Serono, Ipsen Pharma, Grunenthal, Kyowa Kirin, Pfizer and Eisai and received support for attending meetings from Roche, Merck, Eli Lilly, Bristol-Myers Squibb and Nutricia. V.P.B. reports she received a grant as an award from Merck and FSEOM, payment for presentations of Merck, Eli Lilly, Eisai and Pierre Fabre and received support for attending meetings from Roche, Eli Lilly, Bristol-Myers Squibb, Merck, Amgen, Merck Sharp and Dhome, and Nutricia. V.P.B. also reports she participated in an advisory board from advanced accelerator applications, a Novartis company. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Tartarone A, Lerose R, Lettini AR, et al. Current Treatment Approaches for Thymic Epithelial Tumors. Life (Basel) 2023;13:1170. [Crossref] [PubMed]
  2. Scorsetti M, Leo F, Trama A, et al. Thymoma and thymic carcinomas. Crit Rev Oncol Hematol 2016;99:332-50. [Crossref] [PubMed]
  3. Gaude GS, Hattiholi V, Malur PR, et al. Primary neuroendocrine carcinoma of the thymus. Niger Med J 2013;54:68-71. [Crossref] [PubMed]
  4. Zhang Y, Lin D, Aramini B, et al. Thymoma and Thymic Carcinoma: Surgical Resection and Multidisciplinary Treatment. Cancers (Basel) 2023;15:1953. [Crossref] [PubMed]
  5. Girard N, Ruffini E, Marx AESMO Guidelines Committee, et al. Thymic epithelial tumours: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26:v40-v55. [Crossref] [PubMed]
  6. Marchevsky AM, Dikman SH. Mediastinal carcinoid with an incomplete Sipple's syndrome. Cancer 1979;43:2497-501. [Crossref] [PubMed]
  7. Frilling A, Becker H, Roeher HD. Unusual features of multiple endocrine neoplasia. Henry Ford Hosp Med J 1992;40:253-5. [PubMed]
  8. Lang M, Kazdal D, Mohr I, et al. Differences and similarities of GTF2I mutated thymomas in different Eurasian ethnic groups. Transl Lung Cancer Res 2023;12:1842-4. [Crossref] [PubMed]
  9. Yadav S, Salonga R, Tamayo JE, et al. Thymic Carcinoma Presenting as Myasthenia Gravis. Disorders of the Mediastinum. Chest 2016;150:546A. [Crossref]
  10. Sakamoto N, Kurokawa R, Watadani T, et al. Differential diagnosis of thymic epithelial neoplasms on computed tomography using the diameter of the thymic vein. Medicine (Baltimore) 2021;100:e27942. [Crossref] [PubMed]
  11. Bozkurt MF, Virgolini I, Balogova S, et al. Guideline for PET/CT imaging of neuroendocrine neoplasms with (68)Ga-DOTA-conjugated somatostatin receptor targeting peptides and (18)F-DOPA. Eur J Nucl Med Mol Imaging 2017;44:1588-601. [Crossref] [PubMed]
  12. Girard N. Neuroendocrine tumors of the thymus: the oncologist point of view. J Thorac Dis 2017;9:S1491-500. [Crossref] [PubMed]
  13. Karlin K, Michaels PD. Thymic carcinoma: review and update. J Cancer Metastasis Treat 2022;8:15. [Crossref]
  14. World Health Organization. WHO Classification of Tumors Online, Thoracic Tumors, Tumors of the Thymus, 5th ed.; World Heath Organization: Geneva, Switzerland, 2021. Available online: https://tumorclassification.iarc.who.int/chapters/35.
  15. Wang ZM, Xu QR, Kaul D, et al. Significance of tumor mutation burden and immune infiltration in thymic epithelial tumors. Thorac Cancer 2021;12:1995-2006. [Crossref] [PubMed]
  16. Petrini I, Meltzer PS, Kim IK, et al. A specific missense mutation in GTF2I occurs at high frequency in thymic epithelial tumors. Nat Genet 2014;46:844-9. [Crossref] [PubMed]
  17. Marx A, Chan JKC, Chalabreysse L, et al. The 2021 WHO Classification of Tumors of the Thymus and Mediastinum: What Is New in Thymic Epithelial, Germ Cell, and Mesenchymal Tumors? J Thorac Oncol 2022;17:200-13. [Crossref] [PubMed]
  18. Detterbeck FC, Zeeshan A. Thymoma: current diagnosis and treatment. Chin Med J (Engl) 2013;126:2186-91. [Crossref] [PubMed]
  19. Kondo K, Monden Y. Therapy for thymic epithelial tumors: a clinical study of 1,320 patients from Japan. Ann Thorac Surg 2003;76:878-84; discussion 884-5. [Crossref] [PubMed]
  20. Hamaji M, Allen MS, Cassivi SD, et al. The role of surgical management in recurrent thymic tumors. Ann Thorac Surg 2012;94:247-54; discussion 254. [Crossref] [PubMed]
  21. Okumura M, Ohta M, Tateyama H, et al. The World Health Organization histologic classification system reflects the oncologic behavior of thymoma: a clinical study of 273 patients. Cancer 2002;94:624-32. [Crossref] [PubMed]
  22. Kim DJ, Yang WI, Choi SS, et al. Prognostic and clinical relevance of the World Health Organization schema for the classification of thymic epithelial tumors: a clinicopathologic study of 108 patients and literature review. Chest 2005;127:755-61. [Crossref] [PubMed]
  23. Rea F, Marulli G, Girardi R, et al. Long-term survival and prognostic factors in thymic epithelial tumours. Eur J Cardiothorac Surg 2004;26:412-8. [Crossref] [PubMed]
  24. Zhu G, He S, Fu X, et al. Radiotherapy and prognostic factors for thymoma: a retrospective study of 175 patients. Int J Radiat Oncol Biol Phys 2004;60:1113-9. [Crossref] [PubMed]
  25. Regnard JF, Magdeleinat P, Dromer C, et al. Prognostic factors and long-term results after thymoma resection: a series of 307 patients. J Thorac Cardiovasc Surg 1996;112:376-84. [Crossref] [PubMed]
  26. Nakagawa K, Asamura H, Matsuno Y, et al. Thymoma: a clinicopathologic study based on the new World Health Organization classification. J Thorac Cardiovasc Surg 2003;126:1134-40. [Crossref] [PubMed]
  27. Friedant AJ, Handorf EA, Su S, et al. Minimally Invasive versus Open Thymectomy for Thymic Malignancies: Systematic Review and Meta-Analysis. J Thorac Oncol 2016;11:30-8. [Crossref] [PubMed]
  28. Toker A, Sonett J, Zielinski M, et al. Standard terms, definitions, and policies for minimally invasive resection of thymoma. J Thorac Oncol 2011;6:S1739-42. [Crossref] [PubMed]
  29. Agatsuma H, Yoshida K, Yoshino I, et al. Video-Assisted Thoracic Surgery Thymectomy Versus Sternotomy Thymectomy in Patients With Thymoma. Ann Thorac Surg 2017;104:1047-53. [Crossref] [PubMed]
  30. O'Sullivan KE, Kreaden US, Hebert AE, et al. A systematic review of robotic versus open and video assisted thoracoscopic surgery (VATS) approaches for thymectomy. Ann Cardiothorac Surg 2019;8:174-93. [Crossref] [PubMed]
  31. Falkson CB, Bezjak A, Darling G, et al. The management of thymoma: a systematic review and practice guideline. J Thorac Oncol 2009;4:911-9. [Crossref] [PubMed]
  32. Brascia D, De Palma A, Schiavone M, et al. Lymph Nodes Involvement and Lymphadenectomy in Thymic Tumors: Tentative Answers for Unsolved Questions. Cancers (Basel) 2021;13:5085. [Crossref] [PubMed]
  33. Petrella F, Leo F, Veronesi G, et al. "Salvage" surgery for primary mediastinal malignancies: is it worthwhile?. J Thorac Oncol 2008;3:53-8. [Crossref] [PubMed]
  34. Detterbeck FC, Nicholson AG, Kondo K, et al. The Masaoka-Koga stage classification for thymic malignancies: clarification and definition of terms. J Thorac Oncol 2011;6:S1710-6. [Crossref] [PubMed]
  35. Roden AC, Ahmad U, Cardillo GThymic Carcinomas-A Concise Multidisciplinary Update on Recent Developments From the Thymic Carcinoma Working Group of the International Thymic Malignancy Interest Group, et al. J Thorac Oncol 2022;17:637-50. [Crossref] [PubMed]
  36. Bernard C, Frih H, Pasquet F, et al. Thymoma associated with autoimmune diseases: 85 cases and literature review. Autoimmun Rev 2016;15:82-92. [Crossref] [PubMed]
  37. Gilhus NE, Owe JF, Hoff JM, et al. Myasthenia gravis: a review of available treatment approaches. Autoimmune Dis 2011;2011:847393. [Crossref] [PubMed]
  38. Mehran R, Ghosh R, Maziak D, et al. Surgical treatment of thymoma. Can J Surg 2002;45:25-30. [PubMed]
  39. Howard FM Jr, Lennon VA, Finley J, et al. Clinical correlations of antibodies that bind, block, or modulate human acetylcholine receptors in myasthenia gravis. Ann N Y Acad Sci 1987;505:526-38. [Crossref] [PubMed]
  40. Detterbeck FC, Parsons AM. Management of stage I and II thymoma. Thorac Surg Clin 2011;21:59-67. vi-vii. [Crossref] [PubMed]
  41. Masaoka A. Staging system of thymoma. J Thorac Oncol 2010;5:S304-12. [Crossref] [PubMed]
  42. Lewis JE, Wick MR, Scheithauer BW, et al. Thymoma. A clinicopathologic review. Cancer 1987;60:2727-43. [Crossref] [PubMed]
  43. Zhao Y, Shi J, Fan L, et al. Surgical treatment of thymoma: an 11-year experience with 761 patients. Eur J Cardiothorac Surg 2016;49:1144-9. [Crossref] [PubMed]
  44. Detterbeck F, Youssef S, Ruffini E, et al. A review of prognostic factors in thymic malignancies. J Thorac Oncol 2011;6:S1698-704. [Crossref] [PubMed]
  45. Ried M, Potzger T, Sziklavari Z, et al. Extended surgical resections of advanced thymoma Masaoka stages III and IVa facilitate outcome. Thorac Cardiovasc Surg 2014;62:161-8. [PubMed]
  46. Bretti S, Berruti A, Loddo C, et al. Multimodal management of stages III-IVa malignant thymoma. Lung Cancer 2004;44:69-77. [Crossref] [PubMed]
  47. Davenport E, Malthaner RA. The role of surgery in the management of thymoma: a systematic review. Ann Thorac Surg 2008;86:673-84. [Crossref] [PubMed]
  48. Ruffini E, Venuta F. Management of thymic tumors: a European perspective. J Thorac Dis 2014;6:S228-37. [PubMed]
  49. Attaran S, McCormack D, Pilling J, et al. Which stages of thymoma benefit from adjuvant chemotherapy post-thymectomy? Interact Cardiovasc Thorac Surg 2012;15:273-5. [Crossref] [PubMed]
  50. Mornex F, Resbeut M, Richaud P, et al. Radiotherapy and chemotherapy for invasive thymomas: a multicentric retrospective review of 90 cases. The FNCLCC trialists. Fédération Nationale des Centres de Lutte Contre le Cancer. Int J Radiat Oncol Biol Phys 1995;32:651-9. [Crossref] [PubMed]
  51. Myojin M, Choi NC, Wright CD, et al. Stage III thymoma: pattern of failure after surgery and postoperative radiotherapy and its implication for future study. Int J Radiat Oncol Biol Phys 2000;46:927-33. [Crossref] [PubMed]
  52. Basse C, Thureau S, Bota S, et al. Multidisciplinary Tumor Board Decision Making for Postoperative Radiotherapy in Thymic Epithelial Tumors: Insights from the RYTHMIC Prospective Cohort. J Thorac Oncol 2017;12:1715-22. [Crossref] [PubMed]
  53. Kondo K. Optimal therapy for thymoma. J Med Invest 2008;55:17-28. [Crossref] [PubMed]
  54. Tateishi Y, Horita N, Namkoong H, et al. Postoperative Radiotherapy for Completely Resected Masaoka/Masaoka-Koga Stage II/III Thymoma Improves Overall Survival: An Updated Meta-Analysis of 4746 Patients. J Thorac Oncol 2021;16:677-85. [Crossref] [PubMed]
  55. Hamaji M, Shah RM, Ali SO, et al. A Meta-Analysis of Postoperative Radiotherapy for Thymic Carcinoma. Ann Thorac Surg 2017;103:1668-75. [Crossref] [PubMed]
  56. Forquer JA, Rong N, Fakiris AJ, et al. Postoperative radiotherapy after surgical resection of thymoma: differing roles in localized and regional disease. Int J Radiat Oncol Biol Phys 2010;76:440-5. [Crossref] [PubMed]
  57. Korst RJ, Kansler AL, Christos PJ, et al. Adjuvant radiotherapy for thymic epithelial tumors: a systematic review and meta-analysis. Ann Thorac Surg 2009;87:1641-7. [Crossref] [PubMed]
  58. Utsumi T, Shiono H, Kadota Y, et al. Postoperative radiation therapy after complete resection of thymoma has little impact on survival. Cancer 2009;115:5413-20. [Crossref] [PubMed]
  59. Cowen D, Richaud P, Mornex F, et al. Thymoma: results of a multicentric retrospective series of 149 non-metastatic irradiated patients and review of the literature. FNCLCC trialists. Fédération Nationale des Centres de Lutte Contre le Cancer. Radiother Oncol 1995;34:9-16. [Crossref] [PubMed]
  60. Gomez D, Komaki R. Technical advances of radiation therapy for thymic malignancies. J Thorac Oncol 2010;5:S336-43. [Crossref] [PubMed]
  61. Marom EM. Imaging thymoma. J Thorac Oncol 2010;5:S296-303. [Crossref] [PubMed]
  62. Okereke IC, Kesler KA, Freeman RK, et al. Thymic carcinoma: outcomes after surgical resection. Ann Thorac Surg 2012;93:1668-72; discussion 1672-3. [Crossref] [PubMed]
  63. Park S, Park IK, Kim YT, et al. Comparison of Neoadjuvant Chemotherapy Followed by Surgery to Upfront Surgery for Thymic Malignancy. Ann Thorac Surg 2019;107:355-62. [Crossref] [PubMed]
  64. Ruffini E, Guerrera F, Brunelli A, et al. Report from the European Society of Thoracic Surgeons prospective thymic database 2017: a powerful resource for a collaborative global effort to manage thymic tumours. Eur J Cardiothorac Surg 2019;55:601-9. [Crossref] [PubMed]
  65. Kanzaki R, Kanou T, Ose N, et al. Long-term outcomes of advanced thymoma in patients undergoing preoperative chemotherapy or chemoradiotherapy followed by surgery: a 20-year experience. Interact Cardiovasc Thorac Surg 2019;28:360-7. [Crossref] [PubMed]
  66. Riely GJ, Huang J. Induction therapy for locally advanced thymoma. J Thorac Oncol 2010;5:S323-6. [Crossref] [PubMed]
  67. Wright CD, Choi NC, Wain JC, et al. Induction chemoradiotherapy followed by resection for locally advanced Masaoka stage III and IVA thymic tumors. Ann Thorac Surg 2008;85:385-9. [Crossref] [PubMed]
  68. Kim ES, Putnam JB, Komaki R, et al. Phase II study of a multidisciplinary approach with induction chemotherapy, followed by surgical resection, radiation therapy, and consolidation chemotherapy for unresectable malignant thymomas: final report. Lung Cancer 2004;44:369-79. [Crossref] [PubMed]
  69. Hassan M, Seoud DE. Multimodality treatments in locally advanced stage thymomas. Hematol Oncol Stem Cell Ther 2009;2:340-4. [Crossref] [PubMed]
  70. Schmitt J, Loehrer PJ Sr. The role of chemotherapy in advanced thymoma. J Thorac Oncol 2010;5:S357-60. [Crossref] [PubMed]
  71. Rajan A, Giaccone G. Chemotherapy for thymic tumors: induction, consolidation, palliation. Thorac Surg Clin 2011;21:107-14. viii. [Crossref] [PubMed]
  72. Okuma Y, Saito M, Hosomi Y, et al. Key components of chemotherapy for thymic malignancies: a systematic review and pooled analysis for anthracycline-, carboplatin- or cisplatin-based chemotherapy. J Cancer Res Clin Oncol 2015;141:323-31. [Crossref] [PubMed]
  73. Detterbeck FC, Parsons AM. Thymic tumors. Ann Thorac Surg 2004;77:1860-9. [Crossref] [PubMed]
  74. Kelly RJ. Systemic treatment of advanced thymic malignancies. Am Soc Clin Oncol Educ Book 2014;e367-73. [Crossref] [PubMed]
  75. Marx A, Rieker R, Toker A, et al. Thymic carcinoma: is it a separate entity? From molecular to clinical evidence. Thorac Surg Clin 2011;21:25-31. v-vi. [Crossref] [PubMed]
  76. Okuma Y, Hosomi Y, Watanabe K, et al. Clinicopathological analysis of thymic malignancies with a consistent retrospective database in a single institution: from Tokyo Metropolitan Cancer Center. BMC Cancer 2014;14:349. [Crossref] [PubMed]
  77. Litvak AM, Woo K, Hayes S, et al. Clinical characteristics and outcomes for patients with thymic carcinoma: evaluation of Masaoka staging. J Thorac Oncol 2014;9:1810-5. [Crossref] [PubMed]
  78. Ströbel P, Bauer A, Puppe B, et al. Tumor recurrence and survival in patients treated for thymomas and thymic squamous cell carcinomas: a retrospective analysis. J Clin Oncol 2004;22:1501-9. [Crossref] [PubMed]
  79. Jackson MW, Palma DA, Camidge DR, et al. The Impact of Postoperative Radiotherapy for Thymoma and Thymic Carcinoma. J Thorac Oncol 2017;12:734-44. [Crossref] [PubMed]
  80. Boothe D, Orton A, Thorpe C, et al. Postoperative Radiotherapy in Locally Invasive Malignancies of the Thymus: Patterns of Care and Survival. J Thorac Oncol 2016;11:2218-26. [Crossref] [PubMed]
  81. Rimner A, Yao X, Huang J, et al. Postoperative Radiation Therapy Is Associated with Longer Overall Survival in Completely Resected Stage II and III Thymoma-An Analysis of the International Thymic Malignancies Interest Group Retrospective Database. J Thorac Oncol 2016;11:1785-92. [Crossref] [PubMed]
  82. Lim YJ, Kim HJ, Wu HG. Role of Postoperative Radiotherapy in Nonlocalized Thymoma: Propensity-Matched Analysis of Surveillance, Epidemiology, and End Results Database. J Thorac Oncol 2015;10:1357-63. [Crossref] [PubMed]
  83. Omasa M, Date H, Sozu T, et al. Postoperative radiotherapy is effective for thymic carcinoma but not for thymoma in stage II and III thymic epithelial tumors: the Japanese Association for Research on the Thymus Database Study. Cancer 2015;121:1008-16. [Crossref] [PubMed]
  84. National Comprehensive Cancer Network guidelines. Available online: https://www.nccn.org/professionals/physician_gls/pdf/thymic.pdf. (Accessed on September 18, 2023).
  85. Hayes SA, Huang J, Golia Pernicka J, et al. Radiographic Predictors of Resectability in Thymic Carcinoma. Ann Thorac Surg 2018;106:242-8. [Crossref] [PubMed]
  86. Huang J, Rizk NP, Travis WD, et al. Feasibility of multimodality therapy including extended resections in stage IVA thymoma. J Thorac Cardiovasc Surg 2007;134:1477-83; discussion 1483-4. [Crossref] [PubMed]
  87. Hamaji M, Ali SO, Burt BM. A meta-analysis of surgical versus nonsurgical management of recurrent thymoma. Ann Thorac Surg 2014;98:748-55. [Crossref] [PubMed]
  88. Lucchi M, Davini F, Ricciardi R, et al. Management of pleural recurrence after curative resection of thymoma. J Thorac Cardiovasc Surg 2009;137:1185-9. [Crossref] [PubMed]
  89. Okumura M, Shiono H, Inoue M, et al. Outcome of surgical treatment for recurrent thymic epithelial tumors with reference to world health organization histologic classification system. J Surg Oncol 2007;95:40-4. [Crossref] [PubMed]
  90. Hao XJ, Peng B, Zhou Z, et al. Prospective Study of Stereotactic Body Radiation Therapy for Thymoma and Thymic Carcinoma: Therapeutic Effect and Toxicity Assessment. Sci Rep 2017;7:13549. [Crossref] [PubMed]
  91. Teh BT. Thymic carcinoids in multiple endocrine neoplasia type 1. J Intern Med 1998;243:501-4. [Crossref] [PubMed]
  92. Filosso PL, Yao X, Ruffini E, et al. Comparison of outcomes between neuroendocrine thymic tumours and other subtypes of thymic carcinomas: a joint analysis of the European Society of Thoracic Surgeons and the International Thymic Malignancy Interest Group. Eur J Cardiothorac Surg 2016;50:766-71. [Crossref] [PubMed]
  93. Filosso PL, Ruffini E, Solidoro P, et al. Neuroendocrine tumors of the thymus. J Thorac Dis 2017;9:S1484-90. [Crossref] [PubMed]
  94. Economopoulos GC, Lewis JW Jr, Lee MW, et al. Carcinoid tumors of the thymus. Ann Thorac Surg 1990;50:58-61. [Crossref] [PubMed]
  95. Sakuragi T, Rikitake K, Nastuaki M, et al. Complete resection of recurrent thymic carcinoid using cardiopulmonary bypass. Eur J Cardiothorac Surg 2002;21:152-4. [Crossref] [PubMed]
  96. Fukai I, Masaoka A, Fujii Y, et al. Thymic neuroendocrine tumor (thymic carcinoid): a clinicopathologic study in 15 patients. Ann Thorac Surg 1999;67:208-11. [Crossref] [PubMed]
  97. Modh A, Rimner A, Allen PK, et al. Treatment Modalities and Outcomes in Patients With Advanced Invasive Thymoma or Thymic Carcinoma: A Retrospective Multicenter Study. Am J Clin Oncol 2016;39:120-5. [Crossref] [PubMed]
  98. Koizumi T, Takabayashi Y, Yamagishi S, et al. Chemotherapy for advanced thymic carcinoma: clinical response to cisplatin, doxorubicin, vincristine, and cyclophosphamide (ADOC chemotherapy). Am J Clin Oncol 2002;25:266-8. [Crossref] [PubMed]
  99. Merveilleux du Vignaux C, Dansin E, Mhanna L, et al. Systemic Therapy in Advanced Thymic Epithelial Tumors: Insights from the RYTHMIC Prospective Cohort. J Thorac Oncol 2018;13:1762-70. [Crossref] [PubMed]
  100. Giaccone G, Ardizzoni A, Kirkpatrick A, et al. Cisplatin and etoposide combination chemotherapy for locally advanced or metastatic thymoma. A phase II study of the European Organization for Research and Treatment of Cancer Lung Cancer Cooperative Group. J Clin Oncol 1996;14:814-20. [Crossref] [PubMed]
  101. Loehrer PJ Sr, Kim K, Aisner SC, et al. Cisplatin plus doxorubicin plus cyclophosphamide in metastatic or recurrent thymoma: final results of an intergroup trial. The Eastern Cooperative Oncology Group, Southwest Oncology Group, and Southeastern Cancer Study Group. J Clin Oncol 1994;12:1164-8. [Crossref] [PubMed]
  102. Fornasiero A, Daniele O, Ghiotto C, et al. Chemotherapy for invasive thymoma. A 13-year experience. Cancer 1991;68:30-3. [Crossref] [PubMed]
  103. Lemma GL, Lee JW, Aisner SC, et al. Phase II study of carboplatin and paclitaxel in advanced thymoma and thymic carcinoma. J Clin Oncol 2011;29:2060-5. [Crossref] [PubMed]
  104. Loehrer PJ Sr, Jiroutek M, Aisner S, et al. Combined etoposide, ifosfamide, and cisplatin in the treatment of patients with advanced thymoma and thymic carcinoma: an intergroup trial. Cancer 2001;91:2010-5. [Crossref] [PubMed]
  105. Hellyer JA, Ouseph MM, Padda SK, et al. Everolimus in the treatment of metastatic thymic epithelial tumors. Lung Cancer 2020;149:97-102. [Crossref] [PubMed]
  106. Palmieri G, Buonerba C, Ottaviano M, et al. Capecitabine plus gemcitabine in thymic epithelial tumors: final analysis of a Phase II trial. Future Oncol 2014;10:2141-7. [Crossref] [PubMed]
  107. Bluthgen MV, Boutros C, Fayard F, et al. Activity and safety of oral etoposide in pretreated patients with metastatic or recurrent thymic epithelial tumors (TET): A single-institution experience. Lung Cancer 2016;99:111-6. [Crossref] [PubMed]
  108. Zucali PA, De Pas T, Palmieri G, et al. Phase II Study of Everolimus in Patients With Thymoma and Thymic Carcinoma Previously Treated With Cisplatin-Based Chemotherapy. J Clin Oncol 2018;36:342-9. [Crossref] [PubMed]
  109. Thomas A, Rajan A, Berman A, et al. Sunitinib in patients with chemotherapy-refractory thymoma and thymic carcinoma: an open-label phase 2 trial. Lancet Oncol 2015;16:177-86. [Crossref] [PubMed]
  110. Antonarelli G, Corti C, Zucali PA, et al. Continuous sunitinib schedule in advanced platinum refractory thymic epithelial neoplasms: A retrospective analysis from the ThYmic MalignanciEs (TYME) Italian collaborative group. Eur J Cancer 2022;174:31-6. [Crossref] [PubMed]
  111. Gbolahan OB, Porter RF, Salter JT, et al. A Phase II Study of Pemetrexed in Patients with Recurrent Thymoma and Thymic Carcinoma. J Thorac Oncol 2018;13:1940-8. [Crossref] [PubMed]
  112. Liang Y, Padda SK, Riess JW, et al. Pemetrexed in patients with thymic malignancies previously treated with chemotherapy. Lung Cancer 2015;87:34-8. [Crossref] [PubMed]
  113. Longo F, De Filippis L, Zivi A, et al. Efficacy and tolerability of long-acting octreotide in the treatment of thymic tumors: results of a pilot trial. Am J Clin Oncol 2012;35:105-9. [Crossref] [PubMed]
  114. Loehrer PJ Sr, Wang W, Johnson DH, et al. Octreotide alone or with prednisone in patients with advanced thymoma and thymic carcinoma: an Eastern Cooperative Oncology Group Phase II Trial. J Clin Oncol 2004;22:293-9. [Crossref] [PubMed]
  115. Palmieri G, Merola G, Federico P, et al. Preliminary results of phase II study of capecitabine and gemcitabine (CAP-GEM) in patients with metastatic pretreated thymic epithelial tumors (TETs). Ann Oncol 2010;21:1168-72. [Crossref] [PubMed]
  116. Highley MS, Underhill CR, Parnis FX, et al. Treatment of invasive thymoma with single-agent ifosfamide. J Clin Oncol 1999;17:2737-44. [Crossref] [PubMed]
  117. Conforti F, Pala L, Vivanet G, et al. High-dose continuous-infusion ifosfamide in advanced thymic epithelial Tumors: A TYME network study. Lung Cancer 2023;176:98-102. [Crossref] [PubMed]
  118. Girard N, Lal R, Wakelee H, et al. Chemotherapy definitions and policies for thymic malignancies. J Thorac Oncol 2011;6:S1749-55. [Crossref] [PubMed]
  119. Girard N. Chemotherapy and targeted agents for thymic malignancies. Expert Rev Anticancer Ther 2012;12:685-95. [Crossref] [PubMed]
  120. Sato J, Satouchi M, Itoh S, et al. Lenvatinib in patients with advanced or metastatic thymic carcinoma (REMORA): a multicentre, phase 2 trial. Lancet Oncol 2020;21:843-50. [Crossref] [PubMed]
  121. Giaccone G, Kim C, Thompson J, et al. Pembrolizumab in patients with thymic carcinoma: a single-arm, single-centre, phase 2 study. Lancet Oncol 2018;19:347-55. [Crossref] [PubMed]
  122. Cho J, Kim HS, Ku BM, et al. Pembrolizumab for Patients With Refractory or Relapsed Thymic Epithelial Tumor: An Open-Label Phase II Trial. J Clin Oncol 2019;37:2162-70. [Crossref] [PubMed]
  123. Arunachalam A, Zhang I, Zhao B, et al. Efficacy and safety of treatments for advanced thymic carcinoma after failure of first-line platinum-based chemotherapy: A systematic literature review and meta-analysis. Lung Cancer 2023;176:132-9. [Crossref] [PubMed]
  124. Shahabi V, Berman D, Chasalow SD, et al. Gene expression profiling of whole blood in ipilimumab-treated patients for identification of potential biomarkers of immune-related gastrointestinal adverse events. J Transl Med 2013;11:75. [Crossref] [PubMed]
  125. Jaber SH, Cowen EW, Haworth LR, et al. Skin reactions in a subset of patients with stage IV melanoma treated with anti-cytotoxic T-lymphocyte antigen 4 monoclonal antibody as a single agent. Arch Dermatol 2006;142:166-72. [Crossref] [PubMed]
  126. Katsuya Y, Horinouchi H, Seto T, et al. Single-arm, multicentre, phase II trial of nivolumab for unresectable or recurrent thymic carcinoma: PRIMER study. Eur J Cancer 2019;113:78-86. [Crossref] [PubMed]
  127. Rajan A, Heery CR, Thomas A, et al. Efficacy and tolerability of anti-programmed death-ligand 1 (PD-L1) antibody (Avelumab) treatment in advanced thymoma. J Immunother Cancer 2019;7:269. [Crossref] [PubMed]
  128. Tateishi K, Ko R, Shukuya T, et al. Clinical Outcomes of Second-Line Chemotherapy in Patients with Previously Treated Advanced Thymic Carcinoma: A Retrospective Analysis of 191 Patients from the NEJ023 Study. Oncologist 2020;25:e668-74. [Crossref] [PubMed]
  129. Pavel M, de Herder WW. ENETS Consensus Guidelines for the Standard of Care in Neuroendocrine Tumors. Neuroendocrinology 2017;105:193-5. [Crossref] [PubMed]
  130. Rinke A, Müller HH, Schade-Brittinger C, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol 2009;27:4656-63. [Crossref] [PubMed]
  131. Caplin ME, Pavel M, Ćwikła JB, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med 2014;371:224-33. [Crossref] [PubMed]
  132. Yao JC, Fazio N, Singh S, et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet 2016;387:968-77. [Crossref] [PubMed]
  133. Lang M, Hackert T, Anamaterou C. Long-term effect of everolimus in recurrent thymic neuroendocrine neoplasia. Clin Endocrinol (Oxf) 2021;95:744-51. [Crossref] [PubMed]
  134. Ekeblad S, Sundin A, Janson ET, et al. Temozolomide as monotherapy is effective in treatment of advanced malignant neuroendocrine tumors. Clin Cancer Res 2007;13:2986-91. [Crossref] [PubMed]
  135. Crona J, Björklund P, Welin S, et al. Treatment, prognostic markers and survival in thymic neuroendocrine tumours. a study from a single tertiary referral centre. Lung Cancer 2013;79:289-93. [Crossref] [PubMed]
  136. Saranga-Perry V, Morse B, Centeno B, et al. Treatment of metastatic neuroendocrine tumors of the thymus with capecitabine and temozolomide: a case series. Neuroendocrinology 2013;97:318-21. [Crossref] [PubMed]
  137. Strosberg J, El-Haddad G, Wolin E, et al. Phase 3 Trial of (177)Lu-Dotatate for Midgut Neuroendocrine Tumors. N Engl J Med 2017;376:125-35. [Crossref] [PubMed]
  138. van Essen M, Krenning EP, Bakker WH, et al. Peptide receptor radionuclide therapy with 177Lu-octreotate in patients with foregut carcinoid tumours of bronchial, gastric and thymic origin. Eur J Nucl Med Mol Imaging 2007;34:1219-27. [Crossref] [PubMed]
  139. Takahashi T, Hatao K, Yamashita Y, et al. Ectopic ACTH syndrome due to thymic atypical carcinoid treated with combination chemotherapy of cisplatin and etoposide. Intern Med 2003;42:1197-201. [Crossref] [PubMed]
  140. Clinical Trials Home Page. Available online: https://clinicaltrials.gov (accessed on 22 September 2023).
doi: 10.21037/med-23-47
Cite this article as: Cabezón-Gutiérrez L, Pacheco-Barcia V, Carrasco-Valero F, Palka-Kotlowska M, Custodio-Cabello S, Khosravi-Shahi P. Update on thymic epithelial tumors: a narrative review. Mediastinum 2024;8:33.

Download Citation