Next Article in Journal
Global Fight against Malaria: Goals and Achievements 1900–2022
Previous Article in Journal
Biomarkers as Predictors of Mortality in Sepsis and Septic Shock for Patients Admitted to Emergency Department: Who Is the Winner? A Prospective Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunologic and Targeted Molecular Therapies for Chordomas: A Narrative Review

1
Department of Orthopaedic Surgery, Montefiore Einstein, Bronx, NY 10461, USA
2
Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
3
Department of Neurological Surgery, Montefiore Einstein, Bronx, NY 10461, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(19), 5679; https://doi.org/10.3390/jcm13195679
Submission received: 4 September 2024 / Revised: 17 September 2024 / Accepted: 20 September 2024 / Published: 24 September 2024
(This article belongs to the Section Orthopedics)

Abstract

:
Chordomas are rare sarcomas arising from notochordal tissue and occur most commonly in the spine. The standard of care for chordomas without evidence of metastatic disease generally consists of en bloc resection followed by adjuvant radiotherapy. However, long-term (20-year) survival rates are approximately 30%. Chordomas are generally considered as chemo resistant. Therefore, systemic therapies have rarely been employed. Novel immunotherapies, including antibody therapy and tumor vaccines, have shown promise in early trials, leading to extended progression-free survival and symptom relief. However, the outcomes of larger trials using these vectors are heterogeneous. The aim of this review is to summarize novel chordoma treatments in immune-targeted therapies. The current merits, trial outcomes, and toxicities of these novel immune and targeted therapies, including those targeting vascular endothelial growth factor receptor (VEGFR) targets and the epidermal growth factor receptor (EGFR), will be discussed.

1. Introduction

Chordomas are rare, slow-growing malignant tumors that originate from vestigial or ectopic notochord tissue. They constitute 1–4% of all bone tumors and are more frequently observed in males [1,2,3]. United States population-based registries have shown an annual incidence of about 0.08 per 100,000, with median survival times of about 6 years [4]. These tumors typically arise in patients of European descent, with a predilection for patients between 50 and 60 years of age. Chordomas arise from the spine in 61% of cases. Although they most commonly occur in the sacrum, 38% of chordomas arise from the cranium [1]. Chordomas are malignant in nature and have a high predilection for recurrence despite treatment. Typical treatment paradigms include wide surgical excision with radiotherapy, as these tumors are typically chemotherapy-resistant. Local recurrence rates vary and can be up to 50% in 15-year follow-up periods [5]. Poor outcomes are typically associated with younger-age patients, locations within the sacrum, associated bladder or bowel dysfunction, and dedifferentiated histopathology [6].
Current treatment guidelines for chordomas advocate for en bloc surgical resection with wide margins followed by adjuvant radiotherapy. However, this strategy often fails to achieve long-term remission, with a twenty-year survival rate of approximately 30% [7]. The sensitive location of many chordomas often involves vital nerves and blood vessels that cannot be sacrificed without unacceptable morbidity. Surgical efforts are, therefore, highly complicated and commonly leave unclear or positive margins [8,9]. Chordomas also display varying degrees of radioresistance, potentially because of the restricted proliferative capacity of chordoma tumor cells [8,10,11].
Chordomas are resistant to most conventional chemotherapies [11,12,13]. However, immunotherapies and targeted molecular therapies hold significant promise as alternative treatments. Vanderheijden et al. [7], in their review of the genomic characteristics of chordomas, identified numerous markers that may hold prognostic and therapeutic significance. The authors outline cell cycle regulators, tumor microenvironment factors, and immunological aspects of chordoma cells that could be leveraged for more efficient systemic treatments. Adoptive T-cell therapy, the in vitro modification of T-cells followed by their infusion into the patient, also has the potential to elicit a potent host immune response against specific tumor cell markers [11,14].
Molecular Genetic Basis
Various studies have been conducted to investigate the molecular biology and genetics behind chordoma pathology. Studies show that chordoma tumors feature diverse rearrangements, including deletions on chromosome 3p and copy number gains on 7q [15]. Additionally, the brachyury gene, a T-box transcription factor, has been found to be heavily expressed within chordomas [16]. Brachyury is involved in notochord formation in normal embryonic formation. Alterations in the function of this gene may be involved in the prevention of notochord apoptosis and possible chordoma formation. The LYST gene, a known lysosomal regulatory gene responsible for notochord progression, has also been implicated in the development of chordomas [17]. Activation mutations, leading to overexpressions of VEGF, PDGFRA, IGF1R, EGRR, and MGMT genes, have also been implicated in genetic studies of chordomas [18].
A plethora of studies have identified novel immunotherapies and targeted therapies against chordomas over the past 5 years. Particular emphasis has been placed on immune checkpoint inhibitors, such as Programmed Death Protein 1 (PD-1) and its ligand (PD-L1), and oncogenes, such as cyclin-dependent kinases (CDKs). This body of work has laid the foundation for innovative treatment modalities against chordomas [7,11,19,20,21].
This review aims to summarize current advancements in the systemic treatment of chordomas, with a particular emphasis on immunotherapy and targeted molecular therapies (Table 1). This review will particularly focus on works published over the past 5 years, thereby highlighting the most recently published literature.

2. Current Systemic Treatment Strategies for Chordomas

Current treatment guidelines for chordomas recommend en bloc surgical resection with wide margins followed by adjuvant radiotherapy at a dose of 74 GyE, as per the chordoma global consensus group [22]; however, this strategy comes with many potential limitations [10]. Thorough chordoma resection is critical for preventing local tumor recurrence. However, the location of many chordomas within the axial skeleton complicates the oncologic surgeon’s ability to completely remove them [10]. Wound complications and neurologic deficits are often intrinsic to the anatomic resection needed for a partial or complete sacrectomy [23]. Although an Enneking-type resection with clear margins is preferred and most likely to yield optimal local recurrence rates, the resection of specific neurologic and vascular structures that run local to the axial skeleton may result in unacceptable morbidity. For example, the ligation of a number of lumbar and sacral nerve roots may lead to permanent ambulatory deficits and the complete loss of bowel/bladder function [24], outcomes that may be unacceptable to the patient. The anatomic proximity of cervical and cranial chordomas to the spinal cord and vertebral arteries complicates their resection [25,26]. Consequently, a subset of patients with chordomas undergo partial resections or are deemed as non-surgical candidates.
Radiotherapy and systemic therapies are particularly important to those patients who cannot be treated via wide resection. Adjuvant high-dose radiation therapy has been found to improve the survival and decrease the local recurrence of patients with spinal chordomas and is now a part of the standard of care [10,22,26]. The chordoma global consensus group recommends a dose of 74 GyE because of the radioresistant properties of chordomas [22]. However, the 10-year survival rate of patients treated with adjunct radiotherapy is 21–60%, with patients treated with proton radiation therapy having superior survival rates [27]. Numerous complications have also been reported after radiotherapy for chordomas. A systematic review of skull-based chordomas treated with radiotherapy reported that 41% of the patients with recorded complications reported hearing loss, 25.3% reported temporal lobe damage, 9.64% reported visual disturbances, and 7.23% reported pituitary insufficiency [28]. Preoperative radiation, a total radiation dose of over 70 Gy, and proton therapy have been associated with wound complications [29]. Given its risk of complications and relative radioresistance, radiotherapy alone is an insufficient adjunct therapy in the treatment of chordomas.

3. Immunotherapy for Chordomas

Numerous works over the past 5 years have studied the use of immunotherapies targeted against chordomas. The anti-PD-L1 antibodies durvalumab and pembrolizumab were heavily tested, with promising results [30,31]. A phase-2 clinical trial of durvalumab against a wide variety of sarcomas showed that one out of five chordoma patients had a partial response by the end of the 12-week study. The authors concluded that chordoma patients likely require a longer therapeutic window because of pseudo progression [30]. A phase-2 clinical trial of pembrolizumab further supported the hypothesis that a 12-week therapeutic window for chordomas is too short [31]. Pembrolizumab treatment resulted in a median progression-free survival of approximately 6 months with symptomatic improvement. However, 12 out of 34 patients died over a 13-month follow-up.
Vaccines have been used to sensitize the patient’s immune system to treat chordomas [32]. Chordomas universally express brachyury, a gene for the T-box transcription factor [32,33]. A phase-1 trial of the MVA-BN–brachyury–TRICOM vaccine showed promising results, with no dose-limiting toxicities or adverse reactions [32]. The most common adverse events were fevers, chills, and hypotension, and 31% of the patients experienced either grade-1 or -2 cytokine release syndrome in a dose–effect trend. All the reactions resolved after 24 h. One patient had a partial response to the MVA-BN–brachyury–TRICOM vaccine, four had stable disease, and eight had disease progression. Those with stable disease also reported symptom reduction [32]. However, a phase-two trial of the same drug reported that 54% of the patients had disease progression, resulting in the discontinuation of the trial, as there was no significant difference between the placebo and the trial [34]. One case report noted that a patient who had failed checkpoint inhibitor therapy became more responsive to PD-1 immunotherapies following treatment with the oncolytic virus AdAPT-001 [35]. No large-scale trial evaluating this finding has yet been completed.

4. Targeted Therapies against Chordomas

Oncogene addiction is the idea that tumor cells rely on the protein product derived from a mutated oncogene to remain malignant [36]. Targeting these specific oncogenic drivers has demonstrated notable success in the clinical management of various solid tumors [37,38]. Targeted cancer therapy broadly falls into two categories: small molecules and antibodies. Small-molecule inhibitors, owing to their low molecular weight and size, can manifest throughout a broad spectrum of specificities, offering diverse opportunities for targeting both intracellular and extracellular entities [38]. Conversely, antibodies, which are larger, have high specificities for molecular targets that are typically located on the cell surface [38]. Chordoma clinical trials have mainly targeted molecular pathways, such as the platelet-derived growth factor receptor (PDGFR), mammalian target of rapamycin (mTOR), BCR-ABL, KIT, vascular endothelial growth factor receptor (VEGFR), T-box transcription factor T (brachyury protein), epidermal growth factor receptor (EGFR), HER2/NEU, and topoisomerase I [39].
Multiple clinical trials over the past 5 years have studied targeted therapies against chordomas. The most promising involved the vascular endothelial growth factor receptor (VEGFR) [40]. A phase-2 trial of apatinib, a VEGFR2 small-molecule tyrosine kinase inhibitor, resulted in a progression-free survival for a median time of 18 months in 83% of the patients. It is notable that a dose reduction from 500 mg daily to 250 mg alternating with 500 mg or 250 mg daily was required in 87% of the patients because of treatment side-effects, although there were no grade-4 adverse reactions [40]. A phase-2 clinical trial of the proangiogenic vascular endothelial growth factor receptor tyrosine kinase inhibitor sorafenib resulted in progression-free survival after 12 months in 73% of the patients. However, multiple toxicities were reported following sorafenib use, with one patient experiencing a severe adverse reaction of diarrhea with hypokalemia and pancreatitis [41].
The tyrosine kinase inhibitor imatinib has shown promising results in the treatment of chordomas, with a large phase-2 study showing a 64% clinical benefit after six months of treatment, with no unexpected toxicities [42]. When imatinib was combined with everolimus, 37 out of 43 patients had stable disease, and the trial concluded a 76.7% clinical benefit according to the Choi criteria, which require a reduction of at least 10% in the largest tumor diameter or a decrease of at least 15% in the average attenuation calculated for the regions of interest in all the designated lesions [43,44].
A phase-2 clinical trial of lapatinib, an epidermal growth factor inhibitor, resulted in 6/18 chordoma patients tested having a partial response and 7/18 patients with stable disease [45]. Patients were given an average of 1284 mg a day for an average of 4.6 months. The dose of lapatinib had to be reduced in some patients because of adverse events.
The loss of tumor suppressor gene heterozygosity is believed to play a crucial role in the carcinogenesis of chordoma tumor cells. Although the loss of CDKN2A and PTEN expressions is observed in various tumor types, it is thought to play a pivotal role in the pathogenesis of chordomas, with PTEN potentially serving as a prognostic and predictive biomarker [46,47]. In a comparative genomic hybridization study, Le et al. [47] examined 21 samples of sporadic chordoma tumor tissues to conduct a copy number analysis of specific genes. The findings revealed a loss of CDKN2A in 16 out of 20 unique cases (80%) and a one-copy loss of the PTEN locus in 16 out of 20 cases (80%) [47]. Deficiencies of PTEN and CDKN2A are two of the most frequent molecular alterations found in chordoma cases, often being associated with a poor prognosis [48].
Seeling et al. [48] demonstrated that treating PTEN-/p16- (p16 is encoded by CDKN2A) cells with palbociclib, a CDK4/6 inhibitor, in combination with rapamycin, an mTOR inhibitor, resulted in a significant reduction in tumor cell viability. Furthermore, the authors reported that individual inhibition using either palbociclib or rapamycin reduced cell viability but required higher concentrations of these drugs to achieve efficacies comparable with that of the combined therapy [48]. These findings suggest a potentially promising strategy for managing PTEN-/p16- chordoma patients and serve as a possible area for future research.
Unfortunately, not all chordoma therapies have been successful. A phase-2 clinical trial, carried out by Le Cesne et al. [49], of regorafenib, an anti-VEGFR kinase inhibitor, found no significant mortality difference between the placebo and treatment arms at a dose of 160 mg/day [49]. However, another phase-2 trial of regorafenib in the treatment of osteosarcoma has shown promising results [50].
It is reasonable to assume that the patient’s response to immunotherapy and targeted therapy will highly depend on the genetic expression of the host’s immune system and the profile of the specific tumor. Characterizing those markers that have exhibited clinical benefit is crucial to the development of innovative treatment strategies. Moreover, advancements in computer simulation models to predict the course of tumor progression are likely to impact medical care over the next few decades. Ghaith et al. [51] demonstrated that machine-learning algorithms can be used to predict chordoma outcomes with an accuracy of 66% using individual immunohistochemical markers. The presence of pancytokeratin and EMA in tumor cells was associated with a progression risk of 67%, and the presence of the S100 protein represented a 54% risk of progression [51].

5. Future Treatment Directions

Several promising clinical trials for both targeted therapies and immunotherapy are underway to further study treatment options for chordomas. Cetuximab, an EGFR inhibitor, is undergoing a phase-II clinical trial for metastatic chordomas in adults [52]. This is an interventional trial and will use the Choi criteria to evaluate survival, safety, and tolerability and includes an estimated 15 patients. Another phase-II EGFR inhibitor trial is underway for the drug afatinib, also in advanced and metastatic chordomas [53]. This dual-arm study will assess progression-free survival in both a first-line treatment arm and a continuing treatment arm [54]. A phase-I clinical trial for the targeted therapy and established chronic myeloid leukemia drug Nilotinib is underway to treat high-risk chordomas [54]. Preliminary results are positive, with updated data showing good local control and distant metastasis-free survival in the dose escalation arm [54].
For PD-L1-positive chordomas, a phase-II clinical trial utilizing a combined treatment of nivolumab and relatlimab has begun and is estimated to run until April 2025 [55]. Although initial results are promising for benefit with the combined treatment, the population was small, with n = 10 [55]. Nivolumab is also being studied as an adjuvant to stereotactic radiosurgery in a different clinical trial estimated to be completed in November 2024 [56].

6. Conclusions

Chordomas are rare and hard-to-treat malignancies. Consequently, clinical trials of potentially impactful systemic therapies have suffered from the inability to recruit eligible patients. Early anti-PD-L1 trials demonstrated mild improvements in progression-free survival, while the brachyury vaccine appears to result in symptomatic improvement. VEGFR-targeted drugs have produced the most promising results to date. However, often, significant toxicities have been noted. Further investigations into novel systemic therapies with vs. without surgical intervention and radiotherapy are warranted to yield substantive improvements in chordoma survival.
Table 1. Clinical trials, targets, samples, and outcomes.
Table 1. Clinical trials, targets, samples, and outcomes.
PubMed IDTitleType of TherapyTargetStudy TypeEnrollmentOutcome
35934010Durvalumab plus tremelimumab in advanced or metastatic soft-tissue and bone sarcomas: a single-centre phase-2 trial (2022) [30]Immunotherapy Anti-CTLA-4 (tremelimumab), Anti-PD-L1 (durvalumab)Clinical trial (Phase 2)57 in total
Median age: 48 years old
Males: 31
Females: 26
Progression-free survival at 12 weeks was 49% (95% CI: 36–61).
37429302Pembrolizumab in patients with rare and ultra-rare sarcomas (AcSé pembrolizumab): analysis of a subgroup from a non-randomised, open-label, phase-2 basket trial (2023) [31]Immunotherapy Anti-PD-1 Clinical trial (Phase 2)97 in total
Median age: 51 years old
Males: 53
Females: 44
At week 12, objective response rate was 6·2% (95% CI: 2·3–13·0), with no complete responses and six partial responses in the 97 patients.
34479925Phase-1 open-label trial of intravenous administration of MVA-BN–brachyury–TRICOM vaccine in patients with advanced cancer (2021) [32]Immunotherapy BrachyuryClinical trial (Phase 1)13 in total
Mean age: 60 years old
Males: 9
Females: 4
Efficacy analysis of objective response rate per RECIST 1.1 at the end of the study showed one patient with a partial response, four with stable disease, and eight with progressive disease. Three patients with stable disease experienced clinical benefit in the form of improvement in pain.
33594772Randomized, double-blind, placebo-controlled phase-II study of yeast–brachyury vaccine (GI-6301) in combination with standard-of-care radiotherapy in locally advanced, unresectable chordoma (2021) [34]Immunotherapy BrachyuryClinical trial (Phase 2)24 in total
Median age: 61 years old
Males: 16
Females: 8
No difference in the overall response rate was observed, leading to early discontinuation of this trial because of low conditional power to detect a statistical difference at the planned end of the accrual.
32888455Apatinib in patients with advanced chordoma: a single-arm, single-centre, phase-2 study (2020) [40]Targeted therapyVascular endothelial growth factor receptor-2 inhibitorClinical trial (Phase 2)30 in total
Median age: 56 years old
Males: 19
Females: 11
Median progression-free survival was 18 months (95% CI: 3–34) according to RECIST and 18 months (3–33) according to Choi criteria.
22331945Phase-II study of imatinib in advanced chordoma (2012) [42]Targeted therapyPlatelet-derived growth factor βClinical trial (Phase 2)56 in total
Median age: 60 years old
Males: 35
Females: 21
A total of 35 patients with stable disease (SD, 70%) and a 64% clinical benefit rate (i.e., RECIST complete response + PR + SD ≥ 6 months)
30216418Imatinib and everolimus in patients with progressing advanced chordoma: a phase-2 clinical study (2018) [43]Targeted therapyPlatelet-derived growth factor βClinical trial (Phase 2)43 in total
Median age: 64 years old
Males: 28
Females: 15
Imatinib plus everolimus showed limited activity in progressing advanced chordoma. Interestingly, the number of tumor cells activated for mammalian target of rapamycin effectors correlated with the response. Toxicity was not negligible.
37285716Regorafenib in patients with relapsed advanced or metastatic chordoma: results of a non-comparative, randomised, double-blind, placebo-controlled, multicentre phase-II study (2023) [49]Targeted therapysmall-molecule multi-kinase inhibitorRandomized controlled trial23 in total
Median age: 66 years old
Males: 16
Females: 7
According to the study design’s criteria for success, 10 out of 16 progression-free patients at 6 months in the regorafenib arm, according to RECIST 1.1, would have been necessary to consider this study as successful, whereas only 6/14 (40%) patients remained free of disease progression at the 6-month timepoint in the trial.
30477937Efficacy and safety of regorafenib in adult patients with metastatic osteosarcoma: a non-comparative, randomised, double-blind, placebo-controlled phase-2 study (2018) [50]Targeted therapySmall-molecule multi-kinase inhibitorClinical trial (Phase 2)38 in total
Median age: 33 years old
Males: 24
Females: 14
A total of 17 of 26 patients (65%; one-sided 95% CI: 47%) in the regorafenib group were non-progressive at 8 weeks compared with no patients in the placebo group. Ten patients in the placebo group crossed over to receive open-label regorafenib after centrally confirmed disease progression.
31488216Phase-I trial of HuMax-IL8 (BMS-986253), an anti-IL-8 monoclonal antibody, in patients with metastatic or unresectable solid tumors (2019)Targeted therapyIL-8 monoclonal antibodyClinical trial (Phase 1)15 in total
Mean age: 59 years old
Males: 9
Females: 6
HuMax-IL8 is safe and well-tolerated. Ongoing studies are evaluating the combination of the IL-8 blockade and other immunotherapies.

Author Contributions

Conceptualization, M.S.F. and R.G.; methodology, R.G. and A.S.; formal analysis, R.G., R.A., C.J.P., M.B., V.S., A.S. and M.S.F.; investigation, R.G., R.A., C.J.P., M.B., V.S.; resources, R.G., R.A., C.J.P., M.B., V.S., A.S. and M.S.F.; data curation, R.G., R.A., C.J.P., M.B., V.S., A.S. and M.S.F.; writing—original draft preparation, R.G., K.M., E.K., C.J.P., Y.G., S.M., R.D.l.G.R., J.K., A.E. and R.Y.; writing—review and editing, E.M., R.G. and K.M.; visualization, M.S.F.; supervision, M.S.F.; project administration, M.S.F.; funding acquisition, M.S.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The data related to this work can be released by the authors following a reasonable request.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Das, P.; Soni, P.; Jones, J.; Habboub, G.; Barnholtz-Sloan, J.S.; Recinos, P.F.; Kshettry, V.R. Descriptive epidemiology of chordomas in the United States. J. Neurooncol. 2020, 148, 173–178. [Google Scholar] [CrossRef] [PubMed]
  2. Karele, E.N.; Paze, A.N. Chordoma: To know means to recognize. Biochim. Biophys. Acta Rev. Cancer 2022, 1877, 188796. [Google Scholar] [CrossRef] [PubMed]
  3. Tenny, S.; Varacallo, M. Chordoma. In StatPearls; StatPearls Publishing LLC.: St. Petersburg, FL, USA, 2024. [Google Scholar]
  4. McMaster, M.L.; Goldstein, A.M.; Bromley, C.M.; Ishibe, N.; Parry, D.M. Chordoma: Incidence and survival patterns in the United States, 1973–1995. Cancer Causes Control 2001, 12, 1–11. [Google Scholar] [CrossRef] [PubMed]
  5. Stacchiotti, S.; Gronchi, A.; Fossati, P.; Akiyama, T.; Alapetite, C.; Baumann, M.; Blay, J.Y.; Bolle, S.; Boriani, S.; Bruzzi, P.; et al. Best practices for the management of local-regional recurrent chordoma: A position paper by the Chordoma Global Consensus Group. Ann. Oncol. 2017, 28, 1230–1242. [Google Scholar] [CrossRef] [PubMed]
  6. Karpathiou, G.; Dumollard, J.M.; Dridi, M.; Dal Col, P.; Barral, F.G.; Boutonnat, J.; Peoc’h, M. Chordomas: A review with emphasis on their pathophysiology, pathology, molecular biology, and genetics. Pathol. Res. Pract. 2020, 216, 153089. [Google Scholar] [CrossRef]
  7. Vanderheijden, C.; Vaessen, T.; Yakkioui, Y.; Temel, Y.; Hoogland, G.; Hovinga, K. Genes Predicting Survival of Chordoma Patients. World Neurosurg. 2021, 156, 125–132. [Google Scholar] [CrossRef]
  8. Walcott, B.P.; Nahed, B.V.; Mohyeldin, A.; Coumans, J.V.; Kahle, K.T.; Ferreira, M.J. Chordoma: Current concepts, management, and future directions. Lancet Oncol. 2012, 13, e69–e76. [Google Scholar] [CrossRef]
  9. Wang, X.; Chen, Z.; Li, B.; Fan, J.; Xu, W.; Xiao, J. Immunotherapy as a Promising Option for the Treatment of Advanced Chordoma: A Systemic Review. Cancers 2022, 15, 264. [Google Scholar] [CrossRef]
  10. Heery, C.R. Chordoma: The Quest for Better Treatment Options. Oncol. Ther. 2016, 4, 35–51. [Google Scholar] [CrossRef]
  11. Xu, J.; Shi, Q.; Wang, B.; Ji, T.; Guo, W.; Ren, T.; Tang, X. The role of tumor immune microenvironment in chordoma: Promising immunotherapy strategies. Front. Immunol. 2023, 14, 1257254. [Google Scholar] [CrossRef]
  12. Azzarelli, A.; Quagliuolo, V.; Cerasoli, S.; Zucali, R.; Bignami, P.; Mazzaferro, V.; Dossena, G.; Gennari, L. Chordoma: Natural history and treatment results in 33 cases. J. Surg. Oncol. 1988, 37, 185–191. [Google Scholar] [CrossRef] [PubMed]
  13. York, J.E.; Kaczaraj, A.; Abi-Said, D.; Fuller, G.N.; Skibber, J.M.; Janjan, N.A.; Gokaslan, Z.L. Sacral chordoma: 40-year experience at a major cancer center. Neurosurgery 1999, 44, 74–79; discussion 79–80. [Google Scholar] [CrossRef] [PubMed]
  14. Laskowski, T.; Rezvani, K. Adoptive cell therapy: Living drugs against cancer. J. Exp. Med. 2020, 217, e20200377. [Google Scholar] [CrossRef] [PubMed]
  15. Ozair, M.Z.; Shah, P.P.; Mathios, D.; Lim, M.; Moss, N.S. New Prospects for Molecular Targets for Chordomas. Neurosurg. Clin. N. Am. 2020, 31, 289–300. [Google Scholar] [CrossRef] [PubMed]
  16. Henderson, S.R.; Guiliano, D.; Presneau, N.; McLean, S.; Frow, R.; Vujovic, S.; Anderson, J.; Sebire, N.; Whelan, J.; Athanasou, N.; et al. A molecular map of mesenchymal tumors. Genome Biol. 2005, 6, R76. [Google Scholar] [CrossRef]
  17. Tarpey, P.S.; Behjati, S.; Young, M.D.; Martincorena, I.; Alexandrov, L.B.; Farndon, S.J.; Guzzo, C.; Hardy, C.; Latimer, C.; Butler, A.P.; et al. The driver landscape of sporadic chordoma. Nat. Commun. 2017, 8, 890. [Google Scholar] [CrossRef]
  18. Yamaguchi, T.; Imada, H.; Iida, S.; Szuhai, K. Notochordal Tumors: An Update on Molecular Pathology with Therapeutic Implications. Surg. Pathol. Clin. 2017, 10, 637–656. [Google Scholar] [CrossRef]
  19. Memon, H.; Patel, B.M. Immune checkpoint inhibitors in non-small cell lung cancer: A bird’s eye view. Life Sci. 2019, 233, 116713. [Google Scholar] [CrossRef]
  20. Thanindratarn, P.; Dean, D.C.; Feng, W.; Wei, R.; Nelson, S.D.; Hornicek, F.J.; Duan, Z. Cyclin-dependent kinase 12 (CDK12) in chordoma: Prognostic and therapeutic value. Eur. Spine J. 2020, 29, 3214–3228. [Google Scholar] [CrossRef]
  21. Lui, G.Y.L.; Grandori, C.; Kemp, C.J. CDK12: An emerging therapeutic target for cancer. J. Clin. Pathol. 2018, 71, 957–962. [Google Scholar] [CrossRef]
  22. Stacchiotti, S.; Sommer, J. Building a global consensus approach to chordoma: A position paper from the medical and patient community. Lancet Oncol. 2015, 16, e71–e83. [Google Scholar] [CrossRef] [PubMed]
  23. Deskoulidi, P.; Stavrianos, S.D.; Mastorakos, D.; Kontogeorgakos, V.A.; Savvidou, O.; Chrysikos, D.; Samolis, A.; Pappas, N.; Troupis, T.; Papagelopoulos, P.J. Anatomical Considerations and Plastic Surgery Reconstruction Options of Sacral Chordoma Resection. Cureus 2023, 15, e37965. [Google Scholar] [CrossRef] [PubMed]
  24. Bongers, M.E.R.; Dea, N.; Ames, C.P.; Schwab, J.H. Surgical Strategies for Chordoma. Neurosurg. Clin. N. Am. 2020, 31, 251–261. [Google Scholar] [CrossRef] [PubMed]
  25. Alsharif, T.H.; Gronfula, A.G.; Alghdali, L.H.; Hejazi, M.; Alanazi, A.; Wali, S.M.; Alyousef, M. Outcomes of Endoscopic Resection in Pediatric Skull Base Chordoma: A Systematic Review. Cureus 2023, 15, e41487. [Google Scholar] [CrossRef] [PubMed]
  26. Passer, J.Z.; Alvarez-Breckenridge, C.; Rhines, L.; DeMonte, F.; Tatsui, C.; Raza, S.M. Surgical Management of Skull Base and Spine Chordomas. Curr. Treat. Options Oncol. 2021, 22, 40. [Google Scholar] [CrossRef]
  27. Zhou, J.; Yang, B.; Wang, X.; Jing, Z. Comparison of the Effectiveness of Radiotherapy with Photons and Particles for Chordoma After Surgery: A Meta-Analysis. World Neurosurg. 2018, 117, 46–53. [Google Scholar] [CrossRef]
  28. Pahwa, B.; Medani, K.; Lu, V.M.; Elarjani, T. Proton beam therapy for skull base chordomas: A systematic review of tumor control rates and survival rates. Neurosurg. Rev. 2022, 45, 3551–3563. [Google Scholar] [CrossRef]
  29. Houdek, M.T.; Rose, P.S.; Hevesi, M.; Schwab, J.H.; Griffin, A.M.; Healey, J.H.; Petersen, I.A.; DeLaney, T.F.; Chung, P.W.; Yaszemski, M.J.; et al. Low dose radiotherapy is associated with local complications but not disease control in sacral chordoma. J. Surg. Oncol. 2019, 119, 856–863. [Google Scholar] [CrossRef]
  30. Somaiah, N.; Conley, A.P.; Parra, E.R.; Lin, H.; Amini, B.; Solis Soto, L.; Salazar, R.; Barreto, C.; Chen, H.; Gite, S.; et al. Durvalumab plus tremelimumab in advanced or metastatic soft tissue and bone sarcomas: A single-centre phase 2 trial. Lancet Oncol. 2022, 23, 1156–1166. [Google Scholar] [CrossRef]
  31. Blay, J.Y.; Chevret, S.; Le Cesne, A.; Brahmi, M.; Penel, N.; Cousin, S.; Bertucci, F.; Bompas, E.; Ryckewaert, T.; Soibinet, P.; et al. Pembrolizumab in patients with rare and ultra-rare sarcomas (AcSé Pembrolizumab): Analysis of a subgroup from a non-randomised, open-label, phase 2, basket trial. Lancet Oncol. 2023, 24, 892–902. [Google Scholar] [CrossRef]
  32. DeMaria, P.J.; Lee-Wisdom, K.; Donahue, R.N.; Madan, R.A.; Karzai, F.; Schwab, A.; Palena, C.; Jochems, C.; Floudas, C.; Strauss, J.; et al. Phase 1 open-label trial of intravenous administration of MVA-BN-brachyury-TRICOM vaccine in patients with advanced cancer. J. Immunother. Cancer 2021, 9, e003238. [Google Scholar] [CrossRef] [PubMed]
  33. Dridi, M.; Boutonnat, J.; Dumollard, J.M.; Peoc’h, M.; Karpathiou, G. Patterns of brachyury expression in chordomas. Ann. Diagn. Pathol. 2021, 53, 151760. [Google Scholar] [CrossRef] [PubMed]
  34. DeMaria, P.J.; Bilusic, M.; Park, D.M.; Heery, C.R.; Donahue, R.N.; Madan, R.A.; Bagheri, M.H.; Strauss, J.; Shen, V.; Marté, J.L.; et al. Randomized, Double-Blind, Placebo-Controlled Phase II Study of Yeast-Brachyury Vaccine (GI-6301) in Combination with Standard-of-Care Radiotherapy in Locally Advanced, Unresectable Chordoma. Oncologist 2021, 26, e847–e858. [Google Scholar] [CrossRef] [PubMed]
  35. Kesari, S.; Williams, J.; Burbano, E.; Stirn, M.; Caroen, S.; Oronsky, B.; Reid, T.; Larson, C. Case Report of AdAPT-001-Mediated Sensitization to a Previously Failed Checkpoint Inhibitor in a Metastatic Chordoma Patient. Case Rep. Oncol. 2023, 16, 172–176. [Google Scholar] [CrossRef]
  36. Guan, L.Y.; Lu, Y. New developments in molecular targeted therapy of ovarian cancer. Discov. Med. 2018, 26, 219–229. [Google Scholar]
  37. Petroni, G.; Buqué, A.; Coussens, L.M.; Galluzzi, L. Targeting oncogene and non-oncogene addiction to inflame the tumour microenvironment. Nat. Rev. Drug Discov. 2022, 21, 440–462. [Google Scholar] [CrossRef]
  38. Bedard, P.L.; Hyman, D.M.; Davids, M.S.; Siu, L.L. Small molecules, big impact: 20 years of targeted therapy in oncology. Lancet 2020, 395, 1078–1088. [Google Scholar] [CrossRef]
  39. Akinduro, O.O.; Suarez-Meade, P.; Garcia, D.; Brown, D.A.; Sarabia-Estrada, R.; Attia, S.; Gokaslan, Z.L.; Quiñones-Hinojosa, A. Targeted Therapy for Chordoma: Key Molecular Signaling Pathways and the Role of Multimodal Therapy. Target. Oncol. 2021, 16, 325–337. [Google Scholar] [CrossRef]
  40. Liu, C.; Jia, Q.; Wei, H.; Yang, X.; Liu, T.; Zhao, J.; Ling, Y.; Wang, C.; Yu, H.; Li, Z.; et al. Apatinib in patients with advanced chordoma: A single-arm, single-centre, phase 2 study. Lancet Oncol. 2020, 21, 1244–1252. [Google Scholar] [CrossRef]
  41. Bompas, E.; Le Cesne, A.; Tresch-Bruneel, E.; Lebellec, L.; Laurence, V.; Collard, O.; Saada-Bouzid, E.; Isambert, N.; Blay, J.Y.; Amela, E.Y.; et al. Sorafenib in patients with locally advanced and metastatic chordomas: A phase II trial of the French Sarcoma Group (GSF/GETO). Ann. Oncol. 2015, 26, 2168–2173. [Google Scholar] [CrossRef]
  42. Stacchiotti, S.; Longhi, A.; Ferraresi, V.; Grignani, G.; Comandone, A.; Stupp, R.; Bertuzzi, A.; Tamborini, E.; Pilotti, S.; Messina, A.; et al. Phase II study of imatinib in advanced chordoma. J. Clin. Oncol. 2012, 30, 914–920. [Google Scholar] [CrossRef]
  43. Stacchiotti, S.; Morosi, C.; Lo Vullo, S.; Casale, A.; Palassini, E.; Frezza, A.M.; Dinoi, G.; Messina, A.; Gronchi, A.; Cavalleri, A.; et al. Imatinib and everolimus in patients with progressing advanced chordoma: A phase 2 clinical study. Cancer 2018, 124, 4056–4063. [Google Scholar] [CrossRef]
  44. Mitra, A.; Ray, M.K.; Chatterjee, G.C. Role of iron in the enhancement by Agrobacterium tumefaciens infection in mice. Folia Microbiol 1988, 33, 255–260. [Google Scholar] [CrossRef] [PubMed]
  45. Stacchiotti, S.; Tamborini, E.; Lo Vullo, S.; Bozzi, F.; Messina, A.; Morosi, C.; Casale, A.; Crippa, F.; Conca, E.; Negri, T.; et al. Phase II study on lapatinib in advanced EGFR-positive chordoma. Ann. Oncol. 2013, 24, 1931–1936. [Google Scholar] [CrossRef] [PubMed]
  46. Yang, C.; Sun, J.; Yong, L.; Liang, C.; Liu, T.; Xu, Y.; Yang, J.; Liu, X. Deficiency of PTEN and CDKN2A Tumor-Suppressor Genes in Conventional and Chondroid Chordomas: Molecular Characteristics and Clinical Relevance. Onco Targets Ther. 2020, 13, 4649–4663. [Google Scholar] [CrossRef] [PubMed]
  47. Le, L.P.; Nielsen, G.P.; Rosenberg, A.E.; Thomas, D.; Batten, J.M.; Deshpande, V.; Schwab, J.; Duan, Z.; Xavier, R.J.; Hornicek, F.J.; et al. Recurrent chromosomal copy number alterations in sporadic chordomas. PLoS ONE 2011, 6, e18846. [Google Scholar] [CrossRef] [PubMed]
  48. Seeling, C.; Mosca, E.; Mantel, E.; Möller, P.; Barth, T.F.E.; Mellert, K. Prognostic Relevance and In Vitro Targeting of Concomitant PTEN and p16 Deficiency in Chordomas. Cancers 2023, 15, 1977. [Google Scholar] [CrossRef]
  49. Le Cesne, A.; Chevreau, C.; Perrin, C.; Italiano, A.; Hervieu, A.; Blay, J.Y.; Piperno-Neumann, S.; Saada-Bouzid, E.; Bertucci, F.; Firmin, N.; et al. Regorafenib in patients with relapsed advanced or metastatic chordoma: Results of a non-comparative, randomised, double-blind, placebo-controlled, multicentre phase II study. ESMO Open 2023, 8, 101569. [Google Scholar] [CrossRef]
  50. Duffaud, F.; Mir, O.; Boudou-Rouquette, P.; Piperno-Neumann, S.; Penel, N.; Bompas, E.; Delcambre, C.; Kalbacher, E.; Italiano, A.; Collard, O.; et al. Efficacy and safety of regorafenib in adult patients with metastatic osteosarcoma: A non-comparative, randomised, double-blind, placebo-controlled, phase 2 study. Lancet Oncol. 2019, 20, 120–133. [Google Scholar] [CrossRef]
  51. Ghaith, A.K.; Akinduro, O.O.; Perez-Vega, C.; Bon Nieves, A.; Abode-Iyamah, K.; Patel, N.; Kalani, M.; Clarke, M.J.; Rose, P.; Bydon, M. Association between immunohistochemical markers and tumor progression following resection of spinal chordomas: A multicenter study. J. Neurosurg. Spine 2023, 39, 652–660. [Google Scholar] [CrossRef]
  52. A Phase II Trial of Cetuximab for Patients with Advanced or Metastatic Chordoma. Available online: https://clinicaltrials.gov/study/NCT05041127 (accessed on 20 April 2024).
  53. Lipplaa, A.; Strauss, S.J.; Stacchiotti, S.; Kayani, I.; Efthymiadis, K.; Frezza, A.M.; Kranenbarg, E.M.K.; Sommer, J.; Flanagan, A.; Gelderblom, H. A phase 2, single arm, European multi-center trial evaluating the efficacy of afatinib as first line or later line treatment in advanced chordoma. J. Clin. Oncol. 2024, 42 (Suppl. 16), 11517. [Google Scholar] [CrossRef]
  54. Cote, G.M.; DeLaney, T.F.; Miao, R.; Schwab, J.H.; Raskin, K.; Calderón, S.L.; Mullen, J.T.; Haynes, A.B.; Hornicek, F.J.; Chen, Y.-L.; et al. Updated 5-year local control (LC), metastasis-free survival (MFS), and overall survival (OS) data from a phase I study of nilotinib plus radiation (RT) in high-risk chordoma. J. Clin. Oncol. 2020, 38 (Suppl. 15), e23505. [Google Scholar] [CrossRef]
  55. Burkenroad, A.; Singh, A.S.; Chmielowski, B.; Brackert, S.; Federman, N.; Hagopian, A.; Glaspy, J.A.; Kendal, J.; Wessel, L.E.; Hornicek, F.J.; et al. A signal-finding study of nivolumab and relatlimab in patients with advanced chordoma. J. Clin. Oncol. 2024, 42 (Suppl. 16), 11578. [Google Scholar] [CrossRef]
  56. Phase I Safety Study of Stereotactic Radiosurgery with Concurrent and Adjuvant PD-1 Antibody Nivolumab in Subjects with Recurrent or Advanced Chordoma. Available online: https://clinicaltrials.gov/study/NCT02989636 (accessed on 20 April 2024).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Golding, R.; Abuqubo, R.; Pansa, C.J.; Bhatta, M.; Shankar, V.; Mani, K.; Kleinbart, E.; Gelfand, Y.; Murthy, S.; De la Garza Ramos, R.; et al. Immunologic and Targeted Molecular Therapies for Chordomas: A Narrative Review. J. Clin. Med. 2024, 13, 5679. https://doi.org/10.3390/jcm13195679

AMA Style

Golding R, Abuqubo R, Pansa CJ, Bhatta M, Shankar V, Mani K, Kleinbart E, Gelfand Y, Murthy S, De la Garza Ramos R, et al. Immunologic and Targeted Molecular Therapies for Chordomas: A Narrative Review. Journal of Clinical Medicine. 2024; 13(19):5679. https://doi.org/10.3390/jcm13195679

Chicago/Turabian Style

Golding, Regina, Rami Abuqubo, Christopher J. Pansa, Manish Bhatta, Vishal Shankar, Kyle Mani, Emily Kleinbart, Yaroslav Gelfand, Saikiran Murthy, Rafael De la Garza Ramos, and et al. 2024. "Immunologic and Targeted Molecular Therapies for Chordomas: A Narrative Review" Journal of Clinical Medicine 13, no. 19: 5679. https://doi.org/10.3390/jcm13195679

APA Style

Golding, R., Abuqubo, R., Pansa, C. J., Bhatta, M., Shankar, V., Mani, K., Kleinbart, E., Gelfand, Y., Murthy, S., De la Garza Ramos, R., Krystal, J., Eleswarapu, A., Yassari, R., Mostafa, E., Fourman, M. S., & Schlumprecht, A. (2024). Immunologic and Targeted Molecular Therapies for Chordomas: A Narrative Review. Journal of Clinical Medicine, 13(19), 5679. https://doi.org/10.3390/jcm13195679

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop