Skull base chordomas account for less than 0.2% and chondrosarcomas for less than 0.15% of all intracranial tumors. Although their clinical and imaging presentations are similar, they derive from different origins. Chordomas arise from embryonic remnants of the primitive notochord and chondrosarcomas from primitive mesenchymal cells or from the embryonic rest of the cranial cartilaginous matrix. Both entities are characterized by infiltration and destruction of the surrounding bone and soft tissue and a high locoregional recurrence rate. Chondrosarcomas, when treated with similar complex strategies, display a much better prognosis than chordomas. The overall survival is approximately 65% for chordomas and 80% for chondrosarcomas at 5 years and 30 and 50%, respectively, at 10 years. Chordomas are divided into the following 3 histological types: classical (conventional), chondroid, and dedifferentiated. Chondrosarcomas have conventional, mesenchymal, clear cell, and dedifferentiated subgroups. Both tumor entities often present with nonspecific symptoms, and headaches are the most reported initial symptom. Computed tomography and magnetic resonance imaging are required to determine the tumor localization and the extent of tumor growth. The treatment philosophy is to maximize tumor resection, minimize morbidity, and preserve function. Neurosurgical approaches commonly used for the resection of intracranial chordomas and chondrosarcomas are transsphenoidal, transbasal, cranio-orbitozygomatic, transzygomatic extended middle fossa, transcondylar, and transmaxillary approaches. Chordomas and chondrosarcomas are not sensitive to chemotherapy and there are no approved drugs for their treatment. The present treatment concept is a combination of surgical resection with a maximal excision and preserving patients’ quality of life by adjuvant radiotherapy for both chordomas and chondrosarcomas.

Chordomas and chondrosarcomas are a heterogeneous group of primary malignancies that develop within and invade the skull base region. They represent pathologies that exhibit a wide diversity of oncologic behavior. A standard classification is difficult to apply. It appears appropriate to divide the lesions according to their clinical presentation and radiologic characteristics. For this reason, chordomas and chondrosarcomas have been historically grouped together. However, these tumors are distinct clinicopathological entities and vary significantly in terms of their clinical outcome. Chordomas and chondrosarcomas are rare malignant tumors characterized by slow, destructive, and locally invasive growth. Chordomas arise from embryonic remnants of the primitive notochord with a molecular alteration preceding their malignant transformation. On the other hand, chondro-sarcomas are considered to originate from primitive mesenchymal cells or from embryonic remnants of the cartilaginous matrix in the cranium. Furthermore, chordomas and chondrosarcomas share a high propensity for locoregional recurrence with infiltration and destruction of the surrounding bone and soft tissue. Although the metastatic potential of these malignancies is considered to be relatively low, distant metastases has been reported in patients with advanced disease. This is associated with a poor prognosis and a median survival of less than 12 months. Both of these tumors usually display slow growth patterns and cause gradual displacement of neurovascular structures, in turn leading to the manifestation of clinical signs and subsequent diagnosis. The complex structure of the cranial base, together with the close proximity to cranial nerves and vessels, represents a significant challenge in the management of these tumors. It ought to be noted that aggressive surgery is associated with a considerable risk of high morbidity and mortality. Herein, we present a review of the literature summarizing the epidemiology, histopathology, clinical signs, diagnosis and differential diagnosis, imaging characteristics, and various multimodal treatments with related clinical outcomes of skull base chordomas and chondrosarcomas.

Skull base chordomas are rare malignancies and account for less than 0.2% of all intracranial neoplasms [1, 2]. Several large population-based studies have estimated the overall incidence rate of chordomas to be 0.08 per 100,000 persons per year [3-5]. They may occur in all age groups (although most cases are diagnosed during adulthood, with the mean age of affected person being in the fifth and sixth decades of life) [3], rarely affecting children and adolescents (<5% of all chordomas) [6]. The gender distribution has been reported to be nearly equal in skull base lesions. Chordomas have no known causative association with irradiation or other environmental factors. However, there is a very small percentage of cases displaying a familial pattern of inheritance.

Interestingly, chondrosarcomas also represent approximately 0.15% of all intracranial neoplasms [7] and constitute 6% of all skull base tumors [8, 9]. The peak incidence occurs in the fourth and sixth decades of life with no gender predilection. In a study by Bloch et al. [10], 32% of chondrosarcomas involved the clivus and 27% arose at the temporo-occipital junction. It ought to be noted that distant metastases are very rare for both of these intracranial tumors. The outcome is quite different between chordomas and chondrosarcomas, with the latter displaying a much better prognosis when treated with similar complex strategies.

Chordomas

The first macro- and microscopical description of chordomas was provided by the German physician and pathologist Rudolf Virchow [11]. He described on autopsy an incidental, small, slimy growth on the surface of the clivus. Since that time the intracellular, bubble-like vacuole cells referred to by Virchow [11] as “physaliferous” remain a distinguishing, if not pathognomonic, feature of chordomas. Another German pathologist, i.e., Hugo Ribbert, later proposed the term chordoma [12].

The following 3 histopathological types of chordomas have been identified: classic (conventional), chondroid, and dedifferentiated types [13-15]. The histopathological type of a chordoma predicts the prognosis of this tumor. The first 2 forms have a favorable long-term prognosis, with a 3-year overall survival rate of 90% [13]. The dedifferentiated subtype displays aggressive behavior and only a 60% 3-year overall survival rate [13, 16].

A conventional (classic) chordoma consists of cords and islands of eosinophilic and clear vacuolated cells, also called “physaliphorous cells,” suspended in a myxoid-mucous matrix (Fig. 1a, b). The nuclei are round and uniform, although some exhibit considerable pleomorphism. A thin fibrous septum divides groups of these cells into lobules. This classic pattern of chordoma should not show evidence of chondroid or other mesenchymal tissue differentiation. The second type of chordoma or chondroid chordoma shows regions where the stroma resembles hyaline cartilage and neoplastic, sometimes physaliphorous, cells grow in lacunae. The rare dedifferentiated chordomas are characterized by a frankly malignant, mesenchymal component and have a sarcomatoid appearance. They carry a very poor prognosis. Finally, on immunohistochemistry (Fig. 1c–e), chordomas stain positively for S100, vimentin, epithelial membrane antigen, and pan-cytokeratins [6, 17-20]. “Brachyury” is a new, recently discovered specific diagnostic marker for chordomas [21]. A high brachyury expression was found in chordoma tissue samples [22]. There is no correlation between brachyury expression and clinicopathological parameters in chordoma patients [23]. Other tumors do not show expression of this protein [21, 24]. The determination of brachyury can therefore be used to differentiate tumors with a similar histology and geographical location [25]. However, some poorly differentiated and dedifferentiated areas of chordomas may display a loss of brachyury immunoreactivity [21, 24, 26, 27].

Fig. 1.

Characteristic histologic features of chordomas (a–f) and chondrosarcomas (g–k). a, b HE staining of a chordoma showing epitheloid cells arranged in cords (arrow in a) and detectable extracellular mucin (asterisk in a). In other areas a lobular arrangement can be observed (arrows in b). Chordomas show a characteristic immunohistochemical expression of S100 (c), pan-cytokeratins (d), and vimentin (e). Moderate proliferation activity (Ki67 immunostaining in f). g HE staining showing a well-differentiated chondrosarcoma (G1) with moderate cellularity without mitoses or necrosis. Chondrosarkomas show a characteristic immunohistochemical expression of S100 (h) and vimentin (j), while pan-cytokeratins (i) are not expressed. Low proliferation activity (Ki67 immunostaining in k).

Fig. 1.

Characteristic histologic features of chordomas (a–f) and chondrosarcomas (g–k). a, b HE staining of a chordoma showing epitheloid cells arranged in cords (arrow in a) and detectable extracellular mucin (asterisk in a). In other areas a lobular arrangement can be observed (arrows in b). Chordomas show a characteristic immunohistochemical expression of S100 (c), pan-cytokeratins (d), and vimentin (e). Moderate proliferation activity (Ki67 immunostaining in f). g HE staining showing a well-differentiated chondrosarcoma (G1) with moderate cellularity without mitoses or necrosis. Chondrosarkomas show a characteristic immunohistochemical expression of S100 (h) and vimentin (j), while pan-cytokeratins (i) are not expressed. Low proliferation activity (Ki67 immunostaining in k).

Close modal

Chondrosarcomas

The first histological report of an intracranial chondrosarcoma was published in 1899 by the British biochemist and neuropathologist Sir Frederick Walker Mott [28]. Histologically, chondrosarcomas are characterized by an abundant hyaline type of cartilaginous stroma and the presence of a neoplastic chondrocyte population. The chondrocytes have an inconspicuous cytoplasm and a small, dark nucleus with fine chromatin (Fig. 1g). Infiltration of the bony trabeculae is a histological feature of malignancy. Moreover, extension into the surrounding soft tissue may be observed [29, 30]. Chondrosarcomas can be classified into the following 4 histological subgroups: conventional chondrosarcoma, mesenchymal chondrosar-coma, clear cell chondrosarcoma, and dedifferentiated chondrosarcoma [31, 32]. It is well known that chondrosarcomas demonstrate recognizable histological grades of differentiation. Based on cellularity, mitotic activity, atypia, and nuclear size, the WHO introduced the following 3 classes: grade I (well differentiated), grade II (intermediately differentiated), and grade III (poorly differentiated) [33]. Grade I and II chondrosarcomas show a better outcome, whereas grade III chondrosarcomas are associated with a high recurrence rate and even metastases [34]. Chondrosarcomas stain positive for S-100 and vimentin but fail to express epithelial markers such as cytokeratin and epithelial membrane antigen (Fig. 1h, I, j), [35].

Clinical Signs and Symptoms

Patients with chordomas and chondrosarcomas usually present with nonspecific and sometimes confusing symptoms, which markedly delays the diagnosis until later stages of the disease [36-39]. The initial presentation of the tumors can significantly vary depending on the location, extension, and proximity of the lesion to critical structures. Besides headaches typically located in the occipital or retro-orbital region, the most commonly reported signs are neuro-ophthalmological [37-42]. The visual symptoms may include blurred vision or loss of vision, ptosis, and visual field defects related to cranial nerve palsies which could be explained by the location and growth pattern of the tumors [43-47]. It is not unusual for this kind of tumor to invade structures such as the cavernous sinus, the petroclival region, the cerebellopontine angle, the infratemporal fossa, the parapharyngeal space, and the temporal bone [42, 44, 46]. Other signs and symptoms that may present with clival lesions include hearing loss, facial paralysis or hypoesthesia, dysphonia, dysarthria, dysphagia, dyspnea, anosmia, and vertigo [44, 48, 49]. Involvement of the suprasellar space may lead to hypopituitarism and diabetes insipidus through damage of the pituitary gland or the stalk and visual impairment through damage to the optic apparatus, respectively [50, 51]. Larger tumors may also compress the brainstem and cerebellum, causing gait disturbances, ataxia, dysmetria, and motor weakness (e.g., hemiparesis or tetraparesis) [44, 51, 52]. The nature of the tumor cannot be distinguished on the basis of the clinical presentation alone.

Chordomas and chondrosarcomas can arise within the sellar/parasellar region but also extend into this region. In a large transsphenoidal surgical series about 10% of nonpituitary sellar lesions were found to be cartilaginous tumors. Chordomas were diagnosed more frequently than chondrosarcomas [53, 54]. It is well known, that neither tumor entity secretes any bioactive compounds or hormones. Although there are a few published cases about endocrine abnormalities in patients presenting with these tumors, a significant endocrinopathy is highly unusual [53-59]. The most reported complaints were amenorrhea, galactorrhea, and male sexual dysfunction [56, 58-60]. The severity of these symptoms was correlated with the level of hyperprolactinemia secondary to compression of the pituitary stalk. In rare cases, aggressive tumor growth may lead to partial or complete destruction of the pituitary gland, resulting in correspondingly variable degrees of hormonal deficiency [57, 61, 62]. Depending on the imaging peculiarities and presenting symptoms, a lack of systematic measurement of the preoperative hormonal status as part of the diagnostic work-up could lead to the failed diagnosis of not clinically apparent hormonal deficiencies.

Computed tomography (CT) and magnetic resonance imaging (MRI) are the preferred diagnostic tools for the initial evaluation of patients with suspicion of skull base neoplasms [63-65]. Preoperative sagittal and coronal multiplanar reconstructions are essential for evaluation of the detailed tumor’s relationship to adjacent brain structures and hence planning of the optimal surgical treatment. In the follow-up, these planes are needed to detect treatment-related complications and tumor recurrences [66]. Since both tumors display an equally wide diversity in terms of their localization as well as the pattern and extent of invasive growth within the skull base, neither MRI nor CT can be useful for differentiating chordomas from chondrosarcomas preoperatively [63-66].

High-Resolution CT

A high-resolution CT scan is sensitive enough to detect lesions of the skull base and should be performed with intravenous contrast and reviewed in bone and soft-tissue windows. Intracranial chordomas and chondrosarcomas are both delineated in CT scans, as are well-defined expansive tissue masses that arise from the clivus with associated osteolytic transformation with frequent expansion to the adjacent soft tissue [67, 68]. The tumors are seen on noncontrast CT as isodense or slightly hypodense in comparison to normal brain tissue. Intratumoral calcifications are mostly found in chondrosarcomas and are hence suggestive of this diagnosis.

Magnetic Resonance Imaging

MRI provides optimal resolution and soft tissue contrast with exquisite anatomic details in the evaluation of intracranial lesions in comparison to CT scans [69, 70]. The sagittal planar images are indispensable for identification of the posterior tumor margins, as well as their relation to the brainstem, and the coronal planar images are essential for identification of the lateral tumor margins and the tumor extension into the cavernous sinus. Intracranial chordomas and chondrosarcomas are characterized by an intermediate-to-low signal intensity on spin-echo T1-weighted MR sequences and they are recognized within the high signal intensity of the clivus fat tissue. Small foci of hyperintensity recognized in T1-weighted sequences, appearing as bright spots and dark areas on gradient-weighted sequences, represent intratumoral hemorrhage or a mucus pool. Furthermore, the tumors show a high signal intensity on T2-weighted images. The sagittal and axial T1-weighted, postcontrast, fat-suppressed images also help to distinguish tumor from fat in the adjacent bone marrow spaces, since normal bone marrow in adults is characterized by a bright signal intensity on both T1-weighted images and fast spin echo T2-weighted images [71, 72]. Clival chordomas and chondrosarcomas may exhibit similar growth patterns by extending anteriorly, laterally, posteriorly, inferiorly, and superiorly with affection of the sellar/parasellar area, the middle cranial fossa, the prepontine cistern, and the foramen magnum (Fig. 2, 3). Nevertheless, a slight difference in predilection in terms of origin with chordomas tending to arise from the midline as opposed to more laterally for chondrosarcomas may be of use in the initial differential diagnosis on MRI.

Fig. 2.

MRI and CT scans of intracranial chordoma. Axial (a–c), sagittal (e), and coronal (g) T1-weighted and axial T2-weighted (c) MR images show a tumor with clival/retroclival extension. b, e, g Contrast-enhanced images. Axial (d) and sagittal (f) CT scans demonstrate involvement of the clivus.

Fig. 2.

MRI and CT scans of intracranial chordoma. Axial (a–c), sagittal (e), and coronal (g) T1-weighted and axial T2-weighted (c) MR images show a tumor with clival/retroclival extension. b, e, g Contrast-enhanced images. Axial (d) and sagittal (f) CT scans demonstrate involvement of the clivus.

Close modal
Fig. 3.

MRI scans of intracranial chondrosarcoma. Coronal (a, b), sagittal (d), and axial (e, f) T1-weighted and coronal (c) and axial (g) T2-weighted MR images demonstrate a tumor with extension into the posterior fossa. b, e, g Contrast-enhanced images.

Fig. 3.

MRI scans of intracranial chondrosarcoma. Coronal (a, b), sagittal (d), and axial (e, f) T1-weighted and coronal (c) and axial (g) T2-weighted MR images demonstrate a tumor with extension into the posterior fossa. b, e, g Contrast-enhanced images.

Close modal

Surgery

Treatment of intracranial chordomas and chondrosarcomas still represents a big challenge despite advances in neurosurgical techniques [73-77]. The surgery philosophy is to remove as much tumor as possible and to preserve or improve the patient’s status. Macroscopic complete resection with a negative surgical margin is usually impossible due to aggressive and infiltrative tumor growth pattern with involvement of the surrounding neurological structures [51, 73, 78-83]. Many surgical approaches have been described for the resection of skull base chordomas and chondrosarcomas. Taking into consideration the aggressive nature of these tumors and especially the patient’s expectations, the best approaches or even multiple approaches must be selected on an individual basis. It is important to consider the common and serious complications that may occur following the resection of skull base lesions. Many authors report new cranial nerve deficits in up to 80% of the patients, which were frequently transient in the majority of cases. Other complications include cerebrospinal fluid leaks, meningitis, hearing loss, visual decline, and consequences of vascular injury [34, 84]. Neurosurgical approaches often used in the resection of intracranial chordomas and chondrosarcomas include the transsphenoidal, transbasal, cranio-orbitozygomatic, transzygomatic extended middle fossa, transcondylar, and transmaxillary approaches [34, 79-92]. Their applications are summarized as follows.

Transsphenoidal Approach. The transsphenoidal approach is a well-established method for the treatment of tumors limited to the upper clivus, the sellar und suprasellar regions, or the medial aspect of the cavernous sinus. This approach provides visualization to many intracranial anatomical structures (the internal carotid arteries, the pituitary gland, the infundibulum, the optic nerve and chiasm, the mammillary bodies, the basilar artery bifurcation, the hypothalamus etc.). This approach, mostly in an extended fashion, permits tumor resection with minimal displacement or distortion of surrounding tissue. The neurosurgeon can work effectively either above or below the optic chiasm, depending on its situation relative to the lesion. The transsphenoidal approach is the one most often used for the treatment of chordomas and chondrosarcomas in our institute.

Transbasal Approach. There are a number of modifications of the transbasal approach (e.g., uni- or bilateral orbital-cranial subfrontal transbasal, nasofrontal transbasal). It creates the possibility to reach the midline clivus, the medial cavernous sinus, the petrous apex, the occipital condyles, and the foramen magnum. However, this approach is a midline approach between the 2 optic nerves and cannot expose tumors located laterally to the internal carotid artery (ICA). The main disadvantage is the narrow and deep corridor between the optic nerves and the ICA.

Cranio-Orbitozygomatic Approach. This approach provides the shortest route with full exposure of lesions displacing the brain structures superiorly and laterally in the parasellar region. It gives a multidirectional view of the tumor and allows surgical dissection via multiple routes (subfrontal-transbasal, transsylvian, subtemporal, and infratemporal). Many authors have reported using this approach for tumors in the region of the upper clivus with extension lateral to the ICA and into the middle fossa, the petrous apex, and the infratemporal fossa.

Transzygomatic Extended Middle Fossa Approach. This approach has become useful in the treatment of lesions located in the infratemporal, sphenopalatine, and temporal fossae, the orbit, the petrous apex, the upper petroclival region, and the cavernous sinus. It creates a wider surgical corridor offering a direct view of the clivus and brainstem. Tumors extended in the sphenoid sinus can be removed through the Parkinson triangle between the first and second divisions of the fifth cranial nerve.

Transcondylar Approach. Many indications have been reported for the transcondylar approach. In selected cases, it allows approach to chordomas/chondrosarcoma in the region of the inferior clivus with lateral extension to the craniocervical junction or the upper cervical vertebrae. It requires extraperiosteal management of the vertebral artery. This approach can be combined with a transtemporal approach superiorly and a transcervical approach inferiorly. However, the surgery can cause cerebellar damage, vertebral artery injury, or occipito-cervical instability, where extensive condyle resection may even require future occipito-cervical fusion.

Transmaxillary Approach. This technique enables tumor resection located in the clivus area with extension into the nasopharynx or craniocervical junction, inferior to the anterior cranial fossa, superior to the C2-C3 interspace, and medial to the pterygoid plate. The maxillary osteotomy provides a direct view of the clivus region, the posterior nasooropharynx, and the cervical spine. This approach is associated in appropriately selected patients with limited morbidity and mortality. Procedurally related complications include damage to neurovascular structures along the paramedian skull base, including maxillary, mandibular, and vidian neurovascular bundles. The risk of aseptic bone necrosis is increased with multisegmented osteotomies and intraoperative hypotension.

Radiotherapy

The recent literature supports the concept of a combination treatment for chordomas and chondrosarcomas consisting of surgical resection with maximal excision followed by adjuvant radiotherapy [84, 93-97]. It is believed that radiation therapy leads to clearance of the tumor bed from invisible, microscopic tumor nests. The most common adjuvant treatment methods for chordomas and chondrosarcomas include high-dose radiotherapy with proton beam radiation and radiosurgery using a gamma knife and a CyberKnife [98-102]. However, there are no published studies directly comparing these forms of radiation therapy [103]. Retrospective studies have shown a longer local progression-free time following adjuvant radiotherapy. Conventional radiotherapy does not appear to increase the survival duration. The 5-year recurrence-free survival for cranial base chordomas after proton-based therapy ranges between 59 and 73% [94, 100, 104]. In contrast, skull base chondrosarcomas after the same proton-based therapy are characterized by a disease-specific 5-year survival rate of about 99% [102]. Adjuvant radiotherapy is not indicated for chondrosarcomas of WHO grade I and II in case of complete resection, as opposed to chordomas where this is universally advocated.

Chemotherapy

Traditionally, chordomas and chondrosarcomas are not sensitive to chemotherapy [105, 106] and there are no drugs approved for their treatment [106-108]. Dysregulation of different signaling pathways has been found in the development of chordomas and chondrosarcomas [109-119]. Agents that interrupt these signaling pathways could become attractive targets for anticancer therapy. Tyrosine kinase inhibitors (TKI) showed activity against chordomas as a result of targeted inhibition of several intersecting molecular pathways [120]. In a small number of patients treated with the TKI sunitinib, stable disease for at least 16 weeks was achieved [121]. Other studies have demonstrated that TKI, such as imatinib mesylate and erlonitib, and epidermal growth factor receptor inhibitors such as cetuximab and gefitinib, have a beneficial effect on the treatment of advanced or metastatic chordomas [122-124]. TKI, mTOR, hedgehog, and AKT inhibitors have also been tested in clinical studies for chondrosarcoma patients [115-119]. Unfortunately, most of them have failed to demonstrate a clinical benefit in terms of disease progression or outcomes. However, all clinical reports have been limited by a small patient numbers and short follow-ups. Finally, the published studies are clinical phase I/II studies, rendering it difficult to arrive at any conclusions.

It is of interest to note that immunotherapy based on enhancement of the immune system to eliminate cancer cells has developed into a promising modality in cancer therapy [125-127]. As in the case of other human cancers, it has recently been demonstrated that some fractions of chordoma cells escape immune modulated destruction by upregulating the programmed death-1 receptor (PD-1) and programmed cell death ligand 1 (PD-L1) [128-130]. Many checkpoint inhibitor (ICI) antibodies blocking the PD-1/PD-L1 pathways have been developed and introduced into clinical practice [131, 132]. Migliorini et al. [133] have published anecdotal cases of clinical responses to immunotherapy with ICI in relapsing chordomas. Although there are currently several clinical studies on chordomas and ICI (for details, please see https://clinicaltrials.gov), it remains too early to speculate on whether adding ICI antibodies to existing treatment protocols of chordomas could improve the outcome.

There are no reliable markers for the prognosis of skull base chondrosarcomas and chordomas known to date. However, Weber et al. [134] identified histology, gross tumor volume, and brain compression as independent prognostic factors for local control and overall survival (OS) based on multivariate analysis. On the other hand, several authors have considered the prognosis to be based on multiple factors [3-5, 8, 42, 78, 79, 82, 84, 92-94]. Dedifferentiated chordomas exhibit a particularly poor prognosis with an aggressive clinical course. This subtype has a 60% 3-year OS compared to an 89.4% 3-year OS for classical chordomas [10, 15, 16, 79]. Poorly differentiated or grade III chondrosarcomas show aggressive behavior and are associated with the lowest survival rates [10]. Tumor volume has been incorporated in the TNM staging system and can be considered as a potential prognostic factor. Kamrin et al. [135] reported a mean survival time for untreated chordoma patients ranging from 6 to 24 months. According to published data, the median OS of patients with chordomas after surgery and radiation was 6.29 years [3], the 5-year progression-free survival varied from 45 to 56% [3, 8, 9] and 10-year OS rates ranged from 32 to 60% [16]. Metastases from chordomas from all anatomic sites have been estimated to vary from 9 to 41% [78, 136-139]. In a systematic review of 560 patients with intracranial chondrosarcomas, the calculated 5-year mortality was 11.5%, with a median survival time of 24 months [10]. The Surveillance, Epidemiology, and End Results program of the National Cancer Institute shows an OS of 65% for chordomas and 82% for chondrosarcomas at 5 years, as well as rates of 32 and 50% at 10 years, respectively [140].

Skull base chordomas and chondrosarcomas are local invasive and destructive growing tumors with a high locoregional recurrence rate and rare distant metastases. The patients often have nonspecific and confusing symptoms independently of the tumor subtype. CT and MRI still represent equally valuable diagnostic tools for the initial evaluation of patients with a suspected brain disease. Preoperative differentiation between chordomas and chondrosarcomas based on the clinical presentation and clinical and instrument-based diagnostics alone is impossible. The treatment philosophy for both intracranial tumor entities is to maximize tumor resection, minimize morbidity, and preserve function. The appropriate approaches or even multiple approaches must be determined on a case-by-case basis. Surgery should be followed by adjuvant radiotherapy to improve the local progression-free time, where proton-beam therapy has been shown to be more profitable than conventional radiotherapy. To date, there are no drug agents approved for the treatment of chordomas and chondrosarcomas. In recapitulation, from our personal point of view, intracranial skull base chordomas and chondrosarcomas should be subjected to the maximum possible cytoreduction via an appropriate approach without deterioration of the general condition and without incurring in neurological deficits followed by radiotherapy in the form of proton beam therapy.

The authors have no ethical conflicts to disclose.

The authors have no conflict of interests to declare

There was no funding support for this article.

Conception, design, and writing of this paper: N.K. and M.B. Redaction: S.-M.S., R.C., T.G., and T.M.K. All of the authors approved the final submitted version of this work.

1.
Mizerny
BR
,
Kost
KM
.
Chordoma of the cranial base: the McGill experience
.
J Otolaryngol
.
1995
Feb
;
24
(
1
):
14
9
.
[PubMed]
0381-6605
2.
Lee
SY
,
Lim
YC
,
Song
MH
,
Seok
JY
,
Lee
WS
,
Choi
EC
.
Chondrosarcoma of the head and neck
.
Yonsei Med J
.
2005
Apr
;
46
(
2
):
228
32
.
[PubMed]
0513-5796
3.
McMaster
ML
,
Goldstein
AM
,
Bromley
CM
,
Ishibe
N
,
Parry
DM
.
Chordoma: incidence and survival patterns in the United States, 1973-1995
.
Cancer Causes Control
.
2001
Jan
;
12
(
1
):
1
11
.
[PubMed]
0957-5243
4.
Eriksson
B
,
Gunterberg
B
,
Kindblom
LG
.
Chordoma. A clinicopathologic and prognostic study of a Swedish national series
.
Acta Orthop Scand
.
1981
Feb
;
52
(
1
):
49
58
.
[PubMed]
0001-6470
5.
O’Neill
P
,
Bell
BA
,
Miller
JD
,
Jacobson
I
,
Guthrie
W
.
Fifty years of experience with chordomas in southeast Scotland
.
Neurosurgery
.
1985
Feb
;
16
(
2
):
166
70
.
[PubMed]
0148-396X
6.
Hoch
BL
,
Nielsen
GP
,
Liebsch
NJ
,
Rosenberg
AE
.
Base of skull chordomas in children and adolescents: a clinicopathologic study of 73 cases
.
Am J Surg Pathol
.
2006
Jul
;
30
(
7
):
811
8
.
[PubMed]
0147-5185
7.
Cianfriglia
F
,
Pompili
A
,
Occhipinti
E
.
Intracranial malignant cartilaginous tumours. Report of two cases and review of literature
.
Acta Neurochir (Wien)
.
1978
;
45
(
1-2
):
163
75
.
[PubMed]
0001-6268
8.
Harvey
SA
,
Wiet
RJ
,
Kazan
R
.
Chondrosarcoma of the jugular foramen
.
Am J Otol
.
1994
Mar
;
15
(
2
):
257
63
.
[PubMed]
0192-9763
9.
Donaldson
DR
,
Myers
LL
,
Diaz-Ordaz
E
,
Grand
W
,
Paterson
J
,
Wax
MK
.
Pathologic quiz case 2. Chondrosarcoma of the jugular foramen
.
Arch Otolaryngol Head Neck Surg
.
1999
Feb
;
125
(
2
):
229
.
[PubMed]
0886-4470
10.
Bloch
OG
,
Jian
BJ
,
Yang
I
,
Han
SJ
,
Aranda
D
,
Ahn
BJ
, et al
A systematic review of intracranial chondrosarcoma and survival
.
J Clin Neurosci
.
2009
Dec
;
16
(
12
):
1547
51
.
[PubMed]
0967-5868
11.
Virchow
RL
.
Untersuchungen über die Entwickelung des Schädelgrundes im gesunden und krankhaften Zustande: und über den Einfluss derselben auf Schädelform, Gesichtsbildung und Gehirnbau
.
Berlin
:
G Reimer
;
1857
.
12.
Williams
LW
.
The later development of the notochord in mammals
.
Am J Anat
.
1908
;
8
(
1
):
251
84
. 0002-9106
13.
Chugh
R
,
Tawbi
H
,
Lucas
DR
,
Biermann
JS
,
Schuetze
SM
,
Baker
LH
.
Chordoma: the nonsarcoma primary bone tumor
.
Oncologist
.
2007
Nov
;
12
(
11
):
1344
50
.
[PubMed]
1083-7159
14.
Ouyang
T
,
Zhang
N
,
Zhang
Y
,
Jiao
J
,
Ren
J
,
Huang
T
, et al
Clinical characteristics, immunohistochemistry, and outcomes of 77 patients with skull base chordomas
.
World Neurosurg
.
2014
May-Jun
;
81
(
5-6
):
790
7
.
[PubMed]
1878-8750
15.
Menezes
AH
.
Clival and craniovertebral junction chordomas
.
World Neurosurg
.
2014
May-Jun
;
81
(
5-6
):
690
2
.
[PubMed]
1878-8750
16.
Casali
PG
,
Stacchiotti
S
,
Sangalli
C
,
Olmi
P
,
Gronchi
A
.
Chordoma
.
Curr Opin Oncol
.
2007
Jul
;
19
(
4
):
367
70
.
[PubMed]
1040-8746
17.
Abenoza
P
,
Sibley
RK
.
Chordoma: an immunohistologic study
.
Hum Pathol
.
1986
Jul
;
17
(
7
):
744
7
.
[PubMed]
0046-8177
18.
Meis
JM
,
Giraldo
AA
.
Chordoma. An immunohistochemical study of 20 cases
.
Arch Pathol Lab Med
.
1988
May
;
112
(
5
):
553
6
.
[PubMed]
0003-9985
19.
Wick
MR
,
Burgess
JH
,
Manivel
JC
.
A reassessment of “chordoid sarcoma”. Ultrastructural and immunohistochemical comparison with chordoma and skeletal myxoid chondrosarcoma
.
Mod Pathol
.
1988
Nov
;
1
(
6
):
433
43
.
[PubMed]
0893-3952
20.
Walker
WP
,
Landas
SK
,
Bromley
CM
,
Sturm
MT
.
Immunohistochemical distinction of classic and chondroid chordomas
.
Mod Pathol
.
1991
Sep
;
4
(
5
):
661
6
.
[PubMed]
0893-3952
21.
Vujovic
S
,
Henderson
S
,
Presneau
N
,
Odell
E
,
Jacques
TS
,
Tirabosco
R
, et al
Brachyury, a crucial regulator of notochordal development, is a novel biomarker for chordomas
.
J Pathol
.
2006
Jun
;
209
(
2
):
157
65
.
[PubMed]
0022-3417
22.
Romeo
S
,
Hogendoorn
PC
.
Brachyury and chordoma: the chondroid-chordoid dilemma resolved?
J Pathol
.
2006
Jun
;
209
(
2
):
143
6
.
[PubMed]
0022-3417
23.
Zhang
L
,
Guo
S
,
Schwab
JH
,
Nielsen
GP
,
Choy
E
,
Ye
S
, et al
Tissue microarray immunohistochemical detection of brachyury is not a prognostic indicator in chordoma
.
PLoS One
.
2013
Sep
;
8
(
9
):
e75851
.
[PubMed]
1932-6203
24.
Miettinen
M
,
Wang
Z
,
Lasota
J
,
Heery
C
,
Schlom
J
,
Palena
C
.
Nuclear brachyury expression is consistent in chordoma, common in germ cell tumors and small cell carcinomas, and rare in other carcinomas and sarcomas: an immunohistochemical study of 5229 cases
.
Am J Surg Pathol
.
2015
Oct
;
39
(
10
):
1305
12
.
[PubMed]
0147-5185
25.
Barresi
V
,
Ieni
A
,
Branca
G
,
Tuccari
G
.
Brachyury: a diagnostic marker for the differential diagnosis of chordoma and hemangioblastoma versus neoplastic histological mimickers
.
Dis Markers
.
2014
;
2014
:
514753
.
[PubMed]
0278-0240
26.
Jambhekar
NA
,
Rekhi
B
,
Thorat
K
,
Dikshit
R
,
Agrawal
M
,
Puri
A
.
Revisiting chordoma with brachyury, a “new age” marker: analysis of a validation study on 51 cases
.
Arch Pathol Lab Med
.
2010
Aug
;
134
(
8
):
1181
7
.
[PubMed]
1543-2165
27.
Oakley
GJ
,
Fuhrer
K
,
Seethala
RR
.
Brachyury, SOX-9, and podoplanin, new markers in the skull base chordoma vs chondrosarcoma differential: a tissue microarray-based comparative analysis
.
Mod Pathol
.
2008
Dec
;
21
(
12
):
1461
9
.
[PubMed]
0893-3952
28.
Mott
FW
.
Chondrosarcoma springing from the sella turcica
.
Arch Neurol Psychiatry
.
1899
;
1
:
432
3
.0096-6754
29.
Varma
DG
,
Ayala
AG
,
Carrasco
CH
,
Guo
SQ
,
Kumar
R
,
Edeiken
J
.
Chondrosarcoma: MR imaging with pathologic correlation
.
Radiographics
.
1992
Jul
;
12
(
4
):
687
704
.
[PubMed]
0271-5333
30.
Suster
D
,
Hung
YP
,
Nielsen
GP
.
Differential diagnosis of cartilaginous lesions of bone
.
Arch Pathol Lab Med
.
2020
Jan
;
144
(
1
):
71
82
.
[PubMed]
0003-9985
31.
Evans
HL
,
Ayala
AG
,
Romsdahl
MM
.
Prognostic factors in chondrosarcoma of bone: a clinicopathologic analysis with emphasis on histologic grading
.
Cancer
.
1977
Aug
;
40
(
2
):
818
31
.
[PubMed]
0008-543X
32.
Fletcher
CD
.
The evolving classification of soft tissue tumours - an update based on the new 2013 WHO classification
.
Histopathology
.
2014
Jan
;
64
(
1
):
2
11
.
[PubMed]
0309-0167
33.
Hogendoorn
PC
,
Bovee
JV
,
Nielsen
GP
. Chondrosarcoma (grades I-III), including primary and secondary variants and periosteal chondrosarcoma. In:
Fletcher
CD
,
Bridge
JA
,
Hogendoorn
PC
,
Mertens
F
, editors
.
WHO classification of tumours of soft tissue and bone
. 4th ed.
Lyon
:
IARC Press
;
2013
. pp.
264
8
.
34.
Gay
E
,
Sekhar
LN
,
Rubinstein
E
,
Wright
DC
,
Sen
C
,
Janecka
IP
, et al
Chordomas and chondrosarcomas of the cranial base: results and follow-up of 60 patients
.
Neurosurgery
.
1995
May
;
36
(
5
):
887
96
.
[PubMed]
0148-396X
35.
Rosenberg
AE
,
Brown
GA
,
Bhan
AK
,
Lee
JM
.
Chondroid chordoma—a variant of chordoma. A morphologic and immunohistochemical study
.
Am J Clin Pathol
.
1994
Jan
;
101
(
1
):
36
41
.
[PubMed]
0002-9173
36.
Lim
GH
.
Clivus chordoma with unusual bone sclerosis and brainstem invasion. A case report with review of the radiology of cranial chordomas
.
Australas Radiol
.
1975
Sep
;
19
(
3
):
242
50
.
[PubMed]
0004-8461
37.
Roberti
F
,
Sekhar
LN
,
Jones
RV
,
Wright
DC
.
Intradural cranial chordoma: a rare presentation of an uncommon tumor. Surgical experience and review of the literature
.
J Neurosurg
.
2007
Feb
;
106
(
2
):
270
4
.
[PubMed]
0022-3085
38.
Neff
B
,
Sataloff
RT
,
Storey
L
,
Hawkshaw
M
,
Spiegel
JR
.
Chondrosarcoma of the skull base
.
Laryngoscope
.
2002
Jan
;
112
(
1
):
134
9
.
[PubMed]
0023-852X
39.
Lustig
LR
,
Sciubba
J
,
Holliday
MJ
.
Chondrosarcomas of the skull base and temporal bone
.
J Laryngol Otol
.
2007
Aug
;
121
(
8
):
725
35
.
[PubMed]
0022-2151
40.
Volpe
NJ
,
Liebsch
NJ
,
Munzenrider
JE
,
Lessell
S
.
Neuro-ophthalmologic findings in chordoma and chondrosarcoma of the skull base
.
Am J Ophthalmol
.
1993
Jan
;
115
(
1
):
97
104
.
[PubMed]
0002-9394
41.
Bagan
SM
,
Hollenhorst
RW
.
Ocular manifestations of intracranial chordomas
.
Trans Am Ophthalmol Soc
.
1980
;
78
:
148
55
.
[PubMed]
0065-9533
42.
Lanzino
G
,
Sekhar
LN
,
Hirsch
WL
,
Sen
CN
,
Pomonis
S
,
Snyderman
CH
.
Chordomas and chondrosarcomas involving the cavernous sinus: review of surgical treatment and outcome in 31 patients
.
Surg Neurol
.
1993
Nov
;
40
(
5
):
359
71
.
[PubMed]
0090-3019
43.
Masui
K
,
Kawai
S
,
Yonezawa
T
,
Fujimoto
K
,
Nishi
N
.
Intradural retroclival chordoma without bone involvement - case report
.
Neurol Med Chir (Tokyo)
.
2006
Nov
;
46
(
11
):
552
5
.
[PubMed]
0470-8105
44.
Korinth
M
,
Schönrock
L
,
Mayfrank
L
,
Gilsbach
JM
.
Primary intradural pontocerebellar chordoma metastasizing in the subarachnoid spinal canal
.
Zentralbl Neurochir
.
1999
;
60
(
3
):
146
50
.
[PubMed]
0044-4251
45.
Mapstone
TB
,
Wongmongkolrit
T
,
Roessman
U
,
Ratcheson
RA
.
Intradural chondroma: a case report and review of the literature
.
Neurosurgery
.
1983
Jan
;
12
(
1
):
111
4
.
[PubMed]
0148-396X
46.
Wolfe
JT
 3rd
,
Scheithauer
BW
.
“Intradural chordoma” or “giant ecchordosis physaliphora”? Report of two cases
.
Clin Neuropathol
.
1987
May-Jun
;
6
(
3
):
98
103
.
[PubMed]
0722-5091
47.
Tashiro
T
,
Fukuda
T
,
Inoue
Y
,
Nemoto
Y
,
Shakudo
M
,
Katsuyama
J
, et al
Intradural chordoma: case report and review of the literature
.
Neuroradiology
.
1994
May
;
36
(
4
):
313
5
.
[PubMed]
0028-3940
48.
Hardie
RC
.
Magnetic resonance appearance of a rare intradural chordoma
.
Wis Med J
.
1992
Nov
;
91
(
11
):
627
8
.
[PubMed]
0043-6542
49.
Korten
AG
,
ter Berg
HJ
,
Spincemaille
GH
,
van der Laan
RT
,
Van de Wel
AM
.
Intracranial chondrosarcoma: review of the literature and report of 15 cases
.
J Neurol Neurosurg Psychiatry
.
1998
Jul
;
65
(
1
):
88
92
.
[PubMed]
0022-3050
50.
Mathews
W
,
Wilson
CB
.
Ectopic intrasellar chordoma. Case report
.
J Neurosurg
.
1974
Feb
;
40
(
2
):
260
3
.
[PubMed]
0022-3085
51.
Zhang
Z
,
Pang
LJ
,
Wang
N
,
Li
Z
,
Cao
YW
,
Hu
WH
, et al
Low-Grade Chondrosarcoma In The Sellar Area: Case Report And Literature Review
.
OncoTargets Ther
.
2019
Dec
;
12
:
10763
70
.
[PubMed]
1178-6930
52.
Asioli
S
,
Zoli
M
,
Guaraldi
F
,
Sollini
G
,
Bacci
A
,
Gibertoni
D
, et al
Peculiar pathological, radiological and clinical features of skull-base de-differentiated chordomas. Results from a referral centre case-series and literature review
.
Histopathology
.
2020
Apr
;
76
(
5
):
731
9
.
[PubMed]
0309-0167
53.
Freda
PU
,
Wardlaw
SL
,
Post
KD
.
Unusual causes of sellar/parasellar masses in a large transsphenoidal surgical series
.
J Clin Endocrinol Metab
.
1996
Oct
;
81
(
10
):
3455
9
.
[PubMed]
0021-972X
54.
Freda
PU
,
Post
KD
.
Differential diagnosis of sellar masses
.
Endocrinol Metab Clin North Am
.
1999
Mar
;
28
(
1
):
81
117
.
[PubMed]
0889-8529
55.
Thodou
E
,
Kontogeorgos
G
,
Scheithauer
BW
,
Lekka
I
,
Tzanis
S
,
Mariatos
P
, et al
Intrasellar chordomas mimicking pituitary adenoma
.
J Neurosurg
.
2000
Jun
;
92
(
6
):
976
82
.
[PubMed]
0022-3085
56.
Allan
CA
,
Kaltsas
G
,
Evanson
J
,
Geddes
J
,
Lowe
DG
,
Plowman
PN
, et al
Pituitary chondrosarcoma: an unusual cause of a sellar mass presenting as a pituitary adenoma
.
J Clin Endocrinol Metab
.
2001
Jan
;
86
(
1
):
386
91
.
[PubMed]
0021-972X
57.
Hirosawa
RM
,
Santos
AB
,
França
MM
,
Fabris
VE
,
Castro
AV
,
Zanini
MA
, et al
Intrasellar chondroid chordoma: a case report
.
ISRN Endocrinol
.
2011
;
2011
:
259392
.
[PubMed]
2090-4630
58.
Hattori
Y
,
Tahara
S
,
Nakakuki
T
,
Takei
M
,
Ishii
Y
,
Teramoto
A
, et al
Sellar chondroma with endocrine dysfunction that resolved after surgery: case report
.
J Nippon Med Sch
.
2015
;
82
(
3
):
146
50
.
[PubMed]
1345-4676
59.
Cao
J
,
Li
G
,
Sun
Y
,
Hong
X
,
Huang
H
.
Sellar chondrosarcoma presenting with amenorrhea: A case report
.
Medicine (Baltimore)
.
2018
Jul
;
97
(
27
):
e11274
.
[PubMed]
0025-7974
60.
Glezer
A
,
Paraiba
DB
,
Bronstein
MD
.
Rare sellar lesions
.
Endocrinol Metab Clin North Am
.
2008
Mar
;
37
(
1
):
195
211
.
[PubMed]
0889-8529
61.
Abele
TA
,
Yetkin
ZF
,
Raisanen
JM
,
Mickey
BE
,
Mendelsohn
DB
.
Non-pituitary origin sellar tumours mimicking pituitary macroadenomas
.
Clin Radiol
.
2012
Aug
;
67
(
8
):
821
7
.
[PubMed]
0009-9260
62.
Kagawa
T
,
Takamura
M
,
Moritake
K
,
Tsutsumi
A
,
Yamasaki
T
.
A case of sellar chordoma mimicking a non-functioning pituitary adenoma with survival of more than 10 years
.
Noshuyo Byori
.
1993
;
10
(
2
):
103
6
.
[PubMed]
0914-8108
63.
Oot
RF
,
Melville
GE
,
New
PF
,
Austin-Seymour
M
,
Munzenrider
J
,
Pile-Spellman
J
, et al
The role of MR and CT in evaluating clival chordomas and chondrosarcomas
.
AJR Am J Roentgenol
.
1988
Sep
;
151
(
3
):
567
75
.
[PubMed]
0361-803X
64.
Müller
U
,
Kubik-Huch
RA
,
Ares
C
,
Hug
EB
,
Löw
R
,
Valavanis
A
, et al
Is there a role for conventional MRI and MR diffusion-weighted imaging for distinction of skull base chordoma and chondrosarcoma?
Acta Radiol
.
2016
Feb
;
57
(
2
):
225
32
.
[PubMed]
0284-1851
65.
Welzel
T
,
Meyerhof
E
,
Uhl
M
,
Huang
K
,
von Deimling
A
,
Herfarth
K
, et al
Diagnostic accuracy of DW MR imaging in the differentiation of chordomas and chondrosarcomas of the skull base: A 3.0-T MRI study of 105 cases
.
Eur J Radiol
.
2018
Aug
;
105
:
119
24
.
[PubMed]
0720-048X
66.
Doucet
V
,
Peretti-Viton
P
,
Figarella-Branger
D
,
Manera
L
,
Salamon
G
.
MRI of intracranial chordomas. Extent of tumour and contrast enhancement: criteria for differential diagnosis
.
Neuroradiology
.
1997
Aug
;
39
(
8
):
571
6
.
[PubMed]
0028-3940
67.
Meyer
JE
,
Oot
RF
,
Lindfors
KK
.
CT appearance of clival chordomas
.
J Comput Assist Tomogr
.
1986
Jan-Feb
;
10
(
1
):
34
8
.
[PubMed]
0363-8715
68.
Schamschula
RG
,
Soo
MY
.
Clival chordomas
.
Australas Radiol
.
1993
Aug
;
37
(
3
):
259
64
.
[PubMed]
0004-8461
69.
Sze
G
,
Uichanco
LS
 3rd
,
Brant-Zawadzki
MN
,
Davis
RL
,
Gutin
PH
,
Wilson
CB
, et al
Chordomas: MR imaging
.
Radiology
.
1988
Jan
;
166
(
1 Pt 1
):
187
91
.
[PubMed]
0033-8419
70.
Meyers
SP
,
Hirsch
WL
 Jr
,
Curtin
HD
,
Barnes
L
,
Sekhar
LN
,
Sen
C
.
Chordomas of the skull base: MR features
.
AJNR Am J Neuroradiol
.
1992
Nov-Dec
;
13
(
6
):
1627
36
.
[PubMed]
0195-6108
71.
Erdem
E
,
Angtuaco
EC
,
Van Hemert
R
,
Park
JS
,
Al-Mefty
O
.
Comprehensive review of intracranial chordoma
.
Radiographics
.
2003
Jul-Aug
;
23
(
4
):
995
1009
.
[PubMed]
0271-5333
72.
Yeom
KW
,
Lober
RM
,
Mobley
BC
,
Harsh
G
,
Vogel
H
,
Allagio
R
, et al
Diffusion-weighted MRI: distinction of skull base chordoma from chondrosarcoma
.
AJNR Am J Neuroradiol
.
2013
May
;
34
(
5
):
1056
61
.
[PubMed]
0195-6108
73.
al-Mefty
O
,
Borba
LA
.
Skull base chordomas: a management challenge
.
J Neurosurg
.
1997
Feb
;
86
(
2
):
182
9
.
[PubMed]
0022-3085
74.
Abdulrauf
SI
.
Decision-making process for the treatment of intracranial chordomas
.
World Neurosurg
.
2014
Nov
;
82
(
5
):
612
3
.
[PubMed]
1878-8750
75.
Guinto
G
,
Guinto-Nishimura
Y
.
Clivus chordomas: role of surgery
.
World Neurosurg
.
2014
May-Jun
;
81
(
5-6
):
688
9
.
[PubMed]
1878-8750
76.
Tomasello
F
,
Conti
A
.
Chordomas: what’s new?
World Neurosurg
.
2014
Nov
;
82
(
5
):
610
1
.
[PubMed]
1878-8750
77.
Chordoma
AM
.
Acta Neurochir (Wien)
.
2017
Oct
;
159
(
10
):
1869
71
.
[PubMed]
0001-6268
78.
Tzortzidis
F
,
Elahi
F
,
Wright
D
,
Natarajan
SK
,
Sekhar
LN
.
Patient outcome at long-term follow-up after aggressive microsurgical resection of cranial base chordomas
.
Neurosurgery
.
2006
Aug
;
59
(
2
):
230
7
.
[PubMed]
0148-396X
79.
Labidi
M
,
Watanabe
K
,
Bouazza
S
,
Bresson
D
,
Bernat
AL
,
George
B
, et al
Clivus chordomas: a systematic review and meta-analysis of contemporary surgical management
.
J Neurosurg Sci
.
2016
Dec
;
60
(
4
):
476
84
.
[PubMed]
1827-1855
80.
Stippler
M
,
Gardner
PA
,
Snyderman
CH
,
Carrau
RL
,
Prevedello
DM
,
Kassam
AB
.
Endoscopic endonasal approach for clival chordomas
.
Neurosurgery
.
2009
Feb
;
64
(
2
):
268
77
.
[PubMed]
0148-396X
81.
Jho
HD
.
Endoscopic transsphenoidal surgery
.
J Neurooncol
.
2001
Sep
;
54
(
2
):
187
95
.
[PubMed]
0167-594X
82.
Sen
C
,
Triana
AI
,
Berglind
N
,
Godbold
J
,
Shrivastava
RK
.
Clival chordomas: clinical management, results, and complications in 71 patients
.
J Neurosurg
.
2010
Nov
;
113
(
5
):
1059
71
.
[PubMed]
0022-3085
83.
Tan
NC
,
Naidoo
Y
,
Oue
S
,
Alexander
H
,
Robinson
S
,
Wickremesekera
A
, et al
Endoscopic surgery of skull base chordomas
.
J Neurol Surg B Skull Base
.
2012
Dec
;
73
(
6
):
379
86
.
[PubMed]
2193-6331
84.
Samii
A
,
Gerganov
VM
,
Herold
C
,
Hayashi
N
,
Naka
T
,
Mirzayan
MJ
, et al
Chordomas of the skull base: surgical management and outcome
.
J Neurosurg
.
2007
Aug
;
107
(
2
):
319
24
.
[PubMed]
0022-3085
85.
Makhdoomi
R
,
Ramzan
A
,
Khursheed
N
,
Bhat
S
,
Baba
K
,
Mohsin
R
, et al
Clinicopathological characteristics of chordoma: an institutional experience and a review of the literature
.
Turk Neurosurg
.
2013
;
23
(
6
):
700
6
.
[PubMed]
1019-5149
86.
Singh
H
,
Harrop
J
,
Schiffmacher
P
,
Rosen
M
,
Evans
J
.
Ventral surgical approaches to craniovertebral junction chordomas
.
Neurosurgery
.
2010
Mar
;
66
(
3
Suppl
):
96
103
.
[PubMed]
0148-396X
87.
Maira
G
,
Pallini
R
,
Anile
C
,
Fernandez
E
,
Salvinelli
F
,
La Rocca
LM
, et al
Surgical treatment of clival chordomas: the transsphenoidal approach revisited
.
J Neurosurg
.
1996
Nov
;
85
(
5
):
784
92
.
[PubMed]
0022-3085
88.
Sekhar
LN
,
Nanda
A
,
Sen
CN
,
Snyderman
CN
,
Janecka
IP
.
The extended frontal approach to tumors of the anterior, middle, and posterior skull base
.
J Neurosurg
.
1992
Feb
;
76
(
2
):
198
206
.
[PubMed]
0022-3085
89.
Derome
PJ
. The transbasal approach to tumors invading the skullbase. In:
Schmidek
HH
,
Sweet
HW
, editors
.
Operative neurosurgical techniques: indications, methods, and results
.
Philadelphia
:
Saunders
;
1993
. pp.
427
41
.
90.
Menezes
AH
,
Gantz
BJ
,
Traynelis
VC
,
McCulloch
TM
.
Cranial base chordomas
.
Clin Neurosurg
.
1997
;
44
:
491
509
.
[PubMed]
0069-4827
91.
Dehdashti
AR
,
Karabatsou
K
,
Ganna
A
,
Witterick
I
,
Gentili
F
.
Expanded endoscopic endonasal approach for treatment of clival chordomas: early results in 12 patients
.
Neurosurgery
.
2008
Aug
;
63
(
2
):
299
307
.
[PubMed]
0148-396X
92.
Fraser
JF
,
Nyquist
GG
,
Moore
N
,
Anand
VK
,
Schwartz
TH
.
Endoscopic endonasal transclival resection of chordomas: operative technique, clinical outcome, and review of the literature
.
J Neurosurg
.
2010
May
;
112
(
5
):
1061
9
.
[PubMed]
0022-3085
93.
Colli
BO
,
Al-Mefty
O
.
Chordomas of the skull base: follow-up review and prognostic factors
.
Neurosurg Focus
.
2001
Mar
;
10
(
3
):
E1
.
[PubMed]
1092-0684
94.
Castro
JR
,
Linstadt
DE
,
Bahary
JP
,
Petti
PL
,
Daftari
I
,
Collier
JM
, et al
Experience in charged particle irradiation of tumors of the skull base: 1977-1992
.
Int J Radiat Oncol Biol Phys
.
1994
Jul
;
29
(
4
):
647
55
.
[PubMed]
0360-3016
95.
Hug
EB
,
Fitzek
MM
,
Liebsch
NJ
,
Munzenrider
JE
.
Locally challenging osteo- and chondrogenic tumors of the axial skeleton: results of combined proton and photon radiation therapy using three-dimensional treatment planning
.
Int J Radiat Oncol Biol Phys
.
1995
Feb
;
31
(
3
):
467
76
.
[PubMed]
0360-3016
96.
Brackmann
DE
,
Teufert
KB
.
Chondrosarcoma of the skull base: long-term follow-up
.
Otol Neurotol
.
2006
Oct
;
27
(
7
):
981
91
.
[PubMed]
1531-7129
97.
Bloch
O
,
Parsa
AT
.
Skull base chondrosarcoma: evidence-based treatment paradigms
.
Neurosurg Clin N Am
.
2013
Jan
;
24
(
1
):
89
96
.
[PubMed]
1042-3680
98.
Pearlman
AW
,
Friedman
M
.
Radical radiation therapy of chordoma
.
Am J Roentgenol Radium Ther Nucl Med
.
1970
Feb
;
108
(
2
):
332
41
.
[PubMed]
0002-9580
99.
Muthukumar
N
,
Kondziolka
D
,
Lunsford
LD
,
Flickinger
JC
.
Stereotactic radiosurgery for chordoma and chondrosarcoma: further experiences
.
Int J Radiat Oncol Biol Phys
.
1998
May
;
41
(
2
):
387
92
.
[PubMed]
0360-3016
100.
Munzenrider
JE
,
Liebsch
NJ
.
Proton therapy for tumors of the skull base
.
Strahlenther Onkol
.
1999
Jun
;
175
(
S2
Suppl 2
):
57
63
.
[PubMed]
0179-7158
101.
Noël
G
,
Feuvret
L
,
Calugaru
V
,
Dhermain
F
,
Mammar
H
,
Haie-Méder
C
, et al
Chordomas of the base of the skull and upper cervical spine. One hundred patients irradiated by a 3D conformal technique combining photon and proton beams
.
Acta Oncol
.
2005
;
44
(
7
):
700
8
.
[PubMed]
0284-186X
102.
Rosenberg
AE
,
Nielsen
GP
,
Keel
SB
,
Renard
LG
,
Fitzek
MM
,
Munzenrider
JE
, et al
Chondrosarcoma of the base of the skull: a clinicopathologic study of 200 cases with emphasis on its distinction from chordoma
.
Am J Surg Pathol
.
1999
Nov
;
23
(
11
):
1370
8
.
[PubMed]
0147-5185
103.
Amichetti
M
,
Cianchetti
M
,
Amelio
D
,
Enrici
RM
,
Minniti
G
.
Proton therapy in chordoma of the base of the skull: a systematic review
.
Neurosurg Rev
.
2009
Oct
;
32
(
4
):
403
16
.
[PubMed]
0344-5607
104.
Hug
EB
,
Loredo
LN
,
Slater
JD
,
DeVries
A
,
Grove
RI
,
Schaefer
RA
, et al
Proton radiation therapy for chordomas and chondrosarcomas of the skull base
.
J Neurosurg
.
1999
Sep
;
91
(
3
):
432
9
.
[PubMed]
0022-3085
105.
Diaz
RJ
,
Cusimano
MD
.
The biological basis for modern treatment of chordoma
.
J Neurooncol
.
2011
Sep
;
104
(
2
):
411
22
.
[PubMed]
0167-594X
106.
Italiano
A
,
Mir
O
,
Cioffi
A
,
Palmerini
E
,
Piperno-Neumann
S
,
Perrin
C
, et al
Advanced chondrosarcomas: role of chemotherapy and survival
.
Ann Oncol
.
2013
Nov
;
24
(
11
):
2916
22
.
[PubMed]
0923-7534
107.
Stacchiotti
S
,
Sommer
J
;
Chordoma Global Consensus Group
.
Building a global consensus approach to chordoma: a position paper from the medical and patient community
.
Lancet Oncol
.
2015
Feb
;
16
(
2
):
e71
83
.
[PubMed]
1470-2045
108.
Stacchiotti
S
,
Gronchi
A
,
Fossati
P
,
Akiyama
T
,
Alapetite
C
,
Baumann
M
, et al
Best practices for the management of local-regional recurrent chordoma: a position paper by the Chordoma Global Consensus Group
.
Ann Oncol
.
2017
Jun
;
28
(
6
):
1230
42
.
[PubMed]
0923-7534
109.
Xia
M
,
Huang
R
,
Sakamuru
S
,
Alcorta
D
,
Cho
MH
,
Lee
DH
, et al
Identification of repurposed small molecule drugs for chordoma therapy
.
Cancer Biol Ther
.
2013
Jul
;
14
(
7
):
638
47
.
[PubMed]
1538-4047
110.
Weinberger
PM
,
Yu
Z
,
Kowalski
D
,
Joe
J
,
Manger
P
,
Psyrri
A
, et al
Differential expression of epidermal growth factor receptor, c-Met, and HER2/neu in chordoma compared with 17 other malignancies
.
Arch Otolaryngol Head Neck Surg
.
2005
Aug
;
131
(
8
):
707
11
.
[PubMed]
0886-4470
111.
Tamborini
E
,
Miselli
F
,
Negri
T
,
Lagonigro
MS
,
Staurengo
S
,
Dagrada
GP
, et al
Molecular and biochemical analyses of platelet-derived growth factor receptor (PDGFR) B, PDGFRA, and KIT receptors in chordomas
.
Clin Cancer Res
.
2006
Dec
;
12
(
23
):
6920
8
.
[PubMed]
1078-0432
112.
Tatman
PD
,
Osbun
J
,
Yakkioui
Y
,
Kaur
S
,
Parada
C
,
Busald
T
, et al
Kinase activity in recurring primary skull base chordomas and chondrosarcomas: identification of novel pathways of oncogenesis and otential drug targets
.
World Neurosurg
.
2017
Nov
;
107
:
75
81
.
[PubMed]
1878-8750
113.
Presneau
N
,
Shalaby
A
,
Idowu
B
,
Gikas
P
,
Cannon
SR
,
Gout
I
, et al
Potential therapeutic targets for chordoma: PI3K/AKT/TSC1/TSC2/mTOR pathway
.
Br J Cancer
.
2009
May
;
100
(
9
):
1406
14
.
[PubMed]
0007-0920
114.
Akhavan-Sigari
R
,
Gaab
MR
,
Rohde
V
,
Brandis
A
,
Tezval
H
,
Abili
M
, et al
Expression of vascular endothelial growth factor receptor 2 (VEGFR-2), inducible nitric oxide synthase (iNOS), and Ki-M1P in skull base chordoma: a series of 145 tumors
.
Neurosurg Rev
.
2014
Jan
;
37
(
1
):
79
88
.
[PubMed]
0344-5607
115.
Grignani
G
,
Palmerini
E
,
Stacchiotti
S
,
Boglione
A
,
Ferraresi
V
,
Frustaci
S
, et al
A phase 2 trial of imatinib mesylate in patients with recurrent nonresectable chondrosarcomas expressing platelet-derived growth factor receptor-α or -β: An Italian Sarcoma Group study
.
Cancer
.
2011
Feb
;
117
(
4
):
826
31
.
[PubMed]
0008-543X
116.
Schwartz
GK
,
Tap
WD
,
Qin
LX
,
Livingston
MB
,
Undevia
SD
,
Chmielowski
B
, et al
Cixutumumab and temsirolimus for patients with bone and soft-tissue sarcoma: a multicentre, open-label, phase 2 trial
.
Lancet Oncol
.
2013
Apr
;
14
(
4
):
371
82
.
[PubMed]
1470-2045
117.
Thornton
KA
,
Chen
AR
,
Trucco
MM
,
Shah
P
,
Wilky
BA
,
Gul
N
, et al
A dose-finding study of temsirolimus and liposomal doxorubicin for patients with recurrent and refractory bone and soft tissue sarcoma
.
Int J Cancer
.
2013
Aug
;
133
(
4
):
997
1005
.
[PubMed]
0020-7136
118.
Italiano
A
,
Le Cesne
A
,
Bellera
C
,
Piperno-Neumann
S
,
Duffaud
F
,
Penel
N
, et al
GDC-0449 in patients with advanced chondrosarcomas: a French sarcoma group/US and French National Cancer Institute single-arm phase II collaborative study
.
Ann Oncol
.
2013
Nov
;
24
(
11
):
2922
6
.
[PubMed]
0923-7534
119.
Speetjens
FM
,
de Jong
Y
,
Gelderblom
H
,
Bovée
JV
.
Molecular oncogenesis of chondrosarcoma: impact for targeted treatment
.
Curr Opin Oncol
.
2016
Jul
;
28
(
4
):
314
22
.
[PubMed]
1040-8746
120.
Stacchiotti
S
,
Morosi
C
,
Lo Vullo
S
,
Casale
A
,
Palassini
E
,
Frezza
AM
, et al
Imatinib and everolimus in patients with progressing advanced chordoma: A phase 2 clinical study
.
Cancer
.
2018
Oct
;
124
(
20
):
4056
63
.
[PubMed]
0008-543X
121.
Lipplaa
A
,
Dijkstra
S
,
Gelderblom
H
.
Efficacy of pazopanib and sunitinib in advanced axial chordoma: a single reference centre case series
.
Clin Sarcoma Res
.
2016
Nov
;
6
(
1
):
19
.
[PubMed]
2045-3329
122.
Singhal
N
,
Kotasek
D
,
Parnis
FX
.
Response to erlotinib in a patient with treatment refractory chordoma
.
Anticancer Drugs
.
2009
Nov
;
20
(
10
):
953
5
.
[PubMed]
0959-4973
123.
Stacchiotti
S
,
Marrari
A
,
Tamborini
E
,
Palassini
E
,
Virdis
E
,
Messina
A
, et al
Response to imatinib plus sirolimus in advanced chordoma
.
Ann Oncol
.
2009
Nov
;
20
(
11
):
1886
94
.
[PubMed]
0923-7534
124.
Hof
H
,
Welzel
T
,
Debus
J
.
Effectiveness of cetuximab/gefitinib in the therapy of a sacral chordoma
.
Onkologie
.
2006
Dec
;
29
(
12
):
572
4
.
[PubMed]
0378-584X
125.
Kim
S
,
Haas
GP
,
Hillman
GG
.
Development of immunotherapy for the treatment of malignancies refractory to conventional therapies
.
Cytokines Mol Ther
.
1996
Mar
;
2
(
1
):
13
9
.
[PubMed]
1355-6568
126.
Yang
Y
.
Cancer immunotherapy: harnessing the immune system to battle cancer
.
J Clin Invest
.
2015
Sep
;
125
(
9
):
3335
7
.
[PubMed]
0021-9738
127.
Ai
L
,
Xu
A
,
Xu
J
.
Roles of PD-1/PD-L1 Pathway: Signaling, Cancer, and Beyond
.
Adv Exp Med Biol
.
2020
;
1248
:
33
59
.
[PubMed]
0065-2598
128.
Feng
Y
,
Shen
J
,
Gao
Y
,
Liao
Y
,
Cote
G
,
Choy
E
, et al
Expression of programmed cell death ligand 1 (PD-L1) and prevalence of tumor-infiltrating lymphocytes (TILs) in chordoma
.
Oncotarget
.
2015
May
;
6
(
13
):
11139
49
.
[PubMed]
1949-2553
129.
Chovanec
M
,
Cierna
Z
,
Miskovska
V
,
Machalekova
K
,
Svetlovska
D
,
Kalavska
K
, et al
Prognostic role of programmed-death ligand 1 (PD-L1) expressing tumor infiltrating lymphocytes in testicular germ cell tumors
.
Oncotarget
.
2017
Mar
;
8
(
13
):
21794
805
.
[PubMed]
1949-2553
130.
Meng
Y
,
Liang
H
,
Hu
J
,
Liu
S
,
Hao
X
,
Wong
MS
, et al
PD-L1 Expression correlates with tumor infiltrating lymphocytes and response to neoadjuvant chemotherapy in cervical cancer
.
J Cancer
.
2018
Jul
;
9
(
16
):
2938
45
.
[PubMed]
0378-2360
131.
Haanen
JB
,
Robert
C
.
Immune checkpoint inhibitors
.
Prog Tumor Res
.
2015
;
42
:
55
66
.
[PubMed]
2296-1895
132.
Wang
J
,
Yang
T
,
Xu
J
.
Therapeutic development of immune checkpoint inhibitors
.
Adv Exp Med Biol
.
2020
;
1248
:
619
49
.
[PubMed]
0065-2598
133.
Migliorini
D
,
Mach
N
,
Aguiar
D
,
Vernet
R
,
Landis
BN
,
Becker
M
, et al
First report of clinical responses to immunotherapy in 3 relapsing cases of chordoma after failure of standard therapies
.
OncoImmunology
.
2017
Jun
;
6
(
8
):
e1338235
.
[PubMed]
2162-4011
134.
Weber
DC
,
Malyapa
R
,
Albertini
F
,
Bolsi
A
,
Kliebsch
U
,
Walser
M
, et al
Long term outcomes of patients with skull-base low-grade chondrosarcoma and chordoma patients treated with pencil beam scanning proton therapy
.
Radiother Oncol
.
2016
Jul
;
120
(
1
):
169
74
.
[PubMed]
0167-8140
135.
Kamrin
RP
,
Potanos
JN
,
Pool
JL
.
An evaluation of the diagnosis and treatment of chordoma
.
J Neurol Neurosurg Psychiatry
.
1964
Apr
;
27
(
2
):
157
65
.
[PubMed]
0022-3050
136.
Di Maio
S
,
Temkin
N
,
Ramanathan
D
,
Sekhar
LN
.
Current comprehensive management of cranial base chordomas: 10-year meta-analysis of observational studies
.
J Neurosurg
.
2011
Dec
;
115
(
6
):
1094
105
.
[PubMed]
0022-3085
137.
Chambers
PW
,
Schwinn
CP
.
Chordoma. A clinicopathologic study of metastasis
.
Am J Clin Pathol
.
1979
Nov
;
72
(
5
):
765
76
.
[PubMed]
0002-9173
138.
Volpe
R
,
Mazabraud
A
.
A clinicopathologic review of 25 cases of chordoma (a pleomorphic and metastasizing neoplasm)
.
Am J Surg Pathol
.
1983
Mar
;
7
(
2
):
161
70
.
[PubMed]
0147-5185
139.
Yasuda
M
,
Bresson
D
,
Chibbaro
S
,
Cornelius
JF
,
Polivka
M
,
Feuvret
L
, et al
Chordomas of the skull base and cervical spine: clinical outcomes associated with a multimodal surgical resection combined with proton-beam radiation in 40 patients
.
Neurosurg Rev
.
2012
Apr
;
35
(
2
):
171
82
.
[PubMed]
0344-5607
140.
Bohman
LE
,
Koch
M
,
Bailey
RL
,
Alonso-Basanta
M
,
Lee
JY
.
Skull base chordoma and chondrosarcoma: influence of clinical and demographic factors on prognosis: a SEER analysis
.
World Neurosurg
.
2014
Nov
;
82
(
5
):
806
14
.
[PubMed]
1878-8750
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.