Skip to main content

Challenges facing level I evidence in treatment of low-grade gliomas and subsequent uncertainties

Abstract

Background

The current era of molecular characterisation has contributed greatly to our understanding and management of low-grade gliomas (LGGs); however, this has also contributed to a paucity in level 1 evidence.

Review

Diagnostic breakthroughs in LGGs are moving quicker than our experimental capacity can react. The design, analysis, and clinical application of first-level evidence are struggling to compete with the considerable variability in the natural course of LGGs and the rapidly evolving utility of molecular characterisation of tumours. This poses several uncertainties to researchers, clinicians, and more importantly, patients.

Conclusion

Individualised case-by-case decisions based on best available evidence, albeit lacking level 1 evidence, must be made by considering the tumour behaviour, clinical course, and specific patient needs and goals.

Introduction

Low-grade gliomas (LGGs) are a unique group of primary brain tumours encompassing astrocytomas and oligodendrogliomas; they are histologically defined as grade 2 on the traditional World Health Organisation (WHO) classification system. However, this definition is lacking with the advent and rapid expansion of the molecular characterisation of this group of morphological tumours. LGGs have been shown to demonstrate a high risk of malignant transformation and a considerable variability in their natural course [1]. In the era of evidence-based medicine, these factors pose significant challenges to the design, analysis, and application of randomised controlled trials (RCTs). This paper aims to explore the reasons for this conundrum and the subsequent uncertainties faced by researchers, clinicians, and more importantly, patients.

LGGs: a complex definition and a heterogeneous course

The natural history of LGGs is highly variable between patients. There is a latency period of apparent stability, with some gliomas progressing very slowly, while others undergo rapid malignant transformation from the time of diagnosis, even when uniformly treated [2]. Several systems have been used to define a LGG as high risk. The most common definition is that employed in the Radiation Therapy Oncology Group 9802 trial [3]. In this trial, a high-risk patient was defined as being ≥ 40 years of age, or, if younger than 40, as having an incomplete resection. This definition however encompasses a wide range of the grade 2 infiltrating gliomas, does not distinguish astrocytomas from oligodendrogliomas, or consider the molecular characteristics, such as isocitrate dehydrogenase (IDH) mutations, 1p/19q co-deletion, or CDKN2A/B status. These characteristics have been shown to play an important prognostic impact on survival outcomes and contribute to the heterogeneous course of patients with LGGs, see Fig. 1 for a summary of these risk factors [4, 5].

Fig. 1
figure 1

Risk factors associated with more aggressive LGGs and rapid de-differentiation

The absence of this molecular information has been a major limitation of most landmark studies on LGGs over the past decade. Patients have been mostly recruited based on histopathology only, at a time when the utility of molecular biomarkers was not yet established in clinical practice as knowledge of these biomarkers has been rapidly evolving. Consequently, these studies provided limited evidence for molecularly stratified treatments and were found to have flawed recruitment when adjusting for biomarkers. A key example was demonstrated by the European Organisation for Research and Treatment of Cancer (EORTC) 22,033-26033 trial [6]. The EORTC investigators aimed to compare radiotherapy versus temozolomide in LGG patients. Post hoc molecular analysis, inspired by the publication of the revised WHO classification in 2016 at the time, revealed not only a variable mix of IDH-mutant astrocytomas and oligodendrogliomas, but also 15% of patients thought to be LGGs were found to be primary glioblastomas (IDH wild type). Similarly, Jakola et al. (2017) discovered a 26.8% glioblastoma (IDH wild type) inclusion rate upon retrospective molecular analysis of their LGG cohort. A much more recent example is the CODEL trial [8]. The investigators in this trial found a marked disparity in progression-free survival rates between their experimental arms and had to redesign their aim and methodology half-way through the project after adjustment for IDH status.

RCTs: a slow train

The question is whether RCTs will ever align with the rapidly expanding knowledge of prognostic biomarkers. RCTs take several years to develop, making it incredibly challenging to adjust recruitment and assess new therapies in a timely manner. For example, the trial by Buckner et al. [9] looking at radiation alone versus chemo/radiotherapy combination took 12 years to follow-up 251 patients, and a total of 18 years was spent from recruitment to publication. Another very recent trial was the CATNON, published in June 2021 in The Lancet Oncology, looking at concurrent versus adjuvant temozolomide to radiotherapy in adults with newly diagnosed 1p/19q non-co-deleted anaplastic gliomas. It took the investigators eight years to recruit patients and another six years for the results to be published, concluding that adjuvant temozolomide was associated with a survival benefit, while limited benefit was found with concurrent temozolomide [10].

RCTs for surgery in LGGs

No RCT comparing surgical resection with biopsy has been conducted to date in LGG patients. The closest approach to patient randomisation between biopsy and surgery originates from a retrospective population-based study comparing two Norwegian hospitals with two different surgical philosophies—watchful waiting versus early gross total resection (GTR) [7]. A significant overall survival (OS) benefit was found in the surgical group—14.4 years versus 5.8 years in the surveillance group (P < 0.01). While the optimal timing of surgery remains controversial [12], the benefits of surgery on OS and seizure control are clear [13, 14] and have been shown to stand even when adjusted for molecular markers [11, 15].

While British guidelines have acknowledged this evidence, the role of surgery is still not clearly recognised in most international guidelines under the argument that there are no RCTs [2]. This raises the ethical dilemma of whether it is acceptable to enrol a patient in an RCT comparing initial biopsy versus surgery when OS is about 14–15 years with GTR vs 6–7 years with biopsy, only to claim that the benefit of surgery has finally been demonstrated with level I evidence. Additionally, there are several confounding factors that would limit the design of an RCT for surgery, as outcomes in LGGs tend to be highly dependent on tumour location and surgical expertise as these tumours are notoriously known for affecting eloquent brain regions. Another limitation would be keeping the pace with rapid advances in surgical techniques and intraoperative imaging/mapping, as such a randomised trial would last for at least five to ten years [16].

The struggle of LGG patients

The diagnosis of LGGs is often made in young or middle-aged patients who are at a productive time of their lives. This is a time where patients are ascending in their careers, child-bearing, and unrestricted in their everyday activities, such as driving. The challenge with the absence of level I evidence lies in balancing surveillance versus intervention (surgery, chemotherapy, and radiotherapy) and recognising the distinction between treating the tumour and treating the patient. The belief in the 1990s was in favour of surveillance [17]. This was based on the rationale that deferring treatment, such as radiation therapy, might minimise long-term complications such as radiation necrosis and cognitive impairment. This hinges on the hope that intervention will occur before malignant transformation ensues. However, this cannot be guaranteed even with close follow-up, which places patients under substantial psychological distress, and the constant feeling of a “ticking time bomb”. This is particularly challenging in cases where seizures are well controlled and the lesion is static, as there may be no compelling argument for surgery. Hence, it is essential for clinicians to discuss uncertainties in management decision-making and explain why it is incredibly challenging to extrapolate evidence from current studies or recruit patients in a trial with such a variable disease course (see Fig. 2 for a summary of key challenges). Several patients may be understanding, while others may wander around seeking several expert opinions in an attempt to answer a dilemma which cannot be solved [1].

Fig. 2
figure 2

A summary of key challenges limiting level 1 evidence in management of LGGs

Conclusions

Genetic findings and biomarker revolutions in LGGs are moving quicker than our experimental capacity can react. The design, analysis, and clinical application of RCTs must compete with the considerable variability in the natural course of LGGs and the rapidly evolving utility of molecular characterisation of tumours. Well-designed, molecularly enriched RCTs are necessary to inform future treatment. Until then, individualised case-by-case decisions must be made by considering the tumour behaviour, and patient needs. Ultimately, there is a dire need for an intervention to keep LGGs low grade.

Availability of data and materials

Not applicable.

Abbreviations

LGG:

Low-grade glioma

RCT:

Randomised controlled trial

WHO:

World Health Organisation

IDH:

Isocitrate dehydrogenase

EORTC:

European Organisation for Research and Treatment of Cancer

References

  1. Duffau H. Paradoxes of evidence-based medicine in lower-grade glioma: To treat the tumor or the patient? Neurology. 2018;91(14):657–62.

    Article  PubMed  Google Scholar 

  2. Weller M, van den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, Bendszus M, Balana C, Chinot O, Dirven L, French P. EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Rev Clin Oncol. 2021;18(3):170–86.

    Article  PubMed  Google Scholar 

  3. Shaw EG, Wang M, Coons SW, Brachman DG, Buckner JC, Stelzer KJ, Barger GR, Brown PD, Gilbert MR, Mehta MP. Randomized trial of radiation therapy plus procarbazine, lomustine, and vincristine chemotherapy for supratentorial adult low-grade glioma: initial results of RTOG 9802. J Clin Oncol. 2012;30(25):30–65.

    Article  Google Scholar 

  4. Lu VM, O’Connor KP, Shah AH, Eichberg DG, Luther EM, Komotar RJ, Ivan ME. The prognostic significance of CDKN2A homozygous deletion in IDH-mutant lower-grade glioma and glioblastoma: a systematic review of the contemporary literature. J Neurooncol. 2020;148(2):221–9.

    Article  CAS  PubMed  Google Scholar 

  5. Jenkins RB, Blair H, Ballman KV, Giannini C, Arusell RM, Law M, Flynn H, Passe S, Felten S, Brown PD, Shaw EG. A t (1; 19)(q10; p10) mediates the combined deletions of 1p and 19q and predicts a better prognosis of patients with oligodendroglioma. Can Res. 2006;66(20):9852–61.

    Article  CAS  Google Scholar 

  6. Baumert BG, Hegi ME, van den Bent MJ, von Deimling A, Gorlia T, Hoang-Xuan K, Brandes AA, Kantor G, Taphoorn MJ, Hassel MB, Hartmann C. Temozolomide chemotherapy versus radiotherapy in high-risk low-grade glioma (EORTC 22033–26033): a randomised, open-label, phase 3 intergroup study. Lancet Oncol. 2016;17(11):1521–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jakola AS, Skjulsvik AJ, Myrmel KS, Sjåvik K, Unsgård G, Torp SH, Aaberg K, Berg T, Dai HY, Johnsen K, Kloster R. Surgical resection versus watchful waiting in low-grade gliomas. Ann Oncol. 2017;28(8):1942–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jaeckle KA, Ballman KV, van den Bent M, Giannini C, Galanis E, Brown PD, Jenkins RB, Cairncross JG, Wick W, Weller M, Aldape KD. CODEL: phase III study of RT, RT+ TMZ, or TMZ for newly diagnosed 1p/19q codeleted oligodendroglioma, analysis from the initial study design. Neuro Oncol. 2021;23(3):457–67.

    Article  PubMed  Google Scholar 

  9. Buckner JC, Shaw EG, Pugh SL, Chakravarti A, Gilbert MR, Barger GR, Coons S, Ricci P, Bullard D, Brown PD, Stelzer K. Radiation plus procarbazine, CCNU, and vincristine in low-grade glioma. N Engl J Med. 2016;374(14):1344–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bent MJ, Tesileanu CMS, Wick W, Sanson M, Brandes AA, Clement PM, Erridge S, Vogelbaum MA, Nowak AK, Baurain JF, Mason WP. Adjuvant and concurrent temozolomide for 1p/19q non-co-deleted anaplastic glioma (CATNON; EORTC study 26053–22054): second interim analysis of a randomised, open-label, phase 3 study. Lancet Oncol. 2021;22(6):813–23.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Jakola AS, Myrmel KS, Kloster R, Torp SH, Lindal S, Unsgård G, Solheim O. Comparison of a strategy favoring early surgical resection vs a strategy favoring watchful waiting in low-grade gliomas. JAMA. 2012;308(18):1881–8.

    Article  CAS  PubMed  Google Scholar 

  12. Zeng L, Mei Q, Li H, Ke C, Yu J, Chen J. ’A survival analysis of surgically treated incidental low-grade glioma patients’. Sci Rep. 2021;11(1):1–9.

    Google Scholar 

  13. Roelz R, Strohmaier D, Jabbarli R, Kraeutle R, Egger K, Coenen VA, Weyerbrock A, Reinacher PC. Residual tumor volume as best outcome predictor in low grade glioma–a nine-years near-randomized survey of surgery vs. biopsy. Sci Rep. 2016;6(1):1–9.

    Article  Google Scholar 

  14. Solomons MR, Jaunmuktane Z, Weil RS, El-Hassan T, Brandner S, Rees JH. Seizure outcomes and survival in adult low-grade glioma over 11 years: living longer and better. Neuro-oncol Pract. 2020;7(2):196–201.

    Article  Google Scholar 

  15. Cordier D, Gozé C, Schädelin S, Rigau V, Mariani L, Duffau H. A better surgical resectability of WHO grade II gliomas is independent of favorable molecular markers. J Neurooncol. 2015;121(1):185–93.

    Article  CAS  PubMed  Google Scholar 

  16. Hamer PDW, Robles SG, Zwinderman AH, Duffau H, Berger MS. Impact of intraoperative stimulation brain mapping on glioma surgery outcome: a meta-analysis. J Clin Oncol. 2012;30(20):2559–65.

    Article  Google Scholar 

  17. Recht LD, Lew R, Smith TW. Suspected low-grade glioma: is deferring treatment safe? Ann Neurol. 1992;31(4):431–6.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge Dr Ashish Jonathan (Consultant Neurosurgeon at Gold Coast University Hospital) for providing guidance on direction of article.

Funding

No funding required for this submission.

Author information

Authors and Affiliations

Authors

Contributions

(1) PF helped in concept and design, acquisition, drafting of the manuscript, critical review of the manuscript for important intellectual content; (2) TK was involved in acquisition, drafting of the manuscript, and critical review of content. Both authors will review final version of manuscript according to journal edits and review process.

Corresponding author

Correspondence to Peter Fawzy.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

No competing interests by the authors.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fawzy, P., Karpin, T. Challenges facing level I evidence in treatment of low-grade gliomas and subsequent uncertainties. Egypt J Neurosurg 39, 7 (2024). https://doi.org/10.1186/s41984-024-00271-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41984-024-00271-y

Keywords